1
|
Lu C, Lin Q, Guo X, Luo T, Zhou H, Cai Z, Peng C, Yang G, Wang W. Melatonin modulates mitochondrial function and inhibits atherosclerosis progression through NRF2 activation and OPA1 inhibition. Int Immunopharmacol 2025; 160:114960. [PMID: 40449269 DOI: 10.1016/j.intimp.2025.114960] [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: 02/21/2025] [Revised: 05/12/2025] [Accepted: 05/23/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Atherosclerosis (AS), a major cardiovascular disease, is characterized by chronic inflammation and oxidative stress. Melatonin (MLT) has emerged as a potential therapeutic agent due to its anti-inflammatory and antioxidant properties, although the specific mechanisms underlying its action, especially as far as mitochondrial function in AS is concerned, have yet to be fully elucidated. METHODS In this study, ApoE-/- mice were fed a high-fat diet with or without MLT treatment. Aortic tissues were analyzed using hematoxylin and eosin, Masson staining, qPCR, and immunofluorescence. Oxidized low-density lipoprotein-treated RAW264.7 macrophages were assessed for AS progression, mitochondrial function, and oxidative stress using electron microscopy, Seahorse analysis, and molecular docking. RESULTS MLT treatment significantly reduced atherosclerotic plaque formation, systemic and mitochondrial oxidative stress, and inflammation. MLT treatment was found to enhance mitochondrial function through upregulating the expression of key regulators of mitochondrial biogenesis and the activity of mitochondrial respiratory chain complexes, whereas markers of mitochondrial fusion [for example, optic atrophy protein 1 (OPA1)] were downregulated. Mechanistically, MLT was shown to directly interact with nuclear factor erythroid 2-related factor 2 (NRF2), thereby activating its antioxidant pathway, which in turn regulated mitochondrial function. Additionally, OPA1 was identified as a downstream target of MLT, and its inhibition improved mitochondrial function and reduced inflammation. CONCLUSION This study is the first to elucidate that MLT synergistically ameliorates mitochondrial dysfunction through dual mechanisms-activating the NRF2 antioxidant pathway and suppressing OPA1-mediated mitochondrial fusion-providing novel therapeutic targets for AS.
Collapse
Affiliation(s)
- Chengbo Lu
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, China; Department of Cardiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210018, Jiangsu, China
| | - Quan Lin
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150060, China
| | - Xiaoli Guo
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Tan Luo
- Department of Cardiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, China
| | - Han Zhou
- Department of Cardiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, China
| | - Ziteng Cai
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Chaonan Peng
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Guangyuan Yang
- Clinical Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, China
| | - Weiqun Wang
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, China.
| |
Collapse
|
2
|
Park HM, Kim CL, Kong D, Heo SH, Park HJ. Innovations in Vascular Repair from Mechanical Intervention to Regenerative Therapies. Tissue Eng Regen Med 2025; 22:551-567. [PMID: 39921820 PMCID: PMC12122965 DOI: 10.1007/s13770-024-00700-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Vascular diseases, including atherosclerosis and thrombosis, are leading causes of morbidity and mortality worldwide, often resulting in vessel stenosis that impairs blood flow and leads to severe clinical outcomes. Traditional mechanical interventions, such as balloon angioplasty and bare-metal stents, provided initial solutions but were limited by restenosis and thrombosis. The advent of drug-eluting stents improved short-term outcomes by inhibiting vascular smooth muscle cell proliferation, however, they faced challenges including delayed reendothelialization and late-stage thrombosis. METHODS This review highlights the progression from mechanical to biological interventions in treating vascular stenosis and underscores the need for integrated approaches that combine mechanical precision with regenerative therapies. RESULTS To address long-term complications, bioresorbable stents were developed to provide temporary scaffolding that gradually dissolves, yet they still encounter challenges with mechanical integrity and optimal degradation rates. Consequently, emerging therapies now focus on biological approaches, such as gene therapy, extracellular vesicle treatments, and cell therapies, that aim to promote vascular repair at the cellular level. These strategies offer the potential for true vascular regeneration by enhancing endothelialization, modulating immune responses, and stimulating angiogenesis. CONCLUSION Integrating mechanical precision with regenerative biological therapies offers a promising future for treating vascular stenosis. A comprehensive approach combining these modalities could achieve sustainable vascular health.
Collapse
Affiliation(s)
- Hye-Min Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Chae-Lin Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Dasom Kong
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Seon-Hee Heo
- Department of Surgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Hyun-Ji Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
- Advanced College of Bio-Convergence Engineering, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
3
|
Leleu D, Pilot T, Mangin L, Van Dongen K, Proukhnitzky L, Denimal D, Samson M, Laubriet A, Steinmetz E, Rialland M, Pierre L, Groetz E, Pais de Barros JP, Gautier T, Thomas C, Masson D. Inhibition of LXR Signaling in Human Foam Cells Impairs Macrophage-to-Endothelial Cell Cross Talk and Promotes Endothelial Cell Inflammation. Arterioscler Thromb Vasc Biol 2025; 45:910-927. [PMID: 40207367 PMCID: PMC12094261 DOI: 10.1161/atvbaha.125.322448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND During atherogenesis, macrophages turn into foam cells by engulfing lipids present within the atheroma plaques. The shift of foam cells toward proinflammatory or anti-inflammatory phenotypes, a critical step in disease progression, is still poorly understood. LXRs (liver X receptors) play a pivotal role in the macrophage response to lipid, promoting the expression of key genes of cholesterol efflux, mitigating intracellular cholesterol accumulation. LXRs also exert balanced actions on inflammation in human macrophages, displaying both proinflammatory and anti-inflammatory effects. METHODS Our study explored the role of LXRs in the functional response of human macrophage to lipid-rich plaque environment. We used primary human macrophages treated with atheroma plaque extracts and assessed the impact of pharmacological LXR inhibition by GSK2033 on cholesterol homeostasis and inflammatory response. Ultimately, we evaluated macrophage and endothelial cell cross talk by assessing the impact of macrophage-conditioned supernatants on the human endothelial cell. RESULTS LXR inhibition by GSK2033 resulted in increased levels of cholesterol and oxysterols in human macrophages, alongside notable changes in the cholesterol ester profile. This was accompanied by heightened secretion of proinflammatory cytokines such as IL (interleukin)-6 and TNFα (tumor necrosis factor-α), despite a transcriptional repression of IL-1β. Conditioned media from GSK2033-treated macrophages more effectively activated ICAM-1 (intercellular adhesion molecule-1) and CCL2 (C-C motif ligand 2) expression in endothelial cells. CONCLUSIONS Our findings illustrate the intricate relationship between LXR function, cholesterol metabolism, and inflammation in human macrophages. While LXR is required for the proper handling of plaque lipids by macrophages, the differential regulation of IL-1β versus IL-6/TNFα secretion by LXRs could be challenging for potential pharmacological interventions.
Collapse
Affiliation(s)
- Damien Leleu
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
| | - Thomas Pilot
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
| | - Léa Mangin
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
| | - Kevin Van Dongen
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
| | - Lil Proukhnitzky
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- DiviOmics Platform, UMS58 BioSanD, INSERM, Université de Bourgogne Europe, Dijon, France (J.-P.P.B.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
- Department of Internal Medicine (M.S.), CHU Dijon Bourgogne, France
- Department of Cardiovascular Surgery (A.L., E.S.), CHU Dijon Bourgogne, France
| | - Damien Denimal
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
| | - Maxime Samson
- Department of Internal Medicine (M.S.), CHU Dijon Bourgogne, France
| | - Aline Laubriet
- Department of Cardiovascular Surgery (A.L., E.S.), CHU Dijon Bourgogne, France
| | - Eric Steinmetz
- Department of Cardiovascular Surgery (A.L., E.S.), CHU Dijon Bourgogne, France
| | - Mickael Rialland
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- DiviOmics Platform, UMS58 BioSanD, INSERM, Université de Bourgogne Europe, Dijon, France (J.-P.P.B.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
- Department of Internal Medicine (M.S.), CHU Dijon Bourgogne, France
- Department of Cardiovascular Surgery (A.L., E.S.), CHU Dijon Bourgogne, France
| | - Léa Pierre
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- DiviOmics Platform, UMS58 BioSanD, INSERM, Université de Bourgogne Europe, Dijon, France (J.-P.P.B.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
- Department of Internal Medicine (M.S.), CHU Dijon Bourgogne, France
- Department of Cardiovascular Surgery (A.L., E.S.), CHU Dijon Bourgogne, France
| | - Emma Groetz
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- DiviOmics Platform, UMS58 BioSanD, INSERM, Université de Bourgogne Europe, Dijon, France (J.-P.P.B.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
- Department of Internal Medicine (M.S.), CHU Dijon Bourgogne, France
- Department of Cardiovascular Surgery (A.L., E.S.), CHU Dijon Bourgogne, France
| | - Jean-Paul Pais de Barros
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- DiviOmics Platform, UMS58 BioSanD, INSERM, Université de Bourgogne Europe, Dijon, France (J.-P.P.B.)
| | - Thomas Gautier
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
| | - Charles Thomas
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- LipSTIC LabEx, Université Bourgogne-Franche compté, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
| | - David Masson
- Center for Translational and Molecular Medecine (CTM), INSERM, UMR1231, Université Bourgogne Europe, Dijon, France (D.L., T.P., L.M., K.V.D., J.-P.P.B., T.G., C.T., D.M.)
- Department of Clinical Chemistry (D.L., D.D., D.M.), CHU Dijon Bourgogne, France
| |
Collapse
|
4
|
Zhao X, Xiao Y, Jiang M, Cao Y. Pharmacological and toxicological roles of Kruppel-like factors (KLFs) in the cardiovascular system: a review. Mol Biol Rep 2025; 52:506. [PMID: 40418318 DOI: 10.1007/s11033-025-10613-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
Kruppel-like factors (KLFs) are transcription factors (TFs) increasingly implicated in cardiovascular pharmacology and toxicology through molecular mechanisms regulating endothelial function, macrophage polarization, and lipid metabolism. For example, KLF2/4 maintains endothelial homeostasis by modulating endothelial nitric oxide synthase (eNOS) activity and oxidative stress, and KLF4 additionally regulates smooth muscle cell phenotypic switch. KLF6 governs macrophage polarization and pyroptosis, while KLF15 modulates cardiomyocyte lipid metabolism, with dysregulation linked to cardiomyopathy. Not surprisingly, drugs such as statins and phytochemicals, as well as toxicants like ox-LDL, nanomaterials, and radiation, alter KLF expression via non-coding RNA (such as microRNA) or TFs, influencing endothelial cell activation, vascular smooth muscle cell phenotypic switch, macrophage inflammation, and cardiomyocyte apoptosis. KLF-dependent pathways intersect with key toxicological processes, such as autophagy, ferroptosis, and lipid dysregulation, culminating in atherosclerosis and heart failure. Despite preclinical advances demonstrating KLFs as therapeutic targets, clinical translation remains limited, with no KLF-targeted agents in active trials. Future studies should delineate tissue-specific KLF interactions, resolve KLFs' conflicting roles, and explore CRISPR-based KLF-targeting modulation. Bridging molecular mechanisms, such as KLF's regulation of phenotypic transformation pathways in smooth muscle cells, to drug discovery could yield novel therapies for cardiovascular diseases. The present review underscores the need for mechanistic and translational research to harness KLFs in cardiovascular pharmacotherapy and toxicant risk assessment.
Collapse
Affiliation(s)
- Xiaomei Zhao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yangfan Xiao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Miao Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, 421001, China
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
5
|
Borràs C, Rotllan N, Griñán R, Santos D, Solé A, Dong C, Zhao Q, Llorente-Cortes V, Mourín M, Soto B, Camacho M, Tondo M, Canyelles M, Blanco-Vaca F, Escolà-Gil JC. Restoring cholesterol efflux in vascular smooth muscle cells transitioning into foam cells through Liver X receptor activation. Biomed Pharmacother 2025; 188:118178. [PMID: 40403381 DOI: 10.1016/j.biopha.2025.118178] [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: 01/29/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Macrophage foam cells derived from vascular smooth muscle cells (VSMCs) account for 30-70 % of foam cells in atherosclerotic lesions. Liver X receptor (LXR) agonists promote high-density lipoprotein (HDL)-mediated cholesterol efflux from macrophages. This study aimed to investigate the effects of LXR activation on the reverse cholesterol transport (RCT) rate from VSMCs to feces in vivo. Both human and mouse VSMCs exhibited similar levels of cholesterol efflux when exposed to serum and HDL. However, cholesterol efflux was significantly reduced following methyl-β-cyclodextrin (MBD)-cholesterol loading, while treatment with the LXR agonist T090137 markedly enhanced efflux. Radiolabeled foam-like VSMCs injected intraperitoneally into mice exhibited impaired cholesterol transfer to serum, HDL, and feces compared to non-lipid-laden VSMCs. Pre-treatment with the LXR agonist increased radiolabeled cholesterol levels in serum and HDL and doubled its fecal excretion. Furthermore, LXR activation restored RCT from MBD-cholesterol-loaded VSMCs to feces, reaching levels comparable to those of non-lipid-laden cells. Treatment with an acyl-coenzyme A: cholesterol acyltransferase (ACAT) inhibitor fully restored RCT rates in foam-like VSMCs, and the combination of the ACAT inhibitor and the LXR agonist further enhanced RCT. These findings indicate that HDL-mediated cholesterol efflux is significantly impaired during the transition of VSMCs into foam cells. Pharmacological activation of LXR enhances RCT from VSMCs to feces in vivo and restores the impaired RCT from transitioning VSMCs. The combination of LXR agonists and ACAT inhibitors holds promise as a synergistic therapeutic approach to restoring cholesterol homeostasis in lipid-laden VSMCs, offering potential strategies to mitigate atherosclerosis.
