1
|
Yarman Y, Zhao X, Ma P. Nuclear Receptors in Platelet Activation and Thrombosis in Hypercholesterolemia. Circ Res 2025; 136:827-829. [PMID: 40208927 PMCID: PMC11990083 DOI: 10.1161/circresaha.125.326364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Affiliation(s)
- Yanki Yarman
- Cardeza Foundation of Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Xuefei Zhao
- Cardeza Foundation of Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peisong Ma
- Cardeza Foundation of Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Liu W, Li G, Shi J, Gao Y, Fang P, Zhao Y, Zhong F, Guo X, Lyu Y, Da X, Li Z, Fa J, Hu L, Yuan A, Chen L, Liu J, Chen AF, Sheng B, Ji Y, Lu X, Pu J. NR4A1 Acts as a Novel Regulator of Platelet Activation and Thrombus Formation. Circ Res 2025; 136:809-826. [PMID: 40035146 PMCID: PMC11984555 DOI: 10.1161/circresaha.124.325645] [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: 10/02/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Mounting evidence indicates that nuclear receptors play a critical regulatory role in platelet pathophysiology and thrombotic disorders. Although NR4A (the nuclear receptor subfamily 4 group A) plays an important role in cardiovascular pathophysiology, the expression profile and biological function of NR4A member 1 (NR4A1) in platelets have never been reported. METHODS We evaluated the functions and the underlying mechanisms of NR4A1 in platelet activation and thrombus formation using platelet-specific NR4A1-deficient mice and NR4A1-specific agonists. Using a hyperlipidemic mouse model and platelets from patients with hypercholesterolemia, we explored the influence of hypercholesterolemia on NR4A1 expression and the effects of NR4A1-specific agonists on platelet hyperreactivity induced by hypercholesterolemia. RESULTS NR4A1 was expressed in both human and mouse platelets. Platelet-specific NR4A1 deletion accelerated FeCl3-induced carotid arterial occlusive thrombus formation, enhanced collagen/epinephrine-induced pulmonary thromboembolism, and exacerbated microvascular microthrombi obstruction and infarct expansion in an acute myocardial infarction model. NR4A1-deficient platelets exhibited enhanced agonist-induced aggregation responses, integrin αIIbβ3 activation, dense granule release, α-granule release, platelet spreading, and clot retraction. Consistently, pharmacological activation of NR4A1 by specific agonists decreased platelet activation in both mouse and human platelets. Mechanistically, CAP1 (adenylyl cyclase-associated protein 1) was identified as the direct downstream interacting protein of NR4A1. NR4A1 deletion decreased cAMP levels and phosphorylation of VASP (vasodilator-stimulated phosphoprotein), while NR4A1-specific agonists increased cAMP levels and phosphorylation of VASP in platelets. Importantly, NR4A1 expression in platelets was upregulated in the setting of hypercholesterolemia, which was derived from its upregulation in megakaryocytes in a reactive oxygen species-dependent manner. Platelets from hypercholesterolemic patients and mice exhibited hyperreactivity. However, NR4A1-specific agonists significantly inhibited the activation of hypercholesterolemic platelets to the levels of healthy control platelets. CONCLUSIONS We provide the first evidence that nuclear receptor NR4A1 negatively regulates platelet activation and thrombus formation. NR4A1 may serve as a novel therapeutic target for managing thrombosis-based cardiovascular diseases, especially with hypercholesterolemia.
Collapse
MESH Headings
- Animals
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/blood
- Platelet Activation/physiology
- Humans
- Thrombosis/metabolism
- Thrombosis/blood
- Thrombosis/genetics
- Blood Platelets/metabolism
- Mice
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Hypercholesterolemia/blood
- Hypercholesterolemia/genetics
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Gaoxiang Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jianfeng Shi
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yu Gao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Peiliang Fang
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yichao Zhao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Fangyuan Zhong
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiao Guo
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yuyan Lyu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xingwen Da
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Zhaoyan Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jingjing Fa
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Baoshan Branch (J.F.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Liuhua Hu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Ancai Yuan
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Lei Chen
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Junling Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Alex F. Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, China (B.S.)
| | - Yong Ji
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China(Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Heilongjiang, China (Y.J.)
| | - Xiyuan Lu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
3
|
Giakomidi D, Ishola A, Nus M. Targeting gut microbiota to regulate the adaptive immune response in atherosclerosis. Front Cardiovasc Med 2025; 12:1502124. [PMID: 39957996 PMCID: PMC11825770 DOI: 10.3389/fcvm.2025.1502124] [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: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Atherosclerosis, the leading cause of death worldwide, is a chronic inflammatory disease leading to the accumulation of lipid-rich plaques in the intima of large and medium-sized arteries. Accumulating evidence indicates the important regulatory role of the adaptive immune system in atherosclerosis during all stages of the disease. The gut microbiome has also become a key regulator of atherosclerosis and immunomodulation. Whilst existing research extensively explores the impact of the microbiome on the innate immune system, only a handful of studies have explored the regulatory capacity of the microbiome on the adaptive immune system to modulate atherogenesis. Building on these concepts and the pitfalls on the gut microbiota and adaptive immune response interaction, this review explores potential strategies to therapeutically target the microbiome, including the use of prebiotics and vaccinations, which could influence the adaptive immune response and consequently plaque composition and development.
Collapse
Affiliation(s)
- Despina Giakomidi
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Ayoola Ishola
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Yu H, Li X, Zhao J, Wang W, Wei Q, Mao D. NR4A1-mediated regulation of lipid droplets in progesterone synthesis in goat luteal cells†. Biol Reprod 2024; 111:640-654. [PMID: 38936833 DOI: 10.1093/biolre/ioae085] [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: 11/27/2023] [Revised: 04/03/2024] [Indexed: 06/29/2024] Open
Abstract
Nuclear receptor NR4A1 is a key factor in glycolipid metabolism and steroidogenesis, while lipid droplets serve as crucial dynamic organelles for lipid metabolism in luteal cells. To investigate the effects of NR4A1 on lipid droplet metabolism and progesterone (P4) synthesis in goat corpus luteum in vitro, luteal cells from the middle-cyclic corpus luteum were isolated and treated with Cytosporone B (CSNB, an agonist) or siRNA of NR4A1. Results showed that both low (1 μM) and high (50 μM) concentrations of CSNB promoted lipid droplet accumulation, while NR4A1 knockdown reduced lipid droplet content. CSNB increased while siNR4A1 decreased total cholesterol content; however, CSNB and siNR4A1 did not change triglyceride content. CSNB increased the expression of perilipins at mRNA and protein levels, also increased LDLR, SCARB1, SREBFs, and HMGCR mRNA abundance. Treatment with siNR4A1 revealed opposite results of CSNB, except for HMCGR and SREBF2. For steroidogenesis, 1 μM CSNB increased, but 50 μM CSNB inhibited P4 synthesis, NR4A1 knockdown also reduced the P4 level. Further analysis demonstrated that 1 μM CSNB increased the protein levels of StAR, HSD3B, and P-HSL, while 50 μM CSNB decreased StAR, HSD3B, and CYP11A1 protein levels. Moreover, 50 μM CSNB impaired active mitochondria, reduced the BCL2, and increased DRP1, Caspase 3, and cleaved-Caspase 3 protein levels. siNR4A1 consistently downregulated the P-HSL/HSL ratio and the steroidogenic protein levels. In conclusion, NR4A1-mediated lipid droplets are involved in the regulation of progesterone synthesis in goat luteal cells.
