1
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025; 22:399-413. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [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: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Cui J, Zhang Y, Zhang W, Li D, Hong Z, Zhao L, Sun J, Chen Y, Zhang N. Research Hotspots and Development Trends on Apolipoprotein B in the Field of Atherosclerosis: A Bibliometric Analysis. Mol Biotechnol 2025; 67:2204-2222. [PMID: 38963531 DOI: 10.1007/s12033-024-01218-2] [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: 01/03/2024] [Accepted: 04/15/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Cardiovascular diseases caused by atherosclerosis (AS) are the leading causes of disability and death worldwide. Apolipoprotein B (ApoB), the core protein of low-density lipoproteins, is a major contributor to cardiovascular disease-related morbidity and mortality, with apolipoprotein B (ApoB) playing a critical role in its pathogenesis. However, no bibliometric studies on the involvement of ApoB in AS have been published. This study aimed to conduct a comprehensive bibliometric analysis to explore the current and future trends regarding the role of ApoB in AS. METHODS Utilizing the Web of Science Core Collection, a thorough search was conducted for ApoB in AS-related papers related to research on ApoB in the field of AS during 1991-2023. The analysis focused on annual publication trends, leading countries/regions and institutions, influential authors, journal and key journals. CiteSpace and VOSviewer were employed to visualize reference co-citations, and keyword co-occurrences, offering insights into the research landscape and emerging trends. RESULTS This bibliometric analysis employed network diagrams for cluster analysis of a total of 2105 articles and reviews, evidencing a discernible upward trend in annual publication volume. This corpus of research emanates from 76 countries/regions and 2343 organizations, illustrating the widespread international engagement in ApoB-related AS studies. Notably, the United States and the University of California emerge as the most prolific contributors, which underscores their pivotal roles in advancing this research domain. The thematic investigation has increasingly focused on elucidating the mechanistic involvement of ApoB in atherosclerosis, its potential as a diagnostic biomarker, and its implications for therapeutic strategies. CONCLUSION This bibliometric analysis provides the first comprehensive perspective on the evolving promise of ApoB in AS-related research, emphasizing the importance of this molecule in opening up new diagnostic and therapeutic avenues. This study emphasizes the need for continued research and interdisciplinary efforts to strengthen the fight against AS. Furthermore, it emphasizes the critical role of international collaboration and interdisciplinary exploration in leveraging new insights to achieve clinical breakthroughs, thereby addressing the complexities of AS by focusing on ApoB.
Collapse
Affiliation(s)
- Jing Cui
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- Navy Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Yan Zhang
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Wenhong Zhang
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- Navy Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Dongtao Li
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhibo Hong
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Li Zhao
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jiachen Sun
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Yu Chen
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China.
- Navy Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China.
| | - Ningkun Zhang
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
3
|
Ma C, Hua Y, Yang S, Zhao Y, Zhang W, Miao Y, Zhang J, Feng B, Zheng G, Li L, Liu Z, Zhang H, Zhu M, Gao X, Fan G. Wogonin Attenuates Atherosclerosis via KLF11-Mediated Suppression of PPARα-YAP1-Driven Glycolysis and Enhancement of ABCA1/G1-Mediated Cholesterol Efflux. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500610. [PMID: 40397286 DOI: 10.1002/advs.202500610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/04/2025] [Indexed: 05/22/2025]
Abstract
Atherosclerosis, a chronic inflammatory disorder and leading cause of cardiovascular disease, is characterized by macrophage-derived inflammation and foam cell formation. Emerging evidence suggests that metabolic reprogramming of macrophages represents a promising therapeutic approach for atherosclerosis management. In this study, the therapeutic potential of wogonin, a bioactive flavonoid isolated from Scutellaria baicalensis, in modulating macrophage metabolism and attenuating atherogenesis is investigated. Wogonin reduces lesion size and plaque vulnerability, accompanied by a reduction in foam cell formation and inflammation. Mechanistically, wogonin reprogrammes macrophage metabolism from glycolysis to fatty acid oxidation (FAO) by activating the PPARα-CPT1α pathway and acts as a mitochondrial protector by activating PPARα. Wogonin also promotes the KLF11 expression and KLF11 knockout exacerbated atherosclerosis and abolishes the inhibitory effect of wogonin on glycolysis and atherosclerosis. KLF11 forms a transcriptional complex with PPARα and YAP1, serving both as a brake on PPARα-YAP1-mediated glycolysis and a transcriptional activator of ABCA1/G1. Collectively, wogonin reprograms macrophage metabolism from glycolysis to FAO through activation of the PPARα-KLF11-YAP1 pathway, thereby reducing inflammation and foam cell formation, ultimately attenuating atherogenesis.
Collapse
Affiliation(s)
- Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300381, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300381, China
| | - Shu Yang
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen, China, Shenzhen, Guangdong, 518000, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wei Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, P. R. China
| | - Jing Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Boxuan Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Guobin Zheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Han Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Mingjun Zhu
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
| | - Xiumei Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 300193, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300381, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 300193, China
| |
Collapse
|
4
|
Zhou H, Mao Y, Ye M, Zuo Z. Exploring the nonlinear association between cardiometabolic index and hypertension in U.S. Adults: an NHANES-based study. BMC Public Health 2025; 25:1092. [PMID: 40119367 PMCID: PMC11929247 DOI: 10.1186/s12889-025-22231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/07/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Hypertension is a prevalent chronic disease affecting over 1.2 billion people worldwide, representing a major modifiable risk factor for cardiovascular diseases. The Waist-to-Height Ratio (WHtR) and Triglyceride to High-Density Lipoprotein Cholesterol (TG/HDL-C) ratio are established metabolic indicators linked to the risk of cardiovascular and metabolic diseases. Recently, a Cardiometabolic Index (CMI), combining WHtR and TG/HDL-C ratios, has been proposed to provide a comprehensive assessment of metabolic health. This study investigates the association between CMI and hypertension using data from the National Health and Nutrition Examination Survey (NHANES). METHODS The study utilized NHANES data from nine cycles spanning 2001 to 2018, encompassing 20,049 participants aged over 20. Exclusions were made for individuals with incomplete CMI or hypertension data, and pregnant women. CMI was calculated by multiplying the WHtR by the TG/HDL-C ratio. Hypertension was defined according to American Heart Association guidelines. The relationship between CMI and hypertension was evaluated using multivariate logistic regression analyses, with additional subgroup analyses conducted based on demographic factors. Nonlinear relationships were analyzed using smoothing curve fitting techniques. RESULTS The study identified a significant positive correlation between CMI and hypertension risk, with an increase of one unit in CMI associated with a 9% heightened risk of hypertension (OR: 1.09, 95% CI: 1.05, 1.13). The association remained significant across various demographic subgroups. A nonlinear relationship was observed, with a critical CMI threshold of 2.64. Below this threshold, higher CMI values were associated with a progressively higher prevalence of hypertension, whereas beyond this threshold, further increases in CMI did not significantly correlate with an elevated risk of hypertension. CONCLUSION The study demonstrates that CMI is significantly associated with hypertension risk and may serve as a valuable tool for early screening and risk assessment, particularly in identifying individuals at higher risk before reaching the critical CMI threshold. These results underscore the importance of addressing metabolic health in the prevention and management of hypertension. Future research should focus on longitudinal studies to establish causality, explore the clinical utility of CMI in hypertension screening, and examine its applicability in diverse populations.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu Mao
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Hunan Province, No. 139Renmin East Road, Changsha, 410011, People's Republic of China
| | - Muyao Ye
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhongkun Zuo
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Hunan Province, No. 139Renmin East Road, Changsha, 410011, People's Republic of China.
| |
Collapse
|
5
|
Lambaren K, Trac N, Fehrenbach D, Madhur M, Chung EJ. T Cell-Targeting Nanotherapies for Atherosclerosis. Bioconjug Chem 2025; 36:332-346. [PMID: 39979082 DOI: 10.1021/acs.bioconjchem.4c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Cardiovascular diseases remain the leading cause of mortality worldwide. Specifically, atherosclerosis is a primary cause of acute cardiac events. However, current therapies mainly focus on lipid-lowering versus addressing the underlying inflammatory response that leads to its development and progression. Nanoparticle-mediated drug delivery offers a promising approach for targeting and regulating these inflammatory responses. In atherosclerotic lesions, inflammatory cascades result in increased T helper (Th) 1 and Th17 activity and reduced T regulatory activation. The regulation of T cell responses is critical in preventing the inflammatory imbalance in atherosclerosis, making them a key therapeutic target for nanotherapy to achieve precise atherosclerosis treatment. By functionalizing nanoparticles with targeting modalities, therapeutic agents can be delivered specifically to immune cells in atherosclerotic lesions. In this Review, we outline the role of T cells in atherosclerosis, examine current nanotherapeutic strategies for targeting T cells and modulating their differentiation, and provide perspectives for the development of nanoparticles specifically tailored to target T cells for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Karla Lambaren
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Noah Trac
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Daniel Fehrenbach
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Meena Madhur
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Eun Ji Chung
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, California 90089, United States
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089, United States
- Bridge Institute, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
6
|
Nettersheim FS, Brunel S, Sinkovits RS, Armstrong SS, Roy P, Billitti M, Kobiyama K, Alimadadi A, Bombin S, Lu L, Zoccheddu M, Oliaeimotlagh M, Benedict CA, Sette A, Ley K. PD-1 and CD73 on naive CD4 + T cells synergistically limit responses to self. Nat Immunol 2025; 26:105-115. [PMID: 39572641 PMCID: PMC11697576 DOI: 10.1038/s41590-024-02021-6] [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: 03/16/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024]
Abstract
Vaccination with self- and foreign peptides induces weak and strong expansion of antigen-specific CD4+ T cells, respectively, but the mechanism is not known. In the present study, we used computational analysis of the entire mouse major histocompatibility complex class II peptidome to test how much of the naive CD4+ T cell repertoire specific for self-antigens was shaped by negative selection in the thymus and found that negative selection only partially explained the difference between responses to self and foreign. In naive uninfected and unimmunized mice, we identified higher expression of programmed cell death protein 1 (PD-1) and CD73 mRNA and protein on self-specific CD4+ T cells compared with foreign-specific CD4+ T cells. Pharmacological or genetic blockade of PD-1 and CD73 significantly increased the vaccine-induced expansion of self-specific CD4+ T cells and their transcriptomes were similar to those of foreign-specific CD4+ T cells. We concluded that PD-1 and CD73 synergistically limited CD4+ T cell responses to self. These observations have implications for the development of tolerogenic vaccines and cancer immunotherapy.
