1
|
Herrock O, Campbell N, Deer E, Amaral LM, Whitney D, Morris R, Wallace K, Turner T, Cleveland EH, Belk S, Booz GW, Cornelius DC, LaMarca B. Preeclamptic Placental CD19+ B Cells Are Causal to Hypertension During Pregnancy. Hypertension 2025. [PMID: 40171666 DOI: 10.1161/hypertensionaha.124.24552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Patients with preeclampsia exhibit hypertension and chronic inflammation characterized by CD (cluster determinant) 4+T cells, B cells secreting AT1-AA (agonistic autoantibody against the angiotensin II type 1 receptor), inflammatory cytokines, and complement activation. Importantly, a history of COVID-19 during pregnancy is associated with an increased incidence of a preeclampsia-like phenotype and is partly mediated by CD4+T cells. We recently showed pregnant patients with a history of COVID-19 with or without preeclampsia produce AT1-AA, indicating the importance of B lymphocytes in the progression of preeclampsia and possibly of COVID-19. Therefore, we hypothesize that B cells from patients with preeclampsia with or without COVID-19 history induce the preeclampsia phenotype through AT1-AA. METHODS Placental B cells were isolated from normal pregnant, patients with preeclampsia, normotensive COVID-19 history, or preeclampsia COVID-19 history at delivery. Then, 3×105 B cells were transferred intraperitoneally into pregnant athymic rats at gestational day 12. On gestational day 18, carotid catheters were inserted. On gestational day 19, mean arterial pressure was measured, and tissues were collected. RESULTS Preeclampsia B-cell recipients had significantly increased mean arterial pressure, AT1-AA, inflammatory cytokines, and complement activation compared with normal pregnant B-cell recipients. Recipients of B cells with COVID-19 history had markers of inflammation and hypertension but not to the level of significance as recipients of preeclampsia B cells. Inhibition of AT1-AA attenuated the hypertension that occurred in response to preeclampsia or preeclampsia B cells with COVID-19 history. CONCLUSIONS This study demonstrates the important role of B cells in contributing to hypertension and chronic inflammation during preeclampsia with or without COVID-19 history through secretion of AT1-AA.
Collapse
Affiliation(s)
| | - Nathan Campbell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Lorena M Amaral
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Darby Whitney
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Rachael Morris
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson. (R.M., B.L.)
| | - Kedra Wallace
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Ty Turner
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - E Hawthorne Cleveland
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Sheila Belk
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson. (N.C., E.D., L.M.A., D.W., K.W., T.T., E.H.C., S.B., G.W.B., D.C.C., B.L.)
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson. (R.M., B.L.)
| |
Collapse
|
2
|
Bartoloni E, Cacciapaglia F, Erre GL, Gremese E, Manfredi A, Piga M, Sakellariou G, Spinelli FR, Viapiana O, Atzeni F. Immunomodulation for accelerated atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus. Autoimmun Rev 2025; 24:103760. [PMID: 39894242 DOI: 10.1016/j.autrev.2025.103760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
In the last decades, consisting evidence supported a close relationship between both innate and adaptive immune systems and the accelerated cardiovascular (CV) disease characterizing autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Indeed, several cell lines involved in the pathogenesis of these autoimmune diseases, such as macrophages and dendritic cells, as well as different T and B lymphocyte subsets, and inflammatory cytokines, have been demonstrated to be directly involved in the mechanisms underlying early atherosclerotic arterial wall damage. Traditional CV risk factors play a concomitant role but do not sufficiently account for the increased prevalence of CV disease in these patients. Indeed, the pathophysiological link between RA and SLE and atherosclerosis is based on complex inflammatory pathways that interconnect these conditions and may explain the significant morbidity and mortality rates demonstrated in these patients, with consequent significant negative effects on quality of life and long-term survival. Consequently, it is intriguing to hypothesize that immunosuppressive drugs commonly used in the treatment of these pathologies may also exert an immunomodulatory and anti-inflammatory effect in mitigating the atherosclerotic damage that has been demonstrated to occur early in the initial stages of the disease. Recognizing risk factors, predicting occurrences and early intervention to prevent CV disease development have emerged as critical objectives in RA and SLE treatment. In this review, we aimed to provide an updated overview of the atherogenic effects exerted by the immune and inflammatory pathways involved in the pathogenesis of RA and SLE. Moreover, we examined the available evidence which may support the potential effects of immunosuppressive therapies in reducing CV damage and, consequently, CV disease risk in these patients.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Italy
| | - Fabio Cacciapaglia
- Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DePReMeI), University of Bari, Bari, Italy; Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro" Casamassima & Rheumatology Service "Miulli" General Hospital Acquaviva delle Fonti, Bari, Italy
| | - Gian Luca Erre
- Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Italy
| | - Elisa Gremese
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andreina Manfredi
- University of Modena and Reggio Emilia,AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Matteo Piga
- Rheumatology Unit, AOU Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Italy
| | - Garifallia Sakellariou
- Department of Internal Medicine and Therapeutics, University of Pavia, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Francesca Romana Spinelli
- Reumatology Unit, Department of Internal, Anesthesiological, and Cardiovascular Clinical Sciences, Sapienza University of Rome, Rome, Italy
| | - Ombretta Viapiana
- Rheumatology Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Italy.
| |
Collapse
|
3
|
Thangasparan S, Kamisah Y, Ugusman A, Mohamad Anuar NN, Ibrahim N‘I. Unravelling the Mechanisms of Oxidised Low-Density Lipoprotein in Cardiovascular Health: Current Evidence from In Vitro and In Vivo Studies. Int J Mol Sci 2024; 25:13292. [PMID: 39769058 PMCID: PMC11676878 DOI: 10.3390/ijms252413292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular diseases (CVD) are the number one cause of death worldwide, with atherosclerosis, which is the formation of fatty plaques in the arteries, being the most common underlying cause. The activation of inflammatory events and endothelium dysfunction are crucial for the development and pathophysiology of atherosclerosis. Elevated circulating levels of low-density lipoprotein (LDL) have been associated with severity of atherosclerosis. LDL can undergo oxidative modifications, resulting in oxidised LDL (oxLDL). OxLDL has been found to have antigenic potential and contribute significantly to atherosclerosis-associated inflammation by activating innate and adaptive immunity. Various inflammatory stimuli such as interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α) and intercellular adhesion molecule 1 (ICAM-1) play major roles in atherosclerosis. To date, studies have provided valuable insights into the role of oxLDL in the development of atherosclerosis. However, there remains a gap in understanding the specific pathways involved in this process. This review aims to provide and discuss the mechanisms by which oxLDL modulates signalling pathways that cause cardiovascular diseases by providing in vitro and in vivo experimental evidence. Its critical role in triggering and sustaining endothelial dysfunction highlights its potential as a therapeutic target. Advancing the understanding of its atherogenic role and associated signalling pathways could pave the way for novel targeted therapeutic strategies to combat atherosclerosis more effectively.
Collapse
Affiliation(s)
- Sahsikala Thangasparan
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
| | - Azizah Ugusman
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
- Programme of Biomedical Science, Center for Toxicology & Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
| |
Collapse
|
4
|
Juhasz V, Charlier FT, Zhao TX, Tsiantoulas D. Targeting the adaptive immune continuum in atherosclerosis and post-MI injury. Atherosclerosis 2024; 399:118616. [PMID: 39546915 DOI: 10.1016/j.atherosclerosis.2024.118616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024]
Abstract
Atherosclerotic disease is a cholesterol-rich lipoprotein particle-driven disease resulting in the formation of atherosclerotic plaques in large and medium size arteries. Rupture or erosion of atherosclerotic plaques can trigger the formation of a thrombus causing the obstruction of the blood flow in the coronary artery and thereby leading to myocardial infarction (MI). Inflammation is a crucial pillar of the mechanisms leading to atherosclerosis and governing the cardiac repair post-MI. Dissecting the complex and sophisticated networks of the immune responses underlying the formation of atherosclerotic plaques and affecting the healing of the heart after MI will allow the designing of highly precise immunomodulatory therapies for these settings. Notably, MI also accelerates atherosclerosis via modulating the response of the immune system. Therefore, for the identification of effective and safe therapeutic targets, it is critical to consider the inflammatory continuum that interconnects the two pathologies and identify immunomodulatory strategies that confer a protective effect in both settings or at least, affect each pathology independently. Adaptive immunity, which consists of B and T lymphocytes, is a major regulator of atherosclerosis and post-MI cardiac repair. Here, we review and discuss the effect of potential adaptive immunity-targeting therapies, such as cell-depleting therapies, in atherosclerosis and post-MI cardiac injury.
Collapse
Affiliation(s)
- Viktoria Juhasz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Fiona T Charlier
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | | |
Collapse
|
5
|
Zhang L, Li P, Li Y, Qu W, Shi Y, Zhang T, Chen Y. The role of immunoglobins in atherosclerosis development; friends or foe? Mol Cell Biochem 2024:10.1007/s11010-024-05158-y. [PMID: 39592554 DOI: 10.1007/s11010-024-05158-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Coronary artery disease, atherosclerosis, and its life-threatening sequels impose the hugest burden on the healthcare systems throughout the world. The intricate process of atherosclerosis is considered as an inflammatory-based disorder, and therefore, the components of the immune system are involved in different stages from formation of coronary plaques to its development. One of the major effectors in this way are the antibody producing entities, the B cells. These cells, which play a significant and unique role in responding to different stress, injuries, and infections, contribute differently to the development of atherosclerosis, either inhibitory or promoting, depending on the type of subsets. B cells implicate in both systemic and local immune responses of an atherosclerotic artery by cell-cell contact, cytokine production, and antigen presentation. In particular, natural antibodies bind to oxidized lipoproteins and cellular debris, which are abundant during plaque growth. Logically, any defects in B cells and consequent impairment in antibody production may greatly affect the shaping of the plaque and its clinical outcome. In this comprehensive review, we scrutinize the role of B cells and different classes of antibodies in atherosclerosis progression besides current novel B-cell-based therapeutic approaches that aim to resolve this affliction of mankind.
Collapse
Affiliation(s)
- Linlin Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Peize Li
- Department of Orthopedics, Changchun Chinese Medicine Hospital, Changchun, 130022, China
| | - Yuhui Li
- Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Wantong Qu
- Department of Cardiology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yanyu Shi
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Tianyang Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Ying Chen
- Department of Cardiology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
6
|
Wu Y, Xu Y, Xu L. Pharmacological therapy targeting the immune response in atherosclerosis. Int Immunopharmacol 2024; 141:112974. [PMID: 39168023 DOI: 10.1016/j.intimp.2024.112974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by the formation of atherosclerotic plaques that consist of numerous cells including smooth muscle cells, endothelial cells, immune cells, and foam cells. The most abundant innate and adaptive immune cells, including neutrophils, monocytes, macrophages, B cells, and T cells, play a pivotal role in the inflammatory response, lipoprotein metabolism, and foam cell formation to accelerate atherosclerotic plaque formation. In this review, we have discussed the underlying mechanisms of activated immune cells in promoting AS and reviewed published clinical trials for the treatment of AS by suppressing immune cell activation. We have also presented some crucial shortcomings of current clinical trials. Lastly, we have discussed the therapeutic potential of novel compounds, including herbal medicine and dietary food, in alleviating AS in animals. Despite these limitations, further clinical trials and experimental studies will enhance our understanding of the mechanisms modulated by immune cells and promote widespread drug use to treat AS by suppressing immune system-induced inflammation.