Collapse
Affiliation(s)
- Hao Yu
- Animal Reproduction Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaotong Li
- Animal Reproduction Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Zhao
- Animal Reproduction Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Wang
- Animal Reproduction Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Quanwei Wei
- Animal Reproduction Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dagan Mao
- Animal Reproduction Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
5
|
Ransegnola BP, Pattarabanjird T, McNamara CA. Tipping the Scale: Atheroprotective IgM-Producing B Cells in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1906-1915. [PMID: 39022832 PMCID: PMC11338718 DOI: 10.1161/atvbaha.124.319847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease whose progression is fueled by proinflammatory moieties and limited by anti-inflammatory mediators. Whereas oxidative damage and the generation of oxidation-specific epitopes that act as damage-associated molecular patterns are highly inflammatory, IgM antibodies produced by B-1 and marginal zone B cells counteract unrestricted inflammation by neutralizing and encouraging clearance of these proinflammatory signals. In this review, we focus on describing the identities of IgM-producing B cells in both mice and humans, elaborating the mechanisms underlying IgM production, and discussing the potential strategies to augment the production of atheroprotective IgM. In addition, we will discuss promising therapeutic interventions in humans to help tip the scale toward augmentation of IgM production and to provide atheroprotection.
Collapse
Affiliation(s)
- Brett Patrick Ransegnola
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Pathology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tanyaporn Pattarabanjird
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Coleen A. McNamara
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
6
|
Liu T, Chen Y, Hou L, Yu Y, Ma D, Jiang T, Zhao G. Immune cell-mediated features of atherosclerosis. Front Cardiovasc Med 2024; 11:1450737. [PMID: 39234608 PMCID: PMC11371689 DOI: 10.3389/fcvm.2024.1450737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by innate and adaptive immune responses, which seriously threatens human life and health. It is a primary cause of coronary heart disease, myocardial infarction, and peripheral vascular disease. Research has demonstrated that immune cells are fundamental to the development of atherosclerosis and chronic inflammation. Therefore, it is anticipated that immunotherapy targeting immune cells will be a novel technique in the management of atherosclerosis. This article reviews the growth of research on the regulatory role of immune cells in atherosclerosis and targeted therapy approaches. The purpose is to offer new therapeutic approaches for the control and treatment of cardiovascular illnesses caused by atherosclerosis.
Collapse
Affiliation(s)
- Tingting Liu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yanjun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lianjie Hou
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yulu Yu
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dan Ma
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| |
Collapse
|
7
|
Zhou H, Zhang R, Li M, Wang F, Gao Y, Fang K, Zong J, Chang X. Methazolamide Can Treat Atherosclerosis by Increasing Immunosuppressive Cells and Decreasing Expressions of Genes Related to Proinflammation, Calcification, and Tissue Remodeling. J Immunol Res 2024; 2024:5009637. [PMID: 39081633 PMCID: PMC11288698 DOI: 10.1155/2024/5009637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/01/2024] [Accepted: 06/29/2024] [Indexed: 08/02/2024] Open
Abstract
It has been reported that carbonic anhydrase I (CA1) is a target for the diagnosis and therapy of atherosclerosis (AS) since CA1 can promote AS aortic calcification. We also found that methazolamide (MTZ), a drug for glaucoma treatment and an inhibitor of carbonic anhydrases, can treat AS by inhibiting calcification in aortic tissues. This study focused on the therapeutic mechanism of MTZ and the pathogenic mechanism of AS. In this study, a routine AS animal model was established in ApoE-/- mice, which were treated with MTZ. The aortic tissues were analyzed using single-cell sequencing. MTZ significantly increased the proportions of B-1/MZB B cells with high expressions of Nr4A1 and Ccr7, CD8+CD122+ Treg-like cells with high Nr4A1 expression, and smooth muscle cells with high Tpm2 expression. These cells or their marker genes were reported to exert immunosuppressive, anti-proinflammatory, and atheroprotective effects. MTZ also decreased the proportions of endothelial cells with high expressions of Retn, Apoc1, Lcn2, Mt1, Serpina3, Lpl, and Lgals3; nonclassical CD14+CD16++ monocytes with high expressions of Mt1, Tyrobp, Lgals3, and Cxcl2; and Spp1+ macrophages with high expressions of Mmp-12, Trem2, Mt1, Lgals3, Cxcl2, and Lpl. These cells or their marker genes have been reported to promote inflammation, calcification, tissue remodeling, and atherogenesis. A significant decrease in the proportion of CD8+CD183 (CXCR3)+ T cells, the counterpart of murine CD8+CD122+ T cells, was detected in the peripheral blood of newly diagnosed AS patients rather than in that of patients receiving anti-AS treatments. These results suggest that MTZ can treat AS by increasing immunosuppressive cells and decreasing expressions of genes related to inflammation, calcification, and tissue remodeling.
Collapse
Affiliation(s)
- Hongji Zhou
- Medical Research CenterThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
- Department of CardiologyThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
| | - Rui Zhang
- Department of CardiologyThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
| | - Min Li
- Clinical Laboratory and Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Road Renmin 4, Qingdao 266033, Shandong Province, China
| | - Fuyan Wang
- Clinical Laboratory and Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Road Renmin 4, Qingdao 266033, Shandong Province, China
| | - Yuxia Gao
- Shandong Engineering Research Center of Bacterial Anti-tumor Drugs and Cell Therapy, Jingshi Road 7000, Jinan 250000, Shandong Province, China
| | - Kehua Fang
- Clinical LaboratoryThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| | - Jinbao Zong
- Clinical Laboratory and Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Road Renmin 4, Qingdao 266033, Shandong Province, China
| | - Xiaotian Chang
- Medical Research CenterThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
| |
Collapse
|
8
|
Jones PW, Mallat Z, Nus M. T-Cell/B-Cell Interactions in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1502-1511. [PMID: 38813700 PMCID: PMC11208060 DOI: 10.1161/atvbaha.124.319845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Atherosclerosis is a complex inflammatory disease in which the adaptive immune response plays an important role. While the overall impact of T and B cells in atherosclerosis is relatively well established, we are only beginning to understand how bidirectional T-cell/B-cell interactions can exert prominent atheroprotective and proatherogenic functions. In this review, we will focus on these T-cell/B-cell interactions and how we could use them to therapeutically target the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Peter William Jones
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| | - Ziad Mallat
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
- INSERM U970, Paris Cardiovascular Research Centre, France (Z.M.)