Collapse
Affiliation(s)
- Carla Borràs
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Noemí Rotllan
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Raquel Griñán
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David Santos
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Arnau Solé
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Chen Dong
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Qi Zhao
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vicenta Llorente-Cortes
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
| | - Marta Mourín
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Begoña Soto
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Servei d'Angiologia i Cirurgia Vascular i Endovascular, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Mercedes Camacho
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Mireia Tondo
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Servei de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marina Canyelles
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Servei de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | - Francisco Blanco-Vaca
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain; Servei de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | - Joan Carles Escolà-Gil
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain.
| |
Collapse
|
6
|
Wang Y, Dou W, Qian X, Chen H, Zhang Y, Yang L, Wu Y, Xu X. Advancements in the study of short-chain fatty acids and their therapeutic effects on atherosclerosis. Life Sci 2025; 369:123528. [PMID: 40049368 DOI: 10.1016/j.lfs.2025.123528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis (AS) remains a leading cause of cardiovascular disease and mortality globally. This chronic condition is characterized by inflammation, lipid accumulation, and the deposition of cellular components within arterial walls. Emerging evidence has highlighted the multifaceted therapeutic potential of short-chain fatty acids (SCFAs) in mitigating AS progression. SCFAs have demonstrated anti-inflammatory properties and the ability to regulate immune responses, metabolic pathways, vascular integrity, and intestinal barrier function in animal models of AS. Consequently, SCFAs have garnered significant attention as a promising approach for the prevention and treatment of AS. However, further clinical trials and studies are necessary to fully elucidate the underlying mechanisms and effects of SCFAs. Additionally, different types of SCFAs may exert distinct impacts, necessitating more in-depth investigation into their specific roles and mechanisms. This review provides an overview of the diverse cellular mechanisms contributing to AS formation, as well as a discussion of the significance of SCFAs in AS pathogenesis and their multifaceted therapeutic potential. Nonetheless, additional research is warranted to comprehensively understand and harness the potential of various SCFAs in the context of AS.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China; Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China; Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Wei Dou
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xin Qian
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Hao Chen
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Yi Zhang
- Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Liu Yang
- Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xiongfei Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
7
|
Kimura K, Motoyama E, Kanki S, Asano K, Sips P, Sheikh MAA, Clarin MTRDC, Raja E, Takeda M, Ishii R, Murata K, Deleeuw V, Muiño Mosquera L, De Backer J, Mizuno S, Sakai LY, Nakamura T, Yanagisawa H. Novel Aortic Dissection Model Links Endothelial Dysfunction and Immune Infiltration. Circ Res 2025. [PMID: 40365676 DOI: 10.1161/circresaha.125.326230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Aortic dissection (AD) is the separation of medial layers of the aorta and is a major cause of death in patients with connective tissue disorders such as Marfan syndrome. However, molecular triggers instigating AD, its temporospatial progression, and how vascular cells in each vessel layer interact and participate in the pathological process remain incompletely understood. To unravel the underlying molecular mechanisms of AD, we generated a spontaneous AD mouse model. METHODS We incorporated a novel missense variant (p.G234D) in FBN1, the gene for fibrillin-1, identified in a patient with nonsyndromic familial AD into mice using the CRISPR/Cas9 system. We performed molecular pathological analyses of the aortic lesions by histology, immunofluorescence staining, electron microscopy, synchrotron-based imaging, and single-cell RNA sequencing. Biochemical analysis was performed to examine the binding capacity of mutant human FBN1G234D protein to LTBPs (latent TGFβ [transforming growth factor-beta] binding proteins), and signaling pathways in the mutant aortic wall were examined by the Western blot analysis. RESULTS Fifty percent of the Fbn1G234D/G234D mutant mice died within 5 weeks of age from multiple intimomedial tears that expanded longitudinally and progressed to aortic rupture accompanied by massive immune cell infiltration. Fbn1G234D/G234D endothelial cells exhibited altered mechanosensing with loss of parallel alignment to blood flow and upregulation of VCAM-1 and ICAM-1 as early as 1 week of age. Single-cell RNA sequencing, validated by immunostaining, revealed a cluster of monocyte/macrophage predominantly in the intima at 3 weeks of age before the dissection, and the second cluster of macrophages increased during the progression of intimomedial tears, exhibiting strong CCR2+ and both M1- and M2-like features. Consistently, upregulation of MMP2/9 was observed. Biochemically, FBN1G234D lost the ability to bind to LTBP-1, -2, and -4, resulting in the downregulation of TGFβ signaling in the aortic wall. CONCLUSIONS We show that interactions involving endothelial cells and macrophages/monocytes in the intima, where the ECM (extracellular matrix) microenvironment contains reduced TGFβ signaling, contribute to the initiation of AD. Our novel AD mouse model provides a unique opportunity to identify target molecules involved in the intimomedial tears that can be utilized for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Kenichi Kimura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Eri Motoyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Sachiko Kanki
- Thoracic and Cardiovascular Surgery, Osaka Medical and Pharmaceutical University, Japan (S.K.)
| | - Keiichi Asano
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Patrick Sips
- Department of Biomolecular Medicine, Ghent University, Belgium (P.S., V.D.)
| | - Md Al Amin Sheikh
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- School of Integrative and Global Major, University of Tsukuba, Japan. (M.A.A.S., M.T.R.D.C.C.)
| | - Maria Thea Rane Dela Cruz Clarin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- School of Integrative and Global Major, University of Tsukuba, Japan. (M.A.A.S., M.T.R.D.C.C.)
- National Institute for Material Science, Tsukuba, Japan (M.T.R.D.C.C.)
| | - Erna Raja
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
| | - Mariko Takeda
- Department of Pharmacology, Kansai Medical University, Osaka, Japan (M.T., T.N.)
| | - Ryutaro Ishii
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- Faculty of Medicine, University of Tsukuba, Japan. (R.I., H.Y.)
| | - Kazuya Murata
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Japan. (K.M., S.M.)
| | - Violette Deleeuw
- Department of Biomolecular Medicine, Ghent University, Belgium (P.S., V.D.)
| | - Laura Muiño Mosquera
- Department of Pediatric Cardiology and Center for Medical Genetics, Ghent University Hospital, Belgium. (L.M.M.)
| | - Julie De Backer
- Department of Cardiology and Center for Medical Genetics, Ghent University Hospital, Belgium. (J.D.B.)
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Japan. (K.M., S.M.)
| | - Lynn Y Sakai
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland (L.Y.S.)
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Osaka, Japan (M.T., T.N.)
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Japan. (K.K., E.M., K.A., M.A.A.S., M.T.R.D.C.C., E.R., R.I., H.Y.)
- Faculty of Medicine, University of Tsukuba, Japan. (R.I., H.Y.)
| |
Collapse
|
8
|
Nikiforov NG, Chegodaev YS, Verkhova SS, Pudova EA, Popov MA, Tvorogova AV, Zhuravlev AD, Maslennikov RA, Snezhkina AV, Kudryavtseva AV, Yegorov YE, Omelchenko AV, Borodko DD, Zybin DI, Shumakov DV, Orekhov AN. Impaired LPS tolerance in monocytes of coronary atherosclerosis patients is associated with the intermediate subset. J Leukoc Biol 2025; 117:qiaf060. [PMID: 40350260 DOI: 10.1093/jleuko/qiaf060] [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: 01/24/2025] [Revised: 04/03/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025] Open
Abstract
Endotoxin tolerance in monocytes is a mechanism that reduces the secretion of inflammatory cytokines upon repeated pathogen exposure, thereby protecting tissues from hyperinflammation. Previously, we demonstrated that monocytes from patients with asymptomatic carotid atherosclerosis exhibit impaired LPS tolerance. In this study, we aimed to investigate monocyte tolerance impairments in coronary atherosclerosis in greater detail. The study included 46 male patients with ischemic heart disease, divided into two groups based on coronary angiography results with and without coronary atherosclerosis. CD14 + monocytes were isolated from patients' blood and subjected to LPS stimulation on days 1 and 7 of culture. Transcriptomic analysis of monocytes was conducted. Monocyte subpopulations were assessed and sorted based on CD14 and CD16 expression. Patients with coronary atherosclerosis exhibited disrupted inflammatory responses in monocytes, characterized by elevated basal and LPS-induced IL-1β secretion. These patients demonstrated impaired LPS tolerance, as evidenced by increased CCL2 secretion upon repeated stimulation. Transcriptomic analysis revealed upregulation of inflammatory genes, particularly those associated with minor CD16 + monocyte subpopulations. The proportions of non-classical and intermediate monocytes were elevated in patients with atherosclerosis, with IL-1β and CCL2 secretion levels correlating predominantly with the intermediate monocyte subset. Functional analysis revealed that non-classical monocytes from healthy donors developed stable endotoxin tolerance. In contrast, intermediate and classical monocytes from some donors exhibited a non-tolerant response to LPS, as evidenced by secretion of IL-1β, IL-6, and CCL2. The differentiation of classical monocytes into intermediate monocytes may play a key role in the impaired endotoxin tolerance observed in atherosclerosis.
Collapse
Affiliation(s)
- Nikita G Nikiforov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
- Laboratory of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova Street, 119991 Moscow, Russia
| | - Yegor S Chegodaev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Laboratory of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova Street, 119991 Moscow, Russia
| | - Svetlana S Verkhova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Elena A Pudova
- Laboratory of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova Street, 119991 Moscow, Russia
| | - Mikhail A Popov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2 Shchepkina Street, 129110 Moscow, Russia
| | - Anna V Tvorogova
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
| | - Alexander D Zhuravlev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Ruslan A Maslennikov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2 Shchepkina Street, 129110 Moscow, Russia
| | - Anastasiya V Snezhkina
- Laboratory of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova Street, 119991 Moscow, Russia
| | - Anna V Kudryavtseva
- Laboratory of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova Street, 119991 Moscow, Russia
| | - Yegor E Yegorov
- Laboratory of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova Street, 119991 Moscow, Russia
| | - Andrey V Omelchenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
| | - Daria D Borodko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
| | - Dmitry I Zybin
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2 Shchepkina Street, 129110 Moscow, Russia
| | - Dmitry V Shumakov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2 Shchepkina Street, 129110 Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
| |
Collapse
|
9
|
Wang T, Wang X, Ren W, Sun Z, Zhang Y, Wu N, Diao H. Cardiomyocyte proliferation: Advances and insights in macrophage-targeted therapy for myocardial injury. Genes Dis 2025; 12:101332. [PMID: 39935606 PMCID: PMC11810708 DOI: 10.1016/j.gendis.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/20/2024] [Indexed: 02/13/2025] Open
Abstract
In the mammalian heart, cardiomyocytes undergo a transient window of proliferation that leads to regenerative impairment, limiting cardiomyocyte proliferation and myocardial repair capacity. Cardiac developmental patterns exacerbate the progression of heart disease characterized by myocardial cell loss, ultimately leading to cardiac dysfunction and heart failure. Myocardial infarction causes the death of partial cardiomyocytes, which triggers an immune response to remove debris and restore tissue integrity. Interestingly, when transient myocardial injury triggers irreversible loss of cardiomyocytes, the subsequent macrophages responsible for proliferation and regeneration have a unique immune phenotype that promotes the formation of pre-existing new cardiomyocytes. During mammalian regeneration, mononuclear-derived macrophages and self-renewing resident cardiac macrophages provide multiple cytokines and molecular signals that create a regenerative environment and cellular plasticity capacity in postnatal cardiomyocytes, a pivotal strategy for achieving myocardial repair. Consistent with other human tissues, cardiac macrophages originating from the embryonic endothelium produce a hierarchy of contributions to monocyte recruitment and fate specification. In this review, we discuss the novel functions of macrophages in triggering cardiac regeneration and repair after myocardial infarction and provide recent advances and prospective insights into the phenotypic transformation and heterogeneous features involving cardiac macrophages. In conclusion, macrophages contribute critically to regeneration, repair, and remodeling, and are challenging targets for cardiovascular therapeutic interventions.
Collapse
Affiliation(s)
- Tao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Xueyao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Weibin Ren
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yanhui Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
| | - Hongyan Diao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
10
|
Lai Z, Kong D, Li Q, Wang Y, Li K, Duan X, Shao J, Xie Y, Chen J, Zhang T, Feng Y, Deng H, Wang J, Wang C, Shu K, Zhao H, Du H, Jia C, Dai H, Xie L, Liu J, Luo X, Wang L, Xu L, Zhu Z, Lei X, Wang Y, Yang Y, Liu Y, Liang Y, Yang Y, Xie J, Liu B, Deng Z, Liu X. Single-cell spatial transcriptomics of tertiary lymphoid organ-like structures in human atherosclerotic plaques. NATURE CARDIOVASCULAR RESEARCH 2025; 4:547-566. [PMID: 40295810 DOI: 10.1038/s44161-025-00639-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/20/2025] [Indexed: 04/30/2025]
Abstract
Tertiary lymphoid organs have been identified in the arterial adventitia in both mouse models of atherosclerosis and patients with atherosclerosis, yet their role in the disease remains insufficiently explored. Here we present a spatially resolved single-cell transcriptome atlas of human atherosclerotic plaques, identifying 14 distinct cell types and providing evidence of plaque tertiary lymphoid organs (PTLOs). The development of PTLOs was associated with the expression of lymphangiogenic chemokine genes and the adhesion molecule gene in fibroblast-like smooth muscle cells. PTLOs harbor abundant B cells with expanded and diversified B cell receptors, suggesting substantial immune involvement. We also observed that B cells may be exchanged between PTLOs and perivascular adipose tissues. The presence of PTLO-like structures correlates with cerebrovascular events, which may be mediated by PTLO-derived IgG antibodies enhancing macrophage functional activity. Our findings suggest the existence and characteristics of PTLOs in human atherosclerosis, elucidating their cellular functions and clinical implications and offering avenues for understanding, diagnosing and treating this condition.