Collapse
Affiliation(s)
| | - Simon Brunel
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Robert S Sinkovits
- San Diego Supercomputer Center, University of California, La Jolla, CA, USA
| | | | - Payel Roy
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | | | - Kouji Kobiyama
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ahmad Alimadadi
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Sergei Bombin
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Lihui Lu
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | | | | | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
7
|
Jia B, Wei R, Yuan C, Cheng T, Shi S, Chu Y, Hu Y. A bibliometric analysis of vaccination against atherosclerosis. Hum Vaccin Immunother 2024; 20:2365500. [PMID: 39693182 DOI: 10.1080/21645515.2024.2365500] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 12/20/2024] Open
Abstract
A growing body of research indicates the promising potential of vaccines in both preventing and treating atherosclerosis (AS). To gain a comprehensive understanding of the current research landscape and emerging trends in this field, this study conducted a bibliometric analysis of publications on AS vaccines using the Web of Science Core Collection (WoSCC) database, based on the "bibliometric" R package and VOSviewer software. From 1991 to 2024, a total of 462 publications were identified in the WoSCC. The United States appeared as the leading contributor in terms of both total publications and citations. The Vaccine journal exhibited the highest publications output. Nilsson J from the Lund University in Sweden was the author with the most published articles, total citations and Hirsch index (H-index). Keywords analysis and thematic maps analysis revealed the passive immunotherapy (AS protective antibodies vaccines) was a hot mature theme, the active immunotherapy (AS antigens vaccines) was an emerging and booming theme, while the efficacy and safety of AS vaccines was a niche and well-developed theme. These findings offered valuable insights into the AS vaccination and provided guidance for future research in this domain.
Collapse
Affiliation(s)
- Bochao Jia
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Wei
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Chenlu Yuan
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tao Cheng
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuai Shi
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuguang Chu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Tsioulos G, Vallianou NG, Skourtis A, Dalamaga M, Kotsi E, Kargioti S, Adamidis N, Karampela I, Mourouzis I, Kounatidis D. Vaccination as a Promising Approach in Cardiovascular Risk Mitigation: Are We Ready to Embrace a Vaccine Strategy? Biomolecules 2024; 14:1637. [PMID: 39766344 PMCID: PMC11727084 DOI: 10.3390/biom14121637] [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: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and adaptive immune systems play vital roles in its progression, either exacerbating or alleviating disease development. This intricate interplay positions the immune system as a compelling therapeutic target. Consequently, immunomodulatory strategies have gained increasing attention, though none have yet reached widespread clinical adoption. Safety concerns, particularly the suppression of host immune defenses, remain a significant barrier to the clinical application of anti-inflammatory therapies. Recent decades have revealed the significant role of adaptive immune responses to plaque-associated autoantigens in atherogenesis, opening new perspectives for targeted immunological interventions. Preclinical models indicate that vaccines targeting specific atherosclerosis-related autoantigens can slow disease progression while preserving systemic immune function. In this context, numerous experimental studies have advanced the understanding of vaccine development by exploring diverse targeting pathways. Key strategies include passive immunization using naturally occurring immunoglobulin G (IgG) antibodies and active immunization targeting low-density lipoprotein cholesterol (LDL-C) and apolipoproteins, such as apolipoprotein B100 (ApoB100) and apolipoprotein CIII (ApoCIII). Other approaches involve vaccine formulations aimed at proteins that regulate lipoprotein metabolism, including proprotein convertase subtilisin/kexin type 9 (PCSK9), cholesteryl ester transfer protein (CETP), and angiopoietin-like protein 3 (ANGPTL3). Furthermore, the literature highlights the potential for developing non-lipid-related vaccines, with key targets including heat shock proteins (HSPs), interleukins (ILs), angiotensin III (Ang III), and a disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7). However, translating these promising findings into safe and effective clinical therapies presents substantial challenges. This review provides a critical evaluation of current anti-atherosclerotic vaccination strategies, examines their proposed mechanisms of action, and discusses key challenges that need to be overcome to enable clinical translation.
Collapse
Affiliation(s)
- Georgios Tsioulos
- Fourth Department of Internal Medicine, Medical School, Attikon General University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Alexandros Skourtis
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelia Kotsi
- Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio General Hospital, 11527 Athens, Greece;
| | - Sofia Kargioti
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Nikolaos Adamidis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Irene Karampela
- Second Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, 12461 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece;
| |
Collapse
|
9
|
Vasilieva MI, Shatalova RO, Matveeva KS, Shindyapin VV, Minskaia E, Ivanov RA, Shevyrev DV. Senolytic Vaccines from the Central and Peripheral Tolerance Perspective. Vaccines (Basel) 2024; 12:1389. [PMID: 39772050 PMCID: PMC11680330 DOI: 10.3390/vaccines12121389] [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: 11/01/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Preventive medicine has proven its long-term effectiveness and economic feasibility. Over the last century, vaccination has saved more lives than any other medical technology. At present, preventative measures against most infectious diseases are successfully used worldwide; in addition, vaccination platforms against oncological and even autoimmune diseases are being actively developed. At the same time, the development of medicine led to an increase in both life expectancy and the proportion of age-associated diseases, which pose a heavy socio-economic burden. In this context, the development of vaccine-based approaches for the prevention or treatment of age-related diseases opens up broad prospects for extending the period of active longevity and has high economic potential. It is well known that the development of age-related diseases is associated with the accumulation of senescent cells in various organs and tissues. It has been demonstrated that the elimination of such cells leads to the restoration of functions, rejuvenation, and extension of the lives of experimental animals. However, the development of vaccines against senescent cells is complicated by their antigenic heterogeneity and the lack of a unique marker. In addition, senescent cells are the body's own cells, which may be the reason for their low immunogenicity. This mini-review discusses the mechanisms of central and peripheral tolerance that may influence the formation of an anti-senescent immune response and be responsible for the accumulation of senescent cells with age.
Collapse
Affiliation(s)
- Mariia I. Vasilieva
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Rimma O. Shatalova
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Kseniia S. Matveeva
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia;
| | - Vadim V. Shindyapin
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia;
| | - Ekaterina Minskaia
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Roman A. Ivanov
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Daniil V. Shevyrev
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| |
Collapse
|
10
|
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
|
11
|
Horstmann H, Michel NA, Sheng X, Hansen S, Lindau A, Pfeil K, Fernández MC, Marchini T, Winkels H, Mitre LS, Abogunloko T, Li X, Mwinyella TBN, Gissler MC, Bugger H, Heidt T, Buscher K, Hilgendorf I, Stachon P, Piepenburg S, Verheyen N, Rathner T, Gerhardt T, Siegel PM, Oswald WK, Cohnert T, Zernecke A, Madl J, Kohl P, Foks AC, von zur Muehlen C, Westermann D, Zirlik A, Wolf D. Cross-species single-cell RNA sequencing reveals divergent phenotypes and activation states of adaptive immunity in human carotid and experimental murine atherosclerosis. Cardiovasc Res 2024; 120:1713-1726. [PMID: 39041203 PMCID: PMC11587564 DOI: 10.1093/cvr/cvae154] [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: 11/16/2023] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024] Open
Abstract
AIMS The distinct functions of immune cells in atherosclerosis have been mostly defined by pre-clinical mouse studies. Contrastingly, the immune cell composition of human atherosclerotic plaques and their contribution to disease progression are only poorly understood. It remains uncertain whether genetic animal models allow for valuable translational approaches. METHODS AND RESULTS Single-cell RNA-sequencing (scRNA-seq) was performed to define the immune cell landscape in human carotid atherosclerotic plaques. The human immune cell repertoire demonstrated an unexpectedly high heterogeneity and was dominated by cells of the T-cell lineage, a finding confirmed by immunohistochemistry. Bioinformatical integration with 7 mouse scRNA-seq data sets from adventitial and atherosclerotic vascular tissue revealed a total of 51 identities of cell types and differentiation states, of which some were only poorly conserved between species and exclusively found in humans. Locations, frequencies, and transcriptional programmes of immune cells in mouse models did not resemble the immune cell landscape in human carotid atherosclerosis. In contrast to standard mouse models of atherosclerosis, human plaque leucocytes were dominated by several T-cell phenotypes with transcriptional hallmarks of T-cell activation and memory formation, T-cell receptor, and pro-inflammatory signalling. Only mice at the age of 22 months partially resembled the activated T-cell phenotype. In a validation cohort of 43 patients undergoing carotid endarterectomy, the abundance of activated immune cell subsets in the plaque defined by multi-colour flow cytometry associated with the extent of clinical atherosclerosis. CONCLUSION Integrative scRNA-seq reveals a substantial difference in the immune cell composition of murine and human carotid atherosclerosis-a finding that questions the translational value of standard mouse models for adaptive immune cell studies. Clinical associations suggest a specific role for T-cell driven (auto-)immunity in human plaque formation and instability.