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| |
Collapse
|
9
|
Obare LM, Bonami RH, Doran A, Wanjalla CN. B cells and atherosclerosis: A HIV perspective. J Cell Physiol 2024; 239:e31270. [PMID: 38651687 PMCID: PMC11209796 DOI: 10.1002/jcp.31270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/09/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Atherosclerosis remains a leading cause of cardiovascular disease (CVD) globally, with the complex interplay of inflammation and lipid metabolism at its core. Recent evidence suggests a role of B cells in the pathogenesis of atherosclerosis; however, this relationship remains poorly understood, particularly in the context of HIV. We review the multifaceted functions of B cells in atherosclerosis, with a specific focus on HIV. Unique to atherosclerosis is the pivotal role of natural antibodies, particularly those targeting oxidized epitopes abundant in modified lipoproteins and cellular debris. B cells can exert control over cellular immune responses within atherosclerotic arteries through antigen presentation, chemokine production, cytokine production, and cell-cell interactions, actively participating in local and systemic immune responses. We explore how HIV, characterized by chronic immune activation and dysregulation, influences B cells in the context of atherosclerosis, potentially exacerbating CVD risk in persons with HIV. By examining the proatherogenic and antiatherogenic properties of B cells, we aim to deepen our understanding of how B cells influence atherosclerotic plaque development, especially within the framework of HIV. This research provides a foundation for novel B cell-targeted interventions, with the potential to mitigate inflammation-driven cardiovascular events, offering new perspectives on CVD risk management in PLWH.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amanda Doran
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
10
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [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] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
11
|
Yuan F, Wei J, Cheng Y, Wang F, Gu M, Li Y, Zhao X, Sun H, Ban R, Zhou J, Xia Z. SLAMF7 Promotes Foam Cell Formation of Macrophage by Suppressing NR4A1 Expression During Carotid Atherosclerosis. Inflammation 2024; 47:530-542. [PMID: 37971565 DOI: 10.1007/s10753-023-01926-y] [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: 08/02/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Macrophage-derived lipid-laden foam cells from the subendothelium play a crucial role in the initiation and progression of atherosclerosis. However, the molecule mechanism that regulates the formation of foam cells is not completely understood. Here, we found that SLAMF7 was upregulated in mice bone marrow-derived macrophages and RAW264.7 cells stimulated with oxidized low-density lipoprotein (ox-LDL). SLAMF7 promoted ox-LDL-mediated macrophage lipid accumulation and M1-type polarization. SLAMF7 deficiency reduced serum lipid levels and improved the lesions area of carotid plaque and aortic arch in high-fat diet-fed ApoE-/- mice. In response to ox-LDL, SLAMF7 downregulated NR4A1 and upregulated RUNX3 through transcriptome sequencing analysis. Overexpression NR4A1 reversed SLAMF7-induced lipid uptake and M1 polarization via inhibiting RUNX3 expression. Furthermore, RUNX3 enhanced foam cell formation and M1-type polarization. Taken together, the study suggested that SLAMF7 play contributing roles in the pro-atherogenic effects by regulating NR4A1-RUNX3.
Collapse
Affiliation(s)
- Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Jianmei Wei
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Yan Cheng
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Feifei Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Mingliang Gu
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Yanhui Li
- Department of Rehabilitation Medicine, Liaocheng Chinese Medicine Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Xin Zhao
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China
| | - Hao Sun
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China
| | - Ru Ban
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China
| | - Jing Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, People's Republic of China.
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China.
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China.
| |
Collapse
|
12
|
Harrison J, Newland SA, Jiang W, Giakomidi D, Zhao X, Clement M, Masters L, Corovic A, Zhang X, Drago F, Ma M, Ozsvar Kozma M, Yasin F, Saady Y, Kothari H, Zhao TX, Shi GP, McNamara CA, Binder CJ, Sage AP, Tarkin JM, Mallat Z, Nus M. Marginal zone B cells produce 'natural' atheroprotective IgM antibodies in a T cell-dependent manner. Cardiovasc Res 2024; 120:318-328. [PMID: 38381113 PMCID: PMC10939463 DOI: 10.1093/cvr/cvae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The adaptive immune response plays an important role in atherosclerosis. In response to a high-fat/high-cholesterol (HF/HC) diet, marginal zone B (MZB) cells activate an atheroprotective programme by regulating the differentiation and accumulation of 'poorly differentiated' T follicular helper (Tfh) cells. On the other hand, Tfh cells activate the germinal centre response, which promotes atherosclerosis through the production of class-switched high-affinity antibodies. We therefore investigated the direct role of Tfh cells and the role of IL18 in Tfh differentiation in atherosclerosis. METHODS AND RESULTS We generated atherosclerotic mouse models with selective genetic deletion of Tfh cells, MZB cells, or IL18 signalling in Tfh cells. Surprisingly, mice lacking Tfh cells had increased atherosclerosis. Lack of Tfh not only reduced class-switched IgG antibodies against oxidation-specific epitopes (OSEs) but also reduced atheroprotective natural IgM-type anti-phosphorylcholine (PC) antibodies, despite no alteration of natural B1 cells. Moreover, the absence of Tfh cells was associated with an accumulation of MZB cells with substantially reduced ability to secrete antibodies. In the same manner, MZB cell deficiency in Ldlr-/- mice was associated with a significant decrease in atheroprotective IgM antibodies, including natural anti-PC IgM antibodies. In humans, we found a positive correlation between circulating MZB-like cells and anti-OSE IgM antibodies. Finally, we identified an important role for IL18 signalling in HF/HC diet-induced Tfh. CONCLUSION Our findings reveal a previously unsuspected role of MZB cells in regulating atheroprotective 'natural' IgM antibody production in a Tfh-dependent manner, which could have important pathophysiological and therapeutic implications.
Collapse
Affiliation(s)
- James Harrison
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Stephen A Newland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Wei Jiang
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Despoina Giakomidi
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Xiaohui Zhao
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Clement
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Laboratory for Vascular Translational Sciences (LVTS), Université de Paris, INSERM U1148, Paris, France
| | - Leanne Masters
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrej Corovic
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Xian Zhang
- Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabrizio Drago
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Marcella Ma
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, UK
| | - Maria Ozsvar Kozma
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Froher Yasin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Yuta Saady
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hema Kothari
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tian X Zhao
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Coleen A McNamara
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andrew P Sage
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason M Tarkin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Ziad Mallat
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- PARCC Inserm U970, Universite de Paris, Paris, France
| | - Meritxell Nus
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
13
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
14
|
Phelan DE, Reddan B, Shigemura M, Sznajder JI, Crean D, Cummins EP. Orphan Nuclear Receptor Family 4A (NR4A) Members NR4A2 and NR4A3 Selectively Modulate Elements of the Monocyte Response to Buffered Hypercapnia. Int J Mol Sci 2024; 25:2852. [PMID: 38474099 PMCID: PMC10931687 DOI: 10.3390/ijms25052852] [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: 01/12/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Hypercapnia occurs when the partial pressure of carbon dioxide (CO2) in the blood exceeds 45 mmHg. Hypercapnia is associated with several lung pathologies and is transcriptionally linked to suppression of immune and inflammatory signalling through poorly understood mechanisms. Here we propose Orphan Nuclear Receptor Family 4A (NR4A) family members NR4A2 and NR4A3 as potential transcriptional regulators of the cellular response to hypercapnia in monocytes. Using a THP-1 monocyte model, we investigated the sensitivity of NR4A family members to CO2 and the impact of depleting NR4A2 and NR4A3 on the monocyte response to buffered hypercapnia (10% CO2) using RNA-sequencing. We observed that NR4A2 and NR4A3 are CO2-sensitive transcription factors and that depletion of NR4A2 and NR4A3 led to reduced CO2-sensitivity of mitochondrial and heat shock protein (Hsp)-related genes, respectively. Several CO2-sensitive genes were, however, refractory to depletion of NR4A2 and NR4A3, indicating that NR4As regulate certain elements of the cellular response to buffered hypercapnia but that other transcription factors also contribute. Bioinformatic analysis of conserved CO2-sensitive genes implicated several novel putative CO2-sensitive transcription factors, of which the ETS Proto-Oncogene 1 Transcription Factor (ETS-1) was validated to show increased nuclear expression in buffered hypercapnia. These data give significant insights into the understanding of immune responses in patients experiencing hypercapnia.