Collapse
Affiliation(s)
- Zhichao Lai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Deqiang Kong
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Yue Wang
- BGI Research, Beijing, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kang Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaohan Duan
- BGI Research, Beijing, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Shao
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yiyun Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junye Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Tianjing Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuyao Feng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Jiaxian Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chaonan Wang
- Department of Hemangiomas and Vascular Malformations, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Keqiang Shu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongmei Zhao
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Huanyu Dai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lizhi Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Lin Wang
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leyin Xu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhan Zhu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangling Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuru Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yixuan Yang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Jun Xie
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | | | - Xin Liu
- BGI Research, Beijing, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
11
|
Stuttgen GM, Bobek J, Penoske R, Wadding-Lee C, Lam M, Hader SN, Owens AP, Sahoo D. FFAR4 Deficiency Increases Necrotic Cores in Advanced Lesions of ApoE -/- Mice-Brief Report. Arterioscler Thromb Vasc Biol 2025; 45:675-682. [PMID: 40047073 PMCID: PMC12018153 DOI: 10.1161/atvbaha.124.322371] [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: 12/20/2024] [Accepted: 02/20/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND FFAR4 (free fatty acid receptor 4) has emerged as a target for preventing cardiovascular disease through its ability to control macrophage inflammation and foam cell formation. Previous studies have shown that FFAR4 activation can protect against the accumulation of arterial plaque buildup in atherosclerotic animal models. The goal of our study is to test the hypothesis that FFAR4 deficiency will increase atherosclerotic plaque development in apoE-/- mice. METHODS Male and female apoE-/-/Ffar4-/- mice and their apoE-/- controls were fed a Western diet for 8 or 16 weeks to assess early and advanced atherosclerotic lesions, respectively. At the end of each study, atherosclerotic plaque severity was determined by analyzing the aortic sinus lesion area of the heart and the en face lesion area of the aortic arch. RESULTS Following 8 weeks of Western diet feeding, lesions from apoE-/-/Ffar4-/- male and female mice had 33% and 22% decreases, respectively, in the aortic sinus lesion area with no changes in the aortic arch lesion area. After 16 weeks of Western diet feeding, the lesions showed no changes in the area or volume of the aortic sinus between apoE-/-/Ffar4-/- mice and apoE-/- controls. However, male apoE-/-/Ffar4-/- mice had a 27% increase in the plaque lesion area in the aortic arch compared with apoE-/- controls. Despite similar sizes of lesions in the aortic sinus, apoE-/-/Ffar4-/- mice had larger necrotic cores compared with the apoE-/- control mice. In fact, male and female mice had 43% and 37% increases in the necrotic lesion area, respectively. CONCLUSIONS These data suggest a novel role for FFAR4 in reducing necrotic core lesion formation and support a protective role for FFAR4 in stabilizing atherosclerotic plaques.
Collapse
MESH Headings
- Animals
- Female
- Male
- Plaque, Atherosclerotic
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Necrosis
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Disease Models, Animal
- Mice, Knockout, ApoE
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Diet, Western
- Aorta, Thoracic/pathology
- Aorta, Thoracic/metabolism
- Mice, Inbred C57BL
- Apolipoproteins E/genetics
- Apolipoproteins E/deficiency
- Mice
- Sinus of Valsalva/pathology
- Sinus of Valsalva/metabolism
- Mice, Knockout
- Severity of Illness Index
Collapse
Affiliation(s)
- Gage M. Stuttgen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jordan Bobek
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Renee Penoske
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Medicine, Division of Endocrinology & Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Caris Wadding-Lee
- Department of Internal Medicine, Division of Cardiovascular Health & Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Lam
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shelby N. Hader
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - A. Phillip Owens
- Department of Internal Medicine, Division of Cardiovascular Health & Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | - Daisy Sahoo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Medicine, Division of Endocrinology & Molecular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
12
|
Karasawa T, Takahashi M. Inflammasome Activation and Neutrophil Extracellular Traps in Atherosclerosis. J Atheroscler Thromb 2025; 32:535-549. [PMID: 39828369 PMCID: PMC12055512 DOI: 10.5551/jat.rv22033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
The deposition of cholesterol containing cholesterol crystals and the infiltration of immune cells are features of atherosclerosis. Although the role of cholesterol crystals in the progression of atherosclerosis have long remained unclear, recent studies have clarified the involvement of cholesterol crystals in inflammatory responses. Cholesterol crystals activate the NLRP3 inflammasome, a molecular complex involved in the innate immune system. Activation of NLRP3 inflammasomes in macrophages cause pyroptosis, which is accompanied by the release of inflammatory cytokines such as IL-1β and IL-1α. Furthermore, NLRP3 inflammasome activation drives neutrophil infiltration into atherosclerotic plaques. Cholesterol crystals trigger NETosis against infiltrated neutrophils, a form of cell death characterized by the formation of neutrophil extracellular traps (NETs), which, in turn, prime macrophages to enhance inflammasome-mediated inflammatory responses. Colchicine, an anti-inflammatory drug effective in cardiovascular disease, is expected to inhibit cholesterol crystal-induced NLRP3 inflammasome activation and neutrophil infiltration. In this review, we illustrate the reinforcing cycle of inflammation that is amplified by inflammasome activation and NETosis.
Collapse
Affiliation(s)
- Tadayoshi Karasawa
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
13
|
Li Z, Zhang Z, Zhang B, Zhou C, Yu H, Xu L, He Z, Chen P, Peng W, Ye M, Qu G, Zhang X, Song Y, Jin X, Zheng Y. Perfluorinated compounds exposure and atherogenic risk characteristics in a high-fat diet condition: In vitro/in vivo models and population panel study. PNAS NEXUS 2025; 4:pgaf153. [PMID: 40386678 PMCID: PMC12084870 DOI: 10.1093/pnasnexus/pgaf153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Perfluorinated compounds (PFCs) are a well-recognized environmental risk factor for atherosclerosis. However, corresponding atherogenic risk in susceptible populations consuming high-fat diets (HFDs) remains unclear. Here, we found that perfluorooctane sulfonic acid (PFOS), a canonical PFCs, elevated the atherogenic risk in mice fed with HFD, which was characterized by an increased number of pro-inflammatory phenotype macrophages. We also found that macrophages exhibited a metabolic reprogramming to glycolysis, which was attributed to increased intracellular Fe2+ level. Mechanistic investigation revealed that PFOS directly bound to the iron-storage site on the ferritin heavy chain, subsequently weakening the iron-storage function. Notably, PFCs with acidic substituents and short chains had a higher atherogenic risk, as evidenced in the crucial indicators and observed in a population with a high triglyceride level. These findings highlight the potential atherogenic risk posed by PFCs exposure in susceptible populations consuming HFD and provide a potential intervention target.
Collapse
Affiliation(s)
- Ziyuan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Ze Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Biao Zhang
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chengying Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongyan Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Liting Xu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Zhicong He
- School of Water and Environment, Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Key Laboratory of Ecohydrology and Water Security in Arid and Semi-Arid Regions of Ministry of Water Resources, Chang’ an University, Xi’an 710054, China
| | - Pu Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Wei Peng
- Key Laboratory of Aerosol Chemistry and Physics, State Key Laboratory of Loess and Quaternary Geology, Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi’an 710061, China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Xiaoting Jin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yuxin Zheng
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
14
|
He J, Dai Y, Xu F, Huang X, Gao Y, Liu L, Zhang W, Liu J. High-density lipoprotein-based nanoplatforms for macrophage-targeted diagnosis and therapy of atherosclerosis. Int J Biol Macromol 2025; 306:140826. [PMID: 40010459 DOI: 10.1016/j.ijbiomac.2025.140826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/19/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Atherosclerosis, the primary cause of cardiovascular disease, which has the highest mortality worldwide, is a chronic inflammatory disease mainly induced by excessive lipid accumulation in plaque macrophages. Lipid-laden macrophages are crucial at all stages of atherosclerotic lesion progression and are, thus, regarded as popular therapeutic targets for atherosclerosis. High-density lipoprotein (HDL), an endogenous particle with excellent atherosclerotic plaque-homing properties, is considered a potential therapeutic agent for treating atherosclerosis. Based on the excellent properties of HDL, reconstituted HDL (rHDL), with physiological functions similar to those of its natural counterparts, have been successfully prepared as therapeutics and are also recognized as a potential nanoplatform for delivering drugs or contrast agents to atherosclerotic plaques owing to their high biocompatibility, amphiphilic structure, and macrophage-targeting capability. In this review, we focus on the (a) important role of macrophages in atherosclerotic lesions, (b) biological properties of rHDL as a delivery nanoplatform in atherosclerotic diseases, and (c) multiple applications of rHDL in the diagnosis and treatment of atherosclerosis. We systematically summarize the novel applications of rHDL with unique advantages in atherosclerosis, aiming to provide specific insights and inspire additional innovative research in this field.
Collapse
Affiliation(s)
- Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China; School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Fengfei Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xinya Huang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu Gao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Lisha Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
15
|
Zhang J, Xu Y, Han Z, Liu B, Wang M, Bao L, He Y. LACC1 Enhances Polyamine Immunometabolism in Inflammatory Macrophages to Inhibit Atherosclerosis Progression. J Cardiovasc Transl Res 2025:10.1007/s12265-024-10585-9. [PMID: 40293654 DOI: 10.1007/s12265-024-10585-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/25/2024] [Indexed: 04/30/2025]
Abstract
To explore the function and potential mechanism of laccase domain-containing 1 (LACC1) on atherosclerosis (AS). ApoE-/- mice feed with high-fat diet (HFD) were injected with adenovirus shLACC1 (Ad-shLACC1) or Ad-shNC via tail vein. LACC1 was highly expressed in macrophages of atherosclerotic plaque in ApoE-/- mice and ox-LDL-treated Raw264.7 macrophages. LACC1 silencing enhanced AS development and facilitated inflammation in mice. Then, we found that LACC1 silencing facilitated inflammation but repressed polyamine immunometabolism in ox-LDL-treated Raw264.7 macrophages. Through rescue experiments using ornithine or ODC1 inhibitor (DFMO), we further confirmed that LACC1 promoted polyamine immunometabolism to inhibit inflammation in ox-LDL-treated Raw264.7 macrophages. In addition, the observed LACC1 function was dependent on NOS2. In conclusion, we proved that the downregulation of LACC1 promoted AS progression via inhibiting polyamine immunometabolism in inflammatory macrophages, suggesting LACC1 may be a potential therapeutic target for AS.
Collapse
Affiliation(s)
- Jingyong Zhang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Yuan Xu
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zonglin Han
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Bingqi Liu
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Maohua Wang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Lili Bao
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Yuxiang He
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
16
|
Tao Z, Luo Z, Zou Z, Ye W, Hao Y, Li X, Zheng K, Wu J, Xia J, Zhao Y, Wang Y, Zhang X. Novel insights and an updated review of metabolic syndrome in immune-mediated organ transplant rejection. Front Immunol 2025; 16:1580369. [PMID: 40330480 PMCID: PMC12052740 DOI: 10.3389/fimmu.2025.1580369] [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: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic syndrome (MetS) is a group of symptoms that are characterized by abnormal changes in metabolic substances such as glucose, lipids, proteins, and bile acids. MetS is a common complication after organ transplantation and can further affect the survival and physiological function of the graft by reprograming the patient's immune environment. Additionally, MetS can influence the occurrence of post-transplant complications, such as infections. In recent years, research into the epidemiology and mechanisms of MetS has grown significantly. In this review, we summarize the mechanisms of MetS after transplantation and the mechanisms of hyperglycemia, insulin resistance, hyperlipidemia, abnormal bile acids, and abnormal amino acids on the body's immune cells as related to the effect of metabolic disorders on immune rejection after liver, kidney, heart, skin and other organ transplantation. Finally, we provide an overview of current treatment strategies and offer insights into potential future therapies for managing MetS in transplant recipients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yang Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Wiyono AV, Ardinal AP, Raharjo PP. Unraveling the significance of innate inflammation in vascular disease. Int Rev Immunol 2025:1-16. [PMID: 40255209 DOI: 10.1080/08830185.2025.2489346] [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: 04/15/2024] [Revised: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/22/2025]
Abstract
Atheroma formation is initiated by the activation of endothelial and smooth muscle cells, as well as immune cells, including neutrophils, lymphocytes, monocytes, macrophages, and dendritic cells. Monocytes, macrophages, and neutrophils are the innate immune cells that provide a rapid initial line of defence against vascular disease. These cells have a short lifespan and cannot retain memories, making them potential therapeutic targets for the inflammatory process associated with atherosclerosis. In addition, macrophages comprise the majority of vessel wall infiltrates and are, therefore, implicated in all stages of atherosclerosis progression. Neutrophils are the most common type of leukocyte found in circulation, and their high levels of matrix-degrading protease explain their significance in fibrous cap destabilization. However, the activation of immune cells becomes more complex by various microenvironmental stimuli and cytokines, which ultimately transform immune cells into their pro-inflammatory state. Different types of macrophage subsets with distinct functions in inflammation, such as M1 macrophages, cause an increase in pro-inflammatory cytokines and produce reactive oxygen species and nitric oxide, further worsening the disease. This review aims to shed light on immune-mediated inflammation in cardiovascular disease by focusing on the role of macrophage subsets in vascular inflammation and plaque stability, as well as the interaction between neutrophils and monocyte-macrophages.
