1
|
Li M, Wu L, Wen Y, Wang A, Zhou X, Ren L, Lu Q, Li F, Zhu L, Tang C. Dysregulated cholesterol uptake and efflux of bone marrow-derived α-SMA + macrophages contribute to atherosclerotic plaque formation. Cell Mol Life Sci 2025; 82:134. [PMID: 40159437 PMCID: PMC11955436 DOI: 10.1007/s00018-025-05655-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/15/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Macrophages play differential roles in the pathogenesis of atherosclerosis due to their different phenotypes. Although α-SMA+ macrophages have been found to present in bone marrow and atherosclerotic plaques, their role in atherosclerosis remains unclear. By performing partial carotid ligation (PCL) on monocyte/macrophage lineage-tracked mice, we observed bone marrow-derived α-SMA+ macrophages in the subendothelium and atherosclerotic plaques under disturbed flow conditions. The functional role of α-SMA+ macrophages in atherosclerotic plaque formation was examined using macrophage-specific Acta2 knockout (Acta2MKO) mice generated by crossing Acta2f/f transgenic mice with LysM-Cre mice. The size of the aortic plaques was 77.43% smaller in Acta2MKO mice than in Acta2f/f mice following adeno-associated virus-mutant PCSK9 injection and high-fat diet (HFD) feeding for 12 weeks. A significant reduction in lipid deposition, macrophage infiltration and the α-SMA+ area was observed in the aortic roots of Acta2MKO mice compared with Acta2f/f mice. Mechanistically, using Acta2-overexpressing Raw264.7 cells (Acta2hi cells) and bone marrow-derived macrophages (BMDMs) from Acta2MKO mice (Acta2MKO BMDMs), we showed that macrophage α-SMA increased the expression of the scavenger receptor SR-A, induced Ox-LDL binding and uptake, and reduced the level of the cholesterol transporter ABCA1, potentially via the AKT pathway. Together, our results indicate that bone marrow-derived α-SMA+ macrophages contribute to atherosclerotic plaque formation due to dysregulated cholesterol uptake and efflux, providing potential targets for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Menglu Li
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Lili Wu
- Laboratories of Thrombosis and Vascular Biology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Yuxin Wen
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Anni Wang
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Xiao Zhou
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Lijie Ren
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Qiongyu Lu
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China
| | - Fengchan Li
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China.
| | - Li Zhu
- Laboratories of Thrombosis and Vascular Biology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China.
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, Jiangsu, China.
- JinFeng Laboratory, Chongqing, China.
| | - Chaojun Tang
- Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiangsu, China.
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou, Jiangsu, China.
- Department of Cardiology, The First People'S Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Jiangsu, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
2
|
Halmos B, La Rose AM, Methorst D, Groenen AG, Nakládal D, Bazioti V, Koster MH, Kloosterhuis NJ, van Buiten A, Schouten EM, Huijkman NC, Langelaar-Makkinje M, Bongiovanni L, De Neck SM, de Bruin A, Buikema H, Deelman LE, van den Heuvel MC, Kuipers F, de Jong IJ, Sluimer JC, Jørgensen HF, Henning RH, Westerterp M. SMC Abca1 and Abcg1 Deficiency Enhances Urinary Bladder Distension but Not Atherosclerosis. Circ Res 2025; 136:491-507. [PMID: 39931819 PMCID: PMC11867804 DOI: 10.1161/circresaha.124.325103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Smooth muscle cells (SMCs) regulate blood flow distribution via vasoconstriction mediated by α-ARs (α-adrenergic receptors). Plasma membrane cholesterol accumulation affects α1-AR signaling and promotes loss of SMC contractile markers in vitro. ABCA1 and ABCG1 (ATP-binding cassette transporter A1 and G1) mediate cholesterol efflux to HDL (high-density lipoprotein). ABCA1/ABCG1 show high expression in medial and low expression in intimal SMCs of atherosclerotic plaques. The role of ABCA1 and ABCG1 in SMC-mediated vasoconstriction and atherogenesis remains poorly understood. METHODS We generated mice with SMC-specific Abca1/Abcg1 deficiency on the low-density lipoprotein receptor-deficient (Ldlr-/-) background by crossbreeding Abca1fl/flAbcg1fl/flLdlr-/- mice with Myh11CreERT2 transgenic mice. To induce SMC cholesterol accumulation and atherogenesis, we fed