Collapse
Affiliation(s)
- Yirong Wu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China
| | - Yizhou Xu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China.
| | - Linhao Xu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Translational Medicine Research Center, Hangzhou First People's Hospital, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
7
|
Porsch F, Binder CJ. Autoimmune diseases and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:780-807. [PMID: 38937626 DOI: 10.1038/s41569-024-01045-7] [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] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Autoimmune diseases are associated with a dramatically increased risk of atherosclerotic cardiovascular disease and its clinical manifestations. The increased risk is consistent with the notion that atherogenesis is modulated by both protective and disease-promoting immune mechanisms. Notably, traditional cardiovascular risk factors such as dyslipidaemia and hypertension alone do not explain the increased risk of cardiovascular disease associated with autoimmune diseases. Several mechanisms have been implicated in mediating the autoimmunity-associated cardiovascular risk, either directly or by modulating the effect of other risk factors in a complex interplay. Aberrant leukocyte function and pro-inflammatory cytokines are central to both disease entities, resulting in vascular dysfunction, impaired resolution of inflammation and promotion of chronic inflammation. Similarly, loss of tolerance to self-antigens and the generation of autoantibodies are key features of autoimmunity but are also implicated in the maladaptive inflammatory response during atherosclerotic cardiovascular disease. Therefore, immunomodulatory therapies are potential efficacious interventions to directly reduce the risk of cardiovascular disease, and biomarkers of autoimmune disease activity could be relevant tools to stratify patients with autoimmunity according to their cardiovascular risk. In this Review, we discuss the pathophysiological aspects of the increased cardiovascular risk associated with autoimmunity and highlight the many open questions that need to be answered to develop novel therapies that specifically address this unmet clinical need.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Buch MH, Mallat Z, Dweck MR, Tarkin JM, O'Regan DP, Ferreira V, Youngstein T, Plein S. Current understanding and management of cardiovascular involvement in rheumatic immune-mediated inflammatory diseases. Nat Rev Rheumatol 2024; 20:614-634. [PMID: 39232242 DOI: 10.1038/s41584-024-01149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a spectrum of disorders of overlapping immunopathogenesis, with a prevalence of up to 10% in Western populations and increasing incidence in developing countries. Although targeted treatments have revolutionized the management of rheumatic IMIDs, cardiovascular involvement confers an increased risk of mortality and remains clinically under-recognized. Cardiovascular pathology is diverse across rheumatic IMIDs, ranging from premature atherosclerotic cardiovascular disease (ASCVD) to inflammatory cardiomyopathy, which comprises myocardial microvascular dysfunction, vasculitis, myocarditis and pericarditis, and heart failure. Epidemiological and clinical data imply that rheumatic IMIDs and associated cardiovascular disease share common inflammatory mechanisms. This concept is strengthened by emergent trials that indicate improved cardiovascular outcomes with immune modulators in the general population with ASCVD. However, not all disease-modifying therapies that reduce inflammation in IMIDs such as rheumatoid arthritis demonstrate equally beneficial cardiovascular effects, and the evidence base for treatment of inflammatory cardiomyopathy in patients with rheumatic IMIDs is lacking. Specific diagnostic protocols for the early detection and monitoring of cardiovascular involvement in patients with IMIDs are emerging but are in need of ongoing development. This Review summarizes current concepts on the potentially targetable inflammatory mechanisms of cardiovascular pathology in rheumatic IMIDs and discusses how these concepts can be considered for the diagnosis and management of cardiovascular involvement across rheumatic IMIDs, with an emphasis on the potential of cardiovascular imaging for risk stratification, early detection and prognostication.
Collapse
Affiliation(s)
- Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK.
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Ziad Mallat
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Marc R Dweck
- Centre for Cardiovascular Science, Chancellors Building, Little France Crescent, University of Edinburgh, Edinburgh, UK
| | - Jason M Tarkin
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Declan P O'Regan
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Vanessa Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Taryn Youngstein
- National Heart & Lung Institute, Imperial College London, London, UK
- Department of Rheumatology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Sven Plein
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| |
Collapse
|
9
|
Lavillegrand JR, Al-Rifai R, Thietart S, Guyon T, Vandestienne M, Cohen R, Duval V, Zhong X, Yen D, Ozturk M, Negishi Y, Konkel J, Pinteaux E, Lenoir O, Vilar J, Laurans L, Esposito B, Bredon M, Sokol H, Diedisheim M, Saliba AE, Zernecke A, Cochain C, Haub J, Tedgui A, Speck NA, Taleb S, Mhlanga MM, Schlitzer A, Riksen NP, Ait-Oufella H. Alternating high-fat diet enhances atherosclerosis by neutrophil reprogramming. Nature 2024; 634:447-456. [PMID: 39232165 DOI: 10.1038/s41586-024-07693-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/07/2024] [Indexed: 09/06/2024]
Abstract
Systemic immune responses caused by chronic hypercholesterolaemia contribute to atherosclerosis initiation, progression and complications1. However, individuals often change their dietary habits over time2, and the effects of an alternating high-fat diet (HFD) on atherosclerosis remain unclear. Here, to address this relevant issue, we developed a protocol using atherosclerosis-prone mice to compare an alternating versus continuous HFD while maintaining similar overall exposure periods. We found that an alternating HFD accelerated atherosclerosis in Ldlr-/- and Apoe-/- mice compared with a continuous HFD. This pro-atherogenic effect of the alternating HFD was also observed in Apoe-/-Rag2-/- mice lacking T, B and natural killer T cells, ruling out the role of the adaptive immune system in the observed phenotype. Discontinuing the HFD in the alternating HFD group downregulated RUNX13, promoting inflammatory signalling in bone marrow myeloid progenitors. After re-exposure to an HFD, these cells produced IL-1β, leading to emergency myelopoiesis and increased neutrophil levels in blood. Neutrophils infiltrated plaques and released neutrophil extracellular traps, exacerbating atherosclerosis. Specific depletion of neutrophils or inhibition of IL-1β pathways abolished emergency myelopoiesis and reversed the pro-atherogenic effects of the alternating HFD. This study highlights the role of IL-1β-dependent neutrophil progenitor reprogramming in accelerated atherosclerosis induced by alternating HFD.
Collapse
Affiliation(s)
| | - Rida Al-Rifai
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Sara Thietart
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
- Geriatrics Department, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié Salpêtrière, Paris, France
| | - Théo Guyon
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Marie Vandestienne
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Raphael Cohen
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Vincent Duval
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Xiaodan Zhong
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Daniel Yen
- Department of Cell and Developmental Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mumin Ozturk
- Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University FNWI, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yutaka Negishi
- Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University FNWI, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joanne Konkel
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Olivia Lenoir
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Jose Vilar
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Ludivine Laurans
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Bruno Esposito
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Marius Bredon
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), Saint-Cyr-sur-Loire, France
- IMMEDIAB Laboratory, INSERM U1151, Necker Enfants Malades (INEM), Paris, France
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Wuerzburg, Würzburg, Germany
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Wuerzburg, Würzburg, Germany
| | - Jessica Haub
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Alain Tedgui
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Nancy A Speck
- Department of Cell and Developmental Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Soraya Taleb
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France
| | - Musa M Mhlanga
- Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University FNWI, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Hafid Ait-Oufella
- Paris Cardiovascular Research Center, Université Paris Cité, INSERM U970, Paris, France.
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France.
| |
Collapse
|
10
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
11
|
Chen Z, Wang Z, Cui Y, Xie H, Yi L, Zhu Z, Ni J, Du R, Wang X, Zhu J, Ding F, Quan W, Zhang R, Wang Y, Yan X. Serum BAFF level is associated with the presence and severity of coronary artery disease and acute myocardial infarction. BMC Cardiovasc Disord 2024; 24:471. [PMID: 39227771 PMCID: PMC11370111 DOI: 10.1186/s12872-024-04146-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between circulating levels of B cell activating factor (BAFF) and the presence and severity of coronary artery disease (CAD) and acute myocardial infarction (AMI) in humans, as its biological functions in this context remain unclear. METHODS Serum BAFF levels were measured in a cohort of 723 patients undergoing angiography, including 204 patients without CAD (control group), 220 patients with stable CAD (CAD group), and 299 patients with AMI (AMI group). Logistic regression analyses were used to assess the association between BAFF and CAD or AMI. RESULTS Significantly elevated levels of BAFF were observed in patients with CAD and AMI compared to the control group. Furthermore, BAFF levels exhibited a positive correlation with the SYNTAX score (r = 0.3002, P < 0.0001) and the GRACE score (r = 0.5684, P < 0.0001). Logistic regression analysis demonstrated that increased BAFF levels were an independent risk factor for CAD (adjusted OR 1.305, 95% CI 1.078-1.580) and AMI (adjusted OR 2.874, 95% CI 1.708-4.838) after adjusting for confounding variables. Additionally, elevated BAFF levels were significantly associated with a high GRACE score (GRACE score 155 to 319, adjusted OR 4.297, 95% CI 1.841-10.030). BAFF exhibited a sensitivity of 75.0% and specificity of 71.4% in differentiating CAD patients with a high SYNTAX score, and a sensitivity of 75.5% and specificity of 72.8% in identifying AMI patients with a high GRACE score. CONCLUSION Circulating BAFF levels serve as a valuable diagnostic marker for CAD and AMI. Elevated BAFF levels are associated with the presence and severity of these conditions, suggesting its potential as a clinically relevant biomarker in cardiovascular disease.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ziyang Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yuke Cui
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hongyang Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Lei Yi
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Jingwei Ni
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Run Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Xiaoqun Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Jinzhou Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Weiwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
| | - Yueying Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
12
|
Ransegnola BP, Pattarabanjird T, McNamara CA. Tipping the Scale: Atheroprotective IgM-Producing B Cells in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1906-1915. [PMID: 39022832 PMCID: PMC11338718 DOI: 10.1161/atvbaha.124.319847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease whose progression is fueled by proinflammatory moieties and limited by anti-inflammatory mediators. Whereas oxidative damage and the generation of oxidation-specific epitopes that act as damage-associated molecular patterns are highly inflammatory, IgM antibodies produced by B-1 and marginal zone B cells counteract unrestricted inflammation by neutralizing and encouraging clearance of these proinflammatory signals. In this review, we focus on describing the identities of IgM-producing B cells in both mice and humans, elaborating the mechanisms underlying IgM production, and discussing the potential strategies to augment the production of atheroprotective IgM. In addition, we will discuss promising therapeutic interventions in humans to help tip the scale toward augmentation of IgM production and to provide atheroprotection.