Collapse
Affiliation(s)
- Hauke Horstmann
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Division of Cardiology, Department of Medicine, NYU Cardiovascular Research Center, NYU Grossmann School of Medicine, 10016 New York, NY, USA
| | - Nathaly Anto Michel
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Xia Sheng
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sophie Hansen
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexandra Lindau
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Katharina Pfeil
- Division of Cardiology, Department of Medicine, NYU Cardiovascular Research Center, NYU Grossmann School of Medicine, 10016 New York, NY, USA
| | - Marbely C Fernández
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Timoteo Marchini
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50923 Cologne, Germany
| | - Lucia Sol Mitre
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79106 Freiburg, Germany
| | - Tijani Abogunloko
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79106 Freiburg, Germany
| | - Xiaowei Li
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Timothy Bon-Nawul Mwinyella
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Mark Colin Gissler
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Heiko Bugger
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Timo Heidt
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Konrad Buscher
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine, University Hospital of Münster, 48149 Münster, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sven Piepenburg
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nicolas Verheyen
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Thomas Rathner
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Teresa Gerhardt
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, and Berlin Institute of Health (BIH), 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin , 13353 Berlin, Germany
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
| | - Patrick Malcolm Siegel
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | | | - Tina Cohnert
- Department of Vascular Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Josef Madl
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Peter Kohl
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Amanda C Foks
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, 2333 CC Leiden, The Netherlands
| | - Constantin von zur Muehlen
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Andreas Zirlik
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Dennis Wolf
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
12
|
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
|
13
|
Döring Y, van der Vorst EPC, Weber C. Targeting immune cell recruitment in atherosclerosis. Nat Rev Cardiol 2024; 21:824-840. [PMID: 38664575 DOI: 10.1038/s41569-024-01023-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 10/17/2024]
Abstract
Atherosclerosis is the primary underlying cause of myocardial infarction and stroke. Atherosclerotic cardiovascular disease is characterized by a chronic inflammatory reaction in medium-to-large-sized arteries, with its onset and perpetuation driven by leukocytes infiltrating the subendothelial space. Activation of endothelial cells triggered by hyperlipidaemia and lipoprotein retention in the arterial intima initiates the accumulation of pro-inflammatory leukocytes in the arterial wall, fostering the progression of atherosclerosis. This inflammatory response is coordinated by an array of soluble mediators, namely cytokines and chemokines, that amplify inflammation both locally and systemically and are complemented by tissue-specific molecules that regulate the homing, adhesion and transmigration of leukocytes. Despite abundant evidence from mouse models, only a few therapies targeting leukocytes in atherosclerosis have been assessed in humans. The major challenges for the clinical translation of these therapies include the lack of tissue specificity and insufficient selectivity of inhibition strategies. In this Review, we discuss the latest research on receptor-ligand pairs and interactors that regulate leukocyte influx into the inflamed artery wall, primarily focusing on studies that used pharmacological interventions. We also discuss mechanisms that promote the resolution of inflammation and highlight how major findings from these research areas hold promise as potential therapeutic strategies for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Yvonne Döring
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Aachen, Germany.
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany.
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
14
|
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
|
15
|
Yang J, Chen Y, Li X, Qin H, Bao J, Wang C, Dong X, Xu D. Complex Interplay Between Metabolism and CD4 + T-Cell Activation, Differentiation, and Function: a Novel Perspective for Atherosclerosis Immunotherapy. Cardiovasc Drugs Ther 2024; 38:1033-1046. [PMID: 37199882 DOI: 10.1007/s10557-023-07466-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2023] [Indexed: 05/19/2023]
Abstract
Atherosclerosis is a complex pathological process that results from the chronic inflammatory reaction of the blood vessel wall and involves various immune cells and cytokines. An imbalance in the proportion and function of the effector CD4+ T-cell (Teff) and regulatory T-cell (Treg) subsets is an important cause of the occurrence and development of atherosclerotic plaques. Teff cells depend on glycolytic metabolism and glutamine catabolic metabolism for energy, while Treg cells mainly rely on fatty acid oxidation (FAO), which is crucial for determining the fate of CD4+ T cells during differentiation and maintaining their respective immune functions. Here, we review recent research achievements in the field of immunometabolism related to CD4+ T cells, focusing on the cellular metabolic pathways and metabolic reprogramming involved in the activation, proliferation, and differentiation of CD4+ T cells. Subsequently, we discuss the important roles of mTOR and AMPK signaling in regulating CD4+ T-cell differentiation. Finally, we evaluated the links between CD4+ T-cell metabolism and atherosclerosis, highlighting the potential of targeted modulation of CD4+ T-cell metabolism in the prevention and treatment of atherosclerosis in the future.
Collapse
Affiliation(s)
- Jingmin Yang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Yanying Chen
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Xiao Li
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Huali Qin
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Jinghui Bao
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Chunfang Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Xiaochen Dong
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Danyan Xu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China.
| |
Collapse
|
16
|
Tang D, Liu Y, Duan R, Lin R, Li Z, Liu X, Huang J, Zhao M. COL6A6 Peptide Vaccine Alleviates Atherosclerosis through Inducing Immune Response and Regulating Lipid Metabolism in Apoe-/- Mice. Cells 2024; 13:1589. [PMID: 39329770 PMCID: PMC11429512 DOI: 10.3390/cells13181589] [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: 07/11/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Atherosclerosis is an autoimmune disease characterized by lipid imbalances and chronic inflammation within blood vessels, with limited preventive and treatment options currently available. In this study, a vaccine prepared with COL6A6 peptide (named the Pep_A6 vaccine) was administered to immunize Apoe-/- mice, and the immune mechanism of the Pep_A6 vaccine against atherosclerosis was first investigated. The results of arterial oil red O staining demonstrated that the Pep_A6 vaccine significantly reduced the atherosclerotic plaque area in Apoe-/- mice fed with a high-fat diet for 20 weeks. A flow cytometry analysis revealed that the Pep_A6 vaccine inhibited Th1 cell differentiation and increased the proportion of Treg cells. Furthermore, there was a significant increase in Ly6Clow monocytes observed in the vaccinated group. The ELISA results showed that the Pep_A6 vaccine induced a significant expression of Pep_A6-specific antibody IgG and IgG1 in mouse serum. Additionally, we found that the Pep_A6 vaccine significantly decreased serum LDL-C content and regulated the expression of genes related to liver lipid metabolism. Together, our findings suggest that the Pep_A6 vaccine alleviates atherosclerosis by inducing a positive immune response and regulating lipid metabolism, providing new insights into potential prevention strategies for atherosclerosis as an innovative vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ming Zhao
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (D.T.)
| |
Collapse
|
17
|
Khan A, Roy P, Ley K. Breaking tolerance: the autoimmune aspect of atherosclerosis. Nat Rev Immunol 2024; 24:670-679. [PMID: 38472321 PMCID: PMC11682649 DOI: 10.1038/s41577-024-01010-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is a chronic inflammatory disease of the arterial walls and is characterized by the accumulation of lipoproteins that are insufficiently cleared by phagocytes. Following the initiation of atherosclerosis, the pathological progression is accelerated by engagement of the adaptive immune system. Atherosclerosis triggers the breakdown of tolerance to self-components. This loss of tolerance is reflected in defective expression of immune checkpoint molecules, dysfunctional antigen presentation, and aberrations in T cell populations - most notably in regulatory T (Treg) cells - and in the production of autoantibodies. The breakdown of tolerance to self-proteins that is observed in ASCVD may be linked to the conversion of Treg cells to 'exTreg' cells because many Treg cells in ASCVD express T cell receptors that are specific for self-epitopes. Alternatively, or in addition, breakdown of tolerance may trigger the activation of naive T cells, resulting in the clonal expansion of T cell populations with pro-inflammatory and cytotoxic effector phenotypes. In this Perspective, we review the evidence that atherosclerosis is associated with a breakdown of tolerance to self-antigens, discuss possible immunological mechanisms and identify knowledge gaps to map out future research. Rational approaches aimed at re-establishing immune tolerance may become game changers in treating ASCVD and in preventing its downstream sequelae, which include heart attacks and strokes.
Collapse
Affiliation(s)
- Amir Khan
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Payel Roy
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Klaus Ley
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
18
|
Gerdes N, Klingenberg R. Mini-Review: Immunogenic epitopes in apolipoprotein B-100 for atheroprotective immunization. Front Cardiovasc Med 2024; 11:1448664. [PMID: 39211769 PMCID: PMC11357920 DOI: 10.3389/fcvm.2024.1448664] [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: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Here, we provide a concise overview of recent developments in the identification of immunogenic epitopes in human apolipoprotein B-100 for immunization against atherosclerotic cardiovascular disease. Major steps forward toward a clinical application of vaccines include the design of humanized mouse models, tetramer-based identification of antigen-specific T cells, and novel analysis tools, such as single-cell RNA sequencing and cytometry by time of flight, to assess temporal and spatial changes in immune cells in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Roland Klingenberg
- Cardiology, Kerckhoff-Klinik, Bad Nauheim, Campus of the Justus-Liebig-University Giessen, Giessen, Germany
- Department of Cardiology, German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| |
Collapse
|
19
|
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
|
20
|
Kologrivova IV, Naryzhnaya NV, Suslova TE. Thymus in Cardiometabolic Impairments and Atherosclerosis: Not a Silent Player? Biomedicines 2024; 12:1408. [PMID: 39061983 PMCID: PMC11273826 DOI: 10.3390/biomedicines12071408] [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: 05/13/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
The thymus represents a primary organ of the immune system, harboring the generation and maturation of T lymphocytes. Starting from childhood, the thymus undergoes involution, being replaced with adipose tissue, and by an advanced age nearly all the thymus parenchyma is represented by adipocytes. This decline of thymic function is associated with compromised maturation and selection of T lymphocytes, which may directly impact the development of inflammation and induce various autoinflammatory disorders, including atherosclerosis. For a long time, thymus health in adults has been ignored. The process of adipogenesis in thymus and impact of thymic fat on cardiometabolism remains a mysterious process, with many issues being still unresolved. Meanwhile, thymus functional activity has a potential to be regulated, since islets of thymopoeisis remain in adults even at an advanced age. The present review describes the intricate process of thymic adipose involution, focusing on the issues of the thymus' role in the development of atherosclerosis and metabolic health, tightly interconnected with the state of vessels. We also review the recent information on the key molecular pathways and biologically active substances that may be targeted to manipulate both thymic function and atherosclerosis.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (N.V.N.); (T.E.S.)
| | | | | |
Collapse
|
21
|
Xu L, Chen F, Fan W, Saito S, Cao D. The role of γδT lymphocytes in atherosclerosis. Front Immunol 2024; 15:1369202. [PMID: 38774876 PMCID: PMC11106432 DOI: 10.3389/fimmu.2024.1369202] [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: 01/15/2024] [Accepted: 04/18/2024] [Indexed: 05/24/2024] Open
Abstract
Atherosclerosis poses a significant threat to human health, impacting overall well-being and imposing substantial financial burdens. Current treatment strategies mainly focus on managing low-density lipids (LDL) and optimizing liver functions. However, it's crucial to recognize that Atherosclerosis involves more than just lipid accumulation; it entails a complex interplay of immune responses. Research highlights the pivotal role of lipid-laden macrophages in the formation of atherosclerotic plaques. These macrophages attract lymphocytes like CD4 and CD8 to the inflamed site, potentially intensifying the inflammatory response. γδ T lymphocytes, with their diverse functions in innate and adaptive immune responses, pathogen defense, antigen presentation, and inflammation regulation, have been implicated in the early stages of Atherosclerosis. However, our understanding of the roles of γδ T cells in Atherosclerosis remains limited. This mini-review aims to shed light on the characteristics and functions of γδ T cells in Atherosclerosis. By gaining insights into the roles of γδ T cells, we may uncover a promising strategy to mitigate plaque buildup and dampen the inflammatory response, thereby opening new avenues for effectively managing this condition.