Myh11CreERT2Abca1fl/flAbcg1fl/flLdlr-/-, Myh11CreERT2Abca1fl/flLdlr-/-, Myh11CreERT2Abcg1fl/flLdlr-/-, and Myh11CreERT2Ldlr-/- mice Western-type diet for 16 weeks. RESULTS Combined SMC-Abca1/Abcg1 deficiency increased vasoconstriction in aortic rings induced by the α1-AR agonist phenylephrine. Unexpectedly, SMC-Abca1/Abcg1 deficiency induced urinary bladder distension by >20-fold. This was reversed by the α1-AR antagonist tamsulosin, indicating its dependence on bladder neck SMC constriction. Moreover, SMC-Abca1/Abcg1 deficiency decreased contractile markers and increased macrophage and fibroblast markers in bladder SMCs, indicating SMC transdifferentiation. This was accompanied by free cholesterol accumulation and increased endoplasmic reticulum stress. SMC-Abca1/Abcg1 deficiency did not induce thoracic aorta SMC transdifferentiation, presumably due to increased cholesteryl ester accumulation and no endoplasmic reticulum stress in thoracic aorta SMCs. Surprisingly, SMC-Abca1/Abcg1 deficiency did not affect atherosclerotic lesion size or composition in the aortic root or brachiocephalic artery. CONCLUSIONS We uncover a new role of SMC cholesterol efflux pathways in suppressing α1-AR-mediated vasoconstriction and bladder SMC transdifferentiation, decreasing urinary bladder distension. Our data may provide a mechanistic link for the association between urinary bladder distension and diabetes in humans, particularly because diabetes is associated with decreased cholesterol efflux. SMC-Abca1/Abcg1 deficiency did not affect atherosclerotic lesion size or plaque composition, presumably due to low expression of Abca1/Abcg1 in intimal SMCs.
Collapse
MESH Headings
- Animals
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter 1/deficiency
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Vasoconstriction
- Mice, Knockout
- Urinary Bladder/metabolism
- Urinary Bladder/pathology
- Cholesterol/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/metabolism
- Mice, Inbred C57BL
- Male
Collapse
Affiliation(s)
- Benedek Halmos
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Anouk M. La Rose
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Daisey Methorst
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Anouk G. Groenen
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Dalibor Nakládal
- Department of Clinical Pharmacy and Pharmacology (D.N., A.v.B., H.B., L.E.D., R.H.H.), University Medical Center Groningen, University of Groningen, the Netherlands
- Comenius University Science Park, Bratislava, Slovakia (D.N.)
- 5th Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Slovakia (D.N.)
| | - Venetia Bazioti
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Azuwerus van Buiten
- Department of Clinical Pharmacy and Pharmacology (D.N., A.v.B., H.B., L.E.D., R.H.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Elisabeth M. Schouten
- Department of Cardiology (E.M.S.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Nicolette C.A. Huijkman
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Laura Bongiovanni
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, University of Utrecht, the Netherlands (L.B., S.M.D.N., A.d.B.)
- Department of Veterinary Medicine, University of Teramo, Italy (L.B.)
| | - Simon M. De Neck
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, University of Utrecht, the Netherlands (L.B., S.M.D.N., A.d.B.)
| | - Alain de Bruin
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, University of Utrecht, the Netherlands (L.B., S.M.D.N., A.d.B.)
| | - Hendrik Buikema
- Department of Clinical Pharmacy and Pharmacology (D.N., A.v.B., H.B., L.E.D., R.H.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Leo E. Deelman
- Department of Clinical Pharmacy and Pharmacology (D.N., A.v.B., H.B., L.E.D., R.H.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Marius C. van den Heuvel
- Department of Pathology (M.C.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Folkert Kuipers
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
- Department of Laboratory Medicine (F.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Igle Jan de Jong
- Department of Urology (I.J.d.J.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Judith C. Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, the Netherlands (J.C.S.)
- Department of Medical Clinic II for Kidney and Hypertension Diseases, Rheumatological and Immunological Diseases, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Germany (J.C.S.)
- British Heart Foundation (BHF) Centre for Cardiovascular Sciences, University of Edinburgh, United Kingdom (J.C.S.)
| | - Helle F. Jørgensen
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, United Kingdom (H.F.J.)
