1
|
Rodríguez-Fernández MA, Tristán-Flores FE, Casique-Aguirre D, Negrete-Rodríguez MDLLX, Cervantes-Montelongo JA, Conde-Barajas E, Acosta-García G, Silva-Martínez GA. Virtual Screening and Molecular Dynamics of Cytokine-Drug Complexes for Atherosclerosis Therapy. Int J Mol Sci 2025; 26:2931. [PMID: 40243563 PMCID: PMC11988346 DOI: 10.3390/ijms26072931] [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: 03/01/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease remains the leading global cause of mortality, largely driven by atherosclerosis, a chronic inflammatory condition characterized by lipid accumulation and immune-cell infiltration in arterial walls. Macrophages play a central role by forming foam cells and secreting pro-atherogenic cytokines, such as TNF-α, IFN-γ, and IL-1β, which destabilize atherosclerotic plaques, expanding the lipid core and increasing the risk of thrombosis and ischemia. Despite the significant health burden of subclinical atherosclerosis, few targeted therapies exist. Current treatments, including monoclonal antibodies, are limited by high costs and immunosuppressive side effects, underscoring the urgent need for alternative therapeutic strategies. In this study, we employed in silico drug repositioning to identify multitarget inhibitors against TNF-α, IFN-γ, and IL-1β, leveraging a virtual screening of 2750 FDA-approved drugs followed by molecular dynamics simulations to assess the stability of selected cytokine-ligand complexes. This computational approach provides structural insights into potential inhibitors. Additionally, we highlight nutraceutical options, such as fatty acids (oleic, linoleic and eicosapentaenoic acid), which exhibited strong and stable interactions with key cytokine targets. Our study suggests that these bioactive compounds could serve as effective new therapeutic approaches for atherosclerosis.
Collapse
Affiliation(s)
- María Angélica Rodríguez-Fernández
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
| | - Fabiola Estefanía Tristán-Flores
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ciencias Básicas, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico
| | - Diana Casique-Aguirre
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Delegación Puebla, Puebla 06600, Mexico;
- Secretaría de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI), Ciudad de México 03940, Mexico
| | - María de la Luz Xochilt Negrete-Rodríguez
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| | - Juan Antonio Cervantes-Montelongo
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
- Escuela de Medicina, Universidad de Celaya, Celaya 38080, Guanajuato, Mexico
| | - Eloy Conde-Barajas
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| | - Gerardo Acosta-García
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| | - Guillermo Antonio Silva-Martínez
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Secretaría de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI), Ciudad de México 03940, Mexico
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| |
Collapse
|
2
|
Mulumba M, Le C, Schelsohn E, Namkung Y, Laporte SA, Febbraio M, Servant MJ, Chemtob S, Lubell WD, Marleau S, Ong H. Selective Azapeptide CD36 Ligand MPE-298 Regulates oxLDL-LOX-1-Mediated Inflammation and Mitochondrial Oxidative Stress in Macrophages. Cells 2025; 14:385. [PMID: 40072113 PMCID: PMC11898605 DOI: 10.3390/cells14050385] [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: 10/30/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Macrophage mitochondrial dysfunction, caused by oxidative stress, has been proposed as an essential event in the progression of chronic inflammation diseases, such as atherosclerosis. The cluster of differentiation-36 (CD36) and lectin-like oxLDL receptor-1 (LOX-1) scavenger receptors mediate macrophage uptake of oxidized low-density lipoprotein (oxLDL), which contributes to mitochondrial dysfunction by sustained production of mitochondrial reactive oxygen species (mtROS), as well as membrane depolarization. In the present study, the antioxidant mechanisms of action of the selective synthetic azapeptide CD36 ligand MPE-298 have been revealed. After binding to CD36, MPE-298 was rapidly internalized by and simultaneously induced CD36 endocytosis through activation of the Lyn and Syk (spleen) tyrosine kinases. Within this internalized complex, MPE-298 inhibited oxLDL/LOX-1-induced chemokine ligand 2 (CCL2) secretion, abolished the production of mtROS, and prevented mitochondrial membrane potential depolarization in macrophages. This occurred through the inhibition of the multiple-component enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) by oxLDL-activated LOX-1, which was further supported by the reduced recruitment of the p47phox subunit and small GTPase (Rac) 1/2/3 into the plasma membrane. A new mechanism for alleviating oxLDL-induced oxidative stress and inflammation in macrophages is highlighted using the CD36 ligand MPE-298.
Collapse
Affiliation(s)
- Mukandila Mulumba
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Catherine Le
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Emmanuelle Schelsohn
- Institut des Sciences Pharmaceutiques de Suisse Occidentale (ISPSO), Section Sciences Pharmaceutiques, Département des Sciences, Université de Genève, 1205 Genève, Switzerland;
| | - Yoon Namkung
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (Y.N.); (S.A.L.)
| | - Stéphane A. Laporte
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (Y.N.); (S.A.L.)
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, AB T6G 2H5, Canada;
| | - Marc J. Servant
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Sylvain Chemtob
- Faculté de Médecine, Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC H3T 1C5, Canada;
| | - William D. Lubell
- Département de Chimie, Université de Montréal, Montréal, QC H3C 3J7, Canada;
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Huy Ong
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| |
Collapse
|
3
|
Otálora-Alcaraz A, Reilly T, Oró-Nolla M, Sun MC, Costelloe L, Kearney H, Patra PH, Downer EJ. The NLRP3 inflammasome: A central player in multiple sclerosis. Biochem Pharmacol 2025; 232:116667. [PMID: 39647604 DOI: 10.1016/j.bcp.2024.116667] [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: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune condition associated with many symptoms including spasticity, pain, limb numbness and weakness. It is characterised by inflammatory demyelination and axonal degeneration of the brain and spinal cord. A range of disease-modifying therapies (DMTs) are available to suppress inflammatory disease activity in MS, however, there is a pressing need for new therapeutic avenues as DMTs have a limited ability to suppress confirmed disability progression. A body of literature indicates that innate immune inflammation is linked to MS progression. The nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome has a well-established function in innate immunity which is closely associated with the pathogenesis of neuroinflammatory conditions. Evidence suggests that the inflammasome may be a therapeutic target in disorders such as MS and at present, inhibitors of the NLRP3 inflammasome are in pre-clinical development. Therefore, this review systematically highlights the pathogenic role of inflammasomes in MS, presenting an overview of research evidence linking inflammasome-related polymorphisms to MS susceptibility, and gathering evidence investigating NLRP3 biomarkers in MS. The role of the NLRP3 inflammasome in murine models of MS is furthermore discussed. Finally, a significant component of this review focuses on evidence that NLRP3 signalling components are novel drug targets in MS. Overall this review defines the role of the inflammasome in MS pathogenesis and identifies inflammasome inhibitor targets that warrant full investigation in MS and related disorders.
Collapse
Affiliation(s)
- Almudena Otálora-Alcaraz
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Thomas Reilly
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Martí Oró-Nolla
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Melody Cui Sun
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Lisa Costelloe
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Hugh Kearney
- MS Unit, Department of Neurology, St. James's Hospital, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Pabitra H Patra
- Transpharmation Ltd., London Biosciences Innovation Centre, London, United Kingdom
| | - Eric J Downer
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
4
|
Klauzen P, Basovich L, Shishkova D, Markova V, Malashicheva A. Macrophages in Calcific Aortic Valve Disease: Paracrine and Juxtacrine Disease Drivers. Biomolecules 2024; 14:1547. [PMID: 39766254 PMCID: PMC11673549 DOI: 10.3390/biom14121547] [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: 09/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
A significant role in the pathogenesis of CAVD is played by innate immunity cells, such as macrophages. In stenotic valves, macrophages have enhanced inflammatory activity, and the population's balance is shifted toward pro-inflammatory ones. Pro-inflammatory macrophages release cytokines, chemokines, and microRNA, which can directly affect the resident valvular cells and cause valve calcification. In CAVD patients, macrophages may have more pronounced pro-inflammatory properties, enhanced not only by paracrine signals but also by juxtacrine Notch signaling and epigenetic factors, which influence the maturation of macrophages' progenitors. In this review, we observe the accumulated data on the involvement of macrophages in CAVD development via paracrine and juxtacrine interactions.
Collapse
Affiliation(s)
- Polina Klauzen
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Liubov Basovich
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| |
Collapse
|
5
|
Bartoli-Leonard F, Pennel T, Caputo M. Immunotherapy in the Context of Aortic Valve Diseases. Cardiovasc Drugs Ther 2024; 38:1173-1185. [PMID: 39017904 PMCID: PMC11680629 DOI: 10.1007/s10557-024-07608-7] [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] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Aortic valve disease (AVD) affects millions of people around the world, with no pharmacological intervention available. Widely considered a multi-faceted disease comprising both regurgitative pathogenesis, in which retrograde blood flows back through to the left ventricle, and aortic valve stenosis, which is characterized by the thickening, fibrosis, and subsequent mineralization of the aortic valve leaflets, limiting the anterograde flow through the valve, surgical intervention is still the main treatment, which incurs considerable risk to the patient. RESULTS Though originally thought of as a passive degeneration of the valve or a congenital malformation that has occurred before birth, the paradigm of AVD is shifting, and research into the inflammatory drivers of valve disease as a potential mechanism to modulate the pathobiology of this life-limiting pathology is taking center stage. Following limited success in mainstay therapeutics such as statins and mineralisation inhibitors, immunomodulatory strategies are being developed. Immune cell therapy has begun to be adopted in the cancer field, in which T cells (chimeric antigen receptor (CAR) T cells) are isolated from the patient, programmed to attack the cancer, and then re-administered to the patient. Within cardiac research, a novel T cell-based therapeutic approach has been developed to target lipid nanoparticles responsible for increasing cardiac fibrosis in a failing heart. With clonally expanded T-cell populations recently identified within the diseased valve, their unique epitope presentation may serve to identify novel targets for the treatment of valve disease. CONCLUSION Taken together, targeted T-cell therapy may hold promise as a therapeutic platform to target a multitude of diseases with an autoimmune aspect, and this review aims to frame this in the context of cardiovascular disease, delineating what is currently known in the field, both clinically and translationally.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK.
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK.
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa.
| | - Tim Pennel
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
6
|
Onnis C, Virmani R, Madra A, Nardi V, Salgado R, Montisci R, Cau R, Boi A, Lerman A, De Cecco CN, Libby P, Saba L. Whys and Wherefores of Coronary Arterial Positive Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:2416-2427. [PMID: 39479766 PMCID: PMC11594009 DOI: 10.1161/atvbaha.124.321504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Positive remodeling (PR) is an atherosclerotic plaque feature defined as an increase in arterial caliber at the level of an atheroma, in response to increasing plaque burden. The mechanisms that lead to its formation are incompletely understood, but its role in coronary atherosclerosis has major clinical implications. Indeed, plaques with PR have elevated risk of provoking acute cardiac events. Hence, PR figures among the high-risk plaque features that cardiac imaging studies should report. This review aims to provide an overview of the current literature on coronary PR. It outlines the pathophysiology of PR, the different techniques used to assess its presence, and the imaging findings associated to PR, on both noninvasive and invasive studies. This review also summarizes clinical observations, trials, and studies, focused on the impact of PR on clinical outcome.
Collapse
Affiliation(s)
- Carlotta Onnis
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.)
| | - Anna Madra
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.)
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.)
| | - Rodrigo Salgado
- Department of Radiology, Antwerp University Hospital and Antwerp University Lier, Belgium (R.S.)
| | - Roberta Montisci
- Clinical Cardiology, Department of Medical Science and Public Health, University of Cagliari, Italy (R.M.)
| | - Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| | - Alberto Boi
- Department of Cardiology, Azienda Ospedaliera Brotzu, Cagliari, Italy (A.B.)
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.)
| | - Carlo N. De Cecco
- Division of Cardiothoracic Imaging and Biomedical Informatics, Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA (C.N.D.C.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (P.L.)