Collapse
Affiliation(s)
- Alice Valeria Wiyono
- Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| | | | - Pradana Pratomo Raharjo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| |
Collapse
|
18
|
Wang X, Xie Z, Zhang J, Chen Y, Li Q, Yang Q, Chen X, Liu B, Xu S, Dong Y. Interaction between lipid metabolism and macrophage polarization in atherosclerosis. iScience 2025; 28:112168. [PMID: 40201117 PMCID: PMC11978336 DOI: 10.1016/j.isci.2025.112168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory condition associated with lipid deposition. The interaction between abnormal lipid metabolism and the inflammatory response has been identified as the underlying cause of AS. Lipid metabolism disorders are considered the basis of atherosclerotic lesion formation and macrophages are involved in the entire process of AS formation. Macrophages have a high degree of plasticity, and the change of their polarization direction can determine the progress or regression of AS. The disturbances in bioactive lipid metabolism affect the polarization of different phenotypes of macrophages, thus, affecting lipid metabolism and the expression of key signal factors. Therefore, understanding the interaction between lipid metabolism and macrophages as well as their key targets is important for preventing and treating AS and developing new drugs. Recent studies have shown that traditional Chinese medicines play a positive role in the prevention and treatment of AS, providing a basis for clinical individualized treatment.
Collapse
Affiliation(s)
- Xinge Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng Xie
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Chen
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xu Chen
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Bing Liu
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Dong
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
19
|
Gai X, Liu F, Chen Y, Zhang B, Zhang Y, Wu Y, Yang S, Chen L, Deng W, Wang Y, Wang S, Yu C, Du J, Zhang Z, Wang J, Zhang H. GOLM1 Promotes Atherogenesis by Activating Macrophage EGFR-ERK Signaling Cascade. Circ Res 2025; 136:848-861. [PMID: 40026146 DOI: 10.1161/circresaha.124.325880] [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: 11/16/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease. GOLM1 (Golgi membrane protein 1) is an inflammation-responsive protein and a mediator in some inflammation-associated pathological processes. Because we found a positive correlation between GOLM1 expression and atherosclerosis progression by checking the gene expression data set of human atherosclerotic lesions, we explored the potential significance of GOLM1 in atherosclerosis in this study. METHODS GOLM1 levels in serums and lesions of patients with atherosclerosis and mice with atherosclerosis were examined by immunostaining and ELISA. Gain-of-function and loss-of-function approaches were used to study the impacts of GOLM1 in inflammation and atherogenesis of Apoe-/- mice on a Western diet. The effects of GOLM1 on macrophage behaviors were determined by OxLDL (oxidized low-density lipoprotein) uptake assay, single-cell sequencing analysis, global phosphoproteomics analysis, and molecular biological techniques. The therapeutic potential of GOLM1 neutralization for atherosclerosis was evaluated in Apoe-/- mice. RESULTS GOLM1 was elevated in serums and lesions of patients with atherosclerosis and mice with atherosclerosis. Global deletion of GOLM1 ameliorated mouse inflammation and atherosclerosis, while knock-in of GOLM1 exacerbated these pathological manifestations. Furthermore, hepatic GOLM1 deletion reduced circulating GOLM1 and attenuated atherogenesis. Mechanistically, the expression and secretion of GOLM1 were induced in multiple mouse tissues by atherogenic stimulus, leading to the elevation of extracellular GOLM1. Extracellular GOLM1 then stimulated ERK (extracellular signal-regulated kinase) signaling cascade by binding to its putative receptor EGFR (epidermal growth factor receptor) to promote macrophage uptake of LDL (low-density lipoprotein) and enhance the corresponding macrophage immune response. Moreover, neutralizing GOLM1 by an antibody suppressed mouse inflammation and atherogenesis. CONCLUSIONS GOLM1 is an atherogenic mediator and a promising therapeutic target for the intervention of atherosclerotic diseases.
Collapse
Affiliation(s)
- Xiaochen Gai
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Fangming Liu
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Yixin Chen
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.C., S.W., C.Y.)
| | - Baohui Zhang
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning, China (B.Z.)
| | - Yinliang Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Y.Z.)
| | - Yuting Wu
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Shuhui Yang
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | | | - Weiwei Deng
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Beijing, China (Y. Wang, J.D.)
| | - Shuiyun Wang
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.C., S.W., C.Y.)
| | - Cuntao Yu
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.C., S.W., C.Y.)
| | - Jie Du
- Beijing Anzhen Hospital, Beijing, China (Y. Wang, J.D.)
| | - Zhengyi Zhang
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA (Z.Z.)
| | - Jing Wang
- Department of Pathophysiology (J.W.), Hebei University, Baoding, Hebei, China
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem (X.G., F.L., B.Z., Y. Wu, S.Y., W.D., H.Z.), Hebei University, Baoding, Hebei, China
- Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, College of Life Sciences, Hebei University, Baoding, Hebei, China (H.Z.)
| |
Collapse
|
20
|
Huang Z, Shen S, Li W, Wang M, Yang Y, Luo W, Han X, Xu Z, Min J, Long X, Huang W, Wu G, Wang Y, Liang G. Macrophage WEE1 Directly Binds to and Phosphorylates NF-κB p65 Subunit to Induce Inflammatory Response and Drive Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503192. [PMID: 40202104 DOI: 10.1002/advs.202503192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Atherosclerosis has an urgent need for new therapeutic targets. Protein kinases orchestrate multiple cellular events in atherosclerosis and may provide new therapeutic targets for atherosclerosis. Here, a protein kinase, WEE1 G2 checkpoint kinase (WEE1), promoting inflammation in atherosclerosis is identified. Kinase enrichment analysis and experimental evidences reveal macrophage WEE1 phosphorylation at S642 in human and mouse atherosclerotic tissues. RNA-seq analysis, combined with experiment studies using mutant WEE1 plasmids, shows that WEE1 phosphorylation, rather than WEE1 expression, mediated oxLDL-induced inflammation in macrophages. Macrophage-specific deletion of WEE1 or pharmacological inhibition of WEE1 kinase activity attenuates atherosclerosis by reducing inflammation in mice. Mechanistically, RNA-seq and co-immunoprecipitation followed by proteomics analysis are used to explore the mechanism and substrate of WEE1. p-WEE1 promoted inflammatory response through activating NF-κB shown and further revealed that WEE1 can directly bind to the p65 subunit. It is confirmed that p-WEE1 directly interacts with the RHD domain of p65 and phosphorylates p65 at S536, thereby facilitating subsequent NF-κB activation and inflammatory response in macrophages. The findings demonstrate that macrophage WEE1 drives NF-κB activation and atherosclerosis by directly phosphorylating p65 at S536. This study identifies WEE1 as a new upstream kinase of p65 and a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Zhuqi Huang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311000, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Sirui Shen
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Weixin Li
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mengyang Wang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China
| | - Yudie Yang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wu Luo
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China
| | - Zheng Xu
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China
| | - Julian Min
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaohong Long
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311000, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yi Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311000, China
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China
| |
Collapse
|
21
|
Majidiani H, Musavi M, Momtazi-Borojeni AA. New Roles of Artemisinins in Atherosclerosis Progression. Phytother Res 2025; 39:1847-1857. [PMID: 40200587 DOI: 10.1002/ptr.8483] [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: 04/25/2024] [Revised: 10/27/2024] [Accepted: 02/11/2025] [Indexed: 04/10/2025]
Abstract
Artemisinin is a natural compound derived from the Chinese plant Artemisia annua , which was officially approved by the FDA for its antimalarial effects. In recent years, a growing body of studies has shown the novel function of artemisinin in atherosclerosis therapy. In vivo studies have shown that artemisinin can inhibit the progression of atherosclerosis plaque. In the present review, the evidence showing the inhibitory effects of artemisinin on the progression of atherosclerosis plaque and its underlying mechanisms is discussed. Mechanistically, artemisinin and its derivatives act by modulating various atherosclerosis-mediating risk factors, including hyperlipidemia, inflammation, oxidative stress, and malfunctioning vascular smooth muscle cells (VSMCs). Notably, artesunate, but not artemisinin, can attenuate the plasma levels of TG, TC, VLDL-C, and LDL-c, along with a substantial decline in arterial lipid deposition through enhancing the LDPL activity via inducing the KFL2/NRF2/TCF7L2 axis. Artemisinin was found to ameliorate the atherosclerosis plaque inflammation by reducing monocyte adhesion and subsequent transmigration to the intima, via inhibiting the expression of ICAM-1 and VCAM-1, diminishing NLRP3 inflammasome activation, and reducing the expression of inflammatory factors such as IL-1β, IL-18, TNF-α, MCP-1, and TGF-β1 mechanistically and mainly via suppressing the by NF-κB activity. Artemisinin could exert antioxidant effects through activating the PI3K/Akt/eNOS signaling pathway and suppressing the ROS-mediated NF-κB signal pathway. Artemisinin could also improve the VSMC function in the atherosclerosis plaque. These findings can suggest artemisinin as a new therapeutic agent for treating atherosclerosis; however, future clinical trials are warranted to validate its therapeutic efficiency in patients with atherosclerosis.
Collapse
Affiliation(s)
- Hamidreza Majidiani
- Department of Basic Medical Sciences, Faculty of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Maryam Musavi
- Department of Medical Biotechnology, School of Medicine, Neyshabur University of Medical Sciences, Faculty of Medicine,Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Neyshabur University of Medical Sciences, Faculty of Medicine,Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
22
|
Zhang Y, Yan C, Dong Y, Zhao J, Yang X, Deng Y, Su L, Yin J, Zhang Y, Sun F, Feng Y. ANGPTL3 accelerates atherosclerotic progression via direct regulation of M1 macrophage activation in plaque. J Adv Res 2025; 70:125-138. [PMID: 38740260 PMCID: PMC11976407 DOI: 10.1016/j.jare.2024.05.011] [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: 03/01/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION The N-terminal domain of angiopoietin-like protein 3 (ANGPTL3) inhibits lipoprotein lipase activity. Its C-terminal fibrinogen-like (FBN) domain is a ligand of macrophage integrin αvβ3. OBJECTIVES ANGPTL3 might home to plaque where it directly regulates macrophage function via integrin αvβ3 for atherosclerosis progression. METHODS Ldlr-/- mice on a high-fat diet and ApoE-/- mice on a chow diet were received adeno-associated virus (AAV)-mediated Angptl3 gene transfer and followed up for 12 weeks. ApoE-/- mice were injected AAV containing FLAG-tagged Angptl3 cDNA for tracing. Atherosclerotic features were compared between Angptl3-/-ApoE-/- mice and ApoE-/- littermates. THP-1 cells were exposed to 0 or 50 μg/ml ANGPTL3 FBN domain for 24 h to evaluate Toll-like receptor (TLR)4 expression using western blot analysis and circulating cytokine and chemokine profiles by the MILLIPLEX MAP assay. Phospho-proteomic profile was established in ANGPTL3-treated macrophages. Integrin β3 deficient THP-1 cells were obtained by sgRNAs targeting RGD sequence using Lentivirus-Cas9 system. RESULTS Angptl3 overexpression increased atherosclerotic progression and CD68+ macrophages in plaque (p < 0.05 for all). By immunostaining, FLAG+ cells were identified in plaque of gene transferred ApoE-/- mice. Fluorescent immunostaining detected co-localisation of Angptl3 and CD68 in plaque macrophages. Phospho-proteomic analysis revealed that Angptl3 induced phosphorylation of proteins that were involved in the IL-17 signalling pathway in THP-1 cells. In vitro, ANGPTL3 treatment increased the production of interleukin (IL)-1β and tumour necrosis factor-α in THP-1 cells (p < 0.05 for both). Exposure of ANGPTL3 to THP-1 cells induced Akt phosphorylation which was weakened in integrin β3 deficient ones. ANGPTL3 elevated TLR4 expression via Akt phosphorylation. In response to lipopolysaccharide, nuclear factor-κB activity was 2.2-fold higher in THP-1 cells pre-treated with ANGPTL3 than in untreated cells (p < 0.05). CONCLUSIONS Targeting ANGPTL3 could yield a dual benefit of lowering lipid levels in the blood and suppressing macrophage activation in plaque.