Collapse
Affiliation(s)
- Brett Patrick Ransegnola
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Pathology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tanyaporn Pattarabanjird
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Coleen A. McNamara
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
13
|
Wang H, Zhao R, Peng L, Yu A, Wang Y. A Dual-Function CD47-Targeting Nano-Drug Delivery System Used to Regulate Immune and Anti-Inflammatory Activities in the Treatment of Atherosclerosis. Adv Healthc Mater 2024; 13:e2400752. [PMID: 38794825 DOI: 10.1002/adhm.202400752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/15/2024] [Indexed: 05/26/2024]
Abstract
Atherosclerosis is a primary contributor to cardiovascular disease. Current studies have highlighted the association between the immune system, particularly immune cells, and atherosclerosis, although treatment options and clinical trials remain scarce. Immunotherapy for cardiovascular disease is still in its infancy. Bruton's tyrosine kinase (BTK), widely expressed in various immune cells, represents a promising therapeutic target for atherosclerosis by modulating the anti-inflammatory function of immune cells. This study introduces a polydopamine-based nanocarrier system to deliver the BTK inhibitor, ibrutinib, to atherosclerotic plaques with an active targeting property via an anti-CD47 antibody. Leveraging polydopamine's pH-sensitive reversible disassembly, the system offers responsive, controlled release within the pathologic microenvironment. This allows precise and efficient ibrutinib delivery, concurrently inhibiting the activation of the NF-κB pathway in B cells and the NLRP3 inflammasome in macrophages within the plaques. This treatment also modulates both the immune cell microenvironment and inflammatory conditions in atherosclerotic lesions, thereby conveying promising therapeutic effects for atherosclerosis in vivo. This strategy also provides a novel option for atherosclerosis treatment.
Collapse
Affiliation(s)
- Huanhuan Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Runze Zhao
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lei Peng
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ao Yu
- Tianjin Key Laboratory of Molecular Recognition and Biosensing, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yongjian Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
14
|
Ait-Oufella H, Libby P. Inflammation and Atherosclerosis: Prospects for Clinical Trials. Arterioscler Thromb Vasc Biol 2024; 44:1899-1905. [PMID: 39167675 PMCID: PMC11343092 DOI: 10.1161/atvbaha.124.320155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Affiliation(s)
- Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Sorbonne Université, Paris, France
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Ma Z, Liu L, Tian J, Tu C, Zhang D, Zhang M, Zhang H, An Z, Sun M, Zhang H, Song X. Causal Relationships between Lymphocyte Subsets and Risk of Coronary Artery Disease: A Two-Sample Mendelian Randomization Study. Rev Cardiovasc Med 2024; 25:326. [PMID: 39355583 PMCID: PMC11440411 DOI: 10.31083/j.rcm2509326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 10/03/2024] Open
Abstract
Background Currently, the causal relationship between lymphocyte subsets and coronary artery disease (CAD) remains unclear. Therefore, we utilized Mendelian randomization (MR) to assess the association between lymphocyte subsets and CAD. Methods We performed a two-sample MR analysis using publicly available genome-wide association studies (GWAS) datasets. The primary method of analysis to comprehensively evaluate causal effects was the inverse variance-weighted (IVW) method. The four additional MR approaches were MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analysis incorporated Cochran's Q and MR-Egger intercept tests to identify residual heterogeneity and potential horizontal pleiotropy, respectively. The MR-PRESSO distortion test was applied to identify potential pleiotropic outliers. Leave-one-out analysis confirmed that no single single-nucleotide polymorphism (SNP) significantly affected the MR estimate. We conducted reverse MR analysis to investigate the impact of variables correlated with outcomes in forward MR analysis. Results The IVW method revealed a significant positive association between B cell count and CAD (odds ratio (OR) = 1.08 (95% CI: 1.04, 1.11), p = 2.67 × 10-5). A similar association was observed between B cell count and myocardial infarction (MI) (OR = 1.07 (95% CI: 1.03, 1.11), p = 5.69 × 10-4). Sensitivity analyses detected no outliers, heterogeneity, or pleiotropy. The reverse MR analysis was conducted to investigate the impact of CAD and MI on B cell count, and the IVW results showed no statistical significance. Conclusions Our study suggests that a higher absolute B cell count is linked to an increased risk of CAD and MI.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Chenchen Tu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Dongfeng Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Mingduo Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Meichen Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Hongjia Zhang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| |
Collapse
|
16
|
Yang Y, Zhang F, Jiang Z, Du Z, Liu S, Zhang M, Jin Y, Qin Y, Yang X, Wang C, Gao H. Microplastics are associated with elevated atherosclerotic risk and increased vascular complexity in acute coronary syndrome patients. Part Fibre Toxicol 2024; 21:34. [PMID: 39164741 PMCID: PMC11337598 DOI: 10.1186/s12989-024-00596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Microplastics, widely present in the environment, are implicated in disease pathogenesis through oxidative stress and immune modulation. Prevailing research, primarily based on animal and cell studies, falls short in elucidating microplastics' impact on human cardiovascular health. This cross-sectional study detected blood microplastic concentrations in patients presenting with chest pain using pyrolysis-gas chromatography/mass spectrometry and evaluating inflammatory and immune markers through flow cytometry, to explore the potential effects of microplastic on acute coronary syndrome. RESULTS The study included 101 participants, comprising 19 controls and 82 acute coronary syndrome cases. Notably, acute coronary syndrome patients exhibited elevated microplastic concentrations, with those suffering from acute myocardial infarction presenting higher loads compared to those with unstable angina. Furthermore, patients at intermediate to high risk of coronary artery disease displayed significantly higher microplastic accumulations than their low-risk counterparts. A significant relationship was observed between increased microplastic levels and enhanced IL-6 and IL-12p70 contents, alongside elevated B lymphocyte and natural killer cell counts. CONCLUSION These results suggest an association between microplastics and both vascular pathology complexity and immunoinflammatory response in acute coronary syndrome, underscoring the critical need for targeted research to delineate the mechanisms of this association. HIGHLIGHTS 1 Blood microplastic levels escalate from angiographic patency, to angina patients, peaking in myocardial infarction patients. 2 Microplastics in acute coronary syndrome patients are predominantly PE, followed by PVC, PS, and PP. 3 Microplastics may induce immune cell-associated inflammatory responses in acute coronary syndrome patients.
Collapse
Affiliation(s)
- Yunxiao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Feng Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhili Jiang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhiyong Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Sheng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yanyan Jin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yanwen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xiubin Yang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Chenggang Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Hai Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
17
|
Liu T, Chen Y, Hou L, Yu Y, Ma D, Jiang T, Zhao G. Immune cell-mediated features of atherosclerosis. Front Cardiovasc Med 2024; 11:1450737. [PMID: 39234608 PMCID: PMC11371689 DOI: 10.3389/fcvm.2024.1450737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by innate and adaptive immune responses, which seriously threatens human life and health. It is a primary cause of coronary heart disease, myocardial infarction, and peripheral vascular disease. Research has demonstrated that immune cells are fundamental to the development of atherosclerosis and chronic inflammation. Therefore, it is anticipated that immunotherapy targeting immune cells will be a novel technique in the management of atherosclerosis. This article reviews the growth of research on the regulatory role of immune cells in atherosclerosis and targeted therapy approaches. The purpose is to offer new therapeutic approaches for the control and treatment of cardiovascular illnesses caused by atherosclerosis.
Collapse
Affiliation(s)
- Tingting Liu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yanjun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lianjie Hou
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yulu Yu
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dan Ma
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| |
Collapse
|
18
|
Annink ME, Kraaijenhof JM, Stroes ESG, Kroon J. Moving from lipids to leukocytes: inflammation and immune cells in atherosclerosis. Front Cell Dev Biol 2024; 12:1446758. [PMID: 39161593 PMCID: PMC11330886 DOI: 10.3389/fcell.2024.1446758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the most important cause of morbidity and mortality worldwide. While it is traditionally attributed to lipid accumulation in the vascular endothelium, recent research has shown that plaque inflammation is an important additional driver of atherogenesis. Though clinical outcome trials utilizing anti-inflammatory agents have proven promising in terms of reducing ASCVD risk, it is imperative to identify novel actionable targets that are more specific to atherosclerosis to mitigate adverse effects associated with systemic immune suppression. To that end, this review explores the contributions of various immune cells from the innate and adaptive immune system in promoting and mitigating atherosclerosis by integrating findings from experimental studies, high-throughput multi-omics technologies, and epidemiological research.
Collapse
Affiliation(s)
- Maxim E. Annink
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, Netherlands
| |
Collapse
|
19
|
Qi X, Zhang Y, Wang Y, Sun J, Yang R, Wang S, Dong J, Chen W, Ji F, Yu X. Prognostic value of serum immunoglobulin M levels in patients with acute coronary syndrome. Atherosclerosis 2024; 395:117552. [PMID: 38954858 DOI: 10.1016/j.atherosclerosis.2024.117552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND AND AIMS The immuno-inflammatory response is a crucial early step in the development of acute coronary syndrome (ACS). In this study, we investigated whether immunoglobulin M (IgM) in the body's initial immune response can predict the prognosis of patients with ACS. METHODS This prospective cohort study enrolled 1556 ACS patients at Beijing Hospital between March 2017 and October 2020. All patients underwent coronary angiography (CAG). The serum IgM concentration and biochemical indicators were evaluated prior to CAG. The primary endpoint was the composite endpoint of major adverse cardiovascular and cerebrovascular events (MACCEs). Multivariate Cox proportional hazards models was used to explore the association between IgM levels and the endpoint. RESULTS The average serum IgM levels of the population was 61.3 (42.6-88.4) mg/dL. During the median follow-up period of 55 months, 150 MACCEs occurred. Kaplan-Meier analysis showed that low serum IgM levels were associated with occurrence of MACCEs (log-rank p = 0.009). Univariate Cox proportional hazards models showed that low serum IgM (≤78.05 mg/dL) was associated with MACCEs (hazard ratio (HR) 1.648, 95 % confidence interval (CI): 1.129-2.406, p = 0.010). In patients with IgM ≤78.05 mg/dL, the HR for partially adjusted MACCEs events was 1.576 (95 % CI: 1.075-2.310) and 1.930 (95 % CI: 1.080-3.449) after adjusting for multiple covariates. The subgroup analysis showed that for patients in ≤24 BMI, never smoking and non-dyslipidemia subgroup, the lower serum IgM levels was significantly associated with the risk of MACCEs (pinteraction < 0.001, pinteraction = 0.037, pinteraction = 0.024, respectively). CONCLUSIONS Low serum IgM levels was independently associated with MACCEs in ACS patients, especially for patients without obesity, smoking and dyslipidemia.