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| |
Collapse
|
7
|
Rawal S, Randhawa V, Rizvi SHM, Sachan M, Wara AK, Pérez-Cremades D, Weisbrod RM, Hamburg NM, Feinberg MW. miR-369-3p ameliorates diabetes-associated atherosclerosis by regulating macrophage succinate-GPR91 signalling. Cardiovasc Res 2024; 120:1693-1712. [PMID: 38703377 PMCID: PMC11587565 DOI: 10.1093/cvr/cvae102] [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: 10/07/2023] [Revised: 03/04/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024] Open
Abstract
AIMS Diabetes leads to dysregulated macrophage immunometabolism, contributing to accelerated atherosclerosis progression. Identifying critical factors to restore metabolic alterations and promote resolution of inflammation remains an unmet goal. MicroRNAs orchestrate multiple signalling events in macrophages, yet their therapeutic potential in diabetes-associated atherosclerosis remains unclear. METHODS AND RESULTS miRNA profiling revealed significantly lower miR-369-3p expression in aortic intimal lesions from Ldlr-/- mice on a high-fat sucrose-containing (HFSC) diet for 12 weeks. miR-369-3p was also reduced in peripheral blood mononuclear cells from diabetic patients with coronary artery disease (CAD). Cell-type expression profiling showed miR-369-3p enrichment in aortic macrophages. In vitro, oxLDL treatment reduced miR-369-3p expression in mouse bone marrow-derived macrophages (BMDMs). Metabolic profiling in BMDMs revealed that miR-369-3p overexpression blocked the oxidized low density lipoprotein (oxLDL)-mediated increase in the cellular metabolite succinate and reduced mitochondrial respiration (OXPHOS) and inflammation [Interleukin (lL)-1β, TNF-α, and IL-6]. Mechanistically, miR-369-3p targeted the succinate receptor (GPR91) and alleviated the oxLDL-induced activation of inflammasome signalling pathways. Therapeutic administration of miR-369-3p mimics in HFSC-fed Ldlr-/- mice reduced GPR91 expression in lesional macrophages and diabetes-accelerated atherosclerosis, evident by a decrease in plaque size and pro-inflammatory Ly6Chi monocytes. RNA-Seq analyses showed more pro-resolving pathways in plaque macrophages from miR-369-3p-treated mice, consistent with an increase in macrophage efferocytosis in lesions. Finally, a GPR91 antagonist attenuated oxLDL-induced inflammation in primary monocytes from human subjects with diabetes. CONCLUSION These findings establish a therapeutic role for miR-369-3p in halting diabetes-associated atherosclerosis by regulating GPR91 and macrophage succinate metabolism.
Collapse
MESH Headings
- Animals
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Macrophages/metabolism
- Macrophages/pathology
- Signal Transduction
- Humans
- Mice, Knockout
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Lipoproteins, LDL/metabolism
- Succinic Acid/metabolism
- Plaque, Atherosclerotic
- Mice
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Aortic Diseases/immunology
- Cells, Cultured
- Gene Expression Regulation
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/prevention & control
- Female
- Middle Aged
Collapse
Affiliation(s)
- Shruti Rawal
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Vinay Randhawa
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Syed Husain Mustafa Rizvi
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
- Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Madhur Sachan
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Akm Khyrul Wara
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Daniel Pérez-Cremades
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Department of Physiology, University of Valencia, INCLIVA Biomedical Research Institute, Valencia 46010, Spain
| | - Robert M Weisbrod
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
- Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Naomi M Hamburg
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
- Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
8
|
Shamas S, Rahil RR, Kaushal L, Sharma VK, Wani NA, Qureshi SH, Ahmad SF, Attia SM, Zargar MA, Hamid A, Bhat OM. Pyroptosis in Endothelial Cells and Extracellular Vesicle Release in Atherosclerosis via NF-κB-Caspase-4/5-GSDM-D Pathway. Pharmaceuticals (Basel) 2024; 17:1568. [PMID: 39770410 PMCID: PMC11677252 DOI: 10.3390/ph17121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Pyroptosis, an inflammatory cell death, is involved in the progression of atherosclerosis. Pyroptosis in endothelial cells (ECs) and its underlying mechanisms in atherosclerosis are poorly understood. Here, we investigated the role of a caspase-4/5-NF-κB pathway in pyroptosis in palmitic acid (PA)-stimulated ECs and EVs as players in pyroptosis. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured in an endothelial cell medium, treated with Ox-LDL, PA, caspase-4/5 inhibitor, NF-κB inhibitor, and sEV release inhibitor for 24 h, respectively. The cytotoxicity of PA was determined using an MTT assay, cell migration using a scratch-wound-healing assay, cell morphology using bright field microscopy, and lipid deposition using oil red O staining. The mRNA and protein expression of GSDM-D, CASP4, CASP5, NF-κB, NLRP3, IL-1β, and IL-18 were determined with RT-PCR and Western blot. Immunofluorescence was used to determine NLRP3 and ICAM-1 expressions. Extracellular vesicles (EVs) were isolated using an exosome isolation kit and were characterized by Western blot and scanning electron microscopy. Results: PA stimulation significantly changed the morphology of the HUVECs characterized by cell swelling, plasma membrane rupture, and increased LDH release, which are features of pyroptosis. PA significantly increased lipid accumulation and reduced cell migration. PA also triggered inflammation and endothelial dysfunction, as evidenced by NLRP3 activation, upregulation of ICAM-1 (endothelial activation marker), and pyroptotic markers (NLRP3, GSDM-D, IL-1β, IL-18). Inhibition of caspase-4/5 (Ac-FLTD-CMK) and NF-κB (trifluoroacetate salt (TFA)) resulted in a significant reduction in LDH release and expression of caspase-4/5, NF-κB, and gasdermin D (GSDM-D) in PA-treated HUVECs. Furthermore, GW4869, an exosome release inhibitor, markedly reduced LDH release in PA-stimulated HUVECs. EVs derived from PA-treated HUVECs exacerbated pyroptosis, as indicated by significantly increased LDH release and augmented expression of GSDM-D, NF-κB. Conclusions: The present study revealed that inflammatory, non-canonical caspase-4/5-NF-κB signaling may be one of the crucial mechanistic pathways associated with pyroptosis in ECs, and pyroptotic EVs facilitated pyroptosis in normal ECs during atherosclerosis.
Collapse
Affiliation(s)
- Salman Shamas
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Razia Rashid Rahil
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Laveena Kaushal
- Department of Dermatology, Venereology & Leprology, Postgraduate Institute for Medical Education and Research, Chandigarh 160012, India; (L.K.); (V.K.S.)
| | - Vinod Kumar Sharma
- Department of Dermatology, Venereology & Leprology, Postgraduate Institute for Medical Education and Research, Chandigarh 160012, India; (L.K.); (V.K.S.)
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Shabir H. Qureshi
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.F.A.); (S.M.A.)
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.F.A.); (S.M.A.)
| | - Mohammad Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| |
Collapse
|
9
|
Ajoolabady A, Pratico D, Lin L, Mantzoros CS, Bahijri S, Tuomilehto J, Ren J. Inflammation in atherosclerosis: pathophysiology and mechanisms. Cell Death Dis 2024; 15:817. [PMID: 39528464 PMCID: PMC11555284 DOI: 10.1038/s41419-024-07166-8] [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: 02/21/2024] [Revised: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ling Lin
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | | | - Suhad Bahijri
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Jaakko Tuomilehto
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Public Health, University of Helsinki, Helsinki, Finland.
- Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
10
|
Wu Y, Sun X, Cui G, Wang S. Mir-150-5p distinguishes acute pulmonary embolism, predicts the occurrence of pulmonary arterial hypertension, and regulates ox-LDL-induced endothelial cell injury. Hereditas 2024; 161:33. [PMID: 39256826 PMCID: PMC11384695 DOI: 10.1186/s41065-024-00333-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/24/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Acute pulmonary embolism (APE) is a major type of venous thromboembolism (VTE) with a high risk of mortality and disability. There is a lack of biomarkers for APE to indicate deteriorating development and predict adverse outcomes. This study evaluated the significance of miR-150-5p in APE aiming to explore a novel potential biomarker for APE. METHODS The study enrolled APE (n = 137) and deep wein thrombosis (DVT, n = 67) patients and collected plasma samples from all study subjects. The expression of miR-150-5p was analyzed by PCR and its significance in screening APE and pulmonary arterial hypertension (PAH) was assessed by receiver operating curve (ROC) and logistic analyses. The study established oxidized low-density lipoprotein (ox-LDL)-induced human venous endothelial cells (HUVECs). Through cell transfection combined with cell counting kit-8 (CCK8), flow cytometry, and enzyme-linked immunosorbent assay (ELISA), the effect of miR-150-5p on ox-LDL-induced HUVEC injury was evaluated. RESULTS Significant downregulation of miR-150-5p was observed in the plasma of APE patients compared with DVT patients (P < 0.0001). The plasma miR-150-5p levels in APE patients occurred PAH was much lower than in patients without PAH (P < 0.0001). Reducing miR-150-5p distinguished APE patients from DVT patients (AUC = 0.912) and was identified as a risk factor for the occurrence of PAH in APE patients (OR = 0.385, P = 0.010). In HUVECs, oxidized low-density lipoprotein (ox-LDL) caused inhibited cell proliferation, enhanced apoptosis, increased pro-inflammatory cytokines, reactive oxygen species (ROS), malondialdehyde (MDA), and decreased superoxide dismutase (SOD). Overexpressing miR-150-5p could promote proliferation, inhibit apoptosis, and alleviate inflammation and oxidative stress of ox-LDL-treated HUVECs. CONCLUSIONS Downregulated plasma miR-150-5p served as a diagnostic biomarker for APE and predicted the predisposition of PAH in APE patients. Overexpressing miR-150-5p could alleviate ox-LDL-induced endothelial cell injury in HUVECs.
Collapse
Affiliation(s)
- Yue Wu
- Department of Vascular Surgery, Zibo Central Hospital, Zibo, 255020, Zibo, China
| | - Xin Sun
- Department of Cardiothoracic Surgery, Zibo Central Hospital, Zibo, 255020, Zibo, China
| | - Guangqiang Cui
- Department of Cardiothoracic Surgery, Zibo Central Hospital, Zibo, 255020, Zibo, China
| | - Shu Wang
- Department of Respiratory and Critical Care Medicine, Zibo Central Hospital, No. 54, Gongqingtuan West Road, Zhangdian District, Zibo, 255020, Shandong, China.
| |
Collapse
|
11
|
Chambers KL, Myerscough MR, Watson MG, Byrne HM. Blood Lipoproteins Shape the Phenotype and Lipid Content of Early Atherosclerotic Lesion Macrophages: A Dual-Structured Mathematical Model. Bull Math Biol 2024; 86:112. [PMID: 39093509 PMCID: PMC11297092 DOI: 10.1007/s11538-024-01342-9] [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: 04/10/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Macrophages in atherosclerotic lesions exhibit a spectrum of behaviours or phenotypes. The phenotypic distribution of monocyte-derived macrophages (MDMs), its correlation with MDM lipid content, and relation to blood lipoprotein densities are not well understood. Of particular interest is the balance between low density lipoproteins (LDL) and high density lipoproteins (HDL), which carry bad and good cholesterol respectively. To address these issues, we have developed a mathematical model for early atherosclerosis in which the MDM population is structured by phenotype and lipid content. The model admits a simpler, closed subsystem whose analysis shows how lesion composition becomes more pathological as the blood density of LDL increases relative to the HDL capacity. We use asymptotic analysis to derive a power-law relationship between MDM phenotype and lipid content at steady-state. This relationship enables us to understand why, for example, lipid-laden MDMs have a more inflammatory phenotype than lipid-poor MDMs when blood LDL lipid density greatly exceeds HDL capacity. We show further that the MDM phenotype distribution always attains a local maximum, while the lipid content distribution may be unimodal, adopt a quasi-uniform profile or decrease monotonically. Pathological lesions exhibit a local maximum in both the phenotype and lipid content MDM distributions, with the maximum at an inflammatory phenotype and near the lipid content capacity respectively. These results illustrate how macrophage heterogeneity arises in early atherosclerosis and provide a framework for future model validation through comparison with single-cell RNA sequencing data.