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology (D.N., A.v.B., H.B., L.E.D., R.H.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Marit Westerterp
- Department of Pediatrics (B.H., A.M.L.R., D.M., A.G.G., V.B., M.H.K., N.J.K., N.C.A.H., M.L.-M., L.B., S.M.D.N., A.d.B., F.K., M.W.), University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
3
|
Huang S, Lu Y, Fang W, Huang Y, Li Q, Xu Z. Neurodegenerative diseases and neuroinflammation-induced apoptosis. Open Life Sci 2025; 20:20221051. [PMID: 40026360 PMCID: PMC11868719 DOI: 10.1515/biol-2022-1051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/30/2024] [Accepted: 12/27/2024] [Indexed: 03/05/2025] Open
Abstract
Neuroinflammation represents a critical pathway in the brain for the clearance of foreign bodies and the maintenance of homeostasis. When the neuroinflammatory process is dysregulate, such as the over-activation of microglia, which results in the excessive accumulation of free oxygen and inflammatory factors in the brain, among other factors, it can lead to an imbalance in homeostasis and the development of various diseases. Recent research has indicated that the development of numerous neurodegenerative diseases is closely associated with neuroinflammation. The pathogenesis of neuroinflammation in the brain is intricate, involving alterations in numerous genes and proteins, as well as the activation and inhibition of signaling pathways. Furthermore, excessive inflammation can result in neuronal cell apoptosis, which can further exacerbate the extent of the disease. This article presents a summary of recent studies on the relationship between neuronal apoptosis caused by excessive neuroinflammation and neurodegenerative diseases. The aim is to identify the link between the two and to provide new ideas and targets for exploring the pathogenesis, as well as the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shi Huang
- School of Clinical Medicine, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Yaxin Lu
- School of Pharmaceutical Sciences, Wannan Medical College,
241002, Wuhu, Anhui, China
| | - Wanzhen Fang
- School of Stomatology, Wannan Medical College,
241002, Wuhu, Anhui, China
| | - Yanjiao Huang
- Human Anatomy Experimental Training Center, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Qiang Li
- Human Anatomy Experimental Training Center, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
| | - Zhiliang Xu
- Department of Human Anatomy, School of Basic Medical Science, Wannan Medical College, 241002, Wuhu, Anhui, China
- Anhui Province Key Laboratory of Basic Research and Translation of Aging-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, China
| |
Collapse
|
4
|
Li B, Leung S, Elishaev M, Cheng WH, Mocci G, Björkegren JLM, Lai C, Singh A, Wang Y. Spatial Gene Expression of Human Coronary Arteries Revealed the Molecular Features of Diffuse Intimal Thickening in Explanted Hearts. Int J Mol Sci 2025; 26:1949. [PMID: 40076574 PMCID: PMC11900230 DOI: 10.3390/ijms26051949] [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: 01/25/2025] [Revised: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Diffuse intimal thickening (DIT) is a pre-clinical stage of atherosclerosis characterized by thickened intima. The molecular basis of its susceptibility to atherogenesis is unknown, and mechanistic investigations cannot be performed in commonly used mouse models, in which DIT does not exist. Vascular smooth muscle cells (SMCs) are the predominant cell type that occupies the intima and media of DIT. The molecular differences between these two layers may reveal the earliest phenotypic changes in SMCs to promote atherosclerosis. We benchmarked the RNA quality of human coronary arteries from autopsies (n = 7) and explanted hearts (n = 7) and performed Visium spatial gene expression on tissue sections with DIT. Although autopsy samples met the RNA quality standard for Visium (DV200 ≥ 30%), only arteries from explanted hearts exhibited reliable sequencing performance. Genes enriched in TGF-β-mediated remodeling of the extracellular matrix were overrepresented in the intima. SMCs enriched in the intima are dedifferentiated, but unlike those in the atherosclerotic lesions, they are not pro-inflammatory. Our findings indicate that autopsy samples are not ideal to distinguish subtle differences among cell phenotypes. SMCs in thickened intima may lead to lipid retention but not necessarily the onset of atherosclerosis.
Collapse
Affiliation(s)
- Boaz Li
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; (B.L.); (S.L.); (M.E.); (W.H.C.); (C.L.)
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Samuel Leung
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; (B.L.); (S.L.); (M.E.); (W.H.C.); (C.L.)
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Maria Elishaev
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; (B.L.); (S.L.); (M.E.); (W.H.C.); (C.L.)
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Wan Hei Cheng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; (B.L.); (S.L.); (M.E.); (W.H.C.); (C.L.)
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Giuseppe Mocci
- Department of Medicine, Karolinska Institute, 171 77 Solna, Sweden; (G.M.); (J.L.M.B.)
| | - Johan L. M. Björkegren
- Department of Medicine, Karolinska Institute, 171 77 Solna, Sweden; (G.M.); (J.L.M.B.)
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chi Lai
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; (B.L.); (S.L.); (M.E.); (W.H.C.); (C.L.)
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
- Division of Anatomical Pathology, Providence Health Care, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Amrit Singh
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; (B.L.); (S.L.); (M.E.); (W.H.C.); (C.L.)