Collapse
Affiliation(s)
- LiMin Xu
- Department of Neurosurgery, Shenzhen Entry-Exit Frontier Inspection Hospital, Shenzhen, China
| | - Fanfan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - DuoYao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
22
|
Wu R, Sun F, Zhang W, Ren J, Liu GH. Targeting aging and age-related diseases with vaccines. NATURE AGING 2024; 4:464-482. [PMID: 38622408 DOI: 10.1038/s43587-024-00597-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/20/2024] [Indexed: 04/17/2024]
Abstract
Aging is a major risk factor for numerous chronic diseases. Vaccination offers a promising strategy to combat these age-related diseases by targeting specific antigens and inducing immune responses. Here, we provide a comprehensive overview of recent advances in vaccine-based interventions targeting these diseases, including Alzheimer's disease, type II diabetes, hypertension, abdominal aortic aneurysm, atherosclerosis, osteoarthritis, fibrosis and cancer, summarizing current approaches for identifying disease-associated antigens and inducing immune responses against these targets. Further, we reflect on the recent development of vaccines targeting senescent cells, as a strategy for more broadly targeting underlying causes of aging and associated pathologies. In addition to highlighting recent progress in these areas, we discuss important next steps to advance the therapeutic potential of these vaccines, including improving and robustly demonstrating efficacy in human clinical trials, as well as rigorously evaluating the safety and long-term effects of these vaccine strategies.
Collapse
Affiliation(s)
- Ruochen Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Sun
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China.
- Sino-Danish College, School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jie Ren
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China.
- Sino-Danish College, School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
23
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
24
|
Tsiantoulas D, Binder CJ. Identifying the sensor elements of regulatory T cells in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:106-107. [PMID: 39196191 DOI: 10.1038/s44161-023-00416-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
25
|
Bensussen A, Torres-Magallanes JA, Álvarez-Buylla ER, de Álvarez-Buylla ER. Hybrid lineages of CD4 + T cells: a handbook update. Front Immunol 2024; 15:1344078. [PMID: 38312841 PMCID: PMC10834732 DOI: 10.3389/fimmu.2024.1344078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
CD4+ T lymphocytes have been classified into several lineages, according to their gene expression profiles and their effector responses. Interestingly, recent evidence is showing that many lineages could yield hybrid phenotypes with unique properties and functions. It has been reported that such hybrid lineages might underlie pathologies or may function as effector cells with protection capacities against molecular threats. In this work, we reviewed the characteristics of the hybrid lineages reported in the literature, in order to identify the expression profiles that characterize them and the markers that could be used to identify them. We also review the differentiation cues that elicit their hybrid origin and what is known about their physiological roles.
Collapse
Affiliation(s)
- Antonio Bensussen
- Laboratorio de Neuroendocrinología, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | - José Antonio Torres-Magallanes
- Laboratorio de Neuroendocrinología, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | - Elena R. Álvarez-Buylla
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Centro de Ciencias de la Complejidad (C3), Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Elena Roces de Álvarez-Buylla
- Laboratorio de Neuroendocrinología, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| |
Collapse
|
26
|
Chan A, Torelli S, Cheng E, Batchelder R, Waliany S, Neal J, Witteles R, Nguyen P, Cheng P, Zhu H. Immunotherapy-Associated Atherosclerosis: A Comprehensive Review of Recent Findings and Implications for Future Research. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:715-735. [PMID: 38213548 PMCID: PMC10776491 DOI: 10.1007/s11936-023-01024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 01/13/2024]
Abstract
Purpose of the Review Even as immune checkpoint inhibitors (ICIs) have transformed the lifespan of many patients, they may also trigger acceleration of long-term cardiovascular disease. Our review aims to examine the current landscape of research on ICI-mediated atherosclerosis and address key questions regarding its pathogenesis and impact on patient management. Recent Findings Preclinical mouse models suggest that T cell dysregulation and proatherogenic cytokine production are key contributors to plaque development after checkpoint inhibition. Clinical data also highlight the significant burden of atherosclerotic cardiovascular disease (ASCVD) in patients on immunotherapy, although the value of proactively preventing and treating ASCVD in this population remains an open area of inquiry. Current treatment options include dietary/lifestyle modification and traditional medications to manage hypertension, hyperlipidemia, and diabetes risk factors; no current targeted therapies exist. Summary Early identification of high-risk patients is crucial for effective preventive strategies and timely intervention. Future research should focus on refining screening tools, elucidating targetable mechanisms driving ICI atherosclerosis, and evaluating long-term cardiovascular outcomes in cancer survivors who received immunotherapy. Moreover, close collaboration between oncologists and cardiologists is essential to optimize patient outcomes.
Collapse
Affiliation(s)
- Antonia Chan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Evaline Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ryan Batchelder
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Paul Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| |
Collapse
|
27
|
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
|
28
|
Zhang Z, Guo J, Jia R. Treg plasticity and human diseases. Inflamm Res 2023; 72:2181-2197. [PMID: 37878023 DOI: 10.1007/s00011-023-01808-x] [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: 08/07/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
INTRODUCTION As a subset of CD4+ T cells, regulatory T cells (Tregs) with the characteristic expression of transcription factor FOXP3 play a key role in maintaining self-tolerance and regulating immune responses. However, in some inflammatory circumstances, Tregs can express cytokines of other T help (Th) cells by internal reprogramming, which is called Treg plasticity. These reprogrammed Tregs with impaired suppressive ability contribute to the progression of diseases by secreting pro-inflammatory cytokines. However, in the tumor microenvironment (TME), such changes in phenotype rarely occur in Tregs, on the contrary, Tregs usually display a stronger suppressive function and inhibit anti-tumor immunity. It is important to understand the mechanisms of Treg plasticity in inflammatory diseases and cancers. OBJECTIVES In this review, we summarize the characteristics of different Th-like Tregs and discuss the potential mechanisms of these changes in phenotype. Furthermore, we summarize the Treg plasticity in human diseases and discuss the effects of these changes in phenotype on disease progression, as well as the potential application of drugs or reagents that regulate Treg plasticity in human diseases. CONCLUSIONS Treg plasticity is associated with inflammatory diseases and cancers. Regulating Treg plasticity is a promising direction for the treatment of inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Zheng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Jihua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
- Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Rong Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
29
|
Appleton BD, Palmer SA, Smith HP, Stephens LE, Major AS. Oxidized Phospholipid oxPAPC Alters Regulatory T-Cell Differentiation and Decreases Their Protective Function in Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2023; 43:2119-2132. [PMID: 37675632 PMCID: PMC10720352 DOI: 10.1161/atvbaha.123.319674] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) are protective in atherosclerosis but reduced during disease progression due to cell death and loss of stability. However, the mechanisms of Treg dysfunction remain unknown. Oxidized phospholipids are abundant in atherosclerosis and can activate innate immune cells, but little is known regarding their impact on T cells. Given Treg loss during atherosclerosis progression and oxidized phospholipid levels in the plaque microenvironment, we investigated whether oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (oxPAPC), an oxidized phospholipid associated with atherosclerotic plaques, alters Treg differentiation and function. METHODS CD4+ T cells were polarized to Treg, T helper (Th) 1, and Th17 cells with or without oxPAPC and assessed by flow cytometry. Gene expression in oxPAPC-treated Tregs was analyzed by bulk RNA sequencing. Functional studies of oxPAPC-induced Tregs were performed by coculturing Tregs with CellTrace Violet-labeled cells in vitro, and by adoptively transferring Tregs to hyperlipidemic Ldlr-/- mice to measure atherosclerosis progression. RESULTS Compared with controls, oxPAPC-treated Tregs were less viable, but surviving cells expressed higher levels of the Th1-associated markers T-bet, CXCR3, and IFN (interferon)-γ. Th1 and Th17 skewing cultures were unaltered by oxPAPC. IFN-γ is linked to Treg instability, thus Treg polarization experiments were repeated using Ifngr1-/- CD4+ T cells. IFNγR1 (INF gamma receptor 1) deficiency did not improve cell viability in oxPAPC-treated Tregs; however, T-bet and IFN-γ expression was not increased in surviving cells suggesting a role for IFN-γsignaling. OxPAPC-treated Tregs were less suppressive in vitro, and adoptive transfer studies in hyperlipidemic Ldlr-/- mice showed that oxPAPC-induced Tregs possessed altered tissue homing and were insufficient to inhibit atherosclerosis progression. CONCLUSIONS OxPAPC elicits Treg-specific changes altering Treg differentiation and inducing a Th1-like phenotype in surviving cells partially through IFN-γ signaling. This is biologically relevant as oxPAPC-treated Tregs do not reduce atherosclerosis progression in Ldlr-/- mice. This study supports the role of oxidized phospholipids in negatively impacting Treg differentiation and atheroprotective function.
Collapse
Affiliation(s)
- Brenna D. Appleton
- Department of Pathology, Microbiology and Immunology, Vanderbilt University
| | | | | | | | - Amy S. Major
- Department of Pathology, Microbiology and Immunology, Vanderbilt University
- Department of Medicine, Vanderbilt University Medical Center
- Tennessee Valley Health System, Department of Veterans Affairs
| |
Collapse
|
30
|
Della Corte V, Todaro F, Cataldi M, Tuttolomondo A. Atherosclerosis and Its Related Laboratory Biomarkers. Int J Mol Sci 2023; 24:15546. [PMID: 37958528 PMCID: PMC10649778 DOI: 10.3390/ijms242115546] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023] Open
Abstract
Atherosclerosis constitutes a persistent inflammatory ailment, serving as the predominant underlying condition for coronary artery disease (CAD), peripheral artery disease (PAD), and cerebrovascular disease. The progressive buildup of plaques within the walls of medium- and large-caliber arteries characterizes the atherosclerotic process. This accumulation results in significant narrowing that impedes blood flow, leading to critical tissue oxygen deficiency. Spontaneous blockage of thrombotic vessels can precipitate stroke and myocardial infarction, which are complications representing the primary global causes of mortality. Present-day models for predicting cardiovascular risk incorporate conventional risk factors to gauge the likelihood of cardiovascular events over a ten-year span. In recent times, researchers have identified serum biomarkers associated with an elevated risk of atherosclerotic events. Many of these biomarkers, whether used individually or in combination, have been integrated into risk prediction models to assess whether their inclusion enhances predictive accuracy. In this review, we have conducted a comprehensive analysis of the most recently published literature concerning serum biomarkers associated with atherosclerosis. We have explored the potential utility of incorporating these markers in guiding clinical decisions.