Collapse
Affiliation(s)
- David E. Phelan
- School of Medicine, University College Dublin, Dublin 4, Ireland (B.R.)
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Ben Reddan
- School of Medicine, University College Dublin, Dublin 4, Ireland (B.R.)
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Masahiko Shigemura
- Division of Thoracic Surgery, Northwestern University, Chicago, IL 60611, USA;
| | - Jacob I. Sznajder
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Daniel Crean
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
- School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland
| | - Eoin P. Cummins
- School of Medicine, University College Dublin, Dublin 4, Ireland (B.R.)
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
15
|
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
|
16
|
Pattarabanjird T, Nguyen AT, McSkimming C, Dinh HQ, Marshall MA, Ghosheh Y, Gulati R, Durant C, Vallejo J, Saigusa R, Drago F, Guy TV, Premo K, Taylor AM, Paul S, Kundu B, Berr S, Gonen A, Tsimikas S, Miller Y, Pillai S, Ley K, Hedrick CC, McNamara CA. Human circulating CD24 hi marginal zone B cells produce IgM targeting atherogenic antigens and confer protection from vascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1003-1014. [PMID: 39196097 DOI: 10.1038/s44161-023-00356-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 09/26/2023] [Indexed: 08/29/2024]
Abstract
IgMs that inactivate oxidation-specific epitopes (IgMOSE), which are secondary products of lipid peroxidization, protect against inflammatory diseases, including diet-induced atherosclerosis. However, the human B cell subtype that produces IgMOSE remains unknown. In this study, we used single-cell mass cytometry and adoptive transfer of B cell subtypes to NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice to identify B27+IgM+CD24hi cells as the major producers of IgMOSE in humans. Notably, these cells have characteristics of human circulatory marginal zone B (MZB) cells, which are known to be atheoroprotective IgM producers in mice. CD24 antibody treatment to reduce MZB cells and IgM in a hyperlipidemic humanized mouse model provides the evidence that MZB cells protect against vascular inflammation. Consistent with these findings, the frequency of B27+IgM+CD24hi cells (MZB) in patients inversely correlates with coronary artery disease severity.
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Anh Tram Nguyen
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Chantel McSkimming
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
| | - Huy Q Dinh
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine, Madison, WI, USA
| | - Melissa A Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | | | - Fabrizio Drago
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Thomas V Guy
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Angela M Taylor
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Soumen Paul
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Bijoy Kundu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Stuart Berr
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Ayelet Gonen
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Sotirios Tsimikas
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yury Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Klaus Ley
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Coleen A McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA.
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
- Division of Cardiovascular Medicine/Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
17
|
Bending D, Zikherman J. Nr4a nuclear receptors: markers and modulators of antigen receptor signaling. Curr Opin Immunol 2023; 81:102285. [PMID: 36764055 DOI: 10.1016/j.coi.2023.102285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 02/11/2023]
Abstract
Nr4a1-3 encode a small family of orphan nuclear hormone receptors with transcriptional activity. Their expression reflects both acute and chronic antigen-receptor signaling in T and B-cells, and they have been implicated in critical aspects of lymphocyte development, tolerance, and function. These include roles in regulatory T-cell (Treg), thymic-negative selection, humoral responses, anergy, and exhaustion. Here, we review recent advances in this field such as functional roles in B-cells, transcriptional targets, and mechanism of action. We highlight recurrent themes, including integration of antigen-receptor signaling with costimulatory input, as well as unanswered questions and translational applications of this work.
Collapse
Affiliation(s)
- David Bending
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Julie Zikherman
- Division of Rheumatology, Rosalind Russell and Ephraim P. Engleman Arthritis Research Center, Department of Medicine, UCSF, San Francisco, CA 94143, USA.
| |
Collapse
|
18
|
Chen Z, Fan N, Shen G, Yang J. Silencing lncRNA CDKN2B-AS1 Alleviates Childhood Asthma Progression Through Inhibiting ZFP36 Promoter Methylation and Promoting NR4A1 Expression. Inflammation 2023; 46:700-717. [PMID: 36422840 DOI: 10.1007/s10753-022-01766-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 10/10/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022]
Abstract
LncRNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) was found to be upregulated in plasma of patients with bronchial asthma. This study aimed to explore the roles and mechanisms of CDKN2B-AS1 in childhood asthma. We found that CDKN2B-AS1 was upregulated and zinc finger protein 36 (ZFP36) mRNA was downregulated in blood samples of children with asthma compared with healthy controls as measured by RT-qPCR. Human bronchial epithelial cell line BEAS-2B was treated with LPS to induce inflammation model. Small interfering RNA against CDKN2B-AS1 (si-CDKN2B-AS1) was transfected into LPS-treated BEAS-2B cells, and we observed that CDKN2B-AS1 silencing increased cell viability and inhibited apoptosis and inflammation cytokine levels in LPS-treated BEAS-2B cells. Methylation-specific PCR, ChIP, and RIP assays indicated that CDKN2B-AS1 inhibited ZFP36 expression by recruiting DNMT1 to promote ZFP36 promoter methylation. Co-immunoprecipitation (Co-IP) assay verified the interaction between ZFP36 and nuclear receptor subfamily 4 group A member 1 (NR4A1) proteins. Then rescue experiments revealed that ZFP36 knockdown reversed the effects of CDKN2B-AS1 silencing on BEAS-2B cell functions. ZFP36 overexpression facilitated apoptosis, inflammation, and p-p65 expression in BEAS-2B cells, while NR4A1 knockdown reversed these effects. Additionally, CDKN2B-AS1 silencing alleviated airway hyperresponsiveness and inflammation in ovalbumin (OVA)-induced asthma mice. In conclusion, silencing lncRNA CDKN2B-AS1 enhances BEAS-2B cell viability, reduces apoptosis and inflammation in vitro, and alleviated asthma symptoms in OVA-induced asthma mice in vivo through inhibiting ZFP36 promoter methylation and NR4A1-mediated NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhixin Chen
- Department of Pediatrics, Nanyang Central Hospital, No. 312, Gongnong Road, Henan Province, 473000, China.
| | - Nuandong Fan
- Department of Pathology, Nanyang Traditional Chinese Medicine Hospital, Henan Province, 473000, China
| | - Guangsheng Shen
- Department of Pediatrics, Nanyang Central Hospital, No. 312, Gongnong Road, Henan Province, 473000, China
| | - Jing Yang
- Department of Pediatrics, Nanyang Central Hospital, No. 312, Gongnong Road, Henan Province, 473000, China
| |
Collapse
|
19
|
Wang H, Zhang M, Fang F, Xu C, Liu J, Gao L, Zhao C, Wang Z, Zhong Y, Wang X. The nuclear receptor subfamily 4 group A1 in human disease. Biochem Cell Biol 2023; 101:148-159. [PMID: 36861809 DOI: 10.1139/bcb-2022-0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Nuclear receptor 4A1 (NR4A1), a member of the NR4A subfamily, acts as a gene regulator in a wide range of signaling pathways and responses to human diseases. Here, we provide a brief overview of the current functions of NR4A1 in human diseases and the factors involved in its function. A deeper understanding of these mechanisms can potentially improve drug development and disease therapy.