Collapse
Affiliation(s)
- Yuejie Zhang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Cen Yan
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Yuan Dong
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Jiwei Zhao
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Xuanyi Yang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Yalan Deng
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Li Su
- Department of Science and Technology, Beijing Youan Hospital, Beijing 100069, China
| | - Jiming Yin
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China
| | - Yang Zhang
- Neuroscience Research Institute, Peking University Center of Medical and Health Analysis, Peking University, Beijing 100191, China
| | - Fenghui Sun
- Department of Neurology, Beijing Youan Hospital, Beijing 100069, China
| | - Yingmei Feng
- Beijing Institute of Hepatology, Beijing Youan Hospital, Beijing 100069, China; Department of Science and Technology, Beijing Youan Hospital, Beijing 100069, China.
| |
Collapse
|
23
|
Wacker BK, Bi L, Saenz-Pipaon G, Sanford N, Regan AZ, Lim NS, Liu L, Kim F, Dichek DA. Overexpression of ABCA1 in Carotid Endothelium of Hyperlipidemic Rabbits Modulates Vascular Inflammation. Hum Gene Ther 2025; 36:750-764. [PMID: 40111153 PMCID: PMC12042661 DOI: 10.1089/hum.2024.166] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/22/2025] Open
Abstract
Endothelial activation and dysfunction are key early steps in atherogenesis. Vascular gene therapy targeting endothelial inflammation and cholesterol accumulation could decrease atherosclerosis progression. ATP-binding cassette subfamily A member 1 (ABCA1) exhibits anti-inflammatory properties and promotes cholesterol efflux. A mouse model showed that systemic endothelial overexpression of ABCA1 decreased diet-induced atherosclerosis. To test if local ABCA1 endothelial overexpression protects against atherosclerosis, we used helper-dependent adenoviral vectors (HDAd) to express ABCA1 or a "Null" control in the carotid endothelium of hyperlipidemic rabbits. Both ABCA1 mRNA and endothelial protein were increased 3 days after vector infusion. After 24 weeks on a high-fat diet, laser-microdissected endothelium showed increased ABCA1 mRNA expression, but whole-vessel ABCA1 mRNA was decreased with HDAdABCA1. Endothelial ABCA1 protein could not be measured at 24 weeks, so its overexpression may be transient. CD68 expression was decreased (-23%, p < 0.001), but ITGAM (-15%, p = 0.3) was unchanged. Macrophage markers for both M1-like macrophages (IL1B: -44% [p = 0.02]; IL6: -40% [p = 0.02]; CCL2: -25% [p = 0.02]) and M2-like macrophages (ARG1: -27% [p = 0.03]; IL10: -23% [p = 0.09]; TGFB1: -13% [p < 0.001]) were also decreased. The inflammatory cytokines IL6 (-100%; p < 0.001) and TNF (p < 0.05) were significantly decreased in the laser-microdissected endothelium, but VCAM1 (+5%, p = 1.0) was unchanged and ICAM1 (+101%; p = 0.03) increased. Lesion size, intimal lipid, and intimal macrophage content were all unchanged (p > 0.5 for all), and vascular cholesterol measured by mass spectrometry (-11%; p = 0.9) also showed no difference. There was a small decrease in the intimal/medial ratio. scRNAseq revealed that vector transcripts were not restricted to endothelial cells after 24+ weeks but were detected in most cell types. The exception was modulated smooth muscle cells, which were found in substantial numbers in larger lesions. Overall, transient overexpression of ABCA1 in the vascular endothelium subtly alters the expression of inflammatory markers, providing only a modest atheroprotection.
Collapse
Affiliation(s)
- Bradley K. Wacker
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Lianxiang Bi
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Goren Saenz-Pipaon
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nicole Sanford
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Abigail Z. Regan
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Natalie S. Lim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Li Liu
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Francis Kim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - David A. Dichek
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
24
|
Lin M, Zhang C, Li H, Li K, Gou S, He X, Lv C, Gao K. Pyroptosis for osteoarthritis treatment: insights into cellular and molecular interactions inflammatory. Front Immunol 2025; 16:1556990. [PMID: 40236711 PMCID: PMC11996656 DOI: 10.3389/fimmu.2025.1556990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Osteoarthritis (OA) is a widely prevalent chronic degenerative disease often associated with significant pain and disability. It is characterized by the deterioration of cartilage and the extracellular matrix (ECM), synovial inflammation, and subchondral bone remodeling. Recent studies have highlighted pyroptosis-a form of programmed cell death triggered by the inflammasome-as a key factor in sustaining chronic inflammation. Central to this process are the inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18), which play crucial roles mediating intra-articular pyroptosis through the NOD-like receptor protein 3 (NLRP3) inflammasome. This paper investigates the role of the pyroptosis pathway in perpetuating chronic inflammatory diseases and its linkage with OA. Furthermore, it explores the mechanisms of pyroptosis, mediated by nuclear factor κB (NF-κB), the purinergic receptor P2X ligand-gated ion channel 7 (P2X7R), adenosine monophosphate (AMP)-activated protein kinase (AMPK), and hypoxia-inducible factor-1α (HIF-1α). Additionally, it examines the interactions among various cellular components in the context of OA. These insights indicate that targeting the regulation of pyroptosis presents a promising therapeutic approach for the prevention and treatment of OA, offering valuable theoretical perspectives for its effective management.
Collapse
Affiliation(s)
- Minghui Lin
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cunxin Zhang
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Haiming Li
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kang Li
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Shuao Gou
- Jining No.1 People's Hospital, affiliated with Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao He
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
- Medical Integration and Practice Center, Shandong University, Jinan, China
| | - Chaoliang Lv
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Kai Gao
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
25
|
Ping X, Liang X, Xing W, Wang S, Gong F, Cheng Y, Duan S, Lv X, Li X, Zhang T, Chen C, Zhang Y, Yuan C, Liu S, Liu G, Sun B. Deciphering single-cell landscape unravels cell-type-specific functional roles of RNA m 6A modification in atherosclerosis. Theranostics 2025; 15:4785-4807. [PMID: 40225569 PMCID: PMC11984397 DOI: 10.7150/thno.104179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Atherosclerosis is a chronic inflammatory disease that is the major cause of mortality worldwide. Although several studies have assessed the function of m6A (N6-methyladenosine) modification in atherosclerosis, its regulatory mechanism at the single-cell level remains unclear. This study provides a comprehensive single-cell atlas of m6A modification regulating cell-type-specific functions in atherosclerosis. Methods: We analyzed single-cell sequencing data derived from atherosclerosis patients to elucidate the influence of m6A modification on diverse cell types. We demonstrated the potential regulatory functions of m6A regulators across various cell types and key transcription factors involved. Furthermore, we discovered m6A regulators mediated intercellular communication in important biological processes. In vitro experiments were conducted to further investigate the effects of ALKBH5, WTAP and METTL3 on atherosclerosis. Results: ALKBH5 upregulated in endothelial cells induced cell proliferation and migration involved in sprouting angiogenesis. In smooth muscle cells, upregulation of WTAP enhanced proliferation, migration and phenotypic transformation. Upregulation of METTL3 and YTHDF2 promoted macrophage activation and differentiation. Furthermore, we identified abnormally activated transcription factors could regulate m6A regulators in a cell-type-specific manner. Moreover, we revealed that m6A regulators were implicated in dysregulated intercellular communication in atherosclerosis. And a series of experimental validations supported the conclusion that m6A regulators exert cell-type-specific regulatory functions. Conclusion: Our study provided evidence for the roles of ALKBH5, WTAP and METTL3 in orchestrating atherosclerotic cell-type-specific functions, representing promising targets for precision medicine.
Collapse
Affiliation(s)
- Xiaorui Ping
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoyun Liang
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, Hebei 050031, China
- Hebei Engineering Research Center of Intelligent Medical Clinical Application, Shijiazhuang, Hebei 050031, China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, Hebei 050031, China
| | - Wenlu Xing
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Saiqi Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fengcongzhe Gong
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yaqi Cheng
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Songqi Duan
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
- College of Food Science, Sichuan Agricultural University, Sichuan University, Chengdu 610000, China
| | - Xueqi Lv
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xueying Li
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Tianli Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chunxiao Chen
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuxin Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chengzhu Yuan
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shangyu Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, Hebei 050031, China
- Hebei Engineering Research Center of Intelligent Medical Clinical Application, Shijiazhuang, Hebei 050031, China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, Hebei 050031, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, Hebei 050031, China
- Hebei Engineering Research Center of Intelligent Medical Clinical Application, Shijiazhuang, Hebei 050031, China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, Hebei 050031, China
| | - Baofa Sun
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
26
|
Yang L, Li X, Ni L, Lin Y. Treatment of endothelial cell dysfunction in atherosclerosis: a new perspective integrating traditional and modern approaches. Front Physiol 2025; 16:1555118. [PMID: 40206381 PMCID: PMC11979162 DOI: 10.3389/fphys.2025.1555118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Atherosclerosis (AS), a prime causative factor in cardiovascular disease, originates from endothelial cell dysfunction (ECD). Comprising a vital part of the vascular endothelium, endothelial cells play a crucial role in maintaining vascular homeostasis, optimizing redox balance, and regulating inflammatory responses. More evidence shows that ECD not only serves as an early harbinger of AS but also exhibits a strong association with disease progression. In recent years, the treatment strategies for ECD have been continuously evolving, encompassing interventions ranging from lifestyle modifications to traditional pharmacotherapy aimed at reducing risk factors, which also have demonstrated the ability to improve endothelial cell function. Additionally, novel strategies such as promising biotherapy and gene therapy have drawn attention. These methods have demonstrated enormous potential and promising prospects in improving endothelial function and reversing AS. However, it is essential to remain cognizant that the current treatments still present significant challenges regarding therapeutic efficacy, long-term safety, and ethical issues. This article aims to provide a systematic review of these treatment methods, analyze the mechanisms and efficacy of various therapeutic strategies, with the goal of offering insights and guidance for clinical practice, and further advancing the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
27
|
Jha PK, Nakano T, Itto LYU, Barbeiro MC, Lupieri A, Aikawa E, Aikawa M. Vascular inflammation in chronic kidney disease: the role of uremic toxins in macrophage activation. Front Cardiovasc Med 2025; 12:1574489. [PMID: 40201789 PMCID: PMC11975941 DOI: 10.3389/fcvm.2025.1574489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition characterized by the gradual loss of kidney function, leading to the accumulation of uremic toxins in the bloodstream. These toxins play a pivotal role in mediating vascular inflammation, a key contributor to the high cardiovascular morbidity and mortality observed in CKD patients. This review article explores the intricate mechanisms by which uremic toxins accelerate vascular inflammation. Macrophages, as versatile immune cells, are central to the inflammatory response. Evidence suggests that the uremic milieu influences macrophage biology. In this review article, we focus on the signaling through which uremic toxins, particularly indoxyl sulfate-an independent risk factor for cardiovascular complications in CKD patients, modulate macrophage activation and function, and how these changes contribute to vascular inflammation, leading to the increased cardiovascular risk. Investigation of such mechanisms provide molecular bases for the development of new therapies that retard the development of cardiovascular disorders in CKD patients.
Collapse
Affiliation(s)
- Prabhash Kumar Jha
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Toshiaki Nakano
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Lucas Yuji Umesaki Itto
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Miguel Cantadori Barbeiro
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Adrien Lupieri
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Pei X, Cui F, Chen Y, Yang Z, Xie Z, Wen Y. miR-214-3p Promotes ox-LDL-Induced Macrophages Ferroptosis and Inflammation via GPX4. J Inflamm Res 2025; 18:3937-3950. [PMID: 40125091 PMCID: PMC11927573 DOI: 10.2147/jir.s507076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Atherosclerosis (AS) is a chronic inflammatory disease caused by the dysregulation of lipid metabolism. It has been established that oxidized low-density lipoprotein (ox-LDL)-induced macrophage inflammation and ferroptosis play important roles in AS. However, the mechanism by which ox-LDL induces inflammation in macrophages requires further investigation. Materials and Methods A foam cell model derived from ox-LDL-induced macrophages was constructed to study its mechanism of action. The levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were evaluated using an Enzyme-Linked Immunosorbent Assay (ELISA). Oil Red O staining was used to detect intracellular lipid accumulation levels. Lactate dehydrogenase (LDH), malondialdehyde (MDA), reactive oxygen species (ROS), and Fe2+ levels were assessed. Dual-luciferase and RNA-binding protein immunoprecipitation (RIP) experiments validated the binding relationship between microRNA (miR)-214-3p and glutathione peroxidase 4 (GPX4). Results The levels of IL-6, IL-1β, and TNF-α were significantly increased in ox-LDL-induced macrophages, accompanied by increased lipid accumulation, indicating the promotion of foam cell formation. Additionally, ox-LDL increased LDH, MDA, ROS, and Fe2+. The expression of miR-214-3p positively correlated with ox-LDL concentration in macrophages. Treatment with an miR-214-3p inhibitor reduces lipid accumulation, inflammatory responses, and ferroptosis in macrophages. Dual-luciferase and RIP experiments confirmed that miR-214-3p regulates GPX4 transcription. Silenced GPX4 reversed the inflammatory effects of the miR-214-3p inhibitor on ox-LDL-induced macrophages. Low GPX4 expression also increased lipid accumulation and ferroptosis in macrophages. Conclusion miR-214-3p promotes macrophage ferroptosis and inflammation induced by ox-LDL. This mechanism may be associated with miR-214-3p-induced GPX4 expression, which underscores the therapeutic significance of targeting macrophage inflammation and ferroptosis in addressing AS.