Collapse
Affiliation(s)
- Xi Qi
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Yanan Zhang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No.16, Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Yijia Wang
- Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Jiayi Sun
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Ruiyue Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, PR China
| | - Siming Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, PR China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, PR China
| | - Wenxiang Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, PR China
| | - Fusui Ji
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China.
| |
Collapse
|
20
|
O’Brien JW, Case A, Kemper C, Zhao TX, Mallat Z. Therapeutic Avenues to Modulate B-Cell Function in Patients With Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2024; 44:1512-1522. [PMID: 38813699 PMCID: PMC11208059 DOI: 10.1161/atvbaha.124.319844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
The adaptive immune system plays an important role in the development and progression of atherosclerotic cardiovascular disease. B cells can have both proatherogenic and atheroprotective roles, making treatments aimed at modulating B cells important therapeutic targets. The innate-like B-cell response is generally considered atheroprotective, while the adaptive response is associated with mixed consequences for atherosclerosis. Additionally, interactions of B cells with components of the adaptive and innate immune system, including T cells and complement, also represent key points for therapeutic regulation. In this review, we discuss therapeutic approaches based on B-cell depletion, modulation of B-cell survival, manipulation of both the antibody-dependent and antibody-independent B-cell response, and emerging immunization techniques.
Collapse
Affiliation(s)
- James W. O’Brien
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
| | - Ayden Case
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (C.K.)
| | - Tian X. Zhao
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
- Department of Cardiology, Royal Papworth Hospital, Cambridge, United Kingdom (T.X.Z.)
| | - Ziad Mallat
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (J.W.O., A.C., T.X.Z., Z.M.)
- Unversité de Paris, Inserm U970, Paris Cardiovascular Research Centre, France (Z.M.)
| |
Collapse
|
21
|
Hmiel L, Zhang S, Obare LM, Santana MADO, Wanjalla CN, Titanji BK, Hileman CO, Bagchi S. Inflammatory and Immune Mechanisms for Atherosclerotic Cardiovascular Disease in HIV. Int J Mol Sci 2024; 25:7266. [PMID: 39000373 PMCID: PMC11242562 DOI: 10.3390/ijms25137266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Atherosclerotic vascular disease disproportionately affects persons living with HIV (PLWH) compared to those without. The reasons for the excess risk include dysregulated immune response and inflammation related to HIV infection itself, comorbid conditions, and co-infections. Here, we review an updated understanding of immune and inflammatory pathways underlying atherosclerosis in PLWH, including effects of viral products, soluble mediators and chemokines, innate and adaptive immune cells, and important co-infections. We also present potential therapeutic targets which may reduce cardiovascular risk in PLWH.
Collapse
Affiliation(s)
- Laura Hmiel
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Suyu Zhang
- Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Boghuma K. Titanji
- Division of Infectious Diseases, Emory University, Atlanta, GA 30322, USA
| | - Corrilynn O. Hileman
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Shashwatee Bagchi
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
22
|
Jones PW, Mallat Z, Nus M. T-Cell/B-Cell Interactions in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1502-1511. [PMID: 38813700 PMCID: PMC11208060 DOI: 10.1161/atvbaha.124.319845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Atherosclerosis is a complex inflammatory disease in which the adaptive immune response plays an important role. While the overall impact of T and B cells in atherosclerosis is relatively well established, we are only beginning to understand how bidirectional T-cell/B-cell interactions can exert prominent atheroprotective and proatherogenic functions. In this review, we will focus on these T-cell/B-cell interactions and how we could use them to therapeutically target the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Peter William Jones
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| | - Ziad Mallat
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
- INSERM U970, Paris Cardiovascular Research Centre, France (Z.M.)
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| |
Collapse
|
23
|
Meher AK, McNamara CA. B-1 lymphocytes in adipose tissue as innate modulators of inflammation linked to cardiometabolic disease. Immunol Rev 2024; 324:95-103. [PMID: 38747455 PMCID: PMC11262958 DOI: 10.1111/imr.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Fat is stored in distinct depots with unique features in both mice and humans and B cells reside in all adipose depots. We have shown that B cells modulate cardiometabolic disease through activities in two of these key adipose depots: visceral adipose tissue (VAT) and perivascular adipose tissue (PVAT). VAT refers to the adipose tissue surrounding organs, within the abdomen and thorax, and is comprised predominantly of white adipocytes. This depot has been implicated in mediating obesity-related dysmetabolism. PVAT refers to adipose tissue surrounding major arteries. It had long been thought to exist to provide protection and insulation for the vessel, yet recent work demonstrates an important role for PVAT in harboring immune cells, promoting their function and regulating the biology of the underlying vessel. The role of B-2 cells and adaptive immunity in adipose tissue biology has been nicely reviewed elsewhere. Given that, the predominance of B-1 cells in adipose tissue at homeostasis, and the emerging role of B-1 cells in a variety of disease states, we will focus this review on how B-1 cells function in VAT and PVAT depots to promote homeostasis and limit inflammation linked to cardiometabolic disease and factors that regulate this function.
Collapse
Affiliation(s)
- Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Coleen A. McNamara
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
24
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2024:S2090-1232(24)00253-4. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
25
|
Obare LM, Bonami RH, Doran A, Wanjalla CN. B cells and atherosclerosis: A HIV perspective. J Cell Physiol 2024; 239:e31270. [PMID: 38651687 PMCID: PMC11209796 DOI: 10.1002/jcp.31270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/09/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Atherosclerosis remains a leading cause of cardiovascular disease (CVD) globally, with the complex interplay of inflammation and lipid metabolism at its core. Recent evidence suggests a role of B cells in the pathogenesis of atherosclerosis; however, this relationship remains poorly understood, particularly in the context of HIV. We review the multifaceted functions of B cells in atherosclerosis, with a specific focus on HIV. Unique to atherosclerosis is the pivotal role of natural antibodies, particularly those targeting oxidized epitopes abundant in modified lipoproteins and cellular debris. B cells can exert control over cellular immune responses within atherosclerotic arteries through antigen presentation, chemokine production, cytokine production, and cell-cell interactions, actively participating in local and systemic immune responses. We explore how HIV, characterized by chronic immune activation and dysregulation, influences B cells in the context of atherosclerosis, potentially exacerbating CVD risk in persons with HIV. By examining the proatherogenic and antiatherogenic properties of B cells, we aim to deepen our understanding of how B cells influence atherosclerotic plaque development, especially within the framework of HIV. This research provides a foundation for novel B cell-targeted interventions, with the potential to mitigate inflammation-driven cardiovascular events, offering new perspectives on CVD risk management in PLWH.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amanda Doran
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
26
|
Sharma H, Mossman K, Austin RC. Fatal attractions that trigger inflammation and drive atherosclerotic disease. Eur J Clin Invest 2024; 54:e14169. [PMID: 38287209 DOI: 10.1111/eci.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Atherosclerosis is the salient, underlying cause of cardiovascular diseases, such as arrhythmia, coronary artery disease, cardiomyopathy, pulmonary embolism and myocardial infarction. In recent years, atherosclerosis pathophysiology has evolved from a lipid-based to an inflammation-centric ideology. METHODS This narrative review is comprised of review and original articles that were found through the PubMed search engine. The following search terms or amalgamation of terms were used: "cardiovascular disease," "atherosclerosis," "inflammation," "GRP78," "Hsp60," "oxidative low-density lipoproteins," "aldehyde dehydrogenase," "β2-glycoprotein," "lipoprotein lipase A," "human cytomegalovirus." "SARS-CoV-2," "chlamydia pneumonia," "autophagy," "thrombosis" and "therapeutics." RESULTS Emerging evidence supports the concept that atherosclerosis is associated with the interaction between cell surface expression of stress response chaperones, including GRP78 and Hsp60, and their respective autoantibodies. Moreover, various other autoantigens and their autoantibodies have displayed a compelling connection with the development of atherosclerosis, including oxidative low-density lipoproteins, aldehyde dehydrogenase, β2-glycoprotein and lipoprotein lipase A. Atherosclerosis progression is also concurrent with viral and bacterial activators of various diseases. This narrative review will focus on the contributions of human cytomegalovirus as well as SARS-CoV-2 and chlamydia pneumonia in atherosclerosis development. Notably, the interaction of an autoantigen with their respective autoantibodies or the presence of a foreign antigen can enhance inflammation development, which leads to atherosclerotic lesion progression. CONCLUSION We will highlight and discuss the complex role of the interaction between autoantigens and autoantibodies, and the presence of foreign antigens in the development of atherosclerotic lesions in relationship to pro-inflammatory responses.
Collapse
Affiliation(s)
- Hitesh Sharma
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Karen Mossman
- Department of Medicine, Michael DeGroote Institute for Infectious Disease Research and the McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| |
Collapse
|
27
|
Mallat Z, Tedgui A. Century of Milestones and Breakthroughs Related to the Immune Mechanisms of Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1002-1006. [PMID: 38657035 PMCID: PMC11042514 DOI: 10.1161/atvbaha.124.319397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Affiliation(s)
- Ziad Mallat
- Department of Medicine, Section of CardioRespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (Z.M.)
- Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, Université de Paris, France (Z.M., A.T.)
| | - Alain Tedgui
- Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, Université de Paris, France (Z.M., A.T.)
| |
Collapse
|
28
|
Wang M, Zhang X, Fan R, Zhang L. Causal role of immune cell traits in stroke: A Mendelian randomization study. J Stroke Cerebrovasc Dis 2024; 33:107625. [PMID: 38316285 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/04/2024] [Accepted: 02/02/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVES Immune mechanisms play a crucial role in the development of stroke. However, immune-related phenotypes are diverse and their associations with stroke are largely unknown. Here, we aimed to systematically explore the causal role of immune cell traits in stroke and its subtypes by leveraging data from genome-wide association studies (GWASs). MATERIALS AND METHODS Exposure data were obtained from a recent GWAS on 731 immune cell traits profiled by flow cytometry involving 3757 individuals. By conducting two-sample univariable Mendelian randomization (MR) analyses, each immune cell trait was assessed for causal relationships with stroke outcomes from the MEGASTROKE Consortium (40,585 cases and 406,111 controls). The robustness of the MR results was verified by a series of sensitivity analyses. RESULTS We identified three significant associations after Bonferroni correction (P < 1.37E-05). Increased CD27 expression on memory B cell (OR = 1.23, 95% CI = 1.14-1.33, P = 2.78E-08), IgD-CD38dim B cell (OR = 1.16, 95% CI = 1.09-1.23, P = 5.98E-06) and unswitched memory B cell (OR = 1.18, 95% CI = 1.10-1.27, P = 1.09E-05) were associated with a higher risk of large-artery atherosclerotic stroke (LAS). Furthermore, expression quantitative trait loci data also indicated elevated blood CD27 mRNA level was a risk factor for LAS (OR = 1.37, 95% CI = 1.02-1.84, P = 0.037). CONCLUSIONS This study provided genetic evidence of the causal relationship between immune cell traits and stroke, highlighting the role of CD27 on memory B cell as a novel factor for LAS risk.