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
5
|
Zhang Y, Xu Q, Tian X, Xia X, Chen S, Liu F, Wu S, Wang A. Longitudinal changes in remnant cholesterol and the risk of cardiovascular disease. Cardiovasc Diabetol 2025; 24:1. [PMID: 39748387 PMCID: PMC11697916 DOI: 10.1186/s12933-024-02556-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND AND AIM The analyses of longitudinal changes in remnant cholesterol (RC) and cardiovascular disease (CVD) remains are limited. The objective of the study was to investigate the associations of longitudinal changes in RC with the risks of CVD and its subtypes (myocardial infarction [MI] and stroke). METHODS AND RESULTS The participants were enrolled in the Kailuan study. The RC short-term change pattern was defined by RC cutoff points according to equivalent percentiles for low-density lipoprotein cholesterol of 2.6 mmol/L at visits in 2006 and 2008. The RC long-term change pattern was defined as the RC trajectories from 2006 to 2010. Multivariate Cox proportion models were used to calculate hazard ratios (HRs) and their 95% confidence intervals (CIs). The cutoff values of RC were 0.52 mmol/L at the 2006 visit and 0.51 mmol/L at the 2008 visit. In the RC short-term change analysis, the participants in the high stable group had a 31% increased risk of CVD (HR 1.31; 95% CI 1.22-1.41), 73% increased risks of MI (HR 1.73; 95% CI 1.47-2.03), and 21% increased risks of stroke (HR 1.21; 95% CI 1.12-1.31) compared with participants in the low stable group. Three RC trajectories were employed in the RC long-term change analysis. Compared with the low stable group, the high stable group had a 1.34-fold risk of CVD (HR 1.34; 95% CI 1.17-1.53), 1.66-fold risk of MI (HR 1.66; 95% CI 1.24-2.21), and 1.22-fold risk of stroke (HR 1.22; 95% CI 1.05-1.42). CONCLUSIONS The stable high RC was associated with a higher risk of CVD. Maintaining optional RC levels could reduce the lifetime risk of CVD and prolong the year of life free from CVD.
Collapse
Affiliation(s)
- Yijun Zhang
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, No. 119 S 4th Ring W Rd, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing, 100069, China
| | - Qin Xu
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, No. 119 S 4th Ring W Rd, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Xue Tian
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, No. 119 S 4th Ring W Rd, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing, 100069, China
| | - Xue Xia
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, No. 119 S 4th Ring W Rd, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, 57 Xinhua East Rd, Tangshan, 063000, China
| | - Fen Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, No.10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing, 100069, China.
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, 57 Xinhua East Rd, Tangshan, 063000, China.
| | - Anxin Wang
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, No. 119 S 4th Ring W Rd, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Liu Y, Lu K, Zhang R, Hu D, Yang Z, Zeng J, Cai W. Advancements in the Treatment of Atherosclerosis: From Conventional Therapies to Cutting-Edge Innovations. ACS Pharmacol Transl Sci 2024; 7:3804-3826. [PMID: 39698263 PMCID: PMC11651175 DOI: 10.1021/acsptsci.4c00574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Atherosclerosis is a leading cause of morbidity and mortality worldwide, driven by a complex interplay of lipid dysregulation, inflammation, and vascular pathology. Despite advancements in understanding the multifactorial nature of atherosclerosis and improvements in clinical management, existing therapies often fall short in reversing the disease, focusing instead on symptom alleviation and risk reduction. This review highlights recent strides in identifying genetic markers, elucidating inflammatory pathways, and understanding environmental contributors to atherosclerosis. It also evaluates the efficacy and limitations of current pharmacological treatments, revascularization techniques, and the impact of these interventions on patient outcomes. Furthermore, we explore innovative therapeutic strategies, including the promising fields of nanomedicine, nucleic acid-based therapies, and immunomodulation, which offer potential for targeted and effective treatment modalities. However, integrating these advances into clinical practice is challenged by regulatory, economic, and logistical barriers. This review synthesizes the latest research and clinical advancements to provide a comprehensive roadmap for future therapeutic strategies and emphasize the critical need for innovative approaches to fundamentally change the course of atherosclerosis management.