Collapse
Affiliation(s)
- Hongshuang Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Mengjuan Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Fang Fang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chang Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Jiazhi Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Lanjun Gao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chenchen Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.,Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yan Zhong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.,Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China
| |
Collapse
|
20
|
Jing J, Zhu C, Gong R, Qi X, Zhang Y, Zhang Z. Research progress on the active ingredients of traditional Chinese medicine in the intervention of atherosclerosis: A promising natural immunotherapeutic adjuvant. Biomed Pharmacother 2023; 159:114201. [PMID: 36610225 DOI: 10.1016/j.biopha.2022.114201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease caused by disorders of lipid metabolism. Abnormal deposition of low-density lipoproteins in the arterial wall stimulates the activation of immune cells, including the adhesion and infiltration of monocytes, the proliferation and differentiation of macrophages and lymphocytes, and the activation of their functions. The complex interplay between immune cells coordinates the balance between pro- and anti-inflammation and plays a key role in the progression of AS. Therefore, targeting immune cell activity may lead to the development of more selective drugs with fewer side effects to treat AS without compromising host defense mechanisms. At present, an increasing number of studies have found that the active ingredients of traditional Chinese medicine (TCM) can regulate the function of immune cells in multiple ways to against AS, showing great potential for the treatment of AS and promising clinical applications. In this paper, we review the mechanisms of immune cell action in AS lesions and the potential targets and/or pathways for immune cell regulation by the active ingredients of TCM to promote the understanding of the immune system interactions of AS and provide a relevant basis for the use of active ingredients of TCM as natural adjuvants for AS immunotherapy.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Chaojun Zhu
- Surgical Department of Traditional Chinese Medicine, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Rui Gong
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Xue Qi
- Department of General Surgery, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China.
| | - Yue Zhang
- Peripheral Vascular Disease Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Zhaohui Zhang
- Surgical Department of Traditional Chinese Medicine, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
21
|
Subclinical Atherosclerosis Is Associated with Discrepancies in BAFF and APRIL Levels and Altered Breg Potential of Precursor-like Marginal Zone B-Cells in Long-Term HIV Treated Individuals. Vaccines (Basel) 2022; 11:vaccines11010081. [PMID: 36679926 PMCID: PMC9863280 DOI: 10.3390/vaccines11010081] [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: 11/25/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023] Open
Abstract
Chronic inflammation persists in people living with HIV (PLHIV) despite antiretrovial therapy (ART) and is involved in their premature development of cardiovascular diseases (CVD) such as atherosclerosis. We have previously reported that an excess of “B-cell activating factor” (BAFF), an important molecule for the selection and activation of first-line Marginal Zone (MZ) B-cell populations, is associated with deregulations of precursor-like MZ (MZp), whose potent B-cell regulatory (Breg) capacities are altered in PLHIV, early on and despite 1−2 years of ART. Based on these observations, and growing evidence that MZ populations are involved in atherosclerosis control, we designed a cross sectional study to explore the associations between BAFF and its analogue “A proliferation-inducing ligand” (APRIL) with subclinical CVD in long-time-treated individuals of the Canadian HIV and Aging Cohort Study (CHACS) imaging sub-study group. We also characterized the Breg profile of MZp from the blood of these individuals. Results were correlated with the total volume of atherosclerotic plaques (TPV) and with CVD risk factors and biomarkers. TPV was measured using cardiac computerised tomography angiography, and presence of CVD was defined as TPV > 0. We report that blood levels of BAFF are elevated and correlate positively with CVD and its risk factors in PLHIV from the CHACS, in contrast to APRIL levels, which correlate negatively with these factors. The expression levels of Breg markers such as NR4A3, CD39, CD73 and CD83 are significantly lower in PLHIV when compared to those of HIV-uninfected controls. In vitro experiments show that APRIL upregulates the expression of Breg markers by blood MZp from HIV-uninfected individuals, while this modulation is dampened by the addition of recombinant BAFF. Altogether, our observations suggest that strategies viewed to modulate levels of BAFF and/or APRIL could eventually represent a potential treatment target for CVD in PLHIV.
Collapse
|
22
|
Doyon-Laliberté K, Aranguren M, Byrns M, Chagnon-Choquet J, Paniconi M, Routy JP, Tremblay C, Quintal MC, Brassard N, Kaufmann DE, Poudrier J, Roger M. Excess BAFF Alters NR4As Expression Levels and Breg Function of Human Precursor-like Marginal Zone B-Cells in the Context of HIV-1 Infection. Int J Mol Sci 2022; 23:15142. [PMID: 36499469 PMCID: PMC9741410 DOI: 10.3390/ijms232315142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
We have reported excess B-cell activating factor (BAFF) in the blood of HIV-infected progressors, which was concomitant with increased frequencies of precursor-like marginal zone (MZp) B-cells, early on and despite antiretroviral therapy (ART). In controls, MZp possess a strong B-cell regulatory (Breg) potential. They highly express IL-10, the orphan nuclear receptors (NR)4A1, NR4A2 and NR4A3, as well as the ectonucleotidases CD39 and CD73, all of which are associated with the regulation of inflammation. Furthermore, we have shown MZp regulatory function to involve CD83 signaling. To address the impact of HIV infection and excessive BAFF on MZp Breg capacities, we have performed transcriptomic analyses by RNA-seq of sorted MZp B-cells from the blood of HIV-infected progressors. The Breg profile and function of blood MZp B-cells from HIV-infected progressors were assessed by flow-cytometry and light microscopy high-content screening (HCS) analyses, respectively. We report significant downregulation of NR4A1, NR4A2, NR4A3 and CD83 gene transcripts in blood MZp B-cells from HIV-infected progressors when compared to controls. NR4A1, NR4A3 and CD83 protein expression levels and Breg function were also downregulated in blood MZp B-cells from HIV-infected progressors and not restored by ART. Moreover, we observe decreased expression levels of NR4A1, NR4A3, CD83 and IL-10 by blood and tonsillar MZp B-cells from controls following culture with excess BAFF, which significantly diminished their regulatory function. These findings, made on a limited number of individuals, suggest that excess BAFF contributes to the alteration of the Breg potential of MZp B-cells during HIV infection and possibly in other situations where BAFF is found in excess.