Collapse
Affiliation(s)
- Keith L Chambers
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK.
| | - Mary R Myerscough
- School of Mathematics and Statistics, University of Sydney, Carslaw Building, Eastern Avenue, Camperdown, Sydney, NSW, 2006, Australia
| | - Michael G Watson
- School of Mathematics and Statistics, University of New South Wales, Anita B. Lawrence Centre, University Mall, UNSW, Kensington, Sydney, NSW, 2052, Australia
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Headington, Oxford, Oxfordshire, OX3 7DQ, UK
| |
Collapse
|
12
|
Huang Z, Zhou Z, Ma Y, Hu YM. Mito-Tempo alleviates ox-LDL-provoked foam cell formation by regulating Nrf2/NLRP3 signaling. Biosci Biotechnol Biochem 2024; 88:759-767. [PMID: 38719485 DOI: 10.1093/bbb/zbae058] [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: 11/30/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024]
Abstract
Our previous studies have demonstrated that Mito-Tempol (also known as 4-hydroxy-Tempo), a mitochondrial reactive oxygen species scavenger, alleviates oxidized low-density lipoprotein (ox-LDL)-triggered foam cell formation. Given the effect of oxidative stress on activating the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome, which promotes foam cell formation, we aimed to explore whether Mito-Tempo inhibits ox-LDL-triggered foam cell formation by regulating NLRP3 inflammasome. The results revealed that Mito-Tempo re-activated Nrf2 and alleviated macrophage foam cell formation induced by ox-LDL, whereas the effects were reversed by ML385 (a specific Nrf2 inhibitor). Mito-Tempo restored the expression and nuclear translocation of Nrf2 by decreasing ox-LDL-induced ubiquitination. Furthermore, Mito-Tempo suppressed ox-LDL-triggered NLRP3 inflammasome activation and subsequent pyroptosis, whereas the changes were blocked by ML385. Mito-Tempo decreased lipoprotein uptake by inhibiting CD36 expression and suppressed foam cell formation by regulating the NLRP3 inflammasome. Taken together, Mito-Tempo exhibits potent anti-atherosclerotic effects by regulating Nrf2/NLRP3 signaling.
Collapse
Affiliation(s)
- Zhenyu Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhaoli Zhou
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Scientific Research Center, Shanghai University of Medicine & Health Science, Shanghai, China
- Department of Pharmacology, School of Pharmacy, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Ying Ma
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao-Min Hu
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Echrish J, Pasca MI, Cabrera D, Yang Y, Harper AGS. Developing a Biomimetic 3D Neointimal Layer as a Prothrombotic Substrate for a Humanized In Vitro Model of Atherothrombosis. Biomimetics (Basel) 2024; 9:372. [PMID: 38921252 PMCID: PMC11201422 DOI: 10.3390/biomimetics9060372] [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: 04/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Acute cardiovascular events result from clots caused by the rupture and erosion of atherosclerotic plaques. This paper aimed to produce a functional biomimetic hydrogel of the neointimal layer of the atherosclerotic plaque that can support thrombogenesis upon exposure to human blood. A biomimetic hydrogel of the neointima was produced by culturing THP-1-derived foam cells within 3D collagen hydrogels in the presence or absence of atorvastatin. Prothrombin time and platelet aggregation onset were measured after exposure of the neointimal models to platelet-poor plasma and washed platelet suspensions prepared from blood of healthy, medication-free volunteers. Activity of the extrinsic coagulation pathway was measured using the fluorogenic substrate SN-17. Foam cell formation was observed following preincubation of the neointimal biomimetic hydrogels with oxidized LDL, and this was inhibited by pretreatment with atorvastatin. The neointimal biomimetic hydrogel was able to trigger platelet aggregation and blood coagulation upon exposure to human blood products. Atorvastatin pretreatment of the neointimal biomimetic layer significantly reduced its pro-aggregatory and pro-coagulant properties. In the future, this 3D neointimal biomimetic hydrogel can be incorporated as an additional layer within our current thrombus-on-a-chip model to permit the study of atherosclerosis development and the screening of anti-thrombotic drugs as an alternative to current animal models.
Collapse
Affiliation(s)
| | | | - David Cabrera
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK; (D.C.); (Y.Y.)
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK; (D.C.); (Y.Y.)
| | | |
Collapse
|
14
|
Wang J, Cao H, Yang H, Wang N, Weng Y, Luo H. The function of CD36 in Mycobacterium tuberculosis infection. Front Immunol 2024; 15:1413947. [PMID: 38881887 PMCID: PMC11176518 DOI: 10.3389/fimmu.2024.1413947] [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: 04/08/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
CD36 is a scavenger receptor that has been reported to function as a signaling receptor that responds to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) and could integrate metabolic pathways and cell signaling through its dual functions. Thereby influencing activation to regulate the immune response and immune cell differentiation. Recent studies have revealed that CD36 plays critical roles in the process of lipid metabolism, inflammatory response and immune process caused by Mycobacterium tuberculosis infection. This review will comprehensively investigate CD36's functions in lipid uptake and processing, inflammatory response, immune response and therapeutic targets and biomarkers in the infection process of M. tuberculosis. The study also raised outstanding issues in this field to designate future directions.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Hui Cao
- Department of Food and Nutrition Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, China
| | - Hongwei Yang
- Department of Clinical Laboratory, Suzhou BOE Hospital, Suzhou, Jiangsu, China
| | - Nan Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Yiwei Weng
- Department of Clinical Laboratory, The Fourth People’s Hospital of Kunshan, Suzhou, Jiangsu, China
| | - Hao Luo
- Department of Clinical Laboratory, The Second People's Hospital of Kunshan, Suzhou, China
| |
Collapse
|
15
|
Yu X, Qiang W, Gong K, Cao Y, Yan S, Gao G, Tao F, Zhu B. No role of the third-trimester inflammatory factors in the association of gestational diabetes mellitus with postpartum cardiometabolic indicators. BMC Pregnancy Childbirth 2024; 24:361. [PMID: 38750471 PMCID: PMC11095010 DOI: 10.1186/s12884-024-06563-3] [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: 09/26/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND The influence of gestational diabetes mellitus (GDM) on postpartum cardiometabolic indicators is primarily restricted to glucose and lipid metabolism, however the indicators for liver and kidney function have been rarely explored, and the role of the third-trimester inflammatory factors in these associations has never been investigated. METHODS Based on the Ma'anshan birth cohort (MABC), women with or without GDM history were selected and invited to participate in a 6-year postpartum follow-up. The fasting blood samples were collected to measure 16 comprehensive metabolic indicators during a 6-year postpartum follow-up: fasting plasma glucose (FPG), glycosylated hemoglobin (HbA1c), triglycerides (TG), total cholesterol (TC), uric acid (UA), blood urea nitrogen (BUN), serum creatinine (SCR), etc. Seven inflammatory factors, including TNF-α, IFN-γ, IL-1β, IL-6, IL-10, IL-12p70, and IL-17 A, were measured with serum samples collected during the third trimester of pregnancy. Linear regression models were used to analyze the associations between GDM and 6-year postpartum metabolic indicators, GDM and third-trimester inflammatory factors, and the third-trimester inflammatory factors and 6-year postpartum metabolic indicators. Mediating and moderating effect analyses were further performed to explore if the third-trimester inflammatory factors mediate or modify the association between GDM and postpartum cardiometabolic indicators. RESULTS From July 2021 to August 2022, 307 participants have been followed up, with 99 women with a prior GDM history. Compared with those without GDM, individuals with a prior history of GDM had significantly elevated levels of FPG (β = 0.40, 95% CI: 0.18 to 0.62, PFDR < 0.001), HbA1c (β = 0.22, 95% CI: 0.09 to 0.34, PFDR = 0.009), TyG (β = 0.22, 95% CI: 0.07 to 0.37, PFDR = 0.024) at 6 years postpartum, and the association between GDM and SCR (β = 2.43, 95% CI: 0.02 to 4.85, PFDR = 0.144) reached nominal significance level. GDM history was associated with a decreased level of third-trimester IL-17 A (β = -0.58, 95% CI: -0.99 to -0.18, PFDR = 0.035). No significant association between third-trimester inflammatory factors and 6-year postpartum metabolic indicators was observed. And no mediating or moderating effect of third-trimester inflammatory factors was observed in those associations. CONCLUSION A prior history of GDM was significantly associated with elevated FPG, HbA1c, and TyG in women at 6 years postpartum, whereas third-trimester inflammatory factors had no role in mediating or moderating these associations.
Collapse
Affiliation(s)
- Xiayan Yu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Wenjing Qiang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Kexin Gong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Yidan Cao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Shuangqin Yan
- Ma'anshan Maternal and Child Healthcare (MCH) Center, Ma'anshan, 243011, China
| | - Guopeng Gao
- Ma'anshan Maternal and Child Healthcare (MCH) Center, Ma'anshan, 243011, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Beibei Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Anhui Provincial Key Laboratory of Environment and Population Health across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui, 230032, China.
| |
Collapse
|
16
|
Chen J, Sun X, Liu Y, Zhang Y, Zhao M, Shao L. SENP3 attenuates foam cell formation by deSUMOylating NLRP3 in macrophages stimulated with ox-LDL. Cell Signal 2024; 117:111092. [PMID: 38331013 DOI: 10.1016/j.cellsig.2024.111092] [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: 08/22/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
SUMO-specific protease 3 (SENP3) participates in the removal of SUMOylation and maintains the balance of the SUMO system, which ensures normal functioning of substrates and cellular activities. In the present study, we found that SENP3 expression was significantly reduced in ox-LDL-stimulated macrophages. SENP3 overexpression suppressed and SENP3 knockdown promoted macrophage foam cell formation. Moreover, SENP3 inhibited cholesterol uptake, CD36 expression, and NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome activation in ox-LDL-stimulated macrophages. Ox-LDL-stimulated NLRP3 SUMOylation was reduced by SENP3. Blocking NLRP3 SUMOylation inhibited foam cell formation and NLRP3 inflammasome activation. Thus, this study revealed that SENP3 inhibits macrophage foam cell formation by deSUMOylating NLRP3 and regulating NLRP3 inflammasome activation, which may provide a potentially innovative approach to treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jiaojiao Chen
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuan Sun
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuan Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuze Zhang
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Min Zhao
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China.
| | - Luyao Shao
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China.
| |
Collapse
|
17
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
18
|
Li X, Dong X, Zhang L, Zhang S, Huang W, Wang C, Huo Z, Li X, Zhang X, Jia X, Chen G, Kuang B. Astragaloside IV attenuates renal tubule injury in DKD rats via suppression of CD36-mediated NLRP3 inflammasome activation. Front Pharmacol 2024; 15:1285797. [PMID: 38572426 PMCID: PMC10987761 DOI: 10.3389/fphar.2024.1285797] [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: 08/30/2023] [Accepted: 03/01/2024] [Indexed: 04/05/2024] Open
Abstract
Background In recent years, diabetic kidney disease (DKD) has emerged as a prominent factor contributing to end-stage renal disease. Tubulointerstitial inflammation and lipid accumulation have been identified as key factors in the development of DKD. Earlier research indicated that Astragaloside IV (AS-IV) reduces inflammation and oxidative stress, controls lipid accumulation, and provides protection to the kidneys. Nevertheless, the mechanisms responsible for its protective effects against DKD have not yet been completely elucidated. Purpose The primary objective of this research was to examine the protective properties of AS-IV against DKD and investigate the underlying mechanism, which involves CD36, reactive oxygen species (ROS), NLR family pyrin domain containing 3 (NLRP3), and interleukin-1β (IL-1β). Methods The DKD rat model was created by administering streptozotocin along with a high-fat diet. Subsequently, the DKD rats and palmitic acid (PA)-induced HK-2 cells were treated with AS-IV. Atorvastatin was used as the positive control. To assess the therapeutic effects of AS-IV on DKD, various tests including blood sugar levels, the lipid profile, renal function, and histopathological examinations were conducted. The levels of CD36, ROS, NLRP3, Caspase-1, and IL-1β were detected using western blot analysis, PCR, and flow cytometry. Furthermore, adenovirus-mediated CD36 overexpression was applied to explore the underlying mechanisms through in vitro experiments. Results In vivo experiments demonstrated that AS-IV significantly reduced hyperglycemia, dyslipidemia, urinary albumin excretion, and serum creatinine levels in DKD rats. Additionally, it improved renal structural abnormalities and suppressed the expression of CD36, NLRP3, IL-1β, TNF-α, and MCP-1. In vitro experiments showed that AS-IV decreased CD36 expression, lipid accumulation, and lipid ROS production while inhibiting NLRP3 activation and IL-1β secretion in PA-induced HK-2 cells. Conclusion AS-IV alleviated renal tubule interstitial inflammation and tubule epithelial cell apoptosis in DKD rats by inhibiting CD36-mediated lipid accumulation and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xianhong Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Dong
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangyou Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shu Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiying Huang
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, China
| | - Chao Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhihao Huo
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Li
- Shenzhen Luohu District Traditional Chinese Medical Hospital, Shenzhen, China
| | - Xiwen Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaotong Jia
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gangyi Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Kuang
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, China
| |
Collapse
|
19
|
Attiq A, Afzal S, Ahmad W, Kandeel M. Hegemony of inflammation in atherosclerosis and coronary artery disease. Eur J Pharmacol 2024; 966:176338. [PMID: 38242225 DOI: 10.1016/j.ejphar.2024.176338] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/30/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Inflammation drives coronary artery disease and atherosclerosis implications. Lipoprotein entry, retention, and oxidative modification cause endothelial damage, triggering innate and adaptive immune responses. Recruited immune cells orchestrate the early atherosclerotic lesions by releasing proinflammatory cytokines, expediting the foam cell formation, intraplaque haemorrhage, secretion of matrix-degrading enzymes, and lesion progression, eventually promoting coronary artery syndrome via various inflammatory cascades. In addition, soluble mediators disrupt the dynamic anti- and prothrombotic balance maintained by endothelial cells and pave the way for coronary artery disease such as angina pectoris. Recent studies have established a relationship between elevated levels of inflammatory markers, including C-reactive protein (CRP), interleukins (IL-6, IL-1β), and tumour necrosis factor-alpha (TNF-α) with the severity of CAD and the possibility of future cardiovascular events. High-sensitivity C-reactive protein (hs-CRP) is a marker for assessing systemic inflammation and predicting the risk of developing CAD based on its peak plasma levels. Hence, understanding cross-talk interactions of inflammation, atherogenesis, and CAD is highly warranted to recalculate the risk factors that activate and propagate arterial lesions and devise therapeutic strategies accordingly. Cholesterol-inflammation lowering agents (statins), monoclonal antibodies targeting IL-1 and IL-6 (canakinumab and tocilizumab), disease-modifying antirheumatic drugs (methotrexate), sodium-glucose transport protein-2 (SGLT2) inhibitors, colchicine and xanthene oxidase inhibitor (allopurinol) have shown promising results in reducing inflammation, regressing atherogenic plaque and modifying the course of CAD. Here, we review the complex interplay between inflammatory, endothelial, smooth muscle and foam cells. Moreover, the putative role of inflammation in atherosclerotic CAD, underlying mechanisms and potential therapeutic implications are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Sheryar Afzal
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, 31982, Al Ahsa, Saudi Arabia.