Collapse
Affiliation(s)
- Maiqiu Wang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China; Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Hangzhou, China.
| | - Xu Zhang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China; Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Hangzhou, China.
| | - Rongli Fan
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China; Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Hangzhou, China.
| | - Lei Zhang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China; Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Hangzhou, China; School of Information and Electronic Engineering, Zhejiang University of Science and Technology, Hangzhou, China.
| |
Collapse
|
29
|
Wang L, Huang S, Liang X, Zhou J, Han Y, He J, Xu D. Immuno-modulatory role of baicalin in atherosclerosis prevention and treatment: current scenario and future directions. Front Immunol 2024; 15:1377470. [PMID: 38698839 PMCID: PMC11063305 DOI: 10.3389/fimmu.2024.1377470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/27/2024] [Indexed: 05/05/2024] Open
Abstract
Atherosclerosis (AS) is recognized as a chronic inflammatory condition characterized by the accumulation of lipids and inflammatory cells within the damaged walls of arterial vessels. It is a significant independent risk factor for ischemic cardiovascular disease, ischemic stroke, and peripheral arterial disease. Despite the availability of current treatments such as statins, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, and lifestyle modifications for prevention, AS remains a leading cause of morbidity and economic burden worldwide. Thus, there is a pressing need for the development of new supplementary and alternative therapies or medications. Huangqin (Scutellaria baicalensis Georgi. [SBG]), a traditional Chinese medicine, exerts a significant immunomodulatory effect in AS prevention and treatment, with baicalin being identified as one of the primary active ingredients of traditional Chinese medicine. Baicalin offers a broad spectrum of pharmacological activities, including the regulation of immune balance, antioxidant and anti-inflammatory effects, and improvement of lipid metabolism dysregulation. Consequently, it exerts beneficial effects in both AS onset and progression. This review provides an overview of the immunomodulatory properties and mechanisms by which baicalin aids in AS prevention and treatment, highlighting its potential as a clinical translational therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiangshan He
- Department of Traditional Chinese Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Danping Xu
- Department of Traditional Chinese Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
30
|
Snijckers RPM, Foks AC. Adaptive immunity and atherosclerosis: aging at its crossroads. Front Immunol 2024; 15:1350471. [PMID: 38686373 PMCID: PMC11056569 DOI: 10.3389/fimmu.2024.1350471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Adaptive immunity plays a profound role in atherosclerosis pathogenesis by regulating antigen-specific responses, inflammatory signaling and antibody production. However, as we age, our immune system undergoes a gradual functional decline, a phenomenon termed "immunosenescence". This decline is characterized by a reduction in proliferative naïve B- and T cells, decreased B- and T cell receptor repertoire and a pro-inflammatory senescence associated secretory profile. Furthermore, aging affects germinal center responses and deteriorates secondary lymphoid organ function and structure, leading to impaired T-B cell dynamics and increased autoantibody production. In this review, we will dissect the impact of aging on adaptive immunity and the role played by age-associated B- and T cells in atherosclerosis pathogenesis, emphasizing the need for interventions that target age-related immune dysfunction to reduce cardiovascular disease risk.
Collapse
Affiliation(s)
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
31
|
Ambler WG, Kaplan MJ. Vascular damage in systemic lupus erythematosus. Nat Rev Nephrol 2024; 20:251-265. [PMID: 38172627 PMCID: PMC11391830 DOI: 10.1038/s41581-023-00797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/05/2024]
Abstract
Vascular disease is a major cause of morbidity and mortality in patients with systemic autoimmune diseases, particularly systemic lupus erythematosus (SLE). Although comorbid cardiovascular risk factors are frequently present in patients with SLE, they do not explain the high burden of premature vascular disease. Profound innate and adaptive immune dysregulation seems to be the primary driver of accelerated vascular damage in SLE. In particular, evidence suggests that dysregulation of type 1 interferon (IFN-I) and aberrant neutrophils have key roles in the pathogenesis of vascular damage. IFN-I promotes endothelial dysfunction directly via effects on endothelial cells and indirectly via priming of immune cells that contribute to vascular damage. SLE neutrophils are vasculopathic in part because of their increased ability to form immunostimulatory neutrophil extracellular traps. Despite improvements in clinical care, cardiovascular disease remains the leading cause of mortality among patients with SLE, and treatments that improve vascular outcomes are urgently needed. Improved understanding of the mechanisms of vascular injury in inflammatory conditions such as SLE could also have implications for common cardiovascular diseases, such as atherosclerosis and hypertension, and may ultimately lead to personalized therapeutic approaches to the prevention and treatment of this potentially fatal complication.
Collapse
Affiliation(s)
- William G Ambler
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
32
|
Wang T, Xiong X, Xiao N, Yan Y, Liu X, Xie Q, Su X, Chen M, Peng J, Wang S, Mei H, Lin G, Gong F, Cheng L. The therapeutic effect of anti-CD19 antibody on DHEA-induced PCOS mice. Int Immunopharmacol 2024; 130:111711. [PMID: 38428145 DOI: 10.1016/j.intimp.2024.111711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 03/03/2024]
Abstract
Immune dysregulation has been summarized as a critical factor in the occurrence and development of Polycystic ovary syndrome (PCOS), but potential mediators and mechanisms remain unclear. Our previous study showed that CD19+ B cells were involved in the pathogenesis of dehydroepiandrosterone (DHEA)-induced PCOS mice. Here, we studied the therapeutic potential of anti-CD19 antibody (aCD19 Ab) on DHEA-induced PCOS mice. The results showed that aCD19 Ab treatment improved ovarian pathological structure and function of PCOS mice, manifested by an increased number of corpus luteum, a decreased number of cystic follicles and atretic follicles, and regular estrus cycles. The aCD19 Ab treatment reduced the proportion of splenic CD21+ CD23low marginal zone B cells as well as the level of serum IgM and decreased the percentage of peripheral blood and splenic neutrophils. In particular, aCD19 Ab treatment reduced the apoptosis of granulosa cells and macrophage infiltration in ovarian secondary follicles of PCOS mice, as well as the expression of TNF-α in ovarian tissue and serum TNF-α levels. Moreover, we confirmed that TNF-α induced the apoptosis of human ovarian granulosa tumor cell line cells in vitro. Thus, our work demonstrates that aCD19 Ab treatment improves ovarian pathological phenotype and function by reducing local and systemic inflammation in PCOS mice, which may provide a novel insight into PCOS therapy.
Collapse
Affiliation(s)
- Ting Wang
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Xingliang Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Na Xiao
- National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China; Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Yizhong Yan
- National Engineering and Research Center of Human Stem Cells, Changsha, China
| | - Xiaoyang Liu
- Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Qi Xie
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Xian Su
- National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China
| | - Maosheng Chen
- Huaihua City Maternal and Child Health Care Hospital, HuaiHua, China
| | - Jing Peng
- National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China
| | - Siqi Wang
- National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China
| | - Hua Mei
- National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China; National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China; NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China; NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Lamei Cheng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China; National Engineering and Research Center of Human Stem Cells, Changsha, China; Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China; Guangxiu Hospital, Hunan Normal University, Changsha, China; NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
33
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
34
|
Zhang YJ, Huang C, Zu XG, Liu JM, Li YJ. Use of Machine Learning for the Identification and Validation of Immunogenic Cell Death Biomarkers and Immunophenotypes in Coronary Artery Disease. J Inflamm Res 2024; 17:223-249. [PMID: 38229693 PMCID: PMC10790656 DOI: 10.2147/jir.s439315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
Objective Immunogenic cell death (ICD) is part of the immune system's response to coronary artery disease (CAD). In this study, we bioinformatically evaluated the diagnostic and therapeutic utility of immunogenic cell death-related genes (IRGs) and their relationship with immune infiltration features in CAD. Methods We acquired the CAD-related datasets GSE12288, GSE71226, and GSE120521 from the Gene Expression Omnibus (GEO) database and the IRGs from the GeneCards database. After identifying the immune cell death-related differentially expressed genes (IRDEGs), we developed a risk model and detected immune subtypes in CAD. IRDEGs were identified using least absolute shrinkage and selection operator (LASSO) analysis. Using a nomogram, we confirmed that both the LASSO model and ICD signature genes had good diagnostic performance. Results There was a high degree of coincidence and immune representativeness between two CAD groups based on characteristic genes and hub genes. Hub genes were associated with the interaction of neuroactive ligands with receptors and cell adhesion receptors. The two groups differed in terms of adipogenesis, allograft rejection, and apoptosis, as well as the ICD signature and hub gene expression levels. The two CAD-ICD subtypes differed in terms of immune infiltration. Conclusion Quantitative real-time PCR (qRT-PCR) correlated CAD with the expression of OAS3, ITGAV, and PIBF1. The ICD signature genes are candidate biomarkers and reference standards for immune grouping in CAD and can be beneficial in precise immune-targeted therapy.
Collapse
Affiliation(s)
- Yan-jiao Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Chao Huang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Xiu-guang Zu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Jin-ming Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Yong-jun Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| |
Collapse
|
35
|
Gao F, Litchfield B, Wu H. Adipose tissue lymphocytes and obesity. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:5. [PMID: 38455510 PMCID: PMC10919906 DOI: 10.20517/jca.2023.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Obesity is associated with chronic inflammation in adipose tissue (AT), mainly evidenced by infiltration and phenotypic changes of various types of immune cells. Macrophages are the major innate immune cells and represent the predominant immune cell population within AT. Lymphocytes, including T cells and B cells, are adaptive immune cells and constitute another important immune cell population in AT. In obesity, CD8+ effector memory T cells, CD4+ Th1 cells, and B2 cells are increased in AT and promote AT inflammation, while regulatory T cells and Th2 cells, which usually function as immune regulatory or type 2 inflammatory cells, are reduced in AT. Immune cells may regulate the metabolism of adipocytes and other cells through various mechanisms, contributing to the development of metabolic diseases, including insulin resistance and type 2 diabetes. Efforts targeting immune cells and inflammation to prevent and treat obesity-linked metabolic disease have been explored, but have not yielded significant success in clinical studies. This review provides a concise overview of the changes in lymphocyte populations within AT and their potential role in AT inflammation and the regulation of metabolic functions in the context of obesity.