Collapse
Affiliation(s)
- Kim Doyon-Laliberté
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Matheus Aranguren
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Michelle Byrns
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Josiane Chagnon-Choquet
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Matteo Paniconi
- Service d’Aide à la Formation Interdisciplinaire et à la Réussite Étudiante (SAFIRE), Faculté des Arts et Sciences de l’Université de Montréal, Montréal, QC H3T 1N8, Canada
| | - Jean-Pierre Routy
- Department of Medicine, McGill University Health Centre, McGill University, Montréal, QC H4A 3J1, Canada
| | - Cécile Tremblay
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Marie-Claude Quintal
- Centre Hospitalier Ste-Justine de l’Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Nathalie Brassard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Daniel E. Kaufmann
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Médecine de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Johanne Poudrier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Michel Roger
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
23
|
Liu XH, Zhou JT, Yan CX, Cheng C, Fan JN, Xu J, Zheng Q, Bai Q, Li Z, Li S, Li X. Single-cell RNA sequencing reveals a novel inhibitory effect of ApoA4 on NAFL mediated by liver-specific subsets of myeloid cells. Front Immunol 2022; 13:1038401. [PMID: 36426356 PMCID: PMC9678944 DOI: 10.3389/fimmu.2022.1038401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/20/2022] [Indexed: 10/24/2023] Open
Abstract
The liver immune microenvironment is a key element in the development of hepatic inflammation in NAFLD. ApoA4 deficiency increases the hepatic lipid burden, insulin resistance, and metabolic inflammation. However, the effect of ApoA4 on liver immune cells and the precise immune cell subsets that exacerbate fatty liver remain elusive. The aim of this study was to profile the hepatic immune cells affected by ApoA4 in NAFL. We performed scRNA-seq on liver immune cells from WT and ApoA4-deficient mice administered a high-fat diet. Immunostaining and qRT-PCR analysis were used to validate the results of scRNA-seq. We identified 10 discrete immune cell populations comprising macrophages, DCs, granulocytes, B, T and NK&NKT cells and characterized their subsets, gene expression profiles, and functional modules. ApoA4 deficiency led to significant increases in the abundance of specific subsets, including inflammatory macrophages (2-Mφ-Cxcl9 and 4-Mφ-Cxcl2) and activated granulocytes (0-Gran-Wfdc17). Moreover, ApoA4 deficiency resulted in higher Lgals3, Ctss, Fcgr2b, Spp1, Cxcl2, and Elane levels and lower Nr4a1 levels in hepatic immune cells. These genes were consistent with human NAFLD-associated marker genes linked to disease severity. The expression of NE and IL-1β in granulocytes and macrophages as key ApoA4 targets were validate in the presence or absence of ApoA4 by immunostaining. The scRNA-seq data analyses revealed reprogramming of liver immune cells resulted from ApoA4 deficiency. We uncovered that the emergence of ApoA4-associated immune subsets (namely Cxcl9+ macrophage, Cxcl2+ macrophage and Wfdc17+ granulocyte), pathways, and NAFLD-related marker genes may promote the development of NAFL. These findings may provide novel therapeutic targets for NAFL and the foundations for further studying the effects of ApoA4 on immune cells in various diseases.
Collapse
Affiliation(s)
- Xiao-Huan Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jin-Ting Zhou
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Chun-xia Yan
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Department of Pathology, Bio-Evidence Sciences Academy, The Western China Science and Technology Innovation Port, Xi’an Jiaotong University, Xi’an, China
| | - Cheng Cheng
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Jing-Na Fan
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Jing Xu
- Division of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiangsun Zheng
- Division of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiang Bai
- Laboratory of Immunophysiology, GIGA Institute, Liège University, Liège, Belgium
| | - Zongfang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shengbin Li
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
24
|
Ballester-Servera C, Cañes L, Alonso J, Puertas L, Taurón M, Rodríguez C, Martínez-González J. Nuclear receptor NOR-1 (Neuron-derived Orphan Receptor-1) in pathological vascular remodelling and vascular remodelling. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2022; 34:229-243. [PMID: 35581107 DOI: 10.1016/j.arteri.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 06/15/2023]
Abstract
Vascular cells and their interaction with inflammatory cells and the immune system play a key role in pathological vascular remodeling. A large number of genes and proteins regulated in a coordinated manner by a small number of transcription factors are involved in this process. In recent years, research on a small subfamily of transcription factors, the NR4A subfamily, has had a major impact on our understanding of vascular biology. The NR4A1 (Nur77), NR4A2 (Nurr1) and NR4A3 (NOR-1) receptors are products of early response genes whose expression is induced by multiple pathophysiological and physical stimuli. Their wide distribution in different tissues and cells places them in the control of numerous processes such as cell differentiation, proliferation, survival and apoptosis, as well as inflammation and the metabolism of lipids and carbohydrates. This review analyzes the role of these receptors, particularly NOR-1, in pathological vascular remodeling associated with atherosclerosis, abdominal aortic aneurysm and pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España
| | - Lidia Puertas
- Instituto de Investigación Biomédica Sant Pau, Barcelona, España; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, España
| | - Manel Taurón
- Servicio de Cirugía Cardiovascular, Hospital de la Santa Creu i Sant Pau, Barcelona, España
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, España
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, España; Instituto de Investigación Biomédica Sant Pau, Barcelona, España.
| |
Collapse
|
25
|
Hiwa R, Brooks JF, Mueller JL, Nielsen HV, Zikherman J. NR4A nuclear receptors in T and B lymphocytes: Gatekeepers of immune tolerance . Immunol Rev 2022; 307:116-133. [PMID: 35174510 DOI: 10.1111/imr.13072] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 12/21/2022]
Abstract
Random VDJ recombination early in T and B cell development enables the adaptive immune system to recognize a vast array of evolving pathogens via antigen receptors. However, the potential of such randomly generated TCRs and BCRs to recognize and respond to self-antigens requires layers of tolerance mechanisms to mitigate the risk of life-threatening autoimmunity. Since they were originally cloned more than three decades ago, the NR4A family of nuclear hormone receptors have been implicated in many critical aspects of immune tolerance, including negative selection of thymocytes, peripheral T cell tolerance, regulatory T cells (Treg), and most recently in peripheral B cell tolerance. In this review, we discuss important insights from many laboratories as well as our own group into the function and mechanisms by which this small class of primary response genes promotes self-tolerance and immune homeostasis to balance the need for host defense against the inherent risks posed by the adaptive immune system.
Collapse
Affiliation(s)
- Ryosuke Hiwa
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
- Department of Rheumatology and Clinical Immunology, Kyoto University Hospital, Kyoto, Japan
| | - Jeremy F Brooks
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - James L Mueller
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - Hailyn V Nielsen
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - Julie Zikherman
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| |
Collapse
|
26
|
Marginal Zone B-Cell Populations and Their Regulatory Potential in the Context of HIV and Other Chronic Inflammatory Conditions. Int J Mol Sci 2022; 23:ijms23063372. [PMID: 35328792 PMCID: PMC8949885 DOI: 10.3390/ijms23063372] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammation in the context of Human Immunodeficiency Virus (HIV) establishes early and persists beyond antiretroviral therapy (ART). As such, we have shown excess B-cell activating factor (BAFF) in the blood of HIV-infected progressors, as soon as in the acute phase, and despite successful ART. Excess BAFF was associated with deregulation of the B-cell compartment; notably, with increased frequencies of a population sharing features of both transitional immature (TI) and marginal zone (MZ) B-cells, we termed Marginal Zone precursor-like (MZp). We have reported similar observations with HIV-transgenic mice, Simian Immunodeficiency Virus (SIV)-infected macaques, and more recently, with HIV-infected Beninese commercial sex workers, which suggests that excess BAFF and increased frequencies of MZp B-cells are reliable markers of inflammation in the context of HIV. Importantly, we have recently shown that in healthy individuals, MZps present an important regulatory B-cell (Breg) profile and function. Herein, we wish to review our current knowledge on MZ B-cell populations, especially their Breg status, and that of other B-cell populations sharing similar features. BAFF and its analog A Proliferation-Inducing Ligand (APRIL) are important in shaping the MZ B-cell pool; moreover, the impact that excess BAFF—encountered in the context of HIV and several chronic inflammatory conditions—may exert on MZ B-cell populations, Breg and antibody producing capacities is a threat to the self-integrity of their antibody responses and immune surveillance functions. As such, deregulations of MZ B-cell populations contribute to autoimmune manifestations and the development of MZ lymphomas (MZLs) in the context of HIV and other inflammatory diseases. Therefore, further comprehending the mechanisms regulating MZ B-cell populations and their functions could be beneficial to innovative therapeutic avenues that could be deployed to restore MZ B-cell immune competence in the context of chronic inflammation involving excess BAFF.