| | - Waqas Ahmad
- Discipline of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, 31982, Al Ahsa, Saudi Arabia
| |
Collapse
|
20
|
Wu X, Yang J, Wu J, Yang X. Therapeutic potential of MCC950, a specific inhibitor of NLRP3 inflammasome in systemic lupus erythematosus. Biomed Pharmacother 2024; 172:116261. [PMID: 38340397 DOI: 10.1016/j.biopha.2024.116261] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder with a pathogenesis that remains incompletely understood, resulting in limited treatment options. MCC950, a highly specific NLRP3 inflammasome inhibitor, effectively suppresses the activation of NLRP3, thus reducing the production of caspase-1, the pro-inflammatory cytokines IL-1β and IL-18. This review highlights the pivotal role of NLRP3 inflammasome activation pathways in the pathogenesis of SLE and discusses the potential therapeutic application of MCC950 in SLE. Notably, it comprehensively elucidates the mechanism of MCC950 targeting the NLRP3 pathway in SLE treatment, outlining its potential role in regulating autophagy and necroptosis. The insights gained contribute to a deeper understanding of the value of MCC950 in SLE therapy, serving as a robust foundation for further research and potential clinical applications.
Collapse
Affiliation(s)
- Xiaoxiao Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China
| | - Junhao Yang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155North Nanjing Street, Heping District, Shenyang 110001, China
| | - Juanjie Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China
| | - Xuyan Yang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China.
| |
Collapse
|
21
|
Wang H, Wang L, Liu Y, Men W, Hao W, Fang C, Li C, Zhang L. Plasma levels of CD36 and glutathione as biomarkers for ruptured intracranial aneurysm. Open Life Sci 2023; 18:20220757. [PMID: 38196515 PMCID: PMC10775171 DOI: 10.1515/biol-2022-0757] [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: 06/16/2023] [Revised: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 01/11/2024] Open
Abstract
Evidence has proved that intracranial aneurysm (IA) formation and rupture might be closely related to inflammatory response and oxidative stress. Our objective was to evaluate the potential of CD36 and glutathione (GSH) as biomarkers for IA. In this study, the enzyme-linked immunosorbent assay was used to measure the plasma levels of CD36 and GSH in 30 IA patients and 30 healthy controls. Then, correlation analysis, receiver operating characteristic (ROC) curve, and logistic regression analysis were performed. The results showed that the plasma level of CD36 in IA patients was significantly higher than that in the control group (P < 0.0001), and plasma GSH was significantly lower compared with that in the control group (P < 0.0001). ROC analysis showed that CD36 and GSH had high sensitivity (90.0 and 96.6%) and specificity (96.6 and 86.6%) for IA diagnosis. The combined sensitivity and specificity achieved were 100 and 100%, respectively. The plasma levels of CD36 and GSH did not show a significant correlation with age, the Glasgow Coma Scale, Hunter-Hess score, aneurysm size, aneurysm height, aneurysm neck, and aspect ratio. The AUC of the logistic regression model based on CD36 and GSH was 0.505. Our results suggested that the combination of plasma CD36 and GSH could serve as potential biomarkers for IA rupture.
Collapse
Affiliation(s)
- Hanbin Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Luxuan Wang
- Department of Neurological Function Examination, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Yunmei Liu
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Weidong Men
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Wanjiao Hao
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Chuan Fang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Chunhui Li
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| | - Lijian Zhang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Postdoctoral Research Station of Neurosurgery, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, Hebei Province, China
| |
Collapse
|
22
|
Krantz M, Eklund D, Särndahl E, Hedbrant A. A detailed molecular network map and model of the NLRP3 inflammasome. Front Immunol 2023; 14:1233680. [PMID: 38077364 PMCID: PMC10699087 DOI: 10.3389/fimmu.2023.1233680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/16/2023] [Indexed: 12/18/2023] Open
Abstract
The NLRP3 inflammasome is a key regulator of inflammation that responds to a broad range of stimuli. The exact mechanism of activation has not been determined, but there is a consensus on cellular potassium efflux as a major common denominator. Once NLRP3 is activated, it forms high-order complexes together with NEK7 that trigger aggregation of ASC into specks. Typically, there is only one speck per cell, consistent with the proposal that specks form - or end up at - the centrosome. ASC polymerisation in turn triggers caspase-1 activation, leading to maturation and release of IL-1β and pyroptosis, i.e., highly inflammatory cell death. Several gain-of-function mutations in the NLRP3 inflammasome have been suggested to induce spontaneous activation of NLRP3 and hence contribute to development and disease severity in numerous autoinflammatory and autoimmune diseases. Consequently, the NLRP3 inflammasome is of significant clinical interest, and recent attention has drastically improved our insight in the range of involved triggers and mechanisms of signal transduction. However, despite recent progress in knowledge, a clear and comprehensive overview of how these mechanisms interplay to shape the system level function is missing from the literature. Here, we provide such an overview as a resource to researchers working in or entering the field, as well as a computational model that allows for evaluating and explaining the function of the NLRP3 inflammasome system from the current molecular knowledge. We present a detailed reconstruction of the molecular network surrounding the NLRP3 inflammasome, which account for each specific reaction and the known regulatory constraints on each event as well as the mechanisms of drug action and impact of genetics when known. Furthermore, an executable model from this network reconstruction is generated with the aim to be used to explain NLRP3 activation from priming and activation to the maturation and release of IL-1β and IL-18. Finally, we test this detailed mechanistic model against data on the effect of different modes of inhibition of NLRP3 assembly. While the exact mechanisms of NLRP3 activation remains elusive, the literature indicates that the different stimuli converge on a single activation mechanism that is additionally controlled by distinct (positive or negative) priming and licensing events through covalent modifications of the NLRP3 molecule. Taken together, we present a compilation of the literature knowledge on the molecular mechanisms on NLRP3 activation, a detailed mechanistic model of NLRP3 activation, and explore the convergence of diverse NLRP3 activation stimuli into a single input mechanism.
Collapse
Affiliation(s)
- Marcus Krantz
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Daniel Eklund
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Alexander Hedbrant
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| |
Collapse
|
23
|
Hoeffner N, Paul A, Goo YH. Drug screen identifies verteporfin as a regulator of lipid metabolism in macrophage foam cells. Sci Rep 2023; 13:19588. [PMID: 37949969 PMCID: PMC10638409 DOI: 10.1038/s41598-023-46467-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Arterial macrophage foam cells are filled with cholesterol ester (CE) stored in cytosolic lipid droplets (LDs). Foam cells are central players in progression of atherosclerosis as regulators of lipid metabolism and inflammation, two major driving forces of atherosclerosis development. Thus, foam cells are considered plausible targets for intervention in atherosclerosis. However, a compound that directly regulates the lipid metabolism of LDs in the arterial foam cells has not yet been identified. In this study, we screened compounds that inhibit macrophage foam cell formation using a library of 2697 FDA-approved drugs. From the foam cells generated via loading of human oxidized low-density lipoprotein (oxLDL), we found 21 and 6 compounds that reduced and enhanced accumulations of lipids respectively. Among them, verteporfin most significantly reduced oxLDL-induced foam cell formation whereas it did not display a significant impact on foam cell formation induced by fatty acid. Mechanistically our data demonstrate that verteporfin acts via inhibition of oxLDL association with macrophages, reducing accumulation of CE. Interestingly, while other drugs that reduced foam cell formation did not have impact on pre-existing foam cells, verteporfin treatment significantly reduced their total lipids, CE, and pro-inflammatory gene expression. Together, our study identifies verteporfin as a novel regulator of foam cell lipid metabolism and inflammation and a potential compound for intervention in atherosclerosis.
Collapse
Affiliation(s)
- Nicholas Hoeffner
- Molecular and Cellular Physiology Department, Albany Medical College, Albany, NY, 12208, USA
| | - Antoni Paul
- Molecular and Cellular Physiology Department, Albany Medical College, Albany, NY, 12208, USA
| | - Young-Hwa Goo
- Molecular and Cellular Physiology Department, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
24
|
Chen CC, Lii CK, Liu KL, Lin YL, Lo CW, Li CC, Yang YC, Chen HW. Andrographolide Attenuates Oxidized LDL-Induced Activation of the NLRP3 Inflammasome in Bone Marrow-Derived Macrophages and Mitigates HFCCD-Induced Atherosclerosis in Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:2175-2193. [PMID: 37930331 DOI: 10.1142/s0192415x23500933] [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: 11/07/2023]
Abstract
Andrographolide (AND) is a bioactive component of the herb Andrographis paniculata and a well-known anti-inflammatory agent. Atherosclerosis is a chronic inflammatory disease of the vasculature, and oxidized LDL (oxLDL) is thought to contribute heavily to atherosclerosis-associated inflammation. The aim of this study was to investigate whether AND mitigates oxLDL-mediated foam cell formation and diet-induced atherosclerosis (in mice fed a high-fat, high-cholesterol, high-cholic acid [HFCCD] diet) and the underlying mechanisms involved. AND attenuated LPS/oxLDL-mediated foam cell formation, IL-1[Formula: see text] mRNA and protein (p37) expression, NLR family pyrin domain containing 3 (NLRP3) mRNA and protein expression, caspase-1 (p20) protein expression, and IL-1[Formula: see text] release in BMDMs. Treatment with oxLDL significantly induced protein and mRNA expression of CD36, lectin-like oxLDL receptor-1 (LOX-1), and scavenger receptor type A (SR-A), whereas pretreatment with AND significantly inhibited protein and mRNA expression of SR-A only. Treatment with oxLDL significantly induced ROS generation and Dil-oxLDL uptake; however, pretreatment with AND alleviated oxLDL-induced ROS generation and Dil-oxLDL uptake. HFCCD feeding significantly increased aortic lipid accumulation, ICAM-1 expression, and IL-1[Formula: see text] mRNA expression, as well as blood levels of glutamic pyruvic transaminase (GPT), total cholesterol, and LDL-C. AND co-administration mitigated aortic lipid accumulation, the protein expression of ICAM-1, mRNA expression of IL-1[Formula: see text] and ICAM-1, and blood levels of GPT. These results suggest that the working mechanisms by which AND mitigates atherosclerosis involve the inhibition of foam cell formation and NLRP3 inflammasome-dependent vascular inflammation as evidenced by decreased SR-A expression and IL-1[Formula: see text] release, respectively.