Collapse
Affiliation(s)
- Xueliang Pei
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Facai Cui
- Clinical Laboratory, Henan Provincial People’s Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Yu Chen
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Zhiyuan Yang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Zhouliang Xie
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Yongjin Wen
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
29
|
Zhang H, Lv B, Liu K, Du J, Jin H, Huang Y. Sulfur dioxide controls M1 macrophage polarization by sulphenylation of prolyl hydroxylase 2 at cysteine 260. Free Radic Biol Med 2025; 230:33-47. [PMID: 39892500 DOI: 10.1016/j.freeradbiomed.2025.01.054] [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: 10/25/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
M1 macrophage polarization plays a pivotal role in inflammation-related diseases. However, the endogenous regulatory factors and mechanisms underlying M1 macrophage polarization have not been entirely clarified. This study aimed to explore whether endogenous sulfur dioxide (SO2) is involved in M1 macrophage polarization and its mechanism. In the study, we found that the endogenous SO2/aspartate aminotransferase1 (AAT1) pathway was downregulated during M1 polarization of macrophages induced by lipopolysaccharide (LPS) stimulation, and supplementation with SO2 donors or AAT1 overexpression restored SO2 content, suppressed protein expression of inducible nitric oxide synthase, restrained mRNA level of M1 phenotype-related genes tumor necrosis factor α, interleukin-1β and interleukin-12β and decreased the CD86 expression. In addition, AAT1-knockdowned macrophages exhibited reduced level of hypoxia-inducible factor-1α (HIF-1α) hydroxylation, elevated HIF-1α protein level, and polarization into M1-type, while supplementation with SO2 reversed the above effects. Mechanistically, SO2 maintained prolyl hydroxylase (PHD) activity in a thiol-dependent manner. SO2 maintained PHD2 activity by sulphenylating PHD2 at Cys260, thereby reducing HIF-1α protein levels and subsequently inhibiting M1 macrophage polarization. Besides, SO2 enhanced PHD2 sulphenylation, inhibited M1 macrophage polarization, and alleviated lung damage in a mouse model of LPS-induced acute lung injury. These results suggested that downregulation of the endogenous SO2/AAT1 pathway was a pivotal mechanism for M1 macrophage polarization. SO2 maintained PHD2 activity via sulphenylation of Cys260, and promoted HIF-1α hydroxylation and degradation, thereby impeding M1 macrophage polarization.
Collapse
Affiliation(s)
- Han Zhang
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, 100034, Beijing, China
| | - Boyang Lv
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, 100034, Beijing, China
| | - Keyu Liu
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, 100034, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, 100034, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, 100034, Beijing, China; State Key Laboratory of Vascular Homeostasis and Remodeling, 100191, Peking University, Beijing, China.
| | - Yaqian Huang
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, 100034, Beijing, China; State Key Laboratory of Vascular Homeostasis and Remodeling, 100191, Peking University, Beijing, China.
| |
Collapse
|
30
|
Yutani C, Noda H, Iwa N, Komatsu S, Takahashi S, Higuchi Y, Kodama K. Hypothesis on the role of cholesterol crystals in spontaneously ruptured aortic plaques: Potential triggers for inflammation and systemic effects. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2025; 51:100507. [PMID: 39995516 PMCID: PMC11847121 DOI: 10.1016/j.ahjo.2025.100507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025]
Abstract
Cholesterol crystals (CCs) are a key component of atherosclerotic plaques and play a pivotal role in plaque progression, rupture, and the resulting inflammatory responses. CCs emboli trigger proinflammatory cytokines which can potentially lead to organ damage. Spontaneously ruptured aortic plaques (SRAPs) are frequently observed via non-obstructive general angioscopy (NOGA) in patients with or suspected coronary artery disease. The release of CCs from SRAPs can activate the innate immune system and induce neutrophil extracellular trap (NET) formation, further exacerbating inflammation. Inflammation levels in SRAPs vary, and the interleukin (IL)-6 ratio may reflect the degree of inflammation. Systemic inflammation induced by CCs may contribute to conditions that may lead to cerebral infarction, and chronic kidney disease. The effects of anti-inflammatory drugs, including IL-6 inhibitors, IL-1β inhibitors, and colchicine, may be evaluated by measuring the IL-6 ratio in SRAPs. This review examined innate immunity mechanisms associated with CCs in SRAPs sampled via NOGA and discussed their systemic impact and potential therapeutic strategies.
Collapse
Affiliation(s)
- Chikao Yutani
- Division of Pathology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
- Non-Profit Organization Japan Vascular Imaging Research Organization, Osaka, Japan
| | - Hirotaka Noda
- Department of Medical Technology, Morinomiya University of Medical Sciences, Osaka, Japan
- Division Health Sciences, Area of Medical Laboratory Science and Technology/Department of Clinical Laboratory and Biomedical Sciences, Molecular Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Nobuzo Iwa
- Division of Pathology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Sei Komatsu
- Non-Profit Organization Japan Vascular Imaging Research Organization, Osaka, Japan
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Satoru Takahashi
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Yoshiharu Higuchi
- Non-Profit Organization Japan Vascular Imaging Research Organization, Osaka, Japan
- Cardiovascular Division, Osaka Police Hospital, Osaka, Japan
| | - Kazuhisa Kodama
- Non-Profit Organization Japan Vascular Imaging Research Organization, Osaka, Japan
- Department of Cardiology, Cardiovascular Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| |
Collapse
|
31
|
Shi L, Sun T, Huo D, Geng L, Zhao C, Xia W. ETV5-Mediated Transcriptional Repression of DDIT4 Blocks Macrophage Pro-Inflammatory Activation in Diabetic Atherosclerosis. Cardiovasc Toxicol 2025; 25:379-394. [PMID: 39864045 DOI: 10.1007/s12012-024-09956-0] [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: 07/05/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025]
Abstract
Atherosclerosis risk is elevated in diabetic patients, but the underlying mechanism such as the involvement of macrophages remains unclear. Here, we investigated the underlying mechanism related to the pro-inflammatory activation of macrophages in the development of diabetic atherosclerosis. Bioinformatics tools were used to analyze the macrophage-related transcriptome differences in patients with atherosclerosis and diabetic mice. LDLR-/- mice with DDIT4 depletion were generated and fed a Western diet to induce atherosclerosis. DDIT4 expression was elevated in diabetic mice and patients with atherosclerosis. Macrophage proinflammatory factors F4/80, Il-6, and TNFα were reduced in DDIT4-/-LDLR-/- mice and necrotic areas were decreased in the aortic root. Atherosclerotic plaque stability was increased in DDIT4-/-LDLR-/- mice, as evidenced by increased collagen and smooth muscle cell content. DDIT4, regulated by ETV5, acted on macrophages, affecting lipid accumulation, migration capacity, and pro-inflammatory responses. Knockdown of ETV5 increased expression of DDIT4 and pro-inflammatory factors in macrophages, increased necrotic core area in the aortic root, and decreased stability of atherosclerotic plaques in mice, which was abated by DDIT4 knockdown. The findings provide new insight into how diabetes promotes atherosclerosis and support a model wherein loss of ETV5 sustains transcription of DDIT4 and the pro-inflammatory activation of macrophages.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Plaque, Atherosclerotic
- Macrophages/metabolism
- Macrophages/pathology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/deficiency
- Humans
- Mice, Knockout
- Disease Models, Animal
- Inflammation Mediators/metabolism
- Male
- Mice, Inbred C57BL
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Macrophage Activation
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/prevention & control
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
- Signal Transduction
- Aorta/pathology
- Aorta/metabolism
- Transcription, Genetic
- Cytokines/metabolism
- Female
- Mice
Collapse
Affiliation(s)
- Lili Shi
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Tingting Sun
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Di Huo
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Lin Geng
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Chao Zhao
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China
| | - Wenbo Xia
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Harbin, 150001, Heilongjiang, PR China.
| |
Collapse
|
32
|
Shen Z, Zhao M, Lu J, Chen H, Zhang Y, Chen S, Wang Z, Wang M, Liu X, Fu G, Huang H. Integrated multi-omic high-throughput strategies across-species identified potential key diagnostic, prognostic, and therapeutic targets for atherosclerosis under high glucose conditions. Mol Cell Biochem 2025; 480:1785-1805. [PMID: 39223351 DOI: 10.1007/s11010-024-05097-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Diabetes is a well-known risk factor for atherosclerosis (AS), but the underlying molecular mechanism remains unknown. The dysregulated immune response is an important reason. High glucose is proven to induce foam cell formation under lipidemia situations in clinical patients. Exploring the potential regulatory programs of accelerated foam cell formation stimulated by high glucose is meaningful. Macrophage-derived foam cells were induced in vitro, and high-throughput sequencing was performed. Coexpression gene modules were constructed using weighted gene co-expression network analysis (WGCNA). Highly related modules were identified. Hub genes were identified by multiple integrative strategies. The potential roles of selected genes were further validated in bulk-RNA and scRNA datasets of human plaques. By transfection of the siRNA, the role of the screened gene during foam cell formation was further explored. Two modules were found to be both positively related to high glucose and ox-LDL. Further enrichment analyses confirmed the association between the brown module and AS. The high correlation between the brown module and macrophages was identified and 4 hub genes (Aldoa, Creg1, Lgmn, and Pkm) were screened. Further validation in external bulk-RNA and scRNA revealed the potential diagnostic and therapeutic value of selected genes. In addition, the survival analysis confirmed the prognostic value of Aldoa while knocking down Aldoa expression alleviated the foam cell formation in vitro. We systematically investigated the synergetic effects of high glucose and ox-LDL during macrophage-derived foam cell formation and identified that ALDOA might be an important diagnostic, prognostic, and therapeutic target in these patients.
Collapse
Affiliation(s)
- Zhida Shen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Meng Zhao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Jiangting Lu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Huanhuan Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Yicheng Zhang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Songzan Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Zhaojing Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Xianglan Liu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China.
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, China.
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China.
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, China.
| |
Collapse
|
33
|
Zheng Z, Lu H, Wang X, Yang Z, Zhang Y, Li K, Shen C, Yin Z, Sha M, Ye J, Zhu L. Integrative analysis of genes provides insights into the molecular and immune characteristics of mitochondria-related genes in atherosclerosis. Genomics 2025; 117:111013. [PMID: 39914597 DOI: 10.1016/j.ygeno.2025.111013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by lipid accumulation in arterial walls. The role of the interplay between mitochondrial dysfunction and immune inflammation in atherosclerosis is still unclear. This study aimed to investigate the molecular characteristics and immune landscape of mitochondrial hub genes involved in atherosclerosis. Based on bioinformatics analysis, three hub Mitochondria-related DEGs (MitoDEGs), including OXCT1, UCP2, and CPT1B, were screened out and showed good diagnostic performance in identifying atherosclerosis patients and controls. Immune analysis demonstrated strong correlations between hub MitoDEGs and immune cells, such as macrophages and T cells. Additionally, the predicted transcription factors of these hub MitoDEGs were significantly enriched in Th17, Th1 and Th2 cell differentiation signaling pathways. Both cell and animal experiments confirmed the expression trends of OXCT1 and CPT1B observed in the bioinformatics analysis. These hub MitoDEGs may play an important role in coordinating mitochondrial metabolism in the immune inflammation of atherosclerosis.
Collapse
Affiliation(s)
- Zhipeng Zheng
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Huimin Lu
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Xiaowen Wang
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | | | - Yubin Zhang
- Dalian Medical University, Dalian 116000, China
| | - Kaiyuan Li
- Dalian Medical University, Dalian 116000, China
| | - Cheng Shen
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Zhifeng Yin
- Jiangsu Hanjiang Biotechnology Co., LTD, Taizhou 225300, China
| | - Min Sha
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China.
| | - Jun Ye
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China.
| | - Li Zhu
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China; Nanjing University of Chinese Medicine, Nanjing 210023, China; Dalian Medical University, Dalian 116000, China.
| |
Collapse
|
34
|
Huang JXF, Yousaf A, Moon J, Ahmed R, Uppal K, Pemminati S. Recent Advances in the Management of Dyslipidemia: A Systematic Review. Cureus 2025; 17:e81034. [PMID: 40264627 PMCID: PMC12013775 DOI: 10.7759/cureus.81034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2025] [Indexed: 04/24/2025] Open
Abstract
Dyslipidemia refers to abnormal levels of lipids in the bloodstream, typically exhibiting an increased pattern. Total cholesterol, high-density lipoprotein-cholesterol (HDL-C), low-density lipoprotein-cholesterol (LDL-C), and triglycerides (TGs) are all contributing factors to this disorder. This leads to an increased risk of atherosclerosis and cardiovascular diseases, such as coronary artery disease, which elevates the likelihood of morbidity. Dyslipidemia can be managed via the use of numerous classes of drugs and treatments. The conventional pharmacological agents comprising 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, selective cholesterol absorption inhibitors, proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), bile acid sequestrants, monoclonal antibodies, and nutritional supplementation, such as inhibitors of cholesterol synthesis and absorption, and promoters of LDL-C excretion, are also discussed. Furthermore, conventional pharmacological treatment of dyslipidemia may elicit a variety of adverse side effects that are detrimental to the quality of life of the user. These side effects include muscle pain, weakness, liver enzyme elevations, and hyperglycemia. This systematic review further analyzes the pharmacological actions of novel lipid-lowering agents such as adenosine triphosphate-citrate lyase inhibitors (ACLi), selective peroxisome proliferator-activated receptor alpha (PPARα) modulators, cholesteryl ester transfer protein inhibitors (CETPi), antisense oligonucleotides (ASO), and angiopoietin-like protein 3 inhibitors (ANGPTL3i) as well as their efficacy in treating dyslipidemia while sparing the user of potentially severe side effects. Compared to existing treatments, novel therapies have shown significantly greater effectiveness in managing dyslipidemia-related lipid profiles and exhibit fewer systemic adverse effects. Some of the recent therapies discussed are alternative treatments that offer patients promising efficacy and improved tolerability. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed to ensure a robust and transparent search process, aiming to minimize bias and maximize the retrieval of pertinent studies for review. Thus, this systematic review provides an overview of current and novel treatments for dyslipidemia, describing their efficacy, mechanism of action, safety, and side effects. As experimental investigations and clinical research progress, there is a possibility that a combination of newly tested medications and traditional ones may emerge as a promising treatment option for dyslipidemia in the future.