Collapse
|
31
|
Jiang Y, Pan Y, Long T, Qi J, Liu J, Zhang M. Significance of RNA N6-methyladenosine regulators in the diagnosis and subtype classification of coronary heart disease using the Gene Expression Omnibus database. Front Cardiovasc Med 2023; 10:1185873. [PMID: 37928762 PMCID: PMC10621741 DOI: 10.3389/fcvm.2023.1185873] [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: 04/04/2023] [Accepted: 08/21/2023] [Indexed: 11/07/2023] Open
Abstract
Background Many investigations have revealed that alterations in m6A modification levels may be linked to coronary heart disease (CHD). However, the specific link between m6A alteration and CHD warrants further investigation. Methods Gene expression profiles from the Gene Expression Omnibus (GEO) databases. We began by constructing a Random Forest model followed by a Nomogram model, both aimed at enhancing our predictive capabilities on specific m6A markers. We then shifted our focus to identify distinct molecular subtypes based on the key m6A regulators and to discern differentially expressed genes between the unique m6A clusters. Following this molecular exploration, we embarked on an in-depth analysis of the biological characteristics associated with each m6A cluster, revealing profound differences between them. Finally, we delved into the identification and correlation analysis of immune cell infiltration across these clusters, emphasizing the potential interplay between m6A modification and the immune system. Results In this research, 37 important m6Aregulators were identified by comparing non-CHD and CHD patients from the GSE20680, GSE20681, and GSE71226 datasets. To predict the risk of CHD, seven candidate m6A regulators (CBLL1, HNRNPC, YTHDC2, YTHDF1, YTHDF2, YTHDF3, ZC3H13) were screened using the logistic regression model. Based on the seven possible m6A regulators, a nomogram model was constructed. An examination of decision curves revealed that CHD patients could benefit from the nomogram model. On the basis of the selected relevant m6A regulators, patients with CHD were separated into two m6A clusters (cluster1 and cluster2) using the consensus clustering approach. The Single Sample Gene Set Enrichment Analysis (ssGSEA) and CIBERSORT methods were used to estimate the immunological characteristics of two separate m6A Gene Clusters; the results indicated a close association between seven candidate genes and immune cell composition. The drug sensitivity of seven candidate regulators was predicted, and these seven regulators appeared in numerous diseases as pharmacological targets while displaying strong drug sensitivity. Conclusion m6A regulators play crucial roles in the development of CHD. Our research of m6A clusters may facilitate the development of novel molecular therapies and inform future immunotherapeutic methods for CHD.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Cardiovascular Surgery, Yan'an Hospital affiliated to Kunming Medical University, Yunnan, China
| | - Yaqiang Pan
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Tao Long
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Junqing Qi
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jianchao Liu
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Mengya Zhang
- Department of Cardiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| |
Collapse
|
32
|
De Groot AS, Roberts BJ, Mattei A, Lelias S, Boyle C, Martin WD. Immunogenicity risk assessment of synthetic peptide drugs and their impurities. Drug Discov Today 2023; 28:103714. [PMID: 37467878 DOI: 10.1016/j.drudis.2023.103714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Peptide drugs play an important part in medicine owing to their many therapeutic applications. Of the 80 peptide drugs approved for use in humans, at least five are now off-patent and are consequently being developed as generic alternatives to the originator products. To accelerate access to generic products, the FDA has proposed new regulatory pathways that do not require direct comparisons of generics to originators in clinical trials. The 'Abbreviated New Drug Application' (ANDA) pathway recommends that sponsors provide information on any new impurities in the generic drug, compared with the originator product, because the impurities can have potential to elicit unwanted immune responses owing to the introduction of T-cell epitopes. This review describes how peptide drug impurities can elicit unexpected immunogenicity and describes a framework for performing immunogenicity risk assessment of all types of bioactive peptide products. Although this report primarily focuses on generic peptides and their impurities, the approach might also be of interest for developers of novel peptide drugs who are preparing their products for an initial regulatory review.
Collapse
Affiliation(s)
- Anne S De Groot
- EpiVax, 188 Valley Street, Suite 424, Providence, RI, USA; University of Georgia, Center for Vaccines and Immunology, Athens, GA USA.
| | | | - Aimee Mattei
- EpiVax, 188 Valley Street, Suite 424, Providence, RI, USA
| | - Sandra Lelias
- EpiVax, 188 Valley Street, Suite 424, Providence, RI, USA
| | | | | |
Collapse
|
33
|
Freuchet A, Roy P, Armstrong SS, Oliaeimotlagh M, Kumar S, Orecchioni M, Ali AJ, Khan A, Makings J, Lyu Q, Winkels H, Wang E, Durant C, Ghosheh Y, Gulati R, Nettersheim F, Ley K. Identification of human exT reg cells as CD16 +CD56 + cytotoxic CD4 + T cells. Nat Immunol 2023; 24:1748-1761. [PMID: 37563308 PMCID: PMC11022744 DOI: 10.1038/s41590-023-01589-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/13/2023] [Indexed: 08/12/2023]
Abstract
In atherosclerosis, some regulatory T (Treg) cells become exTreg cells. We crossed inducible Treg and exTreg cell lineage-tracker mice (FoxP3eGFP-Cre-ERT2ROSA26CAG-fl-stop-fl-tdTomato) to atherosclerosis-prone Apoe-/- mice, sorted Treg cells and exTreg cells and determined their transcriptomes by bulk RNA sequencing (RNA-seq). Genes that were differentially expressed between mouse Treg cells and exTreg cells and filtered for their presence in a human single-cell RNA-sequencing (scRNA-seq) panel identified exTreg cell signature genes as CST7, NKG7, GZMA, PRF1, TBX21 and CCL4. Projecting these genes onto the human scRNA-seq with CITE-seq data identified human exTreg cells as CD3+CD4+CD16+CD56+, which was validated by flow cytometry. Bulk RNA-seq of sorted human exTreg cells identified them as inflammatory and cytotoxic CD4+T cells that were significantly distinct from both natural killer and Treg cells. DNA sequencing for T cell receptor-β showed clonal expansion of Treg cell CDR3 sequences in exTreg cells. Cytotoxicity was functionally demonstrated in cell killing and CD107a degranulation assays, which identifies human exTreg cells as cytotoxic CD4+T cells.
Collapse
Affiliation(s)
| | - Payel Roy
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Sunil Kumar
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Marco Orecchioni
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Amal J Ali
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Amir Khan
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | | | - Qingkang Lyu
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Holger Winkels
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Erpei Wang
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Yanal Ghosheh
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rishab Gulati
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
34
|
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
|
35
|
Hinkley H, Counts DA, VonCanon E, Lacy M. T Cells in Atherosclerosis: Key Players in the Pathogenesis of Vascular Disease. Cells 2023; 12:2152. [PMID: 37681883 PMCID: PMC10486666 DOI: 10.3390/cells12172152] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques within arterial walls. T cells play a pivotal role in the pathogenesis of atherosclerosis in which they help orchestrate immune responses and contribute to plaque development and instability. Here, we discuss the recognition of atherosclerosis-related antigens that may trigger T cell activation together with additional signaling from co-stimulatory molecules and lesional cytokines. Although few studies have indicated candidates for the antigen specificity of T cells in atherosclerosis, further research is needed. Furthermore, we describe the pro-atherogenic and atheroprotective roles of diverse subsets of T cells such as CD4+ helper, CD8+ cytotoxic, invariant natural killer, and γδ T cells. To classify and quantify T cell subsets in atherosclerosis, we summarize current methods to analyze cellular heterogeneity including single cell RNA sequencing and T cell receptor (TCR) sequencing. Further insights into T cell biology will help shed light on the immunopathology of atherosclerosis, inform potential therapeutic interventions, and pave the way for precision medicine approaches in combating cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Michael Lacy
- Department of Medical Laboratory Sciences, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
36
|
Popa-Fotea NM, Ferdoschi CE, Micheu MM. Molecular and cellular mechanisms of inflammation in atherosclerosis. Front Cardiovasc Med 2023; 10:1200341. [PMID: 37600028 PMCID: PMC10434786 DOI: 10.3389/fcvm.2023.1200341] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Atherosclerosis and its complications are a major cause of morbidity and mortality worldwide in spite of the improved medical and invasive treatment in terms of revascularization. Atherosclerosis is a dynamic, multi-step process in which inflammation is a ubiquitous component participating in the initiation, development, and entanglements of the atherosclerotic plaque. After activation, the immune system, either native or acquired, is part of the atherosclerotic dynamics enhancing the pro-atherogenic function of immune or non-immune cells, such as endothelial cells, smooth muscle cells, or platelets, through mediators such as cytokines or directly by cell-to-cell interaction. Cytokines are molecules secreted by the activated cells mentioned above that mediate the inflammatory component of atherosclerosis whose function is to stimulate the immune cells and the production of further cytokines. This review provides insights of the cell axis activation and specific mechanisms and pathways through which inflammation actuates atherosclerosis.