Collapse
|
27
|
Keeter WC, Moriarty AK, Galkina EV. Role of neutrophils in type 2 diabetes and associated atherosclerosis. Int J Biochem Cell Biol 2021; 141:106098. [PMID: 34655814 PMCID: PMC8962624 DOI: 10.1016/j.biocel.2021.106098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/09/2023]
Abstract
The development of cardiovascular diseases associated with Type-2 diabetes remains one of the most challenging public health burdens in the developed world. Early onset of metabolic deficiencies, namely dysregulated glucose homeostasis, peripheral insulin resistance, and impaired insulin production are accompanied by both innate and adaptive immune responses that culminate in a state of chronic, low-grade inflammation. Neutrophils are a critical component of the innate immune system which offer frontline defense against pathogens through a variety of potent effector functions. Recent data indicate an essential role of neutrophils in various disease processes that contribute to the development of Type-2 diabetes and atherosclerosis. In this brief review, we aim to distill the most relevant clinical and pre-clinical literature that investigates the role of neutrophils as an important mediator for the Type-2 diabetes/atherosclerosis connection.
Collapse
Affiliation(s)
- W Coles Keeter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA 23507, USA
| | - Alina K Moriarty
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA 23507, USA
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA 23507, USA.
| |
Collapse
|
28
|
Martínez-González J, Cañes L, Alonso J, Ballester-Servera C, Rodríguez-Sinovas A, Corrales I, Rodríguez C. NR4A3: A Key Nuclear Receptor in Vascular Biology, Cardiovascular Remodeling, and Beyond. Int J Mol Sci 2021; 22:ijms222111371. [PMID: 34768801 PMCID: PMC8583700 DOI: 10.3390/ijms222111371] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
The mechanisms committed in the activation and response of vascular and inflammatory immune cells play a major role in tissue remodeling in cardiovascular diseases (CVDs) such as atherosclerosis, pulmonary arterial hypertension, and abdominal aortic aneurysm. Cardiovascular remodeling entails interrelated cellular processes (proliferation, survival/apoptosis, inflammation, extracellular matrix (ECM) synthesis/degradation, redox homeostasis, etc.) coordinately regulated by a reduced number of transcription factors. Nuclear receptors of the subfamily 4 group A (NR4A) have recently emerged as key master genes in multiple cellular processes and vital functions of different organs, and have been involved in a variety of high-incidence human pathologies including atherosclerosis and other CVDs. This paper reviews the major findings involving NR4A3 (Neuron-derived Orphan Receptor 1, NOR-1) in the cardiovascular remodeling operating in these diseases.
Collapse
Affiliation(s)
- José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
- Correspondence: (J.M.-G.); (C.R.); Tel.: +34-93-5565896 (J.M.-G.); +34-93-5565897 (C.R.)
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
| | - Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain; (L.C.); (J.A.); (C.B.-S.)
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - Irene Corrales
- Laboratorio de Coagulopatías Congénitas, Banc de Sang i Teixits (BST), 08005 Barcelona, Spain;
- Medicina Transfusional, Vall d’Hebron Institut de Recerca-Universitat Autònoma de Barcelona (VHIR-UAB), 08035 Barcelona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, ISCIII, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica Sant Pau, 08041 Barcelona, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), 08041 Barcelona, Spain
- Correspondence: (J.M.-G.); (C.R.); Tel.: +34-93-5565896 (J.M.-G.); +34-93-5565897 (C.R.)
| |
Collapse
|
29
|
Zhang L, Xue S, Ren F, Huang S, Zhou R, Wang Y, Zhou C, Li Z. An atherosclerotic plaque-targeted single-chain antibody for MR/NIR-II imaging of atherosclerosis and anti-atherosclerosis therapy. J Nanobiotechnology 2021; 19:296. [PMID: 34583680 PMCID: PMC8479957 DOI: 10.1186/s12951-021-01047-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Background Oxidation-specific epitopes (OSEs) are rich in atherosclerotic plaques. Innate and adaptive immune responses to OSEs play an important role in atherosclerosis. The purpose of this study was to develop novel human single-chain variable fragment (scFv) antibody specific to OSEs to image and inhibit atherosclerosis. Results Here, we screened a novel scFv antibody, named as ASA6, from phage-displayed human scFv library. ASA6 can bind to oxidized LDL (Ox-LDL) and atherosclerotic plaques. Meanwhile, ASA6 can also inhibit the uptake of Ox-LDL into macrophage to reduce macrophage apoptosis. The atherosclerotic lesion area of ApoE−/− mice administrated with ASA6 antibody was significantly reduced. Transcriptome analysis reveals the anti-atherosclerosis effect of ASA6 is related to the regulation of fatty acid metabolism and inhibition of M1 macrophage polarization. Moreover, we conjugated ASA6 antibody to NaNdF4@NaGdF4 nanoparticles for noninvasive imaging of atherosclerotic plaques by magnetic resonance (MR) and near-infrared window II (NIR-II) imaging. Conclusions Together, these data demonstrate the potential of ASA6 antibody in targeted therapy and noninvasive imaging for atherosclerosis. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01047-4.
Collapse
Affiliation(s)
- Liwei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Sheng Xue
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Feng Ren
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Siyang Huang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Ruizhi Zhou
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Yu Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Changyong Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
30
|
Nuclear receptor Nur77: its role in chronic inflammatory diseases. Essays Biochem 2021; 65:927-939. [PMID: 34328179 DOI: 10.1042/ebc20210004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
Nur77 is a nuclear receptor that has been implicated as a regulator of inflammatory disease. The expression of Nur77 increases upon stimulation of immune cells and is differentially expressed in chronically inflamed organs in human and experimental models. Furthermore, in a variety of animal models dedicated to study inflammatory diseases, changes in Nur77 expression alter disease outcome. The available studies comprise a wealth of information on the function of Nur77 in diverse cell types and tissues. Negative cross-talk of Nur77 with the NFκB signaling complex is an example of Nur77 effector function. An alternative mechanism of action has been established, involving Nur77-mediated modulation of metabolism in macrophages as well as in T cells. In this review, we summarize our current knowledge on the role of Nur77 in atherosclerosis, inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis, and sepsis. Detailed insight in the control of inflammatory responses will be essential in order to advance Nur77-targeted therapeutic interventions in inflammatory disease.
