1
|
Pradhan P, Vijayan V, Liu B, Martinez-Delgado B, Matamala N, Nikolin C, Greite R, DeLuca DS, Janciauskiene S, Motterlini R, Foresti R, Immenschuh S. Distinct metabolic responses to heme in inflammatory human and mouse macrophages - Role of nitric oxide. Redox Biol 2024; 73:103191. [PMID: 38762951 PMCID: PMC11130737 DOI: 10.1016/j.redox.2024.103191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024] Open
Abstract
Activation of inflammation is tightly associated with metabolic reprogramming in macrophages. The iron-containing tetrapyrrole heme can induce pro-oxidant and pro-inflammatory effects in murine macrophages, but has been associated with polarization towards an anti-inflammatory phenotype in human macrophages. In the current study, we compared the regulatory responses to heme and the prototypical Toll-like receptor (TLR)4 ligand lipopolysaccharide (LPS) in human and mouse macrophages with a particular focus on alterations of cellular bioenergetics. In human macrophages, bulk RNA-sequencing analysis indicated that heme led to an anti-inflammatory transcriptional profile, whereas LPS induced a classical pro-inflammatory gene response. Co-stimulation of heme with LPS caused opposing regulatory patterns of inflammatory activation and cellular bioenergetics in human and mouse macrophages. Specifically, in LPS-stimulated murine, but not human macrophages, heme led to a marked suppression of oxidative phosphorylation and an up-regulation of glycolysis. The species-specific alterations in cellular bioenergetics and inflammatory responses to heme were critically dependent on the availability of nitric oxide (NO) that is generated in inflammatory mouse, but not human macrophages. Accordingly, studies with an inducible nitric oxide synthase (iNOS) inhibitor in mouse, and a pharmacological NO donor in human macrophages, reveal that NO is responsible for the opposing effects of heme in these cells. Taken together, the current findings indicate that NO is critical for the immunomodulatory role of heme in macrophages.
Collapse
Affiliation(s)
- Pooja Pradhan
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Vijith Vijayan
- Department of Pediatrics, Stanford University, Stanford, USA
| | - Bin Liu
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Beatriz Martinez-Delgado
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220, Madrid, Spain
| | - Nerea Matamala
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220, Madrid, Spain
| | - Christoph Nikolin
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Robert Greite
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - David S. DeLuca
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Pulmonary and Infectious Diseases and BREATH German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | | | - Roberta Foresti
- University Paris-Est Créteil, INSERM, IMRB, F-94010, Créteil, France
| | - Stephan Immenschuh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Yeudall S, Upchurch CM, Leitinger N. The clinical relevance of heme detoxification by the macrophage heme oxygenase system. Front Immunol 2024; 15:1379967. [PMID: 38585264 PMCID: PMC10995405 DOI: 10.3389/fimmu.2024.1379967] [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: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Heme degradation by the heme oxygenase (HMOX) family of enzymes is critical for maintaining homeostasis and limiting heme-induced tissue damage. Macrophages express HMOX1 and 2 and are critical sites of heme degradation in healthy and diseased states. Here we review the functions of the macrophage heme oxygenase system and its clinical relevance in discrete groups of pathologies where heme has been demonstrated to play a driving role. HMOX1 function in macrophages is essential for limiting oxidative tissue damage in both acute and chronic hemolytic disorders. By degrading pro-inflammatory heme and releasing anti-inflammatory molecules such as carbon monoxide, HMOX1 fine-tunes the acute inflammatory response with consequences for disorders of hyperinflammation such as sepsis. We then discuss divergent beneficial and pathological roles for HMOX1 in disorders such as atherosclerosis and metabolic syndrome, where activation of the HMOX system sits at the crossroads of chronic low-grade inflammation and oxidative stress. Finally, we highlight the emerging role for HMOX1 in regulating macrophage cell death via the iron- and oxidation-dependent form of cell death, ferroptosis. In summary, the importance of heme clearance by macrophages is an active area of investigation with relevance for therapeutic intervention in a diverse array of human diseases.