Collapse
Affiliation(s)
- Yan Liu
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Kuan Lu
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Ruru Zhang
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Dongliang Hu
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Zhe Yang
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jianfeng Zeng
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Wu Cai
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| |
Collapse
|
7
|
Zhang L, Li J, Kou Y, Shen L, Wang H, Wang Y, Ma R, Wu T, Yang X, Gu Y, Yi L. Mechanisms and treatment of atherosclerosis: focus on macrophages. Front Immunol 2024; 15:1490387. [PMID: 39569201 PMCID: PMC11576186 DOI: 10.3389/fimmu.2024.1490387] [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: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
Macrophages are the basic mediators and coordinators of various types of chronic inflammation and play a crucial role in the formation and development of atherosclerosis (AS). In the complex microenvironment of atherosclerotic plaques, macrophages of different sources are exposed to different signal stimuli and thus polarized into various subpopulations. Various types of macrophages with predominantly M1 and M2 phenotypes also play different regulatory roles in the initiation and progression of AS. Lipid-lowering drugs, mainly statins, are widely used in clinical practice, but the adverse reactions are obvious and there is a lack of personalized treatment. Emerging targeted macrophage and Traditional Chinese medicine (TCM)-related therapies can regulate the cellular microenvironment, inhibit the polarization of M1 macrophages, and promote the activation of M2 macrophages, providing new ideas for the prevention and treatment of AS.
Collapse
Affiliation(s)
- LingNa Zhang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - JiaWei Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YuShun Kou
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - LuFan Shen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Hong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YiYuan Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ruiling Ma
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Tao Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xin Yang
- First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YuanHui Gu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Lin Yi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Chronic Disease Laboratory, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
8
|
Wang C, He Y, Tang J, Mao J, Liang X, Xu M, Zhang Z, Tian J, Jiang J, Li C, Zhou X. Chondroitin sulfate functionalized nanozymes inhibit the inflammation feedback loop for enhanced atherosclerosis therapy by regulating intercellular crosstalk. Int J Biol Macromol 2024; 282:136918. [PMID: 39471920 DOI: 10.1016/j.ijbiomac.2024.136918] [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: 06/25/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
In the inflammatory microenvironment of atherosclerotic plaques, metabolic dysregulation of superoxide anion (O2-) and hydrogen peroxide (H2O2) leads to the activation of feedback mechanisms involving IL-1β, TNF-α, and MCP-1, which triggers inflammatory cascades between macrophages and vascular smooth muscle cells (VSMCs) in atherosclerosis (AS). To address this, a chondroitin sulfate (CS)-functionalized dual-targeted engineered nanozyme, CS-Lip/PB@Rap, was developed by encapsulating mesoporous Prussian blue nanoparticles (PBs) loaded with rapamycin (Rap) within CS-modified liposomes. CS functionalization endowed CS-Lip/PB@Rap with a specific targeting ability for CD44 receptors, thus enabling targeted delivery to inflammatory macrophages and VSMCs. Moreover, its enhanced multiple enzyme-like activities effectively modulated the imbalance of oxidative stress. The underlying mechanism of crosstalk regulation by these engineered nanozymes may inhibit the NF-κB pathway by restoring normal metabolism of O2- and H2O2, thereby blocking the TNF-α, IL-1β, and MCP-1 feedback loops between macrophages and VSMCs. This process reduced the production of inflammatory macrophages and inhibited the VSMC transformation from a contractile phenotype to a synthetic phenotype, preventing the formation of fibrous caps. Furthermore, the elimination of oxidative stress could decrease the production of oxygenized low-density lipoprotein (ox-LDL), which inhibited the formation of foam cells and alleviated the atherogenic progression.