Collapse
Affiliation(s)
- Chih-Chieh Chen
- Department of Sports Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung 406040, Taiwan
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413305, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung 402306, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
| | - Yi-Ling Lin
- Department of Nutrition, China Medical University, Taichung 406040, Taiwan
| | - Chia-Wen Lo
- Department of Nutrition, China Medical University, Taichung 406040, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung 402306, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
| | - Ya-Chen Yang
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413305, Taiwan
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
25
|
Xu Y, Yang Y, Chen X, Jiang D, Zhang F, Guo Y, Hu B, Xu G, Peng S, Wu L, Hu J. NLRP3 inflammasome in cognitive impairment and pharmacological properties of its inhibitors. Transl Neurodegener 2023; 12:49. [PMID: 37915104 PMCID: PMC10621314 DOI: 10.1186/s40035-023-00381-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
Cognitive impairment is a multifactorial and multi-step pathological process that places a heavy burden on patients and the society. Neuroinflammation is one of the main factors leading to cognitive impairment. The inflammasomes are multi-protein complexes that respond to various microorganisms and endogenous danger signals, helping to initiate innate protective responses in inflammatory diseases. NLRP3 inflammasomes produce proinflammatory cytokines (interleukin IL-1β and IL-18) by activating caspase-1. In this review, we comprehensively describe the structure and functions of the NLRP3 inflammasome. We also explore the intrinsic relationship between the NLRP3 inflammasome and cognitive impairment, which involves immune cell activation, cell apoptosis, oxidative stress, mitochondrial autophagy, and neuroinflammation. Finally, we describe NLRP3 inflammasome antagonists as targeted therapies to improve cognitive impairment.
Collapse
Affiliation(s)
- Yi Xu
- The Second Affiliated Hospital of Nanchang University, Department of the Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Yanling Yang
- The Second Affiliated Hospital of Nanchang University, Department of the Second Clinical Medical College of Nanchang University, Nanchang, 330006, China
| | - Xi Chen
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Danling Jiang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fei Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yao Guo
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Bin Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Lidong Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Department of Thyroid and Hernia Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
26
|
Hutton M, Frazer M, Lin A, Patel S, Misra A. New Targets in Atherosclerosis: Vascular Smooth Muscle Cell Plasticity and Macrophage Polarity. Clin Ther 2023; 45:1047-1054. [PMID: 37709601 DOI: 10.1016/j.clinthera.2023.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Despite an increase in treatment options, and substantial reductions in cardiovascular mortality over the past half-century, atherosclerosis remains the most prevalent cause of premature mortality worldwide. The development of innovative new therapies is crucial to further minimize atherosclerosis-related deaths. The diverse array of cell phenotypes derived from vascular smooth muscle cells (SMCs) and macrophages within atherosclerotic plaques are increasingly becoming recognized for their beneficial and detrimental roles in plaque stability and disease burden. This review explores how contemporary transcriptomics and fate-mapping studies have revealed vascular cell plasticity as a relatively unexplored target for therapeutic intervention. METHODS Recent literature for this narrative review was obtained by searching electronic databases (ie, Google Scholar, PubMed). Additional studies were sourced from reference lists and the authors' personal databases. FINDINGS The lipid-rich and inflammatory plaque milieu induces SMC phenotypic switching to both beneficial and detrimental phenotypes. Likewise, macrophage heterogeneity increases with disease burden to a variety of pro-inflammatory and anti-inflammatory activation states. These vascular cell phenotypes are determinants of plaque structure stability, and it is therefore highly likely that they influence clinical outcomes. Development of clinical treatments targeting deleterious phenotypes or promoting pro-healing phenotypes remains in its infancy. However, existing treatments (statins) have shown beneficial effects toward macrophage polarization, providing a rationale for more targeted approaches. In contrast, beneficial SMC phenotypic modulation with these pharmacologic agents has yet to be achieved. The range of modulated vascular cell phenotypes provides a multitude of novel targets and the potential to reduce future adverse events. IMPLICATIONS Vascular cell phenotypic heterogeneity must continue to be explored to lower cardiovascular events in the future. The rapidly increasing weight of evidence surrounding the role of SMC plasticity and macrophage polarity in plaque vulnerability provides a strong foundation upon which development of new therapeutics must follow. This approach may prove to be crucial in reducing cardiovascular events and improving patient benefit in the future.
Collapse
Affiliation(s)
- Michael Hutton
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Madeleine Frazer
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
27
|
Zhao Y, Gan YH. Combination of hyperlipidemia and 17β-Estradiol induces TMJOA-like pathological changes in rats. Oral Dis 2023; 29:3640-3653. [PMID: 35765240 DOI: 10.1111/odi.14294] [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: 01/20/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE We explored whether hyperlipidemia or combination of hyperlipidemia and E2 could induce TMJOA. MATERIALS AND METHODS Four groups of female rats were treated with normal diet, normal diet with E2, high-fat diet, and high-fat diet with E2 (HFD/E2), respectively, to induce TMJOA till 8 weeks. Another three groups were then used for COX2 inhibitor celecoxib to block the induction of TMJOA. Primary condylar chondrocytes were treated with combination of E2, ox-LDL, and corresponding inhibitors for evaluating expressions of related molecules. RESULTS Condylar cartilage proliferation with plenty of chondrocyte apoptosis and increased staining for LOX1, nuclear NF-κB, IL-1β, and COX2 at 4 weeks and decreased condylar cartilage and increased subchondral bone density at 8 weeks were observed only in the HFD/E2 group. Celecoxib significantly alleviated the cartilage proliferation and apoptosis in the HFD/E2 group. Serum ox-LDL increased in both high-fat diet groups, while serum IL-1β increased only in the HFD/E2 group. Combination of E2 and ox-LDL synergistically induced expressions of LOX1, phosphorylated NF-κB, IL-1β, and COX2, while LOX1 inhibitor blocked the induction of phosphorylated NF-κB, and NF-κB inhibitor the induction of IL-1β, and IL-1β inhibitor the induction of COX2. CONCLUSION Combination of hyperlipidemia and E2-induced TMJOA-like pathological changes through LOX1/NF-κB/IL-1β/COX2-signaling pathway.
Collapse
Affiliation(s)
- Yan Zhao
- Central laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Oral & Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ye-Hua Gan
- Central laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Oral & Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Temporomandibular Disorders & Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
28
|
Ye M, Yang M, Dai W, Li H, Zhou X, Chen Y, He L. Targeting Renal Proximal Tubule Cells in Obesity-Related Glomerulopathy. Pharmaceuticals (Basel) 2023; 16:1256. [PMID: 37765062 PMCID: PMC10535317 DOI: 10.3390/ph16091256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
As a metabolic disorder, obesity can cause secondary kidney damage, which is called obesity-related glomerulopathy (ORG). As the incidence of obesity increases worldwide, so does the incidence of end-stage renal disease (ESRD) caused by ORGs. However, there is still a lack of effective strategies to prevent and delay the occurrence and development of ORG. Therefore, a deeper understanding and elaboration of the pathogenesis of ORG is conducive to the development of therapeutic drugs for ORG. Here, we review the characteristics of pathological lesions of ORG and describe the roles of lipid metabolism disorders and mitochondrial oxidative stress in the development of ORG. Finally, we summarize the current available drugs or compounds for the treatment of ORG and suggested that ameliorating renal lipid metabolism and mitochondrial function may be potential therapeutic targets for ORG.
Collapse
Affiliation(s)
- Muyao Ye
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (M.Y.); (M.Y.)
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (M.Y.); (M.Y.)
| | - Wenni Dai
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (M.Y.); (M.Y.)
| | - Hao Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (M.Y.); (M.Y.)
| | - Xun Zhou
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (M.Y.); (M.Y.)
| | - Yinyin Chen
- Department of Nephrology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410081, China
- Changsha Clinical Research, Changsha 410011, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (M.Y.); (M.Y.)
| |
Collapse
|
29
|
Zhang Y, Vandestienne M, Lavillegrand JR, Joffre J, Santos-Zas I, Lavelle A, Zhong X, Le Goff W, Guérin M, Al-Rifai R, Laurans L, Bruneval P, Guérin C, Diedisheim M, Migaud M, Puel A, Lanternier F, Casanova JL, Cochain C, Zernecke A, Saliba AE, Mokry M, Silvestre JS, Tedgui A, Mallat Z, Taleb S, Lenoir O, Vindis C, Camus SM, Sokol H, Ait-Oufella H. Genetic inhibition of CARD9 accelerates the development of atherosclerosis in mice through CD36 dependent-defective autophagy. Nat Commun 2023; 14:4622. [PMID: 37528097 PMCID: PMC10394049 DOI: 10.1038/s41467-023-40216-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 07/17/2023] [Indexed: 08/03/2023] Open
Abstract
Caspase recruitment-domain containing protein 9 (CARD9) is a key signaling pathway in macrophages but its role in atherosclerosis is still poorly understood. Global deletion of Card9 in Apoe-/- mice as well as hematopoietic deletion in Ldlr-/- mice increases atherosclerosis. The acceleration of atherosclerosis is also observed in Apoe-/-Rag2-/-Card9-/- mice, ruling out a role for the adaptive immune system in the vascular phenotype of Card9 deficient mice. Card9 deficiency alters macrophage phenotype through CD36 overexpression with increased IL-1β production, increased lipid uptake, higher cell death susceptibility and defective autophagy. Rapamycin or metformin, two autophagy inducers, abolish intracellular lipid overload, restore macrophage survival and autophagy flux in vitro and finally abolish the pro-atherogenic effects of Card9 deficiency in vivo. Transcriptomic analysis of human CARD9-deficient monocytes confirms the pathogenic signature identified in murine models. In summary, CARD9 is a key protective pathway in atherosclerosis, modulating macrophage CD36-dependent inflammatory responses, lipid uptake and autophagy.
Collapse
Affiliation(s)
- Yujiao Zhang
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Marie Vandestienne
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | | | - Jeremie Joffre
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Sorbonne Université, Paris, France
| | - Icia Santos-Zas
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Aonghus Lavelle
- Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology department, Paris, France
| | - Xiaodan Zhong
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Wilfried Le Goff
- Inserm UMRS1166, ICAN, Institute of CardioMetabolism and Nutrition, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Maryse Guérin
- Inserm UMRS1166, ICAN, Institute of CardioMetabolism and Nutrition, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Rida Al-Rifai
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Ludivine Laurans
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Patrick Bruneval
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Department of Anatomopathology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Coralie Guérin
- Institut Curie, Cytometry Platform, 75006, Paris, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), 37540 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades (INEM), Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, 75015, Paris, France
| | - Melanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Fanny Lanternier
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Imagine Institute, 75015, Paris, France
| | - Clément Cochain
- Comprehensive Heart Failure Center Wuerzburg, University Hospital Wuerzburg, Wuerzburg, Germany
- Institute of Experimental Biomedicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Wuerzburg, Germany
| | - Michal Mokry
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | | | - Alain Tedgui
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Ziad Mallat
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | - Soraya Taleb
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Olivia Lenoir
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | | | - Stéphane M Camus
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
| | - Harry Sokol
- Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology department, Paris, France
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Sorbonne Université, Paris, France.
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France.