Collapse
Affiliation(s)
- Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Clint M. Upchurch
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
3
|
Kiseleva V, Vishnyakova P, Elchaninov A, Fatkhudinov T, Sukhikh G. Biochemical and molecular inducers and modulators of M2 macrophage polarization in clinical perspective. Int Immunopharmacol 2023; 122:110583. [PMID: 37423155 DOI: 10.1016/j.intimp.2023.110583] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/11/2023]
Abstract
Macrophages as innate immune cells with great plasticity are of great interest for cell therapy. There are two main macrophage populations - pro- and anti-inflammatory cells also known as M1 and M2. High potential in cancer research contributed to the in-depth study of the molecular processes leading to the polarization of macrophages into the M1 phenotype, and much less attention has been paid to anti-inflammatory M2 macrophages, which can be successfully used in cell therapy of inflammatory diseases. This review describes ontogenesis of macrophages, main functions of pro- and and-inflammatory cells and four M2 subpopulations characterized by different functionalities. Data on agents (cytokines, microRNAs, drugs, plant extracts) that may induce M2 polarization through the changes in microenvironment, metabolism, and efferocytosis are summarized. Finally, recent attempts at stable macrophage polarization using genetic modifications are described. This review may be helpful for researchers concerned with the problem of M2 macrophage polarization and potential use of these anti-inflammatory cells for the purposes of regenerative medicine.
Collapse
Affiliation(s)
- Viktoriia Kiseleva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia.
| | - Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia; Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
| | - Timur Fatkhudinov
- Peoples' Friendship University of Russia, Moscow, Russia; Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
4
|
Li H, Cao Z, Wang L, Liu C, Lin H, Tang Y, Yao P. Macrophage Subsets and Death Are Responsible for Atherosclerotic Plaque Formation. Front Immunol 2022; 13:843712. [PMID: 35432323 PMCID: PMC9007036 DOI: 10.3389/fimmu.2022.843712] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases, the notorious killer, are mainly caused by atherosclerosis (AS) characterized by lipids, cholesterol, and iron overload in plaques. Macrophages are effector cells and accumulate to the damaged and inflamed sites of arteries to internalize native and chemically modified lipoproteins to transform them into cholesterol-loaded foam cells. Foam cell formation is determined by the capacity of phagocytosis, migration, scavenging, and the features of phenotypes. Macrophages are diverse, and the subsets and functions are controlled by their surrounding microenvironment. Generally, macrophages are divided into classically activated (M1) and alternatively activated (M2). Recently, intraplaque macrophage phenotypes are recognized by the stimulation of CXCL4 (M4), oxidized phospholipids (Mox), hemoglobin/haptoglobin complexes [HA-mac/M(Hb)], and heme (Mhem). The pro-atherogenic or anti-atherosclerotic phenotypes of macrophages decide the progression of AS. Besides, apoptosis, necrosis, ferroptosis, autophagy and pyrotopsis determine plaque formation and cardiovascular vulnerability, which may be associated with macrophage polarization phenotypes. In this review, we first summarize the three most popular hypotheses for AS and find the common key factors for further discussion. Secondly, we discuss the factors affecting macrophage polarization and five types of macrophage death in AS progression, especially ferroptosis. A comprehensive understanding of the cellular and molecular mechanisms of plaque formation is conducive to disentangling the candidate targets of macrophage-targeting therapies for clinical intervention at various stages of AS.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Cao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongkun Lin