Collapse
Affiliation(s)
- Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingying Mao
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
9
|
Arévalo-Martinez M, Ede J, van der Have O, Ritsvall O, Zetterberg FR, Nilsson UJ, Leffler H, Holmberg J, Albinsson S. Myocardin related transcription factor and galectin-3 drive lipid accumulation in human blood vessels. Vascul Pharmacol 2024; 156:107383. [PMID: 38830455 DOI: 10.1016/j.vph.2024.107383] [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: 01/16/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024]
Abstract
OBJECTIVE Diabetes and hypertension are important risk factors for vascular disease, including atherosclerosis. A driving factor in this process is lipid accumulation in smooth muscle cells of the vascular wall. The glucose- and mechano-sensitive transcriptional coactivator, myocardin-related transcription factor A (MRTF-A/MKL1) can promote lipid accumulation in cultured human smooth muscle cells and contribute to the formation of smooth muscle-derived foam cells. The purpose of this study was to determine if intact human blood vessels ex vivo can be used to evaluate lipid accumulation in the vascular wall, and if this process is dependent on MRTF and/or galectin-3/LGALS3. Galectin-3 is an early marker of smooth muscle transdifferentiation and a potential mediator for foam cell formation and atherosclerosis. APPROACH AND RESULTS Human mammary arteries and saphenous veins were exposed to altered cholesterol and glucose levels in an organ culture model. Accumulation of lipids, quantified by Oil Red O, was increased by cholesterol loading and elevated glucose concentrations. Pharmacological inhibition of MRTF with CCG-203971 decreased lipid accumulation, whereas adenoviral-mediated overexpression of MRTF-A had the opposite effect. Cholesterol-induced expression of galectin-3 was decreased after inhibition of MRTF. Importantly, pharmacological inhibition of galectin-3 with GB1107 reduced lipid accumulation in the vascular wall after cholesterol loading. CONCLUSION Ex vivo organ culture of human arteries and veins can be used to evaluate lipid accumulation in the intact vascular wall, as well as adenoviral transduction and pharmacological inhibition. Although MRTF and galectin-3 may have beneficial, anti-inflammatory effects under certain circumstances, our results, which demonstrate a significant decrease in lipid accumulation, support further evaluation of MRTF- and galectin-3-inhibitors for therapeutic intervention against atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Marycarmen Arévalo-Martinez
- Molecular Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84 Lund, Sweden
| | - Jacob Ede
- Department of Clinical Sciences Lund, Department of Cardiothoracic Surgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Oscar van der Have
- Vessel Wall Biology, Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84 Lund, Sweden
| | - Olivia Ritsvall
- Molecular Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84 Lund, Sweden
| | - Fredrik R Zetterberg
- Galecto Biotech AB, Sahlgrenska Science Park, Medicinaregatan 8 A, SE-413 46 Lund, Sweden
| | - Ulf J Nilsson
- Galecto Biotech AB, Sahlgrenska Science Park, Medicinaregatan 8 A, SE-413 46 Lund, Sweden; Department of Chemistry, Lund University, SE-221 00 Lund, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b, Klinikgatan 28, 221 84 Lund, Sweden
| | - Johan Holmberg
- Molecular Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84 Lund, Sweden
| | - Sebastian Albinsson
- Molecular Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84 Lund, Sweden.
| |
Collapse
|
10
|
Annink ME, Kraaijenhof JM, Stroes ESG, Kroon J. Moving from lipids to leukocytes: inflammation and immune cells in atherosclerosis. Front Cell Dev Biol 2024; 12:1446758. [PMID: 39161593 PMCID: PMC11330886 DOI: 10.3389/fcell.2024.1446758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the most important cause of morbidity and mortality worldwide. While it is traditionally attributed to lipid accumulation in the vascular endothelium, recent research has shown that plaque inflammation is an important additional driver of atherogenesis. Though clinical outcome trials utilizing anti-inflammatory agents have proven promising in terms of reducing ASCVD risk, it is imperative to identify novel actionable targets that are more specific to atherosclerosis to mitigate adverse effects associated with systemic immune suppression. To that end, this review explores the contributions of various immune cells from the innate and adaptive immune system in promoting and mitigating atherosclerosis by integrating findings from experimental studies, high-throughput multi-omics technologies, and epidemiological research.
Collapse
Affiliation(s)
- Maxim E. Annink
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, Netherlands
| |
Collapse
|
11
|
Zheng H, Tai L, Xu C, Wang W, Ma Q, Sun W. Microfluidic-based cardiovascular systems for advanced study of atherosclerosis. J Mater Chem B 2024. [PMID: 38948949 DOI: 10.1039/d4tb00756e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Atherosclerosis (AS) is a significant global health concern due to its high morbidity and mortality rates. Extensive efforts have been made to replicate the cardiovascular system and explore the pathogenesis, diagnosis, and treatment of AS. Microfluidics has emerged as a valuable technology for modeling the cardiovascular system and studying AS. Here a brief review of the advances of microfluidic-based cardiovascular systems for AS research is presented. The critical pathogenetic mechanisms of AS investigated by microfluidic-based cardiovascular systems are categorized and reviewed, with a detailed summary of accurate diagnostic methods for detecting biomarkers using microfluidics represented. Furthermore, the review covers the evaluation and screening of AS drugs assisted by microfluidic systems, along with the fabrication of novel drug delivery carriers. Finally, the challenges and future prospects for advancing microfluidic-based cardiovascular systems in AS research are discussed and proposed, particularly regarding new opportunities in multi-disciplinary fundamental research and therapeutic applications for a broader range of disease treatments.
Collapse
Affiliation(s)
- Huiyuan Zheng
- School of Pharmacy, Qingdao University, Qingdao 266071, China.