Collapse
Affiliation(s)
- Feng Gao
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
36
|
Sayer M, Chapman GB, Thomas M, Dhaun N. Cardiovascular Disease in Anti-neutrophil Cytoplasm Antibody-Associated Vasculitis. Curr Rheumatol Rep 2024; 26:12-23. [PMID: 38015334 PMCID: PMC10776689 DOI: 10.1007/s11926-023-01123-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE OF REVIEW Anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) is a rare, multisystem, autoimmune disease characterised by microvascular inflammation. Over the past 20 years, advances in immunological management have improved short-term patient outcomes. Longer-term patient outcomes remain poor with cardiovascular disease now the leading cause of death in AAV. Here, we examine the potential pathways that contribute to the increased risk of cardiovascular disease in AAV and the current evidence to manage this risk. RECENT FINDINGS The incidence of cardiovascular disease in AAV exceeds that expected by traditional risk factors alone, suggesting a contribution from disease-specific factors. Similarly, it is unclear how different immunosuppressive therapies contribute to and modify cardiovascular risk, and there is a paucity of data examining the efficacy of traditional cardioprotective medications in AAV. There is a lack of evidence-based cardiovascular risk assessment tools and cardioprotective therapies in patients with AAV which should be addressed to improve long-term outcomes.
Collapse
Affiliation(s)
- Matthew Sayer
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Gavin B Chapman
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Matthew Thomas
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Neeraj Dhaun
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK.
| |
Collapse
|
37
|
Zhang Y, Jiang Y, Zou Y, Fan Y, Feng P, Fu X, Li K, Zhang J, Dong Y, Yan S, Zhang Y. Peripheral blood CD19 positive B lymphocytes increase after ischemic stroke and correlate with carotid atherosclerosis. Front Neurol 2023; 14:1308041. [PMID: 38221996 PMCID: PMC10784375 DOI: 10.3389/fneur.2023.1308041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024] Open
Abstract
Introduction Atherosclerosis is the primary pathological basis of ischemic stroke, and dyslipidemia is one of its major etiological factors. Acute ischemic stroke patients exhibit imbalances in lymphocyte subpopulations, yet the correlation between these dynamic changes in lymphocyte subpopulations and lipid metabolism disorders, as well as carotid atherosclerosis in stroke patients remains poorly understood. Methods We retrospectively analyzed the demographic data, risk factors of cerebrovascular disease, laboratory examination (lymphocyte subsets, lipid indexes, etc.), clinical features and c;/]-sity from December 2017 to September 2019 and non-stroke patients with dizziness/vertigo during the same period. Results The results showed that peripheral B lymphocyte proportions are elevated in acute ischemic stroke patients compared with those of the control group (13.6 ± 5.3 vs. 11.7 ± 4.4%, p = 0.006). Higher B lymphocyte proportions are associated with concurrent dyslipidemia, increased levels of vascular risk factors including triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and very-low-density lipoprotein cholesterol (VLDL-C), as well as decreased levels of the protective factor high-density lipoprotein cholesterol (HDL-C). Elevated B lymphocyte proportions are independently correlated with carotid atherosclerosis in stroke patients. Discussion We found CD19 positive B Lymphocytes increase after ischemic stroke and correlate with Carotid Atherosclerosis. Lymphocyte subpopulations should be highlighted in stroke patients.
Collapse
Affiliation(s)
- Yuhua Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Jiang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yutian Zou
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurology, Afflliated Changshu Hospital of Nantong University, Changshu, China
| | - Yinyin Fan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ping Feng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Fu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Keru Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinru Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunlei Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shuying Yan
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanlin Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Cheng T, You Y, Jia B, Wang H, Lv M, Zhu X, Hu Y. Knowledge mapping of B cell and atherosclerosis over the past 20 years: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2277567. [PMID: 37953301 PMCID: PMC10760366 DOI: 10.1080/21645515.2023.2277567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023] Open
Abstract
Atherosclerosis (AS) is the main underlying cause of cardiovascular disease, and B cells are considered a key immune cell type to regulate AS. So far, there is no bibliometric study on B cell and AS. This study aims to comprehensively analyze the scientific output about B cell and AS, summarize the literature characteristics, explore research hotspots, and point out emerging trends. We searched the literature from 2003 to 2022 from the Web of Science Core Collection (WoSCC) database. CiteSpace, VOSviewer, and the R package "Bibliometrix" were used for literature analysis and visualization. A total of 1,062 articles and reviews were identified. The number of annual publications generally showed an upward trend. The United States and China were the most productive countries. Medical University of Vienna was the most productive research institution, and Binder Christoph J. was the most productive author, who was also from Medical University of Vienna. "Arteriosclerosis Thrombosis and Vascular Biology" was the most published journal and the most frequently cited journal. The most cited reference was written by Caligiuri G (2002) in "Journal of Clinical Investigation." The most frequent keywords were "inflammation," "macrophages," "cardiovascular disease," "T cells," "apoptosis," "immunity," "cytokines," "lymphocytes," etc. The trend topics were mainly focused on "immune infiltration," "immunoglobulins," and "biomarkers." The complex role of B cell subtypes and a variety of B cell mediators is the main research direction at present. In-depth analysis of B cell-specific targets can provide new ideas and methods for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yaping You
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Bochao Jia
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Wang
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Meng Lv
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xueping Zhu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
39
|
Raposo-Gutiérrez I, Rodríguez-Ronchel A, Ramiro AR. Atherosclerosis antigens as targets for immunotherapy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1129-1147. [PMID: 39196152 DOI: 10.1038/s44161-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 08/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries that can lead to thrombosis, infarction and stroke, underlying the first cause of mortality worldwide. Adaptive immunity plays critical roles in atherosclerosis, and numerous studies have ascribed both atheroprotective and atherogenic functions to specific subsets of T and B cells. However, less is known on how antigen specificity determines the protective or adverse outcome of such adaptive responses. Understanding antigen triggers in atherosclerosis is crucial to delve deeper into mechanisms of disease initiation and progression and to implement specific immunotherapeutic approaches, including vaccination strategies. Here we review the role of adaptive immunity in atherosclerosis and the insights that single-cell technology has provided into the function of distinct immune cell subsets. We outline the most relevant atherosclerosis antigens and antibodies reported to date and examine their immunotherapeutic potential. Finally, we review the most promising vaccination-based clinical trials targeting the adaptive immune system.
Collapse
Affiliation(s)
- Irene Raposo-Gutiérrez
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Ana Rodríguez-Ronchel
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Almudena R Ramiro
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain.
| |
Collapse
|
40
|
Laera N, Malerba P, Vacanti G, Nardin S, Pagnesi M, Nardin M. Impact of Immunity on Coronary Artery Disease: An Updated Pathogenic Interplay and Potential Therapeutic Strategies. Life (Basel) 2023; 13:2128. [PMID: 38004268 PMCID: PMC10672143 DOI: 10.3390/life13112128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. It is a result of the buildup of atherosclerosis within the coronary arteries. The role of the immune system in CAD is complex and multifaceted. The immune system responds to damage or injury to the arterial walls by initiating an inflammatory response. However, this inflammatory response can become chronic and lead to plaque formation. Neutrophiles, macrophages, B lymphocytes, T lymphocytes, and NKT cells play a key role in immunity response, both with proatherogenic and antiatherogenic signaling pathways. Recent findings provide new roles and activities referring to endothelial cells and vascular smooth muscle cells, which help to clarify the intricate signaling crosstalk between the involved actors. Research is ongoing to explore immunomodulatory therapies that target the immune system to reduce inflammation and its contribution to atherosclerosis. This review aims to summarize the pathogenic interplay between immunity and CAD and the potential therapeutic strategies, and explore immunomodulatory therapies that target the immune system to reduce inflammation and its contribution to atherosclerosis.
Collapse
Affiliation(s)
- Nicola Laera
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy;
- Second Medicine Division, Department of Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Paolo Malerba
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy;
- Division of Medicine, Department of Medicine, ASST Spedali Civili di Montichiari, 25018 Montichiari, Italy
| | - Gaetano Vacanti
- Medical Clinic IV, Department of Cardiology, Municipal Hospital, 76133 Karlsruhe, Germany;
| | - Simone Nardin
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, 16126 Genova, Italy
| | - Matteo Pagnesi
- Division of Cardiology, ASST Spedali Civili of Brescia, 25123 Brescia, Italy;
| | - Matteo Nardin
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy;
- Third Medicine Division, Department of Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
41
|
Sun X, Lu Y, Wu J, Wen Q, Li Z, Tang Y, Shi Y, He T, Liu L, Huang W, Weng C, Wu Q, Xiao Q, Yuan H, Xu Q, Cai J. Meta-Analysis of Single-Cell RNA-Seq Data Reveals the Mechanism of Formation and Heterogeneity of Tertiary Lymphoid Organ in Vascular Disease. Arterioscler Thromb Vasc Biol 2023; 43:1867-1886. [PMID: 37589134 PMCID: PMC10521807 DOI: 10.1161/atvbaha.123.318762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Tertiary lymphoid organs (TLOs) are ectopic lymphoid organs developed in nonlymphoid tissues with chronic inflammation, but little is known about their existence in different types of vascular diseases and the mechanism that mediated their development. METHODS To take advantage of single-cell RNA sequencing techniques, we integrated 28 single-cell RNA sequencing data sets containing 5 vascular disease models (atherosclerosis, abdominal aortic aneurysm, intimal hyperplasia, isograft, and allograft) to explore TLOs existence and environment supporting its growth systematically. We also searched Medline, Embase, PubMed, and Web of Science from inception to January 2022 for published histological images of vascular remodeling for histological evidence to support TLO genesis. RESULTS Accumulation and infiltration of innate and adaptive immune cells have been observed in various remodeling vessels. Interestingly, the proportion of such immune cells incrementally increases from atherosclerosis to intimal hyperplasia, abdominal aortic aneurysm, isograft, and allograft. Importantly, we uncovered that TLO structure cells, such as follicular helper T cells and germinal center B cells, present in all remodeled vessels. Among myeloid cells and lymphocytes, inflammatory macrophages, and T helper 17 cells are the major lymphoid tissue inducer cells which were found to be positively associated with the numbers of TLO structural cells in remodeled vessels. Vascular stromal cells also actively participate in vascular TLO genesis by communicating with myeloid cells and lymphocytes via CCLs (C-C motif chemokine ligands), CXCL (C-X-C motif ligand), lymphotoxin, BMP (bone morphogenetic protein) chemotactic, FGF-2 (fibroblast growth factor-2), and IGF (insulin growth factor) proliferation mechanisms, particularly for lymphoid tissue inducer cell aggregation. Additionally, the interaction between stromal cells and immune cells modulates extracellular matrix remodeling. Among TLO structure cells, follicular helper T, and germinal center B cells have strong interactions via TCR (T-cell receptor), CD40 (cluster of differentiation 40), and CXCL signaling, to promote the development and maturation of the germinal center in TLO. Consistently, by reviewing the histological images from the literature, TLO genesis was found in those vascular remodeling models. CONCLUSIONS Our analysis showed the existence of TLOs across 5 models of vascular diseases. The mechanisms that support TLOs formation in different models are heterogeneous. This study could be a valuable resource for understanding and discovering new therapeutic targets for various forms of vascular disease.