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Liu J, Zhu Z, Leung GKK. Erythrophagocytosis by Microglia/Macrophage in Intracerebral Hemorrhage: From Mechanisms to Translation. Front Cell Neurosci 2022; 16:818602. [PMID: 35237132 PMCID: PMC8882619 DOI: 10.3389/fncel.2022.818602] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition characterized by hematoma related mass effect. Microglia/macrophage (M φ) are rapidly recruited in order to remove the red blood cells through erythrophagocytosis. Efficient erythrophagocytosis can detoxify hemolytic products and facilitate neurological recovery after ICH. The underlying mechanisms include modulation of inflammatory response and oxidative stress, among others. It is a dynamic process mediated by a cascade of signal transduction, including “find-me” signals, “eat-me” signals and a set of phagocytotic receptors-ligand pairs that may be exploited as therapeutic targets. This review summarizes mechanistic signaling pathways of erythrophagocytosis and highlights the potential of harnessing M φ-mediated phagocytosis for ICH treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Zhiyuan Zhu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- Department of Functional Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- *Correspondence: Gilberto Ka-Kit Leung,
| |
Collapse
|
6
|
Yan Y, Li T, Li Z, He M, Wang D, Xu Y, Yang X, Bai Y, Lao Y, Zhang Z, Wu W. Metformin Suppresses the Progress of Diabetes-Accelerated Atherosclerosis by Inhibition of Vascular Smooth Muscle Cell Migration Through AMPK-Pdlim5 Pathway. Front Cardiovasc Med 2021; 8:690627. [PMID: 34368251 PMCID: PMC8342753 DOI: 10.3389/fcvm.2021.690627] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Backgrounds: Our previous work revealed that AMP-activated protein kinase (AMPK) activation inhibits vascular smooth muscle cell migration in vitro by phosphorylating PDZ and LIM domain 5 (Pdlim5). As metformin is an AMPK activator, we used a mouse vascular smooth muscle cell (VSMC) line and a Myh11-cre-EGFP mice to investigate whether metformin could inhibit the migration of VSMCs in vitro and in a wire-injury model in vivo. It is recognized that VSMCs contribute to the major composition of atherosclerotic plaques. In order to investigate whether the AMPK–Pdlim5 pathway is involved in the protective function of metformin against atherosclerosis, we utilized ApoE−/− male mice to investigate whether metformin could suppress diabetes-accelerated atherosclerosis by inhibition of VSMC migration via the AMPK–Pdlim5 pathway. Methods: The mouse VSMC cell line was exogenously transfected wild type, phosphomimetic, or unphosphorylatable Pdlim5 mutant before metformin exposure. Myh11-cre-EGFP mice were treated with saline solution or metformin after these were subjected to wire injury in the carotid artery to study whether metformin could inhibit the migration of medial VSMCs into the neo-intima. In order to investigate whether the AMPK–Pdlim5 pathway is involved in the protective function of metformin against atherosclerosis, ApoE−/− male mice were divided randomly into control, streptozocin (STZ), and high-fat diet (HFD)-induced diabetes mellitus; STZ+HFD together with metformin or Pdlim5 mutant carried the adenovirus treatment groups. Results: It was found that metformin could induce the phosphorylation of Pdlim5 and inhibit cell migration as a result. The exogenous expression of phosphomimetic S177D-Pdlim5 inhibits lamellipodia formation and migration in VSMCs. It was also demonstrated that VSMCs contribute to the major composition of injury-induced neointimal lesions, while metformin could alleviate the occlusion of the carotid artery. The data of ApoE−/− mice showed that increased plasma lipids and aggravated vascular smooth muscle cell infiltration into the atherosclerotic lesion in diabetic mice were observed Metformin alleviated diabetes-induced metabolic disorders and atherosclerosis and also reduced VSMC infiltration in atherosclerotic plaques, while the Pdlim5 phospho-abolished mutant that carried adenovirus S177A-Pdlim5 undermines the protective function of metformin. Conclusions: The activation of the AMPK–Pdlim5 pathway by metformin could interrupt the migratory machine of VSMCs and inhibit cell migration in vitro and in vivo. The maintenance of AMPK activity by metformin is beneficial for suppressing diabetes-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Yi Yan
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Ting Li
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Zhonghao Li
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, Southern Medical University, Guangzhou, China