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| | - Lei Tai
- Pharmacy Department, Shandong Qingdao Hospital of Integrated Traditional and Western Medicine, Qingdao 266002, China
| | - Chengbin Xu
- Pharmacy Department, Shandong Qingdao Hospital of Integrated Traditional and Western Medicine, Qingdao 266002, China
| | - Weijiang Wang
- School of Pharmacy, Qingdao University, Qingdao 266071, China.
| | - Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao 266071, China.
| | - Wentao Sun
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| |
Collapse
|
12
|
Elishaev M, Li B, Zhou A, Salim K, Leeper NJ, Francis GA, Lai C, Wang Y. Multiplex Imaging for Cell Phenotyping of Early Human Atherosclerosis. J Am Heart Assoc 2024; 13:e034990. [PMID: 38842292 PMCID: PMC11255771 DOI: 10.1161/jaha.123.034990] [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: 02/29/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Previous studies using animal models and cultured cells suggest that vascular smooth muscle cells (SMCs) and inflammatory cytokines are important players in atherogenesis. Validating these findings in human disease is critical to designing therapeutics that target these components. Multiplex imaging is a powerful tool for characterizing cell phenotypes and microenvironments using biobanked human tissue sections. However, this technology has not been applied to human atherosclerotic lesions and needs to first be customized and validated. METHODS AND RESULTS For validation, we created an 8-plex imaging panel to distinguish foam cells from SMC and leukocyte origins on tissue sections of early human atherosclerotic lesions (n=9). The spatial distribution and characteristics of these foam cells were further analyzed to test the association between SMC phenotypes and inflammation. Consistent with previous reports using human lesions, multiplex imaging showed that foam cells of SMC origin outnumbered those of leukocyte origin and were enriched in the deep intima, where the lipids accumulate in early atherogenesis. This new technology also found that apoptosis or the expression of pro-inflammatory cytokines were not more associated with foam cells than with nonfoam cells in early human lesions. More CD68+ SMCs were present among SMCs that highly expressed interleukin-1β. Highly inflamed SMCs showed a trend of increased apoptosis, whereas leukocytes expressing similar levels of cytokines were enriched in regions of extracellular matrix remodeling. CONCLUSIONS The multiplex imaging method can be applied to biobanked human tissue sections to enable proof-of-concept studies and validate theories based on animal models and cultured cells.
Collapse
Affiliation(s)
- Maria Elishaev
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
| | - Boaz Li
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
| | - Annie Zhou
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
| | - Kevin Salim
- British Columbia Children’s Hospital Research InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular SurgeryStanford University School of MedicineStanfordCAUSA
- Stanford Cardiovascular InstituteStanford UniversityStanfordCAUSA
| | - Gordon A. Francis
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
- Department of MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Chi Lai
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
- Division of Anatomical PathologyProvidence Health Care, St. Paul’s HospitalVancouverBCCanada
| | - Ying Wang
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
| |
Collapse
|
13
|
Xie L, You Q, Mao J, Wu F, Xia C, Hai R, Wei Y, Zhou X. Thyrotropin induces atherosclerosis by upregulating large conductance Ca 2+-activated K + channel subunits. Mol Cell Endocrinol 2024; 583:112145. [PMID: 38184154 DOI: 10.1016/j.mce.2024.112145] [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: 08/19/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
Hypothyroidism is associated with elevated levels of serum thyrotropin (TSH), which have been shown to promote abnormal proliferation of vascular smooth muscle cells and contribute to the development of atherosclerosis. However, the specific mechanisms underlying the TSH-induced abnormal proliferation of vascular smooth muscle cells remain unclear. The objective of this study was to investigate the role of TSH in the progression of atherosclerosis. Our research findings revealed that hypothyroidism can trigger early atherosclerotic changes in the aorta of Wistar rats. In alignment with our in vitro experiments, we observed that TSH induces abnormal proliferation of aortic smooth muscle cells by modulating the expression of α and β1 subunits of large conductance Ca2+-activated K+ (BKCa) channels within these cells via the cAMP/PKA signaling pathway. These results collectively indicate that TSH acts through the cAMP/PKA signaling pathway to upregulate the expression of α and β1 subunits of BKCa channels, thereby promoting abnormal proliferation of arterial smooth muscle cells. These findings may provide a basis for the clinical prevention and treatment of atherosclerosis caused by elevated TSH levels.