Collapse
Affiliation(s)
- Nicoleta-Monica Popa-Fotea
- Department 4 Cardio-Thoracic Pathology, University of Medicine and Pharmacy “Carol Davila,”Bucharest, Romania
- Cardiology Department, Emergency Clinical Hospital, Bucharest, Romania
| | - Corina-Elena Ferdoschi
- Department 4 Cardio-Thoracic Pathology, University of Medicine and Pharmacy “Carol Davila,”Bucharest, Romania
| | | |
Collapse
|
37
|
Gherasie FA, Popescu MR, Bartos D. Acute Coronary Syndrome: Disparities of Pathophysiology and Mortality with and without Peripheral Artery Disease. J Pers Med 2023; 13:944. [PMID: 37373933 DOI: 10.3390/jpm13060944] [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: 03/14/2023] [Revised: 05/25/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
There are a number of devastating complications associated with peripheral artery disease, including limb amputations and acute limb ischemia. Despite the overlap, atherosclerotic diseases have distinct causes that need to be differentiated and managed appropriately. In coronary atherosclerosis, thrombosis is often precipitated by rupture or erosion of fibrous caps around atheromatous plaques, which leads to acute coronary syndrome. Regardless of the extent of atherosclerosis, peripheral artery disease manifests itself as thrombosis. Two-thirds of patients with acute limb ischemia have thrombi associated with insignificant atherosclerosis. A local thrombogenic or remotely embolic basis of critical limb ischemia may be explained by obliterative thrombi in peripheral arteries of patients without coronary artery-like lesions. Studies showed that thrombosis of the above-knee arteries was more commonly due to calcified nodules, which are the least common cause of luminal thrombosis associated with acute coronary events in patients with acute coronary syndrome. Cardiovascular mortality was higher in peripheral artery disease without myocardial infarction/stroke than in myocardial infarction/stroke without peripheral artery disease. The aim of this paper is to gather published data regarding the disparities of acute coronary syndrome with and without peripheral artery disease in terms of pathophysiology and mortality.
Collapse
Affiliation(s)
| | - Mihaela-Roxana Popescu
- Department of Cardiology, University of Medicine and Pharmacy "Carol Davila," 050474 Bucharest, Romania
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 011461 Bucharest, Romania
| | - Daniela Bartos
- Department of Cardiology, University of Medicine and Pharmacy "Carol Davila," 050474 Bucharest, Romania
- Department of Internal Medicine, Clinical University Emergency Hospital, 014461 Bucharest, Romania
| |
Collapse
|
38
|
Moreno-Gonzalez MA, Ortega-Rivera OA, Steinmetz NF. Two decades of vaccine development against atherosclerosis. NANO TODAY 2023; 50:101822. [PMID: 37860053 PMCID: PMC10586238 DOI: 10.1016/j.nantod.2023.101822] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Atherosclerosis is an immune-mediated chronic inflammatory disease that leads to the development of fatty plaques in the arterial walls, ultimately increasing the risk of thrombosis, stroke, and myocardial infarction. The immune response in this complex disease is both atheroprotective and pro-atherogenic, involving both innate and adaptive immunity. Current treatments include the adjustment of lifestyle factors, cholesterol-lowering drugs such as statins, and immunotherapy, whereas vaccine development has received comparatively little attention. In this review, we discuss the potential of antigen-specific vaccination as a preventative approach based on more than 20 years of research and innovation. Vaccination targets include proteins that are more abundant in atherosclerotic patients, such as oxidized low-density lipoprotein (LDL), apolipoprotein B-100, proprotein convertase subtilisin/kexin type-9 serine protease (PCSK9), cholesteryl ester transfer protein (CETP), and heat shock proteins HSP60 and HSP65. Immunization with such proteins or their peptide epitopes has been shown to induce T-cell activation, produce antigen-specific antibodies, reduce the size of atherosclerotic lesions, and/or reduce serum cholesterol levels. Vaccination against atherosclerosis therefore offers a new strategy to address the burden on healthcare systems caused by cardiovascular disease, the leading cause of death worldwide.
Collapse
Affiliation(s)
- Miguel A. Moreno-Gonzalez
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| | - Oscar A. Ortega-Rivera
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla, CA 92039, USA
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92039, USA
- Department of Radiology, University of California-San Diego, La Jolla, CA 92039, USA
- Moores Cancer Center, University of California-San Diego, La Jolla, CA 92039, USA
| |
Collapse
|
39
|
Li Y, Liu Y, Liu S, Gao M, Wang W, Chen K, Huang L, Liu Y. Diabetic vascular diseases: molecular mechanisms and therapeutic strategies. Signal Transduct Target Ther 2023; 8:152. [PMID: 37037849 PMCID: PMC10086073 DOI: 10.1038/s41392-023-01400-z] [Citation(s) in RCA: 207] [Impact Index Per Article: 103.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/19/2023] [Accepted: 02/28/2023] [Indexed: 04/12/2023] Open
Abstract
Vascular complications of diabetes pose a severe threat to human health. Prevention and treatment protocols based on a single vascular complication are no longer suitable for the long-term management of patients with diabetes. Diabetic panvascular disease (DPD) is a clinical syndrome in which vessels of various sizes, including macrovessels and microvessels in the cardiac, cerebral, renal, ophthalmic, and peripheral systems of patients with diabetes, develop atherosclerosis as a common pathology. Pathological manifestations of DPDs usually manifest macrovascular atherosclerosis, as well as microvascular endothelial function impairment, basement membrane thickening, and microthrombosis. Cardiac, cerebral, and peripheral microangiopathy coexist with microangiopathy, while renal and retinal are predominantly microangiopathic. The following associations exist between DPDs: numerous similar molecular mechanisms, and risk-predictive relationships between diseases. Aggressive glycemic control combined with early comprehensive vascular intervention is the key to prevention and treatment. In addition to the widely recommended metformin, glucagon-like peptide-1 agonist, and sodium-glucose cotransporter-2 inhibitors, for the latest molecular mechanisms, aldose reductase inhibitors, peroxisome proliferator-activated receptor-γ agonizts, glucokinases agonizts, mitochondrial energy modulators, etc. are under active development. DPDs are proposed for patients to obtain more systematic clinical care requires a comprehensive diabetes care center focusing on panvascular diseases. This would leverage the advantages of a cross-disciplinary approach to achieve better integration of the pathogenesis and therapeutic evidence. Such a strategy would confer more clinical benefits to patients and promote the comprehensive development of DPD as a discipline.
Collapse
Affiliation(s)
- Yiwen Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yanfei Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
- The Second Department of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Shiwei Liu
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Mengqi Gao
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Wenting Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Keji Chen
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Luqi Huang
- China Center for Evidence-based Medicine of TCM, China Academy of Chinese Medical Sciences, Beijing, 100010, China.
| | - Yue Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
40
|
de Winther MPJ, Bäck M, Evans P, Gomez D, Goncalves I, Jørgensen HF, Koenen RR, Lutgens E, Norata GD, Osto E, Dib L, Simons M, Stellos K, Ylä-Herttuala S, Winkels H, Bochaton-Piallat ML, Monaco C. Translational opportunities of single-cell biology in atherosclerosis. Eur Heart J 2023; 44:1216-1230. [PMID: 36478058 PMCID: PMC10120164 DOI: 10.1093/eurheartj/ehac686] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
The advent of single-cell biology opens a new chapter for understanding human biological processes and for diagnosing, monitoring, and treating disease. This revolution now reaches the field of cardiovascular disease (CVD). New technologies to interrogate CVD samples at single-cell resolution are allowing the identification of novel cell communities that are important in shaping disease development and direct towards new therapeutic strategies. These approaches have begun to revolutionize atherosclerosis pathology and redraw our understanding of disease development. This review discusses the state-of-the-art of single-cell analysis of atherosclerotic plaques, with a particular focus on human lesions, and presents the current resolution of cellular subpopulations and their heterogeneity and plasticity in relation to clinically relevant features. Opportunities and pitfalls of current technologies as well as the clinical impact of single-cell technologies in CVD patient care are highlighted, advocating for multidisciplinary and international collaborative efforts to join the cellular dots of CVD.
Collapse
Affiliation(s)
- Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Magnus Bäck
- Translational Cardiology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- University of Lorraine, INSERM U1116, Nancy University Hospital, Nancy, France
| | - Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Delphine Gomez
- Department of Medicine, Division of Cardiology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Isabel Goncalves
- Cardiovascular Research Translational Studies, Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Helle F Jørgensen
- Cardiorespiratory Medicine Section, Department of Medicine, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Esther Lutgens
- Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilian’s Universität, Munich, Germany
- German Centre of Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Cardiovascular Medicine, Experimental CardioVascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Giuseppe Danilo Norata
- Department of Excellence in Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Center for the Study of Atherosclerosis, SISA, Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Elena Osto
- Institute of Clinical Chemistry and Department of Cardiology, Heart Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Lea Dib
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, OX37FY Oxford, UK
| | - Michael Simons
- Departments of Internal Medicine and Cell Biology, Yale University and Yale Cardiovascular Research Center, 300 George St, New Haven, CT 06511, USA
| | - Konstantinos Stellos
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland and Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Holger Winkels
- Department of Internal Medicine III, Division of Cardiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | | | - Claudia Monaco
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, OX37FY Oxford, UK
| |
Collapse
|
41
|
Duan R, Liu Y, Tang D, Lin R, Huang J, Zhao M. IgG1 Is the Optimal Subtype for Treating Atherosclerosis by Inducing M2 Macrophage Differentiation, and Is Independent of the FcγRIIA Gene Polymorphism. Int J Mol Sci 2023; 24:ijms24065932. [PMID: 36983007 PMCID: PMC10053586 DOI: 10.3390/ijms24065932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
In recent years, it has been established that atherosclerosis is an autoimmune disease. However, little is currently known about the role of FcγRIIA in atherosclerosis. Herein, we sought to investigate the relationship between FcγRIIA genotypes and the effectiveness of different IgG subclasses in treating atherosclerosis. We constructed and produced different subtypes of IgG and Fc-engineered antibodies. In vitro, we observed the effect of different subtypes of IgG and Fc-engineered antibodies on the differentiation of CD14+ monocytes from patients or healthy individuals. In vivo, Apoe-/- mice were fed a high-fat diet (HFD) for 20 weeks and administered injections of different CVI-IgG subclasses or Fc-engineered antibodies. Flow cytometry was used to assess the polarization of monocytes and macrophages. Although CVI-IgG4 reduced the release of MCP-1 compared to the other subtypes, IgG4 did not yield an anti-inflammatory effect by induction of human monocyte and macrophage differentiation in vitro. Furthermore, genetic polymorphisms of FcγRIIA were not associated with different CVI-IgG subclasses during the treatment of atherosclerosis. In vivo, CVI-IgG1 decreased Ly6Chigh monocyte differentiation and promoted M2 macrophage polarization. We also found that the secretion of IL-10 was upregulated in the CVI-IgG1-treated group, whereas V11 and GAALIE exerted no significant effect. These findings highlight that IgG1 is the optimal subtype for treating atherosclerosis, and CVI-IgG1 can induce monocyte/macrophage polarization. Overall, these results have important implications for the development of therapeutic antibodies.