Collapse
Affiliation(s)
- Xuejing Sun
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Yao Lu
- The Center of Clinical Pharmacology (Y.L., H.Y.), Central South University, Changsha, China
| | - Junru Wu
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Qing Wen
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Zhengxin Li
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Yan Tang
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Yunmin Shi
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Tian He
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Lun Liu
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Wei Huang
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Chunyan Weng
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Qing Wu
- The Third Xiangya Hospital and High-Performance Computing Center (Q. Wu), Central South University, Changsha, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
| | - Hong Yuan
- The Center of Clinical Pharmacology (Y.L., H.Y.), Central South University, Changsha, China
| | - Qingbo Xu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (Q. Xu)
| | - Jingjing Cai
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| |
Collapse
|
42
|
Slysz J, Sinha A, DeBerge M, Singh S, Avgousti H, Lee I, Glinton K, Nagasaka R, Dalal P, Alexandria S, Wai CM, Tellez R, Vescovo M, Sunderraj A, Wang X, Schipma M, Sisk R, Gulati R, Vallejo J, Saigusa R, Lloyd-Jones DM, Lomasney J, Weinberg S, Ho K, Ley K, Giannarelli C, Thorp EB, Feinstein MJ. Single-cell profiling reveals inflammatory polarization of human carotid versus femoral plaque leukocytes. JCI Insight 2023; 8:e171359. [PMID: 37471165 PMCID: PMC10544225 DOI: 10.1172/jci.insight.171359] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Femoral atherosclerotic plaques are less inflammatory than carotid plaques histologically, but limited cell-level data exist regarding comparative immune landscapes and polarization at these sites. We investigated intraplaque leukocyte phenotypes and transcriptional polarization in 49 patients undergoing femoral (n = 23) or carotid (n = 26) endarterectomy using single-cell RNA-Seq (scRNA-Seq; n = 13), flow cytometry (n = 24), and IHC (n = 12). Comparative scRNA-Seq of CD45+-selected leukocytes from femoral (n = 9; 35,265 cells) and carotid (n = 4; 30,655 cells) plaque revealed distinct transcriptional profiles. Inflammatory foam cell-like macrophages and monocytes comprised higher proportions of myeloid cells in carotid plaques, whereas noninflammatory foam cell-like macrophages and LYVE1-overexpressing macrophages comprised higher proportions of myeloid cells in femoral plaque (P < 0.001 for all). A significant comparative excess of CCR2+ macrophages in carotid versus plaque was observed by flow cytometry in a separate validation cohort. B cells were more prevalent and exhibited a comparatively antiinflammatory profile in femoral plaque, whereas cytotoxic CD8+ T cells were more prevalent in carotid plaque. In conclusion, human femoral plaques exhibit distinct macrophage phenotypic and transcriptional profiles as well as diminished CD8+ T cell populations compared with human carotid plaques.
Collapse
Affiliation(s)
| | - Arjun Sinha
- Division of Cardiology, Department of Medicine
| | | | | | | | - Inhyeok Lee
- Division of Cardiology, Department of Medicine
| | - Kristofor Glinton
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
| | | | | | - Shaina Alexandria
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| | - Ching Man Wai
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Ricardo Tellez
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
| | | | | | - Xinkun Wang
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Matthew Schipma
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Ryan Sisk
- Division of Cardiology, Department of Medicine
| | - Rishab Gulati
- La Jolla Institute of Immunology, La Jolla, California, USA
| | | | | | - Donald M. Lloyd-Jones
- Division of Cardiology, Department of Medicine
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| | | | | | - Karen Ho
- Division of Vascular Surgery, NUFSM, Chicago, Illinois, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta, Georgia, USA
| | - Chiara Giannarelli
- Department of Medicine and
- Department of Pathology, New York University, New York, New York, USA
| | | | - Matthew J. Feinstein
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| |
Collapse
|
43
|
Bazioti V, Halmos B, Westerterp M. T-cell Cholesterol Accumulation, Aging, and Atherosclerosis. Curr Atheroscler Rep 2023; 25:527-534. [PMID: 37395922 PMCID: PMC10471657 DOI: 10.1007/s11883-023-01125-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW The majority of leukocytes in advanced human atherosclerotic plaques are T-cells. T-cell subsets exert pro- or anti-atherogenic effects largely via the cytokines they secrete. Tregulatory cells (Tregs) are anti-inflammatory, but may lose these properties during atherosclerosis, proposed to be downstream of cholesterol accumulation. Aged T-cells also accumulate cholesterol. The effects of T-cell cholesterol accumulation on T-cell fate and atherosclerosis are not uniform. RECENT FINDINGS T-cell cholesterol accumulation enhances differentiation into pro-atherogenic cytotoxic T-cells and boosts their killing capacity, depending on the localization and extent of cholesterol accumulation. Excessive cholesterol accumulation induces T-cell exhaustion or T-cell apoptosis, the latter decreasing atherosclerosis but impairing T-cell functionality in terms of killing capacity and proliferation. This may explain the compromised T-cell functionality in aged T-cells and T-cells from CVD patients. The extent of T-cell cholesterol accumulation and its cellular localization determine T-cell fate and downstream effects on atherosclerosis and T-cell functionality.
Collapse
Affiliation(s)
- Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, 80336, Munich, Germany
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands.
| |
Collapse
|
44
|
Zhang Y, Vandestienne M, Lavillegrand JR, Joffre J, Santos-Zas I, Lavelle A, Zhong X, Le Goff W, Guérin M, Al-Rifai R, Laurans L, Bruneval P, Guérin C, Diedisheim M, Migaud M, Puel A, Lanternier F, Casanova JL, Cochain C, Zernecke A, Saliba AE, Mokry M, Silvestre JS, Tedgui A, Mallat Z, Taleb S, Lenoir O, Vindis C, Camus SM, Sokol H, Ait-Oufella H. Genetic inhibition of CARD9 accelerates the development of atherosclerosis in mice through CD36 dependent-defective autophagy. Nat Commun 2023; 14:4622. [PMID: 37528097 PMCID: PMC10394049 DOI: 10.1038/s41467-023-40216-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 07/17/2023] [Indexed: 08/03/2023] Open
Abstract
Caspase recruitment-domain containing protein 9 (CARD9) is a key signaling pathway in macrophages but its role in atherosclerosis is still poorly understood. Global deletion of Card9 in Apoe-/- mice as well as hematopoietic deletion in Ldlr-/- mice increases atherosclerosis. The acceleration of atherosclerosis is also observed in Apoe-/-Rag2-/-Card9-/- mice, ruling out a role for the adaptive immune system in the vascular phenotype of Card9 deficient mice. Card9 deficiency alters macrophage phenotype through CD36 overexpression with increased IL-1β production, increased lipid uptake, higher cell death susceptibility and defective autophagy. Rapamycin or metformin, two autophagy inducers, abolish intracellular lipid overload, restore macrophage survival and autophagy flux in vitro and finally abolish the pro-atherogenic effects of Card9 deficiency in vivo. Transcriptomic analysis of human CARD9-deficient monocytes confirms the pathogenic signature identified in murine models. In summary, CARD9 is a key protective pathway in atherosclerosis, modulating macrophage CD36-dependent inflammatory responses, lipid uptake and autophagy.
Collapse
Affiliation(s)
- Yujiao Zhang
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Marie Vandestienne
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | | | - Jeremie Joffre
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Sorbonne Université, Paris, France
| | - Icia Santos-Zas
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Aonghus Lavelle
- Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology department, Paris, France
| | - Xiaodan Zhong
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Wilfried Le Goff
- Inserm UMRS1166, ICAN, Institute of CardioMetabolism and Nutrition, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Maryse Guérin
- Inserm UMRS1166, ICAN, Institute of CardioMetabolism and Nutrition, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Rida Al-Rifai
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Ludivine Laurans
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Patrick Bruneval
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Department of Anatomopathology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Coralie Guérin
- Institut Curie, Cytometry Platform, 75006, Paris, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), 37540 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades (INEM), Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, 75015, Paris, France
| | - Melanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Fanny Lanternier
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
| | - Clément Cochain
- Comprehensive Heart Failure Center Wuerzburg, University Hospital Wuerzburg, Wuerzburg, Germany
- Institute of Experimental Biomedicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Wuerzburg, Germany
| | - Michal Mokry
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | | | - Alain Tedgui
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Ziad Mallat
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | - Soraya Taleb
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Olivia Lenoir
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | | | - Stéphane M Camus
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Harry Sokol
- Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology department, Paris, France
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Sorbonne Université, Paris, France.
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France.
| |
Collapse
|
45
|
Li S, Zhang J, Ni J, Cao J. Hypoxia-associated genes predicting future risk of myocardial infarction: a GEO database-based study. Front Cardiovasc Med 2023; 10:1068782. [PMID: 37465452 PMCID: PMC10351911 DOI: 10.3389/fcvm.2023.1068782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 06/01/2023] [Indexed: 07/20/2023] Open
Abstract
Background Patients with unstable angina (UA) are prone to myocardial infarction (MI) after an attack, yet the altered molecular expression profile therein remains unclear. The current work aims to identify the characteristic hypoxia-related genes associated with UA/MI and to develop a predictive model of hypoxia-related genes for the progression of UA to MI. Methods and results Gene expression profiles were obtained from the GEO database. Then, differential expression analysis and the WGCNA method were performed to select characteristic genes related to hypoxia. Subsequently, all 10 hypoxia-related genes were screened using the Lasso regression model and a classification model was established. The area under the ROC curve of 1 shows its excellent classification performance and is confirmed on the validation set. In parallel, we construct a nomogram based on these genes, showing the risk of MI in patients with UA. Patients with UA and MI had their immunological status determined using CIBERSORT. These 10 genes were primarily linked to B cells and some inflammatory cells, according to correlation analysis. Conclusion Overall, GWAS identified that the CSTF2F UA/MI risk gene promotes atherosclerosis, which provides the basis for the design of innovative cardiovascular drugs by targeting CSTF2F.