| |
Collapse
|
30
|
Zhang X, Evans TD, Chen S, Sergin I, Stitham J, Jeong SJ, Rodriguez-Velez A, Yeh YS, Park A, Jung IH, Diwan A, Schilling JD, Rom O, Yurdagul A, Epelman S, Cho J, Lodhi IJ, Mittendorfer B, Razani B. Loss of Macrophage mTORC2 Drives Atherosclerosis via FoxO1 and IL-1β Signaling. Circ Res 2023; 133:200-219. [PMID: 37350264 PMCID: PMC10527041 DOI: 10.1161/circresaha.122.321542] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/12/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND The mTOR (mechanistic target of rapamycin) pathway is a complex signaling cascade that regulates cellular growth, proliferation, metabolism, and survival. Although activation of mTOR signaling has been linked to atherosclerosis, its direct role in lesion progression and in plaque macrophages remains poorly understood. We previously demonstrated that mTORC1 (mTOR complex 1) activation promotes atherogenesis through inhibition of autophagy and increased apoptosis in macrophages. METHODS Using macrophage-specific Rictor- and mTOR-deficient mice, we now dissect the distinct functions of mTORC2 pathways in atherogenesis. RESULTS In contrast to the atheroprotective effect seen with blockade of macrophage mTORC1, macrophage-specific mTORC2-deficient mice exhibit an atherogenic phenotype, with larger, more complex lesions and increased cell death. In cultured macrophages, we show that mTORC2 signaling inhibits the FoxO1 (forkhead box protein O1) transcription factor, leading to suppression of proinflammatory pathways, especially the inflammasome/IL (interleukin)-1β response, a key mediator of vascular inflammation and atherosclerosis. In addition, administration of FoxO1 inhibitors efficiently rescued the proinflammatory response caused by mTORC2 deficiency both in vitro and in vivo. Interestingly, collective deletion of macrophage mTOR, which ablates mTORC1- and mTORC2-dependent pathways, leads to minimal change in plaque size or complexity, reflecting the balanced yet opposing roles of these signaling arms. CONCLUSIONS Our data provide the first mechanistic details of macrophage mTOR signaling in atherosclerosis and suggest that therapeutic measures aimed at modulating mTOR need to account for its dichotomous functions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Trent D. Evans
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Sunny Chen
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Ismail Sergin
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | | | - Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - In-Hyuk Jung
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Joel D. Schilling
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Slava Epelman
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network and University of Toronto, Toronto, Canada
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Geriatrics and Nutritional Science, and Washington University School of Medicine, St Louis, MO, USA
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| |
Collapse
|
31
|
Wang L, Zhao X, Ding J, Liu Y, Liu H, Zheng L, Zhao H, Sun Z, Li K, Cai J, Qiao T. Oridonin attenuates the progression of atherosclerosis by inhibiting NLRP3 and activating Nrf2 in apolipoprotein E-deficient mice. Inflammopharmacology 2023:10.1007/s10787-023-01161-9. [PMID: 37155118 DOI: 10.1007/s10787-023-01161-9] [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: 11/16/2022] [Accepted: 02/08/2023] [Indexed: 05/10/2023]
Abstract
Oridonin, a well-known traditional Chinese herbal medicinal product isolated from Isodon rubescens (Hemsl.) H.Hara, has many potential properties, including anti-inflammatory and antioxidant activities. However, there is no evidence whether oridonin have a protective effect on atherosclerosis. This study focused on the effects of oridonin on oxidative stress and inflammation generated from atherosclerosis. The therapeutic effect on atherosclerosis was evaluated by intraperitoneal injection of oridonin in a high-fat fed ApoE-/- mouse model. We isolated mouse peritoneal macrophages and detected the effect of oridonin on oxidized low-density lipoprotein-induced lipid deposition. Oil red O staining, Masson's staining, dihydroethidium fluorescence staining, immunohistochemical staining, western blotting analysis, immunofluorescence, enzyme-linked immunosorbent assay and quantitative real-time PCR were used to evaluate the effect on atherosclerosis and explore the mechanisms. Oridonin treatment significantly alleviated the progression of atherosclerosis, reduced macrophage infiltration and stabilized plaques. Oridonin could significantly inhibit inflammation associated with NLRP3 activation. Oridonin significantly reduced oxidative stress by blocking Nrf2 ubiquitination and degradation. We also found that oridonin could prevent the formation of foam cells by increasing lipid efflux protein and reducing lipid uptake protein in macrophages. Oridonin has a protective effect on atherosclerosis in ApoE-/- mice, which may be related to the inhibition of NLRP3 and the stabilization of Nrf2. Therefore, oridonin may be a potential therapeutic agent for atherosclerosis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Xiaoqi Zhao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Jiawen Ding
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Yutong Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Han Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Lei Zheng
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Hongting Zhao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Zichen Sun
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Jing Cai
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China.
| | - Tong Qiao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, People's Republic of China.
| |
Collapse
|
32
|
Bartoszewicz M, Rać M. Prognostic Value of the Selected Polymorphisms in the CD36 Gene in the Domain-Encoding Lipid-Binding Region at a 10-Year Follow-Up for Early-Onset CAD Patients. Biomedicines 2023; 11:biomedicines11051332. [PMID: 37239003 DOI: 10.3390/biomedicines11051332] [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: 03/23/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The polymorphism of the CD36 gene may have a decisive impact on the formation and progression of atherosclerotic changes. The aim of the study was to confirm the prognostic values of the previously studied polymorphisms in the CD36 gene within a 10-year follow-up period. This is the first published report confirming the long-term observation of patients with CAD. The study group covered 100 early-onset CAD patients. It included 26 women not older than 55 years and 74 men not older than 50 years, tested in a ten-year study as a long-term follow-up after the first cardiovascular episode. There are no notable differences between the CD36 variants and the number of fatalities during observation, fatalities due to cardiological reasons, cases of myocardial infarction within a ten-year observation period, hospitalizations for cardiovascular issues, all cardiovascular occurrences, and the number of months lived. We have shown that the CD36 variants analyzed in this study do not appear to be related to the risk of early CAD occurrence in the Caucasian population in long-term observation.
Collapse
Affiliation(s)
- Michał Bartoszewicz
- Psychosocial and Medical Rehabilitation Centre, West Pomeranian Centre Oncology, 71-730 Szczecin, Poland
| | - Monika Rać
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-204 Szczecin, Poland
| |
Collapse
|
33
|
Alijagic A, Hedbrant A, Persson A, Larsson M, Engwall M, Särndahl E. NLRP3 inflammasome as a sensor of micro- and nanoplastics immunotoxicity. Front Immunol 2023; 14:1178434. [PMID: 37143682 PMCID: PMC10151538 DOI: 10.3389/fimmu.2023.1178434] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Micro- and nanoplastics (MNPs) are emerging pollutants with scarcely investigated effects on human innate immunity. If they follow a similar course of action as other, more thoroughly investigated particulates, MNPs may penetrate epithelial barriers, potentially triggering a cascade of signaling events leading to cell damage and inflammation. Inflammasomes are intracellular multiprotein complexes and stimulus-induced sensors critical for mounting inflammatory responses upon recognition of pathogen- or damage-associated molecular patterns. Among these, the NLRP3 inflammasome is the most studied in terms of activation via particulates. However, studies delineating the ability of MNPs to affect NLRP3 inflammasome activation are still rare. In this review, we address the issue of MNPs source and fate, highlight the main concepts of inflammasome activation via particulates, and explore recent advances in using inflammasome activation for assessment of MNP immunotoxicity. We also discuss the impact of co-exposure and MNP complex chemistry in potential inflammasome activation. Development of robust biological sensors is crucial in order to maximize global efforts to effectively address and mitigate risks that MNPs pose for human health.
Collapse
Affiliation(s)
- Andi Alijagic
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Alexander Hedbrant
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Alexander Persson
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Maria Larsson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Magnus Engwall
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, Sweden
| | - Eva Särndahl
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
34
|
The Development of Dyslipidemia in Chronic Kidney Disease and Associated Cardiovascular Damage, and the Protective Effects of Curcuminoids. Foods 2023; 12:foods12050921. [PMID: 36900438 PMCID: PMC10000737 DOI: 10.3390/foods12050921] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Chronic kidney disease (CKD) is a health problem that is constantly growing. This disease presents a diverse symptomatology that implies complex therapeutic management. One of its characteristic symptoms is dyslipidemia, which becomes a risk factor for developing cardiovascular diseases and increases the mortality of CKD patients. Various drugs, particularly those used for dyslipidemia, consumed in the course of CKD lead to side effects that delay the patient's recovery. Therefore, it is necessary to implement new therapies with natural compounds, such as curcuminoids (derived from the Curcuma longa plant), which can cushion the damage caused by the excessive use of medications. This manuscript aims to review the current evidence on the use of curcuminoids on dyslipidemia in CKD and CKD-induced cardiovascular disease (CVD). We first described oxidative stress, inflammation, fibrosis, and metabolic reprogramming as factors that induce dyslipidemia in CKD and their association with CVD development. We proposed the potential use of curcuminoids in CKD and their utilization in clinics to treat CKD-dyslipidemia.
Collapse
|
35
|
Werner LE, Wagner U. Calcium-sensing receptor-mediated NLRP3 inflammasome activation in rheumatoid arthritis and autoinflammation. Front Physiol 2023; 13:1078569. [PMID: 36685206 PMCID: PMC9854345 DOI: 10.3389/fphys.2022.1078569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/17/2022] [Indexed: 01/09/2023] Open
Abstract
The calcium-sensing receptor (CaSR) is expressed in many cell types - including immune cells and in particular circulating monocytes. Here, the receptor plays an important physiological role as a regulator of constitutive macropinocytosis. This review article provides an overview of the literature on the role of the calcium sensing receptor in the context of inflammatory processes. Special emphasis is laid upon the importance for monocytes in the context of rheumatoid arthritis. We have shown previously, that stimulation of the receptor by increased extracellular Ca2+ ([Ca2+]ex) triggers a pro-inflammatory response due to NLRP3 inflammasome assembly and interleukin (IL)-1β release. The underlying mechanism includes macropinocytosis of calciprotein particles (CPPs), which are taken up in a [Ca2+]ex-induced, CaSR dependent manner, and leads to strong IL-1β release. In rheumatoid arthritis (RA), this uptake and the resulting IL-1β release is significantly increased due to increased expression of the receptor. Moreover, increased [Ca2+]ex-induced CPP uptake and IL-1β release is associated with more active disease, while CaSR overexpression has been reported to be associated with cardiovascular complications of RA. Most importantly, however, in animal experiments with arthritic mice, increased local calcium concentrations are present, which in combination with release of fetuin-A from eroded bone could contribute to formation of CPPs. We propose, that increased [Ca2+]ex, CPPs and pro-inflammatory cytokines drive a vicious cycle of inflammation and bone destruction which in turn offers new potential therapeutic approaches.
Collapse
|
36
|
Wang Y, Fang D, Yang Q, You J, Wang L, Wu J, Zeng M, Luo M. Interactions between PCSK9 and NLRP3 inflammasome signaling in atherosclerosis. Front Immunol 2023; 14:1126823. [PMID: 36911736 PMCID: PMC9992811 DOI: 10.3389/fimmu.2023.1126823] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Atherosclerosis is an early pathological basis of numerous cardiovascular events that result in death or disability. Recent studies have described PCSK9 as a novel target for the treatment of atherosclerosis; PCSK9 is capable of degrading LDLR on the surface of hepatocytes through the regulation of lipid metabolism, and it can function as a novel inflammatory modulator in atherosclerosis. Inflammasomes are important intracellular multiprotein complexes that promote the inflammatory response in atherosclerosis. Among inflammasomes, the NLRP3 inflammasome is particularly notable because of its important role in the development of atherosclerotic disease. After activation, NLRP3 forms a complex with ASC and pro-caspase-1, converting pro-caspase-1 into activated caspase-1, which may trigger the release of IL-1β and IL-18 and contribute to the inflammatory response. Several recent studies have indicated that there may be interactions between PCSK9 and the NLRP3 inflammasome, which may contribute to the inflammatory response that drives atherosclerosis development and progression. On the one hand, the NLRP3 inflammasome plays an important role via IL-1β in regulating PCSK9 secretion. On the other hand, PCSK9 regulates caspase-1-dependent pyroptosis by initiating mtDNA damage and activating NLRP3 inflammasome signaling. This paper reviews the mechanisms underlying PCSK9 and NLRP3 inflammasome activation in the context of atherosclerosis. Furthermore, we describe the current understanding of the specific molecular mechanism underlying the interactions between PCSK9 and NLRP3 inflammasome signaling as well as the drug repositioning events that influence vascular cells and exert beneficial antiatherosclerotic effects. This review may provide a new therapeutic direction for the effective prevention and treatment of atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Yanan Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Qinzhi Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jingcan You
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Liqun Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Min Zeng
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China.,Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
37
|
Yang R, Liu Q, Zhang M. The Past and Present Lives of the Intraocular Transmembrane Protein CD36. Cells 2022; 12:cells12010171. [PMID: 36611964 PMCID: PMC9818597 DOI: 10.3390/cells12010171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cluster of differentiation 36 (CD36) belongs to the B2 receptors of the scavenger receptor class B family, which is comprised of single-chain secondary transmembrane glycoproteins. It is present in a variety of cell types, including monocytes, macrophages, microvascular endothelial cells, adipocytes, hepatocytes, platelets, skeletal muscle cells, kidney cells, cardiomyocytes, taste bud cells, and a variety of other cell types. CD36 can be localized on the cell surface, mitochondria, endoplasmic reticulum, and endosomes, playing a role in lipid accumulation, oxidative stress injury, apoptosis, and inflammatory signaling. Recent studies have found that CD36 is expressed in a variety of ocular cells, including retinal pigment epithelium (RPE), retinal microvascular endothelial cells, retinal ganglion cells (RGC), Müller cells, and photoreceptor cells, playing an important role in eye diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma. Therefore, a comprehensive understanding of CD36 function and downstream signaling pathways is of great significance for the prevention and treatment of eye diseases. This article reviews the molecular characteristics, distribution, and function of scavenger receptor CD36 and its role in ophthalmology in order to deepen the understanding of CD36 in eye diseases and provide new ideas for treatment strategies.