Collapse
Affiliation(s)
- Jacky Xiao Feng Huang
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Adil Yousaf
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Julie Moon
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Ramiz Ahmed
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Krishma Uppal
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| | - Sudhakar Pemminati
- Department of Biomedical Education, California Health Sciences University College of Osteopathic Medicine, Clovis, USA
| |
Collapse
|
35
|
Zhang Q, Zheng P, Pan Y, Zhou H, Fu Y, Jia E. Phosphoglycerate Mutase 5 Is Important Mediator for Instigating Arterial Lipid Accumulation and Aggravating Atherosclerosis. JACC Basic Transl Sci 2025. [DOI: 10.1016/j.jacbts.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
36
|
Bonacina F, Zhang X, Manel N, Yvan-Charvet L, Razani B, Norata GD. Lysosomes in the immunometabolic reprogramming of immune cells in atherosclerosis. Nat Rev Cardiol 2025; 22:149-164. [PMID: 39304748 PMCID: PMC11835540 DOI: 10.1038/s41569-024-01072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Lysosomes have a central role in the disposal of extracellular and intracellular cargo and also function as metabolic sensors and signalling platforms in the immunometabolic reprogramming of macrophages and other immune cells in atherosclerosis. Lysosomes can rapidly sense the presence of nutrients within immune cells, thereby switching from catabolism of extracellular material to the recycling of intracellular cargo. Such a fine-tuned degradative response supports the generation of metabolic building blocks through effectors such as mTORC1 or TFEB. By coupling nutrients to downstream signalling and metabolism, lysosomes serve as a crucial hub for cellular function in innate and adaptive immune cells. Lysosomal dysfunction is now recognized to be a hallmark of atherogenesis. Perturbations in nutrient-sensing and signalling have profound effects on the capacity of immune cells to handle cholesterol, perform phagocytosis and efferocytosis, and limit the activation of the inflammasome and other inflammatory pathways. Strategies to improve lysosomal function hold promise as novel modulators of the immunoinflammatory response associated with atherosclerosis. In this Review, we describe the crosstalk between lysosomal biology and immune cell function and polarization, with a particular focus on cellular immunometabolic reprogramming in the context of atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU), Oncoage, Nice, France
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Giuseppe D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
37
|
Jin M, Chen X, Zheng L, Peng Y, Lin M, Liang K, Liu X, Xu Z, Yang Y, Wei B, Wan J. Astaxanthin-loaded polylactic acid-glycolic acid nanoparticles alleviates atherosclerosis by suppressing macrophage ferroptosis via the NRF2/SLC7A11/GPX4 pathway. Arch Biochem Biophys 2025; 765:110316. [PMID: 39848420 DOI: 10.1016/j.abb.2025.110316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/11/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Astaxanthin (ASX), a fat-soluble carotenoid mainly sourced from Haematococcus pluvialis, shows promise for clinical applications in chronic inflammatory diseases. This study investigates whether ASX can mitigate atherosclerosis (AS) by modulating macrophage ferroptosis and provides astaxanthin-loaded polylactic acid-glycolic acid nanoparticles (ASX-PLGA NPs) as comparison. METHOD ApoE-/- mice were fed a high-fat diet with ASX or statin intervention. Plaque area, lipid aggregation, collagen content, and ferroptosis-related indicators were assessed. Moreover, ASX-PLGA NPs were synthesized and characterized and were used to pretreat macrophages induced with oxidized low-density lipoprotein (ox-LDL). Indicators linked to ferroptosis and oxidative stress were detected. Finally, the expression of nuclear factor erythroid -related factor 2 (NRF2) was evaluated. RESULTS ASX intervention significantly delayed the progression of AS plaques, characterized by reductions in plaque area and increased collagen fibers. The observed improvements in AS were consistent with statins. ASX-PLGA NPs demonstrate good safety and stability and have better therapeutic effects than ASX alone. Indicators linked to ferroptosis and oxidative stress were significantly improved in groups containing ASX in vivo and vitro. Additionally, ASX facilitated the nuclear translocation of NRF2, which could be attenuated with ML385, a specific inhibitor of NRF2. CONCLUSION ASX-PLGA NPs have better therapeutic effects than ASX alone. The regulation of NRF2/SLC7A11/GPX4 represents a novel mechanism by which ASX can counteract ferroptosis and impede AS progression.
Collapse
Affiliation(s)
- Mengying Jin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Xiao Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Lanzhuoying Zheng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Yuanyuan Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Mingying Lin
- Department of Cardiology, Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China.
| | - Ke Liang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Xinran Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Zihan Xu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Yiming Yang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Baozhu Wei
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| |
Collapse
|
38
|
Li H, Ye B, Tian J, Wang B, Zha Y, Zheng S, Ma T, Zhuang W, Park WS, Liang J. Monotropein resists atherosclerosis by reducing inflammation, oxidative stress, and abnormal proliferation and migration of vascular smooth muscle cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:245-255. [PMID: 39972674 PMCID: PMC11842295 DOI: 10.4196/kjpp.24.352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 02/21/2025]
Abstract
Monotropein is a compound classified into iridoid which is found in herbaceous plants Morindae officinalis. It possesses anti-inflammatory, antioxidant, and anti-osteoarthritic activities. Previous study indicates that monotropein may have the potential to combat cardiovascular disease, although the related mechanism remains unclear. In this study, we constructed the model of atherosclerosis by oxidized low density lipoprotein-induced vascular smooth muscle cells and LDLR-/- mice given high-fat diet to investigate the effects of monotropein on atherosclerosis. Our results showed that monotropein treatment significantly reduced the area of atherosclerotic plaques and necrotic cores in mice, inhibited the proliferation and migration of vascular smooth muscle cells, and reduced inflammatory responses and oxidative stress, which in turn alleviated atherosclerosis. In addition, we found that monotropein reduced the expression levels of P-NF-κB and P-AP-1. In conclusion, our data suggest that monotropein inhibited the proliferation and migration of vascular smooth muscle cells by mediating the activity of NF-κB, AP-1, reducing the level of inflammation and oxidative stress, and thus resisting the development of atherosclerosis. These findings demonstrate the efficacious therapeutic impact of monotropein on atherosclerosis and elucidate its specific target.
Collapse
Affiliation(s)
- Hongliang Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Bingqian Ye
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Jiping Tian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Bofan Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Yiwen Zha
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Shuying Zheng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Wenwen Zhuang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Won Sun Park
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225001, Jiangsu, China
| |
Collapse
|
39
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11443-11468. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
40
|
Zhang S, Ji Y, Xu B, Hu D, Zhang X, Song Y, Chen K, Wen Y, He X, Chen Y, Zheng T. Study on the use of black phosphorus quantum dots in the treatment of atherosclerosis. Aging (Albany NY) 2025; 17:563-587. [PMID: 39998897 PMCID: PMC11892921 DOI: 10.18632/aging.206205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/01/2025] [Indexed: 02/27/2025]
Abstract
Atherosclerosis is the pathological basis of cardiovascular disease, and there are no clinical drugs that can safely and efficiently remove atherosclerotic plaques. In this study, black phosphorus quantum dots (BPQDs) were applied to the treatment of atherosclerosis in high fat diet ApoE-/- model mice that BPQDs were given every other day for 3 weeks without changing the high-fat diet. 45.3% atherosclerotic plaque was cleared efficiently within 3 weeks in BPQDs intravenous administration way every other day. The treatment was more effective than traditional statins. The findings suggest that BPQDs have great potential to be applied for clinical prevention and treatment of AS that does not require dietary changes.
Collapse
Affiliation(s)
- Shengwei Zhang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
- Department of Ultrasound, Xiaolan People’s Hospital of Zhongshan, Zhongshan 528415, Guangdong, P.R. China
| | - Yiran Ji
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Bingxuan Xu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Die Hu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Xue Zhang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Yujian Song
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
- Ultrasound Diagnosis and Treatment Center of the First People’s Hospital of Foshan, Foshan 528000, Guangdong, P.R. China
| | - Keke Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
- Department of Ultrasound, Nanjing Drum Tower Hospital, Nanjing 210000, Jiangsu, P.R. China
| | - Yilin Wen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Xiaoxin He
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Yun Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Tingting Zheng
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| |
Collapse
|
41
|
Yu W, Zhao Y, Ilyas I, Wang L, Little PJ, Xu S. The natural polyphenol fisetin in atherosclerosis prevention: a mechanistic review. J Pharm Pharmacol 2025; 77:206-221. [PMID: 38733634 DOI: 10.1093/jpp/rgae053] [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: 10/09/2023] [Accepted: 04/22/2024] [Indexed: 05/13/2024]
Abstract
The incidence and mortality rate of atherosclerotic cardiovascular disease (ASCVD) is increasing yearly worldwide. Recently, a growing body of evidence has unveiled the anti-atherosclerotic properties of fisetin, a natural polyphenol compound. In this article, we reviewed the pharmacologic actions of fisetin on experimental atherosclerosis and its protective effects on disease-relevant cell types such as endothelial cells, macrophages, vascular smooth muscle cells, and platelets. Based on its profound cardiovascular actions, fisetin holds potential for clinical translation and could be developed as a potential therapeutic option for atherosclerosis and its related complications. Large-scale randomized clinical trials are warranted to ascertain the safety and efficacy of fisetin in patients with or high risk for ASCVD.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
- Anhui Renovo Pharmaceutical Co., Ltd, Hefei, Anhui, 230001, China
- Anhui Guozheng Pharmaceutical Co., Ltd, Hefei, Anhui, 230041, China
| | - Yaping Zhao
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Peter J Little
- Department of Pharmacy, Guangzhou Xinhua University, No. 721, Guangshan Road 1, Tianhe District, Guangzhou, 510520, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
42
|
Wei J, Peng MY, Lu HX. Functional transformation of macrophage mitochondria in cardiovascular diseases. Mol Cell Biochem 2025; 480:747-757. [PMID: 38884847 DOI: 10.1007/s11010-024-05049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024]
Abstract
Mitochondria are pivotal in the modulation of macrophage activation, differentiation, and survival. Furthermore, macrophages are instrumental in the onset and progression of cardiovascular diseases. Hence, it is imperative to investigate the role of mitochondria within macrophages in the context of cardiovascular disease. In this review, we provide an updated description of the origin and classification of cardiac macrophages and also focused on the relationship between macrophages and mitochondria in cardiovascular diseases with respect to (1) proinflammatory or anti-inflammatory macrophages, (2) macrophage apoptosis, (3) macrophage pyroptosis, and (4) macrophage efferocytosis. Clarifying the relationship between mitochondria and macrophages can aid the exploration of novel therapeutic strategies for cardiovascular disease.
Collapse
Affiliation(s)
- Jing Wei
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing, 211100, China
| | - Ming-Yu Peng
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing, 211100, China
| | - Hong-Xiang Lu
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing, 211100, China.
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing, 211100, China.
| |
Collapse
|
43
|
Yuan X, Shen G, Xiao H, Wang Z, Ma Y, Qin X. Netrin-1 and RGMa: Novel Regulators of Atherosclerosis-Related Diseases. Cardiovasc Drugs Ther 2025; 39:211-219. [PMID: 37439909 DOI: 10.1007/s10557-023-07478-5] [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] [Accepted: 05/26/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUNDS Neuronal guidance proteins (NGPs) have been demonstrated to guide the elongation of neuronal axonal growth cones in the developing central nervous system. Non-neuronal functions of NGPs have also been described, especially in relation to atherosclerosis. FINDINGS Netrin-1 and repulsive guidance molecule a (RGMa) are NGPs that have been shown to regulate endothelial cell adhesion and angiogenesis, macrophage migration and apoptosis, smooth muscle cells (SMCs) phenotypic dedifferentiation and mobility, chemokine activities, and inflammatory responses during atherosclerosis initiation and progression. PURPOSES However, mechanistic studies have generated controversy about the specific role of Netrin-1 in atherosclerosis due to the diversity of its structure, receptors and cell sources, and the actions of RGMa in atherosclerosis have not been reported in previous reviews. Therefore, the current work reviews the evidence for roles of Netrin-1 and RGMa in the initiation and progression of atherosclerosis and discusses potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Xiaofan Yuan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Guanru Shen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Hongmei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Zijie Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Yue Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Chongqing, Yuzhong District, China.
| |
Collapse
|
44
|
Liang X, Tian S, Zhang H, Sun S, Zhang P, Li J, Li Y, Zhang Y, Liu Z. Efferocytosis: A new star of atherosclerotic plaques reversal. Int Immunopharmacol 2025; 146:113904. [PMID: 39724733 DOI: 10.1016/j.intimp.2024.113904] [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: 03/21/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Efferocytosis is considered the key to eliminate apoptotic cells (ACs) under physiological and pathological conditions in vivo, mainly through different types of macrophages to achieve this process. Especially, tissue-resident macrophages (TRMs) are very significant for inflammation regression and maintenance of homeostasis in vivo. Abnormal efferocytosis will lead to the accumulation of ACs and the release of a variety of pro-inflammatory factors, which mediates the occurrence of many inflammatory diseases, including atherosclerosis (AS). AS is a chronic inflammatory vascular disease with the participation of the immune system. Defective efferocytosis will accelerate the progress of AS to a certain extent. Therefore, it is of great significance to understand the mechanism of efferocytosis and realize the prevention and treatment of AS through efferocytosis. In this review, we will briefly describe the specific process of efferocytosis, deeply discuss the possible molecular mechanism of impaired efferocytosis promoting the development of AS, and summarize the ways to prevent and treat AS through efferocytosis intervention therapy.