Collapse
Affiliation(s)
- Rui Duan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Liu
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Dongmei Tang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Run Lin
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinrong Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ming Zhao
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
42
|
Brown PA, Brown PD. Extracellular vesicles and atherosclerotic peripheral arterial disease. Cardiovasc Pathol 2023; 63:107510. [PMID: 36460259 DOI: 10.1016/j.carpath.2022.107510] [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: 07/23/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Atherogenesis involves a complex multifactorial process including chronic inflammation that requires the participation of several cell types and molecules. In addition to their role in vascular homeostasis, extracellular vesicles also appear to play an important role in atherogenesis, including monocyte transmigration and foam cell formation, SMC proliferation and migration, leukocyte transmigration, and thrombosis. Peripheral arterial disease, a major form of peripheral vascular disease, is characterized by structural or functional impairment of peripheral arterial supply, often secondary to atherosclerosis. Elevated levels of extracellular vesicles have been demonstrated in patients with peripheral arterial disease and implicated in the development of atherosclerosis within peripheral vascular beds. However, extracellular vesicles also appear capable of delivering cargo with atheroprotective effects. This capability has been exploited in vesicles engineered to carry content capable of neovascularization, suggesting potential for therapeutic angiogenesis. This dual capacity holds substantial promise for diagnosis and therapy, including possibly limb- and life-saving options for peripheral arterial disease management.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica.
| | - Paul D Brown
- Department of Basic Medical Sciences, University of the West Indies, Mona, Jamaica
| |
Collapse
|
43
|
Wang Z, Zhang X, Lu S, Zhang C, Ma Z, Su R, Li Y, Sun T, Li Y, Hong M, Deng X, Monjezi MR, Hristov M, Steffens S, Santovito D, Dornmair K, Ley K, Weber C, Mohanta SK, Habenicht AJR, Yin C. Pairing of single-cell RNA analysis and T cell antigen receptor profiling indicates breakdown of T cell tolerance checkpoints in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2023; 2:290-306. [PMID: 37621765 PMCID: PMC10448629 DOI: 10.1038/s44161-023-00218-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 01/18/2023] [Indexed: 08/26/2023]
Abstract
Atherosclerotic plaques form in the inner layer of arteries triggering heart attacks and strokes. Although T cells have been detected in atherosclerosis, tolerance dysfunction as a disease driver remains unexplored. Here we examine tolerance checkpoints in atherosclerotic plaques, artery tertiary lymphoid organs and lymph nodes in mice burdened by advanced atherosclerosis, via single-cell RNA sequencing paired with T cell antigen receptor sequencing. Complex patterns of deteriorating peripheral T cell tolerance were observed being most pronounced in plaques followed by artery tertiary lymphoid organs, lymph nodes and blood. Affected checkpoints included clonal expansion of CD4+, CD8+ and regulatory T cells; aberrant tolerance-regulating transcripts of clonally expanded T cells; T cell exhaustion; Treg-TH17 T cell conversion; and dysfunctional antigen presentation. Moreover, single-cell RNA-sequencing profiles of human plaques revealed that the CD8+ T cell tolerance dysfunction observed in mouse plaques was shared in human coronary and carotid artery plaques. Thus, our data support the concept of atherosclerosis as a bona fide T cell autoimmune disease targeting the arterial wall.
Collapse
Affiliation(s)
- Zhihua Wang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- These authors contributed equally: Zhihua Wang, Xi Zhang, Shu Lu, Andreas J. R. Habenicht, Changjun Yin
| | - Xi Zhang
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- These authors contributed equally: Zhihua Wang, Xi Zhang, Shu Lu, Andreas J. R. Habenicht, Changjun Yin
| | - Shu Lu
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- These authors contributed equally: Zhihua Wang, Xi Zhang, Shu Lu, Andreas J. R. Habenicht, Changjun Yin
| | - Chuankai Zhang
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhe Ma
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Rui Su
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Yuanfang Li
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Ting Sun
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Yutao Li
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Mingyang Hong
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Xinyi Deng
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Mohammad Rafiee Monjezi
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy
| | - Klaus Dornmair
- Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University, Munich, Germany
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Klaus Ley
- Immunology Center of Georgia (IMMCG), Augusta University, Augusta, GA, USA
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University, Munich, Germany
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Sarajo K. Mohanta
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Andreas J. R. Habenicht
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- These authors contributed equally: Zhihua Wang, Xi Zhang, Shu Lu, Andreas J. R. Habenicht, Changjun Yin
| | - Changjun Yin
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- These authors contributed equally: Zhihua Wang, Xi Zhang, Shu Lu, Andreas J. R. Habenicht, Changjun Yin
| |
Collapse
|
44
|
Ma Z, Mao C, Chen X, Yang S, Qiu Z, Yu B, Jia Y, Wu C, Wang Y, Wang Y, Gu R, Yu F, Yin Y, Wang X, Xu Q, Liu C, Liao Y, Zheng J, Fu Y, Kong W. Peptide Vaccine Against ADAMTS-7 Ameliorates Atherosclerosis and Postinjury Neointima Hyperplasia. Circulation 2023; 147:728-742. [PMID: 36562301 DOI: 10.1161/circulationaha.122.061516] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The metalloprotease ADAMTS-7 (a disintegrin and metalloproteinase with thrombospondin type 1 motif 7) is a novel locus associated with human coronary atherosclerosis. ADAMTS-7 deletion protects against atherosclerosis and vascular restenosis in rodents. METHODS We designed 3 potential vaccines consisting of distinct B cell epitopic peptides derived from ADAMTS-7 and conjugated with the carrier protein KLH (keyhole limpet hemocyanin) as well as aluminum hydroxide as an adjuvant. Arterial ligation or wire injury was used to induce neointima in mice, whereas ApoE-/- and LDLR-/- (LDLR [low-density lipoprotein receptor]) mice fed a high-fat diet were applied to assess atherosclerosis. In addition, coronary stent implantation was performed on vaccine-immunized Bama miniature pigs, followed by optical coherence tomography to evaluate coronary intimal hyperplasia. RESULTS A vaccine, ATS7vac, was screened out from 3 candidates to effectively inhibit intimal thickening in murine carotid artery ligation models after vaccination. As well, immunization with ATS7vac alleviated neointima formation in murine wire injury models and mitigated atherosclerotic lesions in both hyperlipidemic ApoE-/- and LDLR-/- mice without lowering lipid levels. Preclinically, ATS7vac markedly impeded intimal hyperplasia in swine stented coronary arteries, but without significant immune-related organ injuries. Mechanistically, ATS7vac vaccination produced specific antibodies against ADAMTS-7, which markedly repressed ADAMTS-7-mediated COMP (cartilage oligomeric matrix protein) and TSP-1 (thrombospondin-1) degradation and subsequently inhibited vascular smooth muscle cell migration but promoted re-endothelialization. CONCLUSIONS ATS7vac is a novel atherosclerosis vaccine that also alleviates in-stent restenosis. The application of ATS7vac would be a complementary therapeutic avenue to the current lipid-lowering strategy for atherosclerotic disease.
Collapse
Affiliation(s)
- Zihan Ma
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China.,Beijing Institute of Biotechnology, Beijing, China (C.M.)
| | - Xiao Chen
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Zhihua Qiu
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Baoqi Yu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (B.Y.), Ministry of Education, Beijing, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (B.Y.)
| | - Yiting Jia
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Chao Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (C.W., J.Z.)
| | - Yiyi Wang
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Yuhui Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education (Yuhui Wang), Peking University, Beijing, China
| | - Rui Gu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Yanhui Yin
- Department of Immunology (Y.Y.), Peking University, Beijing, China.,Key Laboratory of Medical Immunology of Ministry of Health (Y.Y.), Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Qingbo Xu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (Q.X.).,Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q.X.)
| | - Chuanju Liu
- Department of Orthopedic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY (C.L.)
| | - Yuhua Liao
- Department of Cardiology and Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.).,Key Laboratory of Molecular Biological Targeted Therapies, Ministry of Education, Wuhan, China (X.C., Z.Q., Yiyi Wang, Y.L.)
| | - Jingang Zheng
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China (C.W., J.Z.)