Collapse
Affiliation(s)
- Rucui Yang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
- Department of Ophthalmology, Shantou University Medical College, Shantou University, Shantou 515041, China
| | - Qingping Liu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| |
Collapse
|
38
|
Liao M, Li Y, Xiao A, Lu Q, Zeng H, Qin H, Zheng E, Luo X, Chen L, Ruan XZ, Yang P, Chen Y. HIF-2α-induced upregulation of CD36 promotes the development of ccRCC. Exp Cell Res 2022; 421:113389. [PMID: 36252650 DOI: 10.1016/j.yexcr.2022.113389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 12/29/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by the abundance of lipid droplets and the activation of the hypoxia-inducible factor (HIF) signaling pathway. However, the lipid reprogramming induced by HIF signaling in ccRCC is not fully understood. In this study, we found that the fatty acid receptor CD36 was highly expressed in human ccRCC tissues and ccRCC cell lines. CD36 overexpression increased fatty acid uptake and lipid droplet formation, and enhanced the proliferation and migration of ccRCC cells in a DGAT1-dependent manner. In contrast, the disruption of endogenous CD36 showed the opposite effects. The upregulated expression of CD36 in ccRCC was associated with hypoxia and HIF-2α activation. Furthermore, we identified CD36 as a new target of the transcription factor HIF-2α. The knockdown of CD36 in ccRCC cells reduced lipid accumulation and also blocked the tumor-promoting effects induced by HIF-2α under hypoxia. Our findings suggest that hypoxia-dependent HIF-2α promotes the remodeling of lipid metabolism and the malignant phenotype of ccRCC via CD36, providing a certain theoretical basis for clarifying the mechanism of ccRCC.
Collapse
Affiliation(s)
- Meng Liao
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Yiyu Li
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Anhua Xiao
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Qianlan Lu
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Han Zeng
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Qin
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Enze Zheng
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoqing Luo
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Lin Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Xiong Z Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China; John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, London, NW3 2PF, United Kingdom
| | - Ping Yang
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Yaxi Chen
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
39
|
Herrera-Martínez AD, Herrero-Aguayo V, Pérez-Gómez JM, Gahete MD, Luque RM. Inflammasomes: Cause or consequence of obesity-associated comorbidities in humans. Obesity (Silver Spring) 2022; 30:2351-2362. [PMID: 36415999 DOI: 10.1002/oby.23581] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022]
Abstract
Inflammasomes are multiprotein intracellular complexes composed of innate immune system receptors and sensors; they activate the inflammatory cascade in response to infectious microbes and/or molecules derived from host proteins. Because of cytokine secretion, inflammasomes can induce amplified systemic responses, its dysregulation can exacerbate symptoms in infectious diseases, and it has been related to the development of autoimmune diseases, inflammatory disorders, and even cancer. Obesity is associated with a chronic low-grade inflammation, in which circulating proinflammatory cytokines are elevated. Some publications describe changes in inflammation markers as a consequence of obesity, but others suggest that chronic inflammation might cause obesity (e.g., C-reactive protein): these assumptions reflect the difficulty of identifying the appropriate role of inflammation as cause or consequence of obesity and its related complications. Obesity is recognized as a clinical risk factor for developing cardiovascular diseases including atherosclerosis, metabolic syndrome, insulin resistance, and diabetes mellitus. Changes in the expression of inflammasomes are described in some of these obesity-related complications, and moreover, its modulation might exert a beneficial effect in some cases. Despite some contradictory results, most publications suggest a promising clinical effect based on in vitro and in vivo experiments. In this review, we summarized recent publications about inflammasome dysregulation in humans and its relationship with obesity-related comorbidities.
Collapse
Affiliation(s)
- Aura D Herrera-Martínez
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
- Maimonides Institute for Biomedical Research of Córdoba, Córdoba, Spain
| | - Vicente Herrero-Aguayo
- Maimonides Institute for Biomedical Research of Córdoba, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Córdoba, Spain
| | - Jesús M Pérez-Gómez
- Maimonides Institute for Biomedical Research of Córdoba, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Córdoba, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Córdoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Córdoba, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Córdoba, Spain
| |
Collapse
|
40
|
Margiana R, Alsaikhan F, Al-Awsi GRL, Patra I, Sivaraman R, Fadhil AA, Al-Baghdady HFA, Qasim MT, Hameed NM, Mustafa YF, Hosseini-Fard S. Functions and therapeutic interventions of non-coding RNAs associated with TLR signaling pathway in atherosclerosis. Cell Signal 2022; 100:110471. [PMID: 36122884 DOI: 10.1016/j.cellsig.2022.110471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022]
Abstract
Nowadays, emerging data demonstrate that the toll-like receptor (TLR) signaling pathway plays an important role in the progression of inflammatory atherosclerosis. Indeed, dysregulated TLR signaling pathway could be a cornerstone of inflammation and atherosclerosis, which contributes to the development of cardiovascular diseases. It is interesting to note that this pathway is heavily controlled by several mechanisms, such as epigenetic factors in which the role of non-coding RNAs (ncRNAs), particularly microRNAs and long noncoding RNAs as well as circular RNAs in the pathogenesis of atherosclerosis has been well studied. Recent years have seen a significant surge in the amount of research exploring the interplay between ncRNAs and TLR signaling pathway downstream targets in the development of atherosclerosis; however, there is still considerable room for improvement in this field. The current study was designed to review underlying mechanisms of TLR signaling pathway and ncRNA interactions to shed light on therapeutic implications in patients with atherosclerosis.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Dr. Soetomo General Academic Hospital, Surabaya, Jakarta, Indonesia
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | | | - Indrajit Patra
- An Independent Researcher, PhD from NIT Durgapur, Durgapur, West Bengal, India
| | - Ramaswamy Sivaraman
- Dwaraka Doss Goverdhan Doss Vaishnav College, University of Madras, Arumbakkam, Chennai, India
| | | | | | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Noora M Hameed
- Anesthesia techniques, Al-Nisour University College, Baghdad, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Seyedreza Hosseini-Fard
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Tetramethylpyrazine and Paeoniflorin Synergistically Attenuate Cholesterol Efflux in Macrophage Cells via Enhancing ABCA1 and ABCG1 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4304790. [PMID: 36387364 PMCID: PMC9653297 DOI: 10.1155/2022/4304790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/28/2022] [Accepted: 10/29/2022] [Indexed: 11/08/2022]
Abstract
The formation of foam cells is a characteristic of the occurrence and development of atherosclerosis. ATP-binding cassette subfamily A1 and G1 (ABCA1 and ABCG1) and scavenger receptor B1 (SR-B1) play critical roles in promoting intracellular cholesterol efflux to high-density lipoprotein (HDL) or apolipoprotein A1 (apoA1). We attempted to test the effect of the tetramethylpyrazine-paeoniflorin pair (TP) on cholesterol outflow in foam cells derived from macrophages. In this study, RAW264.7 macrophages were treated with 80 mg/L oxidized low-density lipoprotein (ox-LDL) for 24 h to obtain foam cells. Then they were intervened with TP (tetramethylpyrazine 40 ug/ml plus paeoniflorin 80 ug/ml) for additional 24 h. The distribution of cholesterol in foam cells was evaluated by oil red O staining. The contents of total cholesterol (TC) and free cholesterol (FC) were assessed with commercial kits. Fluorescent imaging was observed with a fluorescent inverted microscope. The capacity of cholesterol efflux was measured with a fluorescent plate reader, and the transcript and protein levels of ABCA1, ABCG1, and SR-B1 were detected by Western blot and quantitative polymerase chain reactions (Q-PCRs). Cytokines in the medium were detected by ELISA and adjusted by total cellular proteins. The results showed that TP decreased ox-LDL-induced cholesterol deposition and foam cell formation by promoting cholesterol efflux to apoA1, which was related to the upregulation of ABCA1 and ABCG1. Moreover, TP decreased the secretion of ox-LDL-induced tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), and monocyte chemotactic protein-1 (MCP-1), an important profoam cell cytokine in atherosclerosis.
Collapse
|
42
|
Ruan S, Zha L. Moronic acid improves intestinal inflammation in mice with chronic colitis by inhibiting intestinal macrophage polarization. J Biochem Mol Toxicol 2022; 36:e23188. [PMID: 35924425 DOI: 10.1002/jbt.23188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/06/2022]
Abstract
This study focuses on exploring the role and mechanism of moronic acid (MOA), a small triterpenoid molecule, against inflammatory bowel disease (IBD). Intestinal macrophages were cultured in vitro, and their M1 polarization was induced by lipopolysaccharide (LPS) and interferon gamma (IFN-γ). After intervention with MOA, the proportion of M1 macrophages was detected, and the levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β) were examined by ELISA. IFA staining was performed to determine the P50 and CD86 expressions, while DCFH-DA was used to determine the reactive oxygen species (ROS) level, as well as the p-P50 and NLRP3 protein levels. Additionally, we also used N-acetylcysteine, a ROS inhibitor, to further explore the association between MOA and ROS-NF-κB signaling. In murine experimentation, colitis was induced in mice with DSS. After MOA intervention, we assessed the mucosal barrier damage, tissue ROS, as well as protein and inflammatory cytokine levels. MOA could inhibit the M1 polarization of intestinal macrophages, suppress the expressions of inflammatory cytokines, and reduce the level of ROS-NF-κB-NLRP3 signaling. After inhibiting ROS through NAC treatment, the effect of MOA was evidently weakened. Clearly, MOA exerted its activity via ROS. In the murine model, MOA could lower the CD86 level in the intestinal tissues, inhibit the M1 polarization of macrophages, and reduce the tissue levels of inflammatory cytokines. This study finds that MOA can regulate ROS-NF-κB-NLRP3 signaling by inhibiting ROS, thereby suppressing the M1 polarization of intestinal macrophages, which plays a protective role in IBD.
Collapse
Affiliation(s)
- Shuiliang Ruan
- Department of Gastroenterology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Li Zha
- Department of Gastroenterology, the Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
43
|
Ficus hirta Vahl. Ameliorates Nonalcoholic Fatty Liver Disease through Regulating Lipid Metabolism and Gut Microbiota. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3474723. [PMID: 35592528 PMCID: PMC9113867 DOI: 10.1155/2022/3474723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has gradually become one of the most serious liver diseases threatening human health in the world. Currently, Chinese herbal medicine is a potentially important treatment option for NAFLD, and the development of effective Chinese herbal medicine has a good prospect. Previous studies have suggested that Ficus hirta Vahl. (FV) has various protective effects on the liver. In this study, we investigated the therapeutic outcomes of FV treatment for the liver disease and its underlying mechanism using HepG2 cell lines induced by palmitate (PA) and mouse model fed with high-fat diet (HFD). FV mainly exerts pharmacological effects by mediating lipid metabolism and inflammation. During the lipid metabolism regulation process, CD36, SREBP-1, SCD1, PPAR γ, ACOX1, and CPT1α are the key factors related to the healing effects of FV on NAFLD. During the inflammation process, the downregulation of IL-6, IL-1β, and TNF-α is involved in alleviation of NAFLD. Furthermore, CD36 overexpression promotes lipid abnormal metabolism and inflammation in PA-induced HepG2 cells, while CD36 knockdown and FV supplementation reverse these responses. In addition, FV also modulates gut microbiota composition, such as Allobaculum, Faecalibaculum, and Butyricicoccus in HFD-fed mice. In summary, our findings demonstrated that FV exerted a beneficial preventive and therapeutic effect on NAFLD by improving lipid metabolism and inflammation as well as regulating the structure of gut microbiota, and therefore, FV may be a candidate for the treatment of NAFLD.