Collapse
Affiliation(s)
- Linjun Xie
- Department of Thyroid and Breast Surgery, The First People's Hospital of Zigong, Zigong, Sichuan, China; Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Qian You
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Breast, Thyroid and Vessel Surgery, The Neijiang First People's Hospital, Neijiang, 641000, China.
| | - Jingying Mao
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Fei Wu
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Chengwei Xia
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Rui Hai
- Department of Vascular, Breast, Thyroid Surgery, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan, China.
| | - Yan Wei
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
14
|
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024; 9:53. [PMID: 38433280 PMCID: PMC10910037 DOI: 10.1038/s41392-024-01757-9] [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: 10/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
NF-κB signaling has been discovered for nearly 40 years. Initially, NF-κB signaling was identified as a pivotal pathway in mediating inflammatory responses. However, with extensive and in-depth investigations, researchers have discovered that its role can be expanded to a variety of signaling mechanisms, biological processes, human diseases, and treatment options. In this review, we first scrutinize the research process of NF-κB signaling, and summarize the composition, activation, and regulatory mechanism of NF-κB signaling. We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog, and TLR signaling. The physiological and pathological states of NF-κB signaling, as well as its intricate involvement in inflammation, immune regulation, and tumor microenvironment, are also explicated. Additionally, we illustrate how NF-κB signaling is involved in a variety of human diseases, including cancers, inflammatory and autoimmune diseases, cardiovascular diseases, metabolic diseases, neurological diseases, and COVID-19. Further, we discuss the therapeutic approaches targeting NF-κB signaling, including IKK inhibitors, monoclonal antibodies, proteasome inhibitors, nuclear translocation inhibitors, DNA binding inhibitors, TKIs, non-coding RNAs, immunotherapy, and CAR-T. Finally, we provide an outlook for research in the field of NF-κB signaling. We hope to present a stereoscopic, comprehensive NF-κB signaling that will inform future research and clinical practice.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiaomin Ye
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xin Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Oladosu O, Chin E, Barksdale C, Powell RR, Bruce T, Stamatikos A. Inhibition of miR-33a-5p in Macrophage-like Cells In Vitro Promotes apoAI-Mediated Cholesterol Efflux. PATHOPHYSIOLOGY 2024; 31:117-126. [PMID: 38535619 PMCID: PMC10976131 DOI: 10.3390/pathophysiology31010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 04/01/2024] Open
Abstract
Atherosclerosis is caused by cholesterol accumulation within arteries. The intima is where atherosclerotic plaque accumulates and where lipid-laden foam cells reside. Intimal foam cells comprise of both monocyte-derived macrophages and macrophage-like cells (MLC) of vascular smooth muscle cell (VSMC) origin. Foam cells can remove cholesterol via apoAI-mediated cholesterol efflux and this process is regulated by the transporter ABCA1. The microRNA miR-33a-5p is thought to be atherogenic via silencing ABCA1 which promotes cholesterol retention and data has shown inhibiting miR-33a-5p in macrophages may be atheroprotective via enhancing apoAI-mediated cholesterol efflux. However, it is not entirely elucidated whether precisely inhibiting miR-33a-5p in MLC also increases ABCA1-dependent cholesterol efflux. Therefore, the purpose of this work is to test the hypothesis that inhibition of miR-33a-5p in cultured MLC enhances apoAI-mediated cholesterol efflux. In our study, we utilized the VSMC line MOVAS cells in our experiments, and cholesterol-loaded MOVAS cells to convert this cell line into MLC. Inhibition of miR-33a-5p was accomplished by transducing cells with a lentivirus that expresses an antagomiR directed at miR-33a-5p. Expression of miR-33a-5p was analyzed by qRT-PCR, ABCA1 protein expression was assessed via immunoblotting, and apoAI-mediated cholesterol efflux was measured using cholesterol efflux assays. In our results, we demonstrated that lentiviral vector-mediated knockdown of miR-33a-5p resulted in decreasing expression of this microRNA in cultured MLC. Moreover, reduction of miR-33a-5p in cultured MLC resulted in de-repression of ABCA1 expression, which caused ABCA1 protein upregulation in cultured MLC. Additionally, this increase in ABCA1 protein expression resulted in enhancing ABCA1-dependent cholesterol efflux through increasing apoAI-mediated cholesterol efflux in cultured MLC. From these findings, we conclude that inhibiting miR-33a-5p in MLC may protect against atherosclerosis by promoting ABCA1-dependent cholesterol efflux.
Collapse
Affiliation(s)
- Olanrewaju Oladosu
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (E.C.); (C.B.)
| | - Emma Chin
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (E.C.); (C.B.)
| | - Christian Barksdale
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (E.C.); (C.B.)
| | - Rhonda R. Powell
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Terri Bruce
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (O.O.); (E.C.); (C.B.)
| |
Collapse
|