| | - Yi Fu
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science (Z.M., C.M., S.Y., Y.J., R.G., F.Y., X.W., Y.F., W.K.), Ministry of Education, Beijing, China
| |
Collapse
|
45
|
Lyu Q, Ley K. How Lymphatic Endothelial Cells Destabilize Regulatory T Cells. Arterioscler Thromb Vasc Biol 2023; 43:215-217. [PMID: 36579643 PMCID: PMC10108378 DOI: 10.1161/atvbaha.122.318849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Qingkang Lyu
- Immunology Center of Georgia (Q.L., K.L.), Augusta University
| | - Klaus Ley
- Immunology Center of Georgia (Q.L., K.L.), Augusta University.,Department of Physiology (K.L.), Augusta University
| |
Collapse
|
46
|
Nettersheim FS, Ghosheh Y, Winkels H, Kobiyama K, Durant C, Armstrong SS, Brunel S, Roy P, Dileepan T, Jenkins MK, Zajonc DM, Ley K. Single-cell transcriptomes and T cell receptors of vaccine-expanded apolipoprotein B-specific T cells. Front Cardiovasc Med 2023; 9:1076808. [PMID: 36684560 PMCID: PMC9849899 DOI: 10.3389/fcvm.2022.1076808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerotic cardiovascular diseases are the major cause of death worldwide. CD4 T cells responding to Apolipoprotein B (ApoB), the core protein of most lipoproteins, have been identified as critical disease modulators. In healthy individuals, ApoB-reactive (ApoB+) CD4 T cells are mostly regulatory T cells (Tregs), which exert anti-inflammatory effects. Yet, they may obtain pro-inflammatory features and thus become proatherogenic. Evidence from animal studies suggests that vaccination against certain major histocompatibility complex (MHC) II-binding ApoB peptides induces an expansion of ApoB+ Tregs and thus confers atheroprotection. To date, in-depth phenotyping of vaccine-expanded ApoB+ T cells has not yet been performed. To this end, we vaccinated C57BL/6J mice with the ApoB-peptide P6 (ApoB978-993 TGAYSNASSTESASY) and performed single-cell RNA sequencing of tetramer-sorted P6+ T cells. P6+ cells were clonally expanded (one major, two minor clones) and formed a transcriptional cluster distinct from clusters mainly containing non-expanded P6+ and P6- cells. Transcriptomic profiling revealed that most expanded P6+ cells had a strong Treg signature and highly expressed genes mediating suppressive functions. Yet, some expanded P6+ cells only had a residual Treg signature and expressed genes related to T helper 1 (TH1) cells, which are proatherogenic. Modeling the T cell receptor (TCR) and P6:MHC-II interaction showed that only three amino acid residues in the α and β chain contact the P6 peptide in the MHC-II groove and thus determine the specificity of this TCR to P6. Our data begin to reveal the vaccination-induced response to an ApoB epitope.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Yanal Ghosheh
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Holger Winkels
- La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kouji Kobiyama
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | | | - Simon Brunel
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Payel Roy
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Thamotharampillai Dileepan
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Marc K. Jenkins
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Dirk M. Zajonc
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
- Immunology Center of Georgia (IMMCG), Augusta University, Augusta, GA, United States
| |
Collapse
|
47
|
Orecchioni M, Wolf D, Suryawanshi V, Winkels H, Kobiyama K, Makings J, Kiosses WB, Ley K. Deleting interleukin-10 from myeloid cells exacerbates atherosclerosis in Apoe -/- mice. Cell Mol Life Sci 2022; 80:10. [PMID: 36496494 PMCID: PMC10141524 DOI: 10.1007/s00018-022-04649-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/05/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is initiated by subendothelial retention of lipoproteins and cholesterol, which triggers a non-resolving inflammatory process that over time leads to plaque progression in the artery wall. Myeloid cells and in particular macrophages are the primary drivers of the inflammatory response and plaque formation. Several immune cells including macrophages, T cells and B cells secrete the anti-inflammatory cytokine IL-10, known to be essential for the atherosclerosis protection. The cellular source of IL-10 in natural atherosclerosis progression is unknown. This study aimed to determine the main IL10-producing cell type in atherosclerosis. To do so, we crossed VertX mice, in which IRES-green fluorescent protein (eGFP) was placed downstream of exon 5 of the Il10 gene, with atherosclerosis-prone Apoe-/- mice. We found that myeloid cells express high levels of IL-10 in VertX Apoe-/- mice in both chow and western-diet fed mice. By single cell RNA sequencing and flow cytometry analysis, we identified resident and inflammatory macrophages in atherosclerotic plaques as the main IL-10 producers. To address whether IL-10 secreted by myeloid cells is essential for the protection, we utilized LyzMCre+Il10fl/fl mice crossed into the Apoe-/- background and confirmed that macrophages were unable to secrete IL-10. Chow and western diet-fed LyzMCre+Il10fl/fl Apoe-/- mice developed significantly larger atherosclerotic plaques as measured by en face morphometry than LyzMCre-Il10 fl/flApoe-/-. Flow cytometry and cytokine measurements suggest that the depletion of IL-10 in myeloid cells increases Th17 cells with elevated CCL2, and TNFα in blood plasma. We conclude that macrophage-derived IL-10 is critical for limiting atherosclerosis in mice.
Collapse
Affiliation(s)
- Marco Orecchioni
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA.
| | - Dennis Wolf
- Cardiology and Angiology I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vasantika Suryawanshi
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jeffrey Makings
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA
| | - William B Kiosses
- Histology and Microscopy Core Facility, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Immunology Center of Georgia (IMMCG), Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
48
|
ANGPTL3 deficiency associates with the expansion of regulatory T cells with reduced lipid content. Atherosclerosis 2022; 362:38-46. [PMID: 36253169 DOI: 10.1016/j.atherosclerosis.2022.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Angiopoietin-like 3 (ANGPTL3) regulates lipid and glucose metabolism. Loss-of-function mutations in its gene, leading to ANGPTL3 deficiency, cause in humans the familial combined hypolipidemia type 2 (FHBL2) phenotype, characterized by very low concentrations of circulating lipoproteins and reduced risk of atherosclerotic cardiovascular disease. Whether this condition is accompanied by immune dysfunctions is unknown. Regulatory T cells (Tregs) are CD4 T lymphocytes endowed with immune suppressive and atheroprotective functions and sensitive to metabolic signals. By investigating FHBL2, we explored the hypothesis that Tregs expand in response to extreme hypolipidemia, through a modulation of the Treg-intrinsic lipid metabolism. METHODS Treg frequency, phenotype, and intracellular lipid content were assessed ex vivo from FHBL2 subjects and age- and sex-matched controls, through multiparameter flow cytometry. The response of CD4 T cells from healthy controls to marked hypolipidemia was tested in vitro in low-lipid culture conditions. RESULTS The ex vivo analysis revealed that FHBL2 subjects showed higher percentages of Tregs with a phenotype undistinguishable from controls and with a lower lipid content, which directly correlated with the concentrations of circulating lipoproteins. In vitro, lipid restriction induced the upregulation of genes of the mevalonate pathway, including those involved in isoprenoid biosynthesis, and concurrently increased the expression of the Treg markers FOXP3 and Helios. The latter event was found to be prenylation-dependent, and likely related to increased IL-2 production and signaling. CONCLUSIONS Our study demonstrates that FHBL2 is characterized by high Treg frequencies, a feature which may concur to the reduced atherosclerotic risk in this condition. Mechanistically, hypolipidemia may directly favor Treg expansion, through the induction of the mevalonate pathway and the prenylation of key signaling proteins.
Collapse
|
49
|
Lyu P, Song Y, Bi R, Li Z, Wei Y, Huang Q, Cui C, Song D, Zhou X, Fan Y. Protective Actions in Apical Periodontitis: The Regenerative Bioactivities Led by Mesenchymal Stem Cells. Biomolecules 2022; 12:1737. [PMID: 36551165 PMCID: PMC9776067 DOI: 10.3390/biom12121737] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Resulting from bacterial infection, apical periodontitis (AP) is a common inflammatory disease of the periapical region of the tooth. The regeneration of the destroyed periapical alveolar bone and the surrounding periodontium tissues has long been a difficult task in clinical practice. These lesions are closely related to pathogen invasion and an overreactive immune response. It is worth noting that the protective healing process occurs simultaneously, in which mesenchymal stem cells (MSCs) have a crucial function in mediating the immune system and promoting regeneration. Here, we review the recent studies related to AP, with a focus on the regulatory network of MSCs. We also discuss the potential therapeutic approaches of MSCs in inflammatory diseases to provide a basis for promoting tissue regeneration and modulating inflammation in AP. A deeper understanding of the protective action of MSCs and the regulatory networks will help to delineate the underlying mechanisms of AP and pave the way for stem-cell-based regenerative medicine in the future.
Collapse
Affiliation(s)
- Ping Lyu
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yiming Song
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ruiye Bi
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zucen Li
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yali Wei
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qin Huang
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chen Cui
- Guangdong Province Key Laboratory of Stomatology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou 510055, China
| | - Dongzhe Song
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xuedong Zhou
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yi Fan
- National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
50
|
Fernández-Gallego N, Castillo-González R, Méndez-Barbero N, López-Sanz C, Obeso D, Villaseñor A, Escribese MM, López-Melgar B, Salamanca J, Benedicto-Buendía A, Jiménez-Borreguero LJ, Ibañez B, Sastre J, Belver MT, Vega F, Blanco C, Barber D, Sánchez-Madrid F, de la Fuente H, Martín P, Esteban V, Jiménez-Saiz R. The impact of type 2 immunity and allergic diseases in atherosclerosis. Allergy 2022; 77:3249-3266. [PMID: 35781885 DOI: 10.1111/all.15426] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 01/28/2023]
Abstract
Allergic diseases are allergen-induced immunological disorders characterized by the development of type 2 immunity and IgE responses. The prevalence of allergic diseases has been on the rise alike cardiovascular disease (CVD), which affects arteries of different organs such as the heart, the kidney and the brain. The underlying cause of CVD is often atherosclerosis, a disease distinguished by endothelial dysfunction, fibrofatty material accumulation in the intima of the artery wall, smooth muscle cell proliferation, and Th1 inflammation. The opposed T-cell identity of allergy and atherosclerosis implies an atheroprotective role for Th2 cells by counteracting Th1 responses. Yet, the clinical association between allergic disease and CVD argues against it. Within, we review different phases of allergic pathology, basic immunological mechanisms of atherosclerosis and the clinical association between allergic diseases (particularly asthma, atopic dermatitis, allergic rhinitis and food allergy) and CVD. Then, we discuss putative atherogenic mechanisms of type 2 immunity and allergic inflammation including acute allergic reactions (IgE, IgG1, mast cells, macrophages and allergic mediators such as vasoactive components, growth factors and those derived from the complement, contact and coagulation systems) and late phase inflammation (Th2 cells, eosinophils, type 2 innate-like lymphoid cells, alarmins, IL-4, IL-5, IL-9, IL-13 and IL-17).
Collapse
Affiliation(s)
- Nieves Fernández-Gallego
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Raquel Castillo-González
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Pathology, Hospital 12 de Octubre, Madrid, Spain
| | - Nerea Méndez-Barbero
- Vascular Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Celia López-Sanz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - David Obeso
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Department of Chemistry and Biochemistry, Faculty of Pharmacy, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alma Villaseñor
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Department of Chemistry and Biochemistry, Faculty of Pharmacy, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María M Escribese
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Beatriz López-Melgar
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Jorge Salamanca
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Amparo Benedicto-Buendía
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Luis Jesús Jiménez-Borreguero
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Borja Ibañez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Cardiology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Joaquín Sastre
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - María Teresa Belver
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Francisco Vega
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Carlos Blanco
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Domingo Barber
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Hortensia de la Fuente
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Martín
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Rodrigo Jiménez-Saiz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
- Department of Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|