Collapse
|
44
|
Wang P, Yin B, Zhang Z, Mao S, Bao W, Lian W, Fan Y, Hong C, Su Y, Jia C. Foamy macrophages potentially inhibit tuberculous wound healing by inhibiting the TLRs/NF-κB signalling pathway. Wound Repair Regen 2022; 30:376-396. [PMID: 35384137 DOI: 10.1111/wrr.13006] [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: 07/12/2021] [Revised: 01/26/2022] [Accepted: 03/20/2022] [Indexed: 11/29/2022]
Abstract
To characterise the distribution, classification, and quantity of foamy macrophages (FMs) in tuberculous wound tissue and the relationship between FM and delayed healing of tuberculous wounds. Morphological studies were performed to explore the distribution of FM and Mycobacterium tuberculosis (Mtb) in tuberculous wounds, with acute and chronic wounds included for comparison. Phorbol-12-myristate-13-acetate stimulation-differentiated THP-1 cells were treated with Mtb to induce their differentiation into FM with oxidised low-density lipoprotein treatment serving as a control. Relative cytokine levels were determined by quantitative PCR and Western blotting. Varied co-culture combinations of Mtb, THP-1, FM, and fibroblasts were performed, and proliferation, migration, ability to contract collagen gel, and protein levels of the chemokines in the supernatants of the fibroblasts were assessed. The differentially expressed genes in human skin fibroblasts (HSFs) after co-culture with or without FM were identified using microarray. Many FM were found in the tissues of tuberculous wounds. The FM that did not engulf Mtb (NM-FM) were mainly distributed in tissues surrounding tuberculous wounds, whereas the FM that engulfed Mtb (M-FM) were dominantly located within granulomatous tissues. Co-culture experiments showed that, with the Mtb co-culture, the portions of NM-FM in the total FM grew over time. The migration, proliferation, chemokine secretion, and the ability of fibroblasts to contract collagen gel were inhibited when co-cultured with Mtb, FM, or a combination of the two. Further investigation showed that the TLRs/NF-κB signalling pathway is involved in fibroblast function under the stimulation of FM. TLRs and NF-κB agonists could reverse the phenotypic changes in HSFs after co-culture with FM. The tuberculous wound microenvironment composed of Mtb and FM may affect wound healing by inhibiting the functions of fibroblasts. FM potentially inhibit fibroblasts' function by inhibiting the TLRs/NF-κB signalling pathway in tuberculous wounds.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bin Yin
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zexin Zhang
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shuting Mao
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wu Bao
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenqin Lian
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yueying Fan
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chao Hong
- Xiamen Center for Disease Control and Prevention, Xiamen, China
| | - Yingjun Su
- Department of Burns and Plastic Surgery, Plastic Surgery Hospital of Xi'an International Medical Center, Xi'an, China
| | - Chiyu Jia
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
45
|
Xu W, Qian L, Yuan X, Lu Y. MicroRNA-223-3p inhibits oxidized low-density lipoprotein-mediated NLRP3 inflammasome activation via directly targeting NLRP3 and FOXO3. Clin Hemorheol Microcirc 2022; 81:241-253. [PMID: 35275525 DOI: 10.3233/ch-211232] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) have emerged as crucial players in the initiation and development of atherosclerosis (AS), and the low miR-223-3p level is observed in AS patients. However, the function and mechanism behind miR-223-3p in AS progression have not been fully elucidated. METHOD In the present study, THP-1 cells treated with oxidized low-density lipoprotein (ox-LDL) were employed as the cell model of AS. The expression levels of miR-223-3p, NLR family pyrin domain containing 3 (NLRP3), caspase-1, pro-caspase-1, cleaved interleukin 18 (IL-18), cleaved IL-1β, and forkhead box O3 (FOXO3) were measured by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot (WB) analyses. The relationship between miR-223-3p and FOXO3 or NLRP3 was determined using a dual-luciferase reporter assay. The production of IL-1β, IL-18, IL-6, and TNF-α was examined by Enzyme-linked immunosorbent assay (ELISA). RESULTS MiR-223-3p was decreased in AS patients and ox-LDL-induced THP-1 cells, and its upregulation downregulated the abundance of NLRP3, caspase-1, cleaved IL-18, cleaved IL-1β, IL-1β, IL-6, and TNF-α in THP-1 cells treated with ox-LDL or not, and the depletion of miR-223-3p revealed an opposite phenomenon. NLPR3 and FOXO3 were identified as two authentic targets of miR-223-3p. Knockdown of NLRP3 or FOXO3 reversed the stimulatory effect of the miR-223-3p inhibitor on the inflammatory responses of THP-1 cells. CONCLUSIONS Our data indicate that miR-223-3p inhibited ox-LDL-mediated NLRP3 inflammasome activation via directly targeting NLRP3 and FOXO3 in THP-1 cells, which offered a prospective therapeutic target for AS therapy.
Collapse
Affiliation(s)
- Wei Xu
- Heart Rehabilitation Center, Department of Cardiology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Lei Qian
- Heart Rehabilitation Center, Department of Cardiology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Xiaoyan Yuan
- Heart Rehabilitation Center, Department of Cardiology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Yong Lu
- Heart Rehabilitation Center, Department of Cardiology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
46
|
Tian Y, Chang S, Xu J, Gong P, Yu B, Qi J. Investigation of the effective components inhibited macrophage foam cell formation in Ophiopogonis Radix. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114678. [PMID: 34563614 DOI: 10.1016/j.jep.2021.114678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/12/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ophiopogonis Radix, the commonly used traditional Chinese medicine in clinic for treating cardiovascular diseases, is returned to the stomach, lung and heart meridian. It is reported to nourish yin, moisten lung and is used to treat heart yin deficiency syndromes and asthenia of heart and lung, which indicated that Ophiopogonis Radix may have a protective effect on heart disorders. Atherosclerosisis is an important process in the development of cardiovascular diseases and abnormal lipid deposition induced macrophage foam cells is its crucial foundation. Our previous study showed the extract of Ophiopogonis Radix (EOR) ameliorates atherosclerosis in vitro. However, it may protect against cardiovascular diseases through inhibiting macrophage foam cell formation and its potential effective components and mechanisms are still unclear. AIM OF THE STUDY Our study aimed to investigate the effect of Ophiopogonis Radix on macrophage foam cell formation and its potential active constituents and mechanisms. MATERIALS AND METHODS Ox-LDL induced macrophage cells were employed to evaluate the effect of Ophiopogonis Radix on macrophage foam cell formation. Then the potential active constituents inhibited formation of macrophage foam cells were screened by biospecific cell extraction and its underlying mechanisms were also explored by Western blot. RESULTS The extract of Ophiopogonis Radix was found to significantly inhibit macrophage foam cell formation, evidenced by the decrease of TG and TC and Oil Red O staining analysis in macrophage cells, which indicated that EOR reduced the formation of macrophage foam cells. At the same time, EOR was showed to increase antioxidant capacity in macrophage cells. After treatment with EOR, two potential active components interacted with macrophage foam cells specifically were identified to inhibit macrophage foam cell formation including methylophiopogonanone A and methylophiopogonanone B. Methylophiopogonanone A was then proved to decrease the expression of CD36, Lox-1 and SREBP2, increase the expression of ABCA1 obviously, while the expression of ABCG1 and SREBP1 had no changes. CONCLUSIONS In our study, Ophiopogonis Radix was found to protect against atherosclerosis through suppressing ox-LDL induced macrophage foam cell formation and two potential compounds were identified by biospecific cell extraction including methylophiopogonanone A and methylophiopogonanone B. Moreover, methylophiopogonanone A was proved to inhibit foam cells through reducing uptake, synthesis and increasing efflux, which may provide guidance and reference for application of Ophiopogonis Radix and investigation of the effective components of TCMs.
Collapse
Affiliation(s)
- YuShan Tian
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Shanquan Chang
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Juntao Xu
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Puyang Gong
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - BoYang Yu
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Jin Qi
- Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| |
Collapse
|
47
|
Phage Display Preparation of Specific Polypeptides in Atherosclerotic Foam Cells. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12020562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Atherosclerosis and related complications are the most common causes of death in modern societies. Macrophage-derived foam cells play critical roles in the initiation and progression of atherosclerosis. Effective, rapid, and instrument-independent detection in the early stage of chronic atherosclerosis progression could provide an opportunity for early intervention and treatment. Therefore, as a starting point, in this study, we aimed to isolate and prepare foam cell-specific polypeptides using a phage display platform. The six target polypeptides, which were acquired in this study, were evaluated by ELISA and showed strong specificity with foam cells. Streptavidin coupled quantum dots (QDs) were used as fluorescence developing agents, and images of biotin-modified polypeptides specifically binding with foam cells were clearly observed. The polypeptides obtained in this study could lay the foundation for developing a rapid detection kit for early atherosclerosis lesions and could provide new materials for research on the mechanisms of foam cell formation and the development of blocking drugs.
Collapse
|
48
|
Lipids in Pathophysiology and Development of the Membrane Lipid Therapy: New Bioactive Lipids. MEMBRANES 2021; 11:membranes11120919. [PMID: 34940418 PMCID: PMC8708953 DOI: 10.3390/membranes11120919] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Membranes are mainly composed of a lipid bilayer and proteins, constituting a checkpoint for the entry and passage of signals and other molecules. Their composition can be modulated by diet, pathophysiological processes, and nutritional/pharmaceutical interventions. In addition to their use as an energy source, lipids have important structural and functional roles, e.g., fatty acyl moieties in phospholipids have distinct impacts on human health depending on their saturation, carbon length, and isometry. These and other membrane lipids have quite specific effects on the lipid bilayer structure, which regulates the interaction with signaling proteins. Alterations to lipids have been associated with important diseases, and, consequently, normalization of these alterations or regulatory interventions that control membrane lipid composition have therapeutic potential. This approach, termed membrane lipid therapy or membrane lipid replacement, has emerged as a novel technology platform for nutraceutical interventions and drug discovery. Several clinical trials and therapeutic products have validated this technology based on the understanding of membrane structure and function. The present review analyzes the molecular basis of this innovative approach, describing how membrane lipid composition and structure affects protein-lipid interactions, cell signaling, disease, and therapy (e.g., fatigue and cardiovascular, neurodegenerative, tumor, infectious diseases).
Collapse
|
49
|
Bartoli-Leonard F, Zimmer J, Aikawa E. Innate and adaptive immunity: the understudied driving force of heart valve disease. Cardiovasc Res 2021; 117:2506-2524. [PMID: 34432007 PMCID: PMC8783388 DOI: 10.1093/cvr/cvab273] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific aortic valve disease (CAVD), and its clinical manifestation that is calcific aortic valve stenosis, is the leading cause for valve disease within the developed world, with no current pharmacological treatment available to delay or halt its progression. Characterized by progressive fibrotic remodelling and subsequent pathogenic mineralization of the valve leaflets, valve disease affects 2.5% of the western population, thus highlighting the need for urgent intervention. Whilst the pathobiology of valve disease is complex, involving genetic factors, lipid infiltration, and oxidative damage, the immune system is now being accepted to play a crucial role in pathogenesis and disease continuation. No longer considered a passive degenerative disease, CAVD is understood to be an active inflammatory process, involving a multitude of pro-inflammatory mechanisms, with both the adaptive and the innate immune system underpinning these complex mechanisms. Within the valve, 15% of cells evolve from haemopoietic origin, and this number greatly expands following inflammation, as macrophages, T lymphocytes, B lymphocytes, and innate immune cells infiltrate the valve, promoting further inflammation. Whether chronic immune infiltration or pathogenic clonal expansion of immune cells within the valve or a combination of the two is responsible for disease progression, it is clear that greater understanding of the immune systems role in valve disease is required to inform future treatment strategies for control of CAVD development.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jonas Zimmer
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
50
|
The Potential Role of Electronegative High-Density Lipoprotein H5 Subfraction in RA-Related Atherosclerosis. Int J Mol Sci 2021; 22:ijms222111419. [PMID: 34768851 PMCID: PMC8584111 DOI: 10.3390/ijms222111419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 01/23/2023] Open
Abstract
Although the heterogeneity of high-density lipoprotein-cholesterol (HDL-c) composition is associated with atherosclerotic cardiovascular risk, the link between electronegative subfractions of HDL-c and atherosclerosis in rheumatoid arthritis (RA) remains unknown. We examined the association of the percentage of the most electronegative subfraction of HDL-c (H5%) and RA-related atherosclerosis. Using anion-exchange purification/fast-protein liquid chromatography, we demonstrated significantly higher H5% in patients (median, 7.2%) than HC (2.8%, p < 0.005). Multivariable regression analysis revealed H5% as a significant predictor for subclinical atherosclerosis. We subsequently explored atherogenic role of H5 using cell-based assay. The results showed significantly higher levels of IL-1β and IL-8 mRNA in H5-treated (mean ± SD, 4.45 ± 1.22 folds, 6.02 ± 1.43-folds, respectively) than H1-treated monocytes (0.89 ± 0.18-folds, 1.03 ± 0.26-folds, respectively, both p < 0.001). In macrophages, H5 upregulated the mRNA and protein expression of IL-1β and IL-8 in a dose-dependent manner, and their expression levels were significantly higher than H1-treated macrophages (all p < 0.001). H5 induced more foam cell formation compared with H1-treated macrophages (p < 0.005). In addition, H5 has significantly lower cholesterol efflux capacity than H1 (p < 0.005). The results of nanoLC-MS/MS approach reveal that the best discriminator between high-H5% and normal-H5% is Apo(a), the main constituent of Lp(a). Moreover, Lp(a) level is a significant predictor for high-H5%. These observations suggest that H5 is involved in RA-related atherosclerosis.
Collapse
|