Collapse
Affiliation(s)
- Xiangyu Liang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Shuoqi Tian
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Han Zhang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Shusen Sun
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Peixiang Zhang
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Jiameng Li
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| | - Yong Li
- Beijing Yongkang Nian Health Technology Co., Ltd., Beijing, China.
| | - Yanfen Zhang
- Technology Transfer Center, Hebei University, Baoding, China.
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Baoding, China.
| |
Collapse
|
45
|
Wang Z, Li X, Moura AK, Hu JZ, Wang YT, Zhang Y. Lysosome Functions in Atherosclerosis: A Potential Therapeutic Target. Cells 2025; 14:183. [PMID: 39936975 PMCID: PMC11816498 DOI: 10.3390/cells14030183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Lysosomes in mammalian cells are recognized as key digestive organelles, containing a variety of hydrolytic enzymes that enable the processing of both endogenous and exogenous substrates. These organelles digest various macromolecules and recycle them through the autophagy-lysosomal system. Recent research has expanded our understanding of lysosomes, identifying them not only as centers of degradation but also as crucial regulators of nutrient sensing, immunity, secretion, and other vital cellular functions. The lysosomal pathway plays a significant role in vascular regulation and is implicated in diseases such as atherosclerosis. During atherosclerotic plaque formation, macrophages initially engulf large quantities of lipoproteins, triggering pathogenic responses that include lysosomal dysfunction, foam cell formation, and subsequent atherosclerosis development. Lysosomal dysfunction, along with the inefficient degradation of apoptotic cells and the accumulation of modified low-density lipoproteins, negatively impacts atherosclerotic lesion progression. Recent studies have highlighted that lysosomal dysfunction contributes critically to atherosclerosis in a cell- and stage-specific manner. In this review, we discuss the mechanisms of lysosomal biogenesis and its regulatory role in atherosclerotic lesions. Based on these lysosomal functions, we propose that targeting lysosomes could offer a novel therapeutic approach for atherosclerosis, shedding light on the connection between lysosomal dysfunction and disease progression while offering new insights into potential anti-atherosclerotic strategies.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| |
Collapse
|
46
|
Liu Z, Liu J, Wang X, Zhang Y, Ma Y, Guan G, Yuwen Y, He N, Liu H, Yu X, Ma S, Wang J, Zhang J, Zhu L, Zhang Y. N ε-carboxyethyl-lysin influences atherosclerotic plaque stability through ZKSCAN3 acetylation-regulated macrophage autophagy via the RAGE/LKB1/AMPK1/SIRT1 pathway. Cardiovasc Diabetol 2025; 24:36. [PMID: 39844245 PMCID: PMC11755919 DOI: 10.1186/s12933-025-02586-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory condition characterized by plaque formation, often leads to instability, particularly under Type 2 diabetes mellitus (T2DM) conditions, which exacerbate cardiovascular risks. However, the molecular mechanisms underlying this process remain incompletely understood. In this study, we investigated the correlation between acute coronary syndrome (ACS) and serum levels of Nε-carboxyethyl-lysin (CEL), a prominent advanced glycation end product (AGE) elevated in T2DM, in a cohort of 225 patients with coronary artery disease. Using a murine model of atherosclerosis complicated by T2DM, we examined the effects of CEL on plaque stability and macrophage autophagy. Our findings revealed that elevated serum CEL levels are independently associated with increased ACS incidence. Metabolomic profiling identified CEL as a key AGE contributing to plaque instability in diabetic conditions. Mechanistically, CEL disrupted macrophage autophagy and plaque stability by perturbing the Receptor for Advanced Glycation End products (RAGE)/Liver Kinase B1 (LKB1)/AMP-activated Protein Kinase 1 (AMPK1)/Sirtuin 1 (SIRT1) signaling cascade. This pathway further regulated autophagic activity through SIRT1-mediated acetylation of Zinc Finger with KRAB and SCAN Domains 3 (ZKSCAN3). These findings highlight CEL's critical role in promoting plaque instability in T2DM by impairing key molecular pathways that regulate autophagy, offering potential therapeutic targets for managing atherosclerosis in diabetic patients.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Department of Technology Transfer and Management, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
- School of Medicine, Xizangminzu University, Xianyang, 712082, China
- School of Medicine, Yan'an University, Yan'an, 716000, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Jing Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Xiqiang Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Yanpeng Ma
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Gongchang Guan
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Ya Yuwen
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
- School of Medicine, Xizangminzu University, Xianyang, 712082, China
| | - Ni He
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Hanxiu Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Department of Technology Transfer and Management, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Xingfeng Yu
- Department of Nursing, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Sen Ma
- Department of Technology Transfer and Management, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China.
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China.
| | - Jin Zhang
- Department of Otolaryngology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
| | - Ling Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China.
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China.
| | - Yulian Zhang
- The Director's Office, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
| |
Collapse
|
47
|
Ansari A, Yadav PK, Zhou L, Prakash B, Ijaz L, Christiano A, Ahmad S, Rimbert A, Hussain MM. Casz1 and Znf101/Zfp961 differentially regulate apolipoproteins A1 and B, alter plasma lipoproteins, and reduce atherosclerosis. JCI Insight 2025; 10:e182260. [PMID: 39782688 PMCID: PMC11721306 DOI: 10.1172/jci.insight.182260] [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: 04/19/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
High apolipoprotein B-containing (apoB-containing) low-density lipoproteins (LDLs) and low apoA1-containing high-density lipoproteins (HDLs) are associated with atherosclerotic cardiovascular diseases. In search of a molecular regulator that could simultaneously and reciprocally control both LDL and HDL levels, we screened a microRNA (miR) library using human hepatoma Huh-7 cells. We identified miR-541-3p that both significantly decreases apoB and increases apoA1 expression by inducing mRNA degradation of 2 different transcription factors, Znf101 and Casz1. We found that Znf101 enhances apoB expression, while Casz1 represses apoA1 expression. The hepatic knockdown of Casz1 in mice increased plasma apoA1, HDL, and cholesterol efflux capacity. The hepatic knockdown of Zfp961, an ortholog of Znf101, reduced lipogenesis and production of triglyceride-rich lipoproteins and atherosclerosis, without causing hepatic lipid accumulation. This study identifies hepatic Znf101/Zfp961 and Casz1 as potential therapeutic targets to alter plasma lipoproteins and reduce atherosclerosis without causing liver steatosis.
Collapse
Affiliation(s)
- Abulaish Ansari
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Liye Zhou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Binu Prakash
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Laraib Ijaz
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Amanda Christiano
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Sameer Ahmad
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
- VA New York Harbor Healthcare System, Brooklyn, New York, USA
| |
Collapse
|
48
|
Shimizu Y, Kawashiri SY, Yamanashi H, Nakamichi S, Hayashida N, Nagata Y, Maeda T. Beneficial influence of low-density lipoprotein cholesterol on the endothelium in relation to endothelial repair. Environ Health Prev Med 2025; 30:24. [PMID: 40189259 PMCID: PMC11986262 DOI: 10.1265/ehpm.24-00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDLc) is regarded as a risk factor for endothelial dysfunction. However, LDLc stimulates the proliferation of hematopoietic stem cells (CD34-positive cells), which contribute to endothelial repair. Therefore, LDLc may have a beneficial influence on the endothelium of individuals with lower endothelial repair activity. METHODS This cross-sectional study included 245 men aged 60-69 years. Endothelial repair activity was categorized by the circulating levels of CD34-positive cells based on median values. The status of endothelium was evaluated using the cardio-ankle vascular index (CAVI). RESULTS Among individuals with low levels of circulating CD34-positive cells, LDL-c levels were significantly inversely correlated with CAVI and positively correlated with circulating CD34-positive cells. No significant correlations were observed among the participants with high levels of circulating CD34-positive cells. Among low levels of CD34-positive cells, the adjusted standardized parameter (β) and p value were -0.24 (p = 0.021) for CAVI and 0.41 (p < 0.001) for CD34-positive cells, whereas among high levels of CD34-positive cells, the corresponding values were 0.03 (p = 0.738) and -0.09 (p = 0.355). CONCLUSION LDLc has a beneficial influence on endothelial health among individuals with low endothelial repair activity, possibly by stimulating the proliferation of hematopoietic stem cells.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Seiko Nakamichi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naomi Hayashida
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Division of Strategic Collaborative Research, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Yasuhiro Nagata
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Islands and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
49
|
Bamgbose TT, Schilke RM, Igiehon OO, Nkadi EH, Binwal M, Custis D, Bharrhan S, Schwarz B, Bohrnsen E, Bosio CM, Scott RS, Yurdagul Jr. A, Finck BN, Woolard MD. Lipin-1 restrains macrophage lipid synthesis to promote inflammation resolution. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:85-103. [PMID: 40073265 PMCID: PMC11844145 DOI: 10.1093/jimmun/vkae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/24/2024] [Indexed: 03/14/2025]
Abstract
Macrophages are critical to maintaining and restoring tissue homeostasis during inflammation. The lipid metabolic state of macrophages influences their function and polarization, which is crucial to the resolution of inflammation. The contribution of lipid synthesis to proinflammatory macrophage responses is well understood. However, how lipid synthesis regulates proresolving macrophage responses needs to be better understood. Lipin-1 is a phosphatidic acid phosphatase with a transcriptional coregulatory activity that regulates lipid metabolism. We previously demonstrated that lipin-1 supports proresolving macrophage responses, and here, myeloid-associated lipin-1 is required for inflammation resolution, yet how lipin-1-regulated cellular mechanisms promote macrophage proresolution responses is unknown. We demonstrated that the loss of lipin-1 in macrophages led to increased free fatty acid, neutral lipid, and ceramide content and increased phosphorylation of acetyl-CoA carboxylase. The inhibition of the first step of lipid synthesis, the transport of citrate from the mitochondria, reduced lipid content and restored efferocytosis and inflammation resolution in lipin-1mKO mice and macrophages. Our findings suggest macrophage-associated lipin-1 restrains lipid synthesis, promoting proresolving macrophage function in response to proresolving stimuli.
Collapse
Affiliation(s)
- Temitayo T Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Oluwakemi O Igiehon
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ebubechukwu H Nkadi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Monika Binwal
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - David Custis
- Research Core Facility, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sushma Bharrhan
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Benjamin Schwarz
- Proteins and Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Eric Bohrnsen
- Proteins and Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Rona S Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Arif Yurdagul Jr.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Brian N Finck
- Division of Nutritional Sciences and Obesity Medicine, Washington University School of Medicine in St. Louis, St Louis, MO, United States
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
50
|
Wehbe Z, Wehbe M, Al Khatib A, Dakroub AH, Pintus G, Kobeissy F, Eid AH. Emerging understandings of the role of exosomes in atherosclerosis. J Cell Physiol 2025; 240:e31454. [PMID: 39370679 PMCID: PMC11730360 DOI: 10.1002/jcp.31454] [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: 01/23/2024] [Revised: 08/20/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Atherosclerosis remains a major contributor to cardiovascular disease, the leading cause of global morbidity and mortality. Despite the elucidation of several molecular, biochemical, and cellular aspects that contribute to the etio-pathogenesis of atherosclerosis, much remains to be understood about the onset and progression of this disease. Emerging evidence supports a role for exosomes in the cellular basis of atherosclerosis. Indeed, exosomes of activated monocytes seem to accentuate a positive feedback loop that promotes recruitment of pro-inflammatory leukocytes. Moreover, in addition to their role in promoting proliferation and invasion of vascular smooth muscle cells, exosomes can also induce neovascularization within lesions and increase endothelial permeability, two important features of fibrous plaques. Depending on their sources and cargo, exosomes can also induce clot formation and contribute to other hallmarks of atherosclerosis. Taken together, it is becoming increasingly evident that a better understanding of exosome biology is integral to elucidating the pathogenesis of atherosclerosis, and may thus provide insight into a potentially new therapeutic target for this disease.
Collapse
Affiliation(s)
- Zena Wehbe
- Vascular Biology Research Centre, Molecular and Clinical Research InstituteSt. George's University of LondonLondonUnited Kingdom
| | - Maya Wehbe
- Oxford University HospitalsOxfordUnited Kingdom
| | - Ali Al Khatib
- Department of Nutrition and Food SciencesLebanese International UniversityBeirutLebanon
| | - Ali H. Dakroub
- Departments of Medicine (Cardiology) and Population Health Science and Policy, Blavatnik Family Research InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Gianfranco Pintus
- Department of Biomedical SciencesUniversity of Sassari, Viale San PietroSassari07100Italy
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of MedicineCenter for Neurotrauma, Multiomics & Biomarkers (CNMB)AtlantaGAUSA
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of MedicineQU Health, Qatar UniversityDohaP.O. Box 2713Qatar
| |
Collapse
|