Collapse
Affiliation(s)
- Shaohua Li
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Junwen Zhang
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingwei Ni
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiumei Cao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Ebrahimian T, Dierick F, Ta V, Kotsiopriftis M, O'Connor Miranda J, Mann KK, Orthwein A, Lehoux S. B cell-specific knockout of AID protects against atherosclerosis. Sci Rep 2023; 13:8723. [PMID: 37253865 DOI: 10.1038/s41598-023-35980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/26/2023] [Indexed: 06/01/2023] Open
Abstract
Antigen-naive IgM-producing B cells are atheroprotective, whereas mature B cells producing class-switched antibodies promote atherosclerosis. Activation-induced cytidine deaminase (AID), which mediates class switch recombination (CSR), would thus be expected to foster atherosclerosis. Yet, AID also plays a major role in the establishment of B cell tolerance. We sought to define whether AID affects atherosclerotic plaque formation. We generated Ldlr-/- chimeras transplanted with bone marrow from Aicda-/- or wild-type (WT) mice, fed a HFD for 14 weeks. Decreased B cell maturation in Ldlr-/-Aicda-/- mice was demonstrated by 50% reduction in splenic and aortic BAFFR expression, a key signaling component of B2 cell maturation. This was associated with increased plasma IgM in Ldlr-/-Aicda-/- compared with Ldlr-/-WT animals. Importantly, Ldlr-/-Aicda-/- mice had reduced atherosclerotic lesion area (0.20 ± 0.03mm2) compared with Ldlr-/-WT (0.30 ± 0.04mm2, P < 0.05), although no differences in plaque composition were noted between groups. In addition, immunofluorescence analysis revealed increased splenic B and T cell areas independent of cell number. AID depletion directly inhibits atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Talin Ebrahimian
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada.
| | - France Dierick
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Vincent Ta
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Maria Kotsiopriftis
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | | | - Koren K Mann
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Alexandre Orthwein
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada
| | - Stephanie Lehoux
- Lady Davis Institute for Medical Research, 3755, Cote Ste Catherine, Montreal, QC, H3T 1E2, Canada.
| |
Collapse
|
47
|
van Os BW, Kusters PJH, den Toom M, Beckers L, van Tiel CM, Vos WG, de Jong E, Kieser A, van Roomen C, Binder CJ, Reiche ME, de Winther MP, Bosmans LA, Lutgens E. Deficiency of germinal center kinase TRAF2 and NCK-interacting kinase (TNIK) in B cells does not affect atherosclerosis. Front Cardiovasc Med 2023; 10:1171764. [PMID: 37215541 PMCID: PMC10196212 DOI: 10.3389/fcvm.2023.1171764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 05/24/2023] Open
Abstract
Background Atherosclerosis is the underlying cause of many cardiovascular diseases, such as myocardial infarction or stroke. B cells, and their production of pro- and anti-atherogenic antibodies, play an important role in atherosclerosis. In B cells, TRAF2 and NCK-interacting Kinase (TNIK), a germinal center kinase, was shown to bind to TNF-receptor associated factor 6 (TRAF6), and to be involved in JNK and NF-κB signaling in human B cells, a pathway associated with antibody production. Objective We here investigate the role of TNIK-deficient B cells in atherosclerosis. Results ApoE-/-TNIKfl/fl (TNIKBWT) and ApoE-/-TNIKfl/flCD19-cre (TNIKBKO) mice received a high cholesterol diet for 10 weeks. Atherosclerotic plaque area did not differ between TNIKBKO and TNIKBWT mice, nor was there any difference in plaque necrotic core, macrophage, T cell, α-SMA and collagen content. B1 and B2 cell numbers did not change in TNIKBKO mice, and marginal zone, follicular or germinal center B cells were unaffected. Total IgM and IgG levels, as well as oxidation specific epitope (OSE) IgM and IgG levels, did not change in absence of B cell TNIK. In contrast, plasma IgA levels were decreased in TNIKBKO mice, whereas the number of IgA+ B cells in intestinal Peyer's patches increased. No effects could be detected on T cell or myeloid cell numbers or subsets. Conclusion We here conclude that in hyperlipidemic ApoE-/- mice, B cell specific TNIK deficiency does not affect atherosclerosis.
Collapse
Affiliation(s)
- Bram W. van Os
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Pascal J. H. Kusters
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Claudia M. van Tiel
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Winnie G. Vos
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Elize de Jong
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Arnd Kieser
- Research Unit Signaling and Translation, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Cindy van Roomen
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Myrthe E. Reiche
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Menno P. de Winther
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Laura A. Bosmans
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands
- Amsterdam Immunity and Infection, Amsterdam UMC, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Ludwig-Maximilians-Universität München, Germany
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
48
|
Hemme E, Biskop D, Depuydt MAC, Smit V, Delfos L, Bernabé Kleijn MNA, Foks AC, Kuiper J, Bot I. Bruton's Tyrosine Kinase inhibition by Acalabrutinib does not affect early or advanced atherosclerotic plaque size and morphology in Ldlr-/- mice. Vascul Pharmacol 2023; 150:107172. [PMID: 37075932 DOI: 10.1016/j.vph.2023.107172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/31/2023] [Accepted: 04/16/2023] [Indexed: 04/21/2023]
Abstract
Atherosclerosis is characterized by the accumulation of lipids and immune cells, including mast cells and B cells, in the arterial wall. Mast cells contribute to atherosclerotic plaque growth and destabilization upon active degranulation. The FcεRI-IgE pathway is the most prominent mast cell activation route. Bruton's Tyrosine Kinase (BTK) is involved in FcεRI-signaling and may be a potential therapeutic target to limit mast cell activation in atherosclerosis. Additionally, BTK is crucial in B cell development and B-cell receptor signaling. In this project, we aimed to assess the effects of BTK inhibition on mast cell activation and B cell development in atherosclerosis. In human carotid artery plaques, we showed that BTK is primarily expressed on mast cells, B cells and myeloid cells. In vitro, BTK inhibitor Acalabrutinib dose-dependently inhibited IgE mediated activation of mouse bone marrow derived mast cells. In vivo, male Ldlr-/- mice were fed a high-fat diet for eight weeks, during which mice were treated with Acalabrutinib or control solvent. In Acalabrutinib treated mice, B cell maturation was reduced compared to control mice, showing a shift from follicular II towards follicular I B cells. Mast cell numbers and activation status were not affected. Acalabrutinib treatment did not affect atherosclerotic plaque size or morphology. In advanced atherosclerosis, where mice were first fed a high-fat diet for eight weeks before receiving treatment, similar effects were observed. Conclusively, BTK inhibition by Acalabrutinib alone did neither affect either mast cell activation nor early- and advanced atherosclerosis, despite the effects on follicular B cell maturation.
Collapse
Affiliation(s)
- Esmeralda Hemme
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Danique Biskop
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Marie A C Depuydt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Virginia Smit
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Lucie Delfos
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Mireia N A Bernabé Kleijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands..
| |
Collapse
|
49
|
Atzeni F, Maiani S, Corda M, Rodríguez-Carrio J. Diagnosis and management of cardiovascular risk in rheumatoid arthritis: main challenges and research agenda. Expert Rev Clin Immunol 2023; 19:279-292. [PMID: 36651086 DOI: 10.1080/1744666x.2023.2170351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) exhibit a cardiovascular (CV) risk that is 1.5-2.0 times higher compared to the general population. This CV risk excess is likely caused by the involvement of chronic inflammation and immune dysregulation. Therefore, conventional algorithms and imaging techniques fail to fully account for this risk excess and provide a suboptimal risk stratification, hence limiting clinical management in this setting. AREAS COVERED Compelling evidence has suggested a role for adaptations of conventional algorithms (Framingham, SCORE, AHA, etc) or the development of RA-specific algorithms, as well as the use of a number of several, noninvasive imaging techniques to improve CV risk assessment in RA populations. Similarly, in-depth analyses of atherosclerosis pathogenesis in RA patients have shed new light into a plethora of soluble biomarkers (such as inflammatory cytokines, vascular remodeling mediators or autoantibodies) that may provide incremental value for CV risk stratification. EXPERT OPINION Extensive research has demonstrated a lack of performance of chart adaptations in capturing real CV risk in RA population, as well as for RA-specific algorithms. Similarly, limitations have been detected in the use of soluble mediators. The development of a novel, RA-specific algorithm including classical and non-traditional risk factors may be advisable.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Silvia Maiani
- Clinical Cardiology, Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Marco Corda
- S.C. Cardiologia UTIC, ARNAS, G.Brotzu, Cagliari, Italy
| | - Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
50
|
Wang Z, Wang Y, Cui Y, Chen Z, Yi L, Zhu Z, Ni J, Du R, Wang X, Zhu J, Ding F, Quan W, Zhang R, Hu J, Yan X. Association of Serum BAFF Levels with Cardiovascular Events in ST-Segment Elevation Myocardial Infarction. J Clin Med 2023; 12:jcm12041692. [PMID: 36836225 PMCID: PMC9964977 DOI: 10.3390/jcm12041692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
OBJECTIVES The B cell activating factor (BAFF) is a B cell survival factor involved in atherosclerosis and ischemia-reperfusion (IR) injury. This study sought to investigate whether BAFF is a potential predictor of poor outcomes in patients with ST-segment elevation myocardial infarction (STEMI). METHODS We prospectively enrolled 299 patients with STEMI, and serum levels of BAFF were measured. All subjects were followed for three years. The primary endpoint was major adverse cardiovascular events (MACEs), including cardiovascular death, nonfatal reinfarction, hospitalization for heart failure (HF), and stroke. Multivariable Cox proportional hazards models were constructed to analyze the predictive value of BAFF for MACEs. RESULTS In multivariate analysis, BAFF was independently associated with risk of MACEs (adjusted HR 1.525, 95% CI 1.085-2.145; p = 0.015) and cardiovascular death (adjusted hazard ratio [HR] 3.632, 95% confidence interval [CI] 1.132-11.650, p = 0.030) after adjustment for traditional risk factors. Kaplan-Meier survival curves demonstrated that patients with BAFF levels above the cut-off value (1.46 ng/mL) were more likely to have MACEs (log-rank p < 0.0001) and cardiovascular death (log-rank p < 0.0001). In subgroup analysis, the impact of high BAFF on MACEs development was stronger in patients without dyslipidemia. Furthermore, the C-statistic and Integrated Discrimination Improvement (IDI) values for MACEs were improved with BAFF as an independent risk factor or when combined with cardiac troponin I. CONCLUSIONS This study suggests that higher BAFF levels in the acute phase are an independent predictor of the incidence of MACEs in patients with STEMI.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yueying Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuke Cui
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiyong Chen
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Yi
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jingwei Ni
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Run Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoqun Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinzhou Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (R.Z.); (J.H.); (X.Y.); Tel./Fax: +86-21-6445-7177 (R.Z. & J.H. & X.Y.)
| | - Jian Hu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (R.Z.); (J.H.); (X.Y.); Tel./Fax: +86-21-6445-7177 (R.Z. & J.H. & X.Y.)
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (R.Z.); (J.H.); (X.Y.); Tel./Fax: +86-21-6445-7177 (R.Z. & J.H. & X.Y.)
| |
Collapse
|