Collapse
|
31
|
Chen S, Duan Y, Wu Y, Yang D, An J. A Novel Integrated Metabolism-Immunity Gene Expression Model Predicts the Prognosis of Lung Adenocarcinoma Patients. Front Pharmacol 2021; 12:728368. [PMID: 34393804 PMCID: PMC8361602 DOI: 10.3389/fphar.2021.728368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although multiple metabolic pathways are involved in the initiation, progression, and therapy of lung adenocarcinoma (LUAD), the tumor microenvironment (TME) for immune cell infiltration that is regulated by metabolic enzymes has not yet been characterized. Methods: 517 LUAD samples and 59 non-tumor samples were obtained from The Cancer Genome Atlas (TCGA) database as the training cohort. Kaplan-Meier analysis and Univariate Cox analysis were applied to screen the candidate metabolic enzymes for their role in relation to survival rate in LUAD patients. A prognostic metabolic enzyme signature, termed the metabolic gene risk score (MGRS), was established based on multivariate Cox proportional hazards regression analysis and was verified in an independent test cohort, GSE31210. In addition, we analyzed the immune cell infiltration characteristics in patients grouped by their Risk Score. Furthermore, the prognostic value of these four enzymes was verified in another independent cohort by immunohistochemistry and an optimized model of the metabolic-immune protein risk score (MIPRS) was constructed. Results: The MGRS model comprising 4 genes (TYMS, NME4, LDHA, and SMOX) was developed to classify patients into high-risk and low-risk groups. Patients with a high-risk score had a poor prognosis and exhibited activated carbon and nucleotide metabolism, both of which were associated with changes to TME immune cell infiltration characteristics. In addition, the optimized MIPRS model showed more accurate predictive power in prognosis of LUAD. Conclusion: Our study revealed an integrated metabolic enzyme signature as a reliable prognostic tool to accurately predict the prognosis of LUAD.
Collapse
Affiliation(s)
- Songming Chen
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanhao Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Desong Yang
- Thoracic Surgery Department II, Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jian An
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Clinical Research Center for Respiratory Diseases, Changsha, China
| |
Collapse
|
32
|
Masago K, Fujita S. Novel NR4A1 Arg293Ser Mutation in Patients With Familial Crohn's Disease. In Vivo 2021; 35:2135-2140. [PMID: 34182489 DOI: 10.21873/invivo.12483] [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: 03/26/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The underlying etiology of Crohn's disease remains unknown. The aim of this study was to identify genomic alterations associated with the development of Crohn's disease in one Japanese family with a family history of Crohn's disease. MATERIALS AND METHODS We performed whole-exome sequence and pedigree analysis of a Japanese family in which both sisters developed Crohn's disease. Whole-exome sequencing was performed using the Ion Torrent Proton™ system. Data from the Proton runs were initially processed using the Ion Torrent platform-specific pipeline software Ion Reporter. An autosomal dominant mode of inheritance was assumed, and stringent selection criteria were applied. RESULTS A substitution in the NR4A1 gene at codon 293 resulting in an amino acid change from arginine to serine was identified only in the affected sisters. CONCLUSION The impaired DNA-binding capacity of the NR4A1 protein due to an NR4A1 germline mutation may be a possible cause of Crohn's disease.
Collapse
Affiliation(s)
- Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Aichi, Japan; .,Division of Integrated Oncology, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Shiro Fujita
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Aichi, Japan.,Division of Integrated Oncology, Institute of Biomedical Research and Innovation, Kobe, Japan
| |
Collapse
|
33
|
Phelan DE, Shigemura M, Aldhafiri S, Mota C, Hall TJ, Sznajder JI, Murphy EP, Crean D, Cummins EP. Transcriptional Profiling of Monocytes Deficient in Nuclear Orphan Receptors NR4A2 and NR4A3 Reveals Distinct Signalling Roles Related to Antigen Presentation and Viral Response. Front Immunol 2021; 12:676644. [PMID: 34248958 PMCID: PMC8267906 DOI: 10.3389/fimmu.2021.676644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022] Open
Abstract
The nuclear receptor sub-family 4 group A (NR4A) family are early response genes that encode proteins that are activated in several tissues/cells in response to a variety of stressors. The NR4A family comprises NR4A1, NR4A2 and NR4A3 of which NR4A2 and NR4A3 are under researched and less understood, particularly in the context of immune cells. NR4A expression is associated with multiple diseases e.g. arthritis and atherosclerosis and the development of NR4A-targetting molecules as therapeutics is a current focus in this research field. Here, we use a combination of RNA-sequencing coupled with strategic bioinformatic analysis to investigate the down-stream effects of NR4A2 and NR4A3 in monocytes and dissect their common and distinct signalling roles. Our data reveals that NR4A2 and NR4A3 depletion has a robust and broad-reaching effect on transcription in both the unstimulated state and in the presence of LPS. Interestingly, many of the genes affected were present in both the unstimulated and stimulated states revealing a previously unappreciated role for the NR4As in unstimulated cells. Strategic clustering and bioinformatic analysis identified both distinct and common transcriptional roles for NR4A2 and NR4A3 in monocytes. NR4A2 notably was linked by both bioinformatic clustering analysis and transcription factor interactome analysis to pathways associated with antigen presentation and regulation of MHC genes. NR4A3 in contrast was more closely linked to pathways associated with viral response. Functional studies further support our data analysis pointing towards preferential/selective roles for NR4A2 in the regulation of antigen processing with common roles for NR4A2 and NR4A3 evident with respect to cell migration. Taken together this study provides novel mechanistic insights into the role of the enigmatic nuclear receptors NR4A2 and NR4A3 in monocytes.
Collapse
Affiliation(s)
- David E Phelan
- School of Medicine, University College Dublin, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Masahiko Shigemura
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Sarah Aldhafiri
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,Animal Genomics Laboratory, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Catarina Mota
- School of Medicine, University College Dublin, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Thomas J Hall
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Evelyn P Murphy
- School of Medicine, University of Limerick, Limerick, Ireland
| | - Daniel Crean
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,Animal Genomics Laboratory, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Eoin P Cummins
- School of Medicine, University College Dublin, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Functional Role of B Cells in Atherosclerosis. Cells 2021; 10:cells10020270. [PMID: 33572939 PMCID: PMC7911276 DOI: 10.3390/cells10020270] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of blood vessels, and both innate and adaptive immune responses are involved in its development. The impact of B cells on atherosclerosis has been demonstrated in numerous studies and B cells have been found in close proximity to atherosclerotic plaques in humans and mice. B cells exert both atheroprotective and pro-atherogenic functions, which have been associated with their B cell subset attribution. While B1 cells and marginal zone B cells are considered to protect against atherosclerosis, follicular B cells and innate response activator B cells have been shown to promote atherosclerosis. In this review, we shed light on the role of B cells from a different, functional perspective and focus on the three major B cell functions: antibody production, antigen presentation/T cell interaction, and the release of cytokines. All of these functions have the potential to affect atherosclerosis by multiple ways and are dependent on the cellular milieu and the activation status of the B cell. Moreover, we discuss B cell receptor signaling and the mechanism of B cell activation under atherosclerosis-prone conditions. By summarizing current knowledge of B cells in and beyond atherosclerosis, we are pointing out open questions and enabling new perspectives.
Collapse
|