| | - Mingyuan He
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Dejiang Wang
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingyi Xu
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuewen Yang
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuanyuan Bai
- Guangzhou First People's Hospital, Guangzhou, China
| | - Yi Lao
- Department of Cardiology, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, China
| | - Zhiyong Zhang
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Wu
- Department of Cardiology, Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, Weng J. Metformin, Macrophage Dysfunction and Atherosclerosis. Front Immunol 2021; 12:682853. [PMID: 34163481 PMCID: PMC8215340 DOI: 10.3389/fimmu.2021.682853] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Metformin is one of the most widely prescribed hypoglycemic drugs and has the potential to treat many diseases. More and more evidence shows that metformin can regulate the function of macrophages in atherosclerosis, including reducing the differentiation of monocytes and inhibiting the inflammation, oxidative stress, polarization, foam cell formation and apoptosis of macrophages. The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-κB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. On the basis of summarizing these studies, we further discussed the future research directions of metformin: single-cell RNA sequencing, neutrophil extracellular traps (NETs), epigenetic modification, and metformin-based combination drugs. In short, macrophages play an important role in a variety of diseases, and improving macrophage dysfunction may be an important mechanism for metformin to expand its pleiotropic pharmacological profile. In addition, the combination of metformin with other drugs that improve the function of macrophages (such as SGLT2 inhibitors, statins and IL-1β inhibitors/monoclonal antibodies) may further enhance the pleiotropic therapeutic potential of metformin in conditions such as atherosclerosis, obesity, cancer, dementia and aging.
Collapse
Affiliation(s)
- Xiaojun Feng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Wenxu Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Peter J. Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| |
Collapse
|
8
|
Babusha Wega A, Kelta Wabalo E, Kenenisa Edae C, Bogale Awgichew G. Cardiac Troponin-I Status of Type-2 Diabetic Patients on Anti-Diabetic Drugs Treatment at Jimma Medical Center, Jimma, Southwest Ethiopia. RESEARCH REPORTS IN CLINICAL CARDIOLOGY 2021. [DOI: 10.2147/rrcc.s313432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
9
|
Additive contribution of microRNA-34a/b/c to human arterial ageing and atherosclerosis. Atherosclerosis 2021; 327:49-58. [PMID: 34038763 DOI: 10.1016/j.atherosclerosis.2021.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Preclinical data suggest that the ageing-induced miR-34a regulates vascular senescence. Herein we sought to assess whether the miR-34 family members miR-34a, miR-34b and miR-34c are involved in human arterial disease. METHODS Expression levels of miR-34a/b/c were quantified by TaqMan assay in peripheral blood mononuclear cells (PBMCs) derived from a consecutive cohort of 221 subjects who underwent cardiovascular risk assessment and thorough vascular examination for aortic stiffness and extent of arterial atherosclerosis. RESULTS High miR-34a was independently associated with the presence of CAD [OR (95%C.I.): 3.87 (1.56-9.56); p = 0.003] and high miR-34c with the number of diseased arterial beds [OR (95%C.I.): 1.88 (1.034-3.41); p = 0.038], while concurrent high expression of miR-34-a/c or all three miR-34a/b/c was associated with aortic stiffening (miR-34a/c: p = 0.022; miR-34a/b/c: p = 0.041) and with the extent of atherosclerosis [OR (95%C.I.) for number of coronary arteries [miR-34a/c: 3.29 (1.085-9.95); miR-34a/b/c: 6.06 (1.74-21.2)] and number of diseased arterial beds [miR-34a/c: 3.51 (1.45-8.52); miR-34a/b/c: 2.89 (1.05-7.92)] after controlling for possible confounders (p < 0.05 for all). Mechanistically, the increased levels of miR-34a or miR-34c were inversely associated with expression of SIRT1 or JAG1, NOTCH2, CTNNB1 and ATF1, respectively. The association of miR-34a/c or miR-34a/b/c with CAD was mainly mediated through SIRT1 and to a lesser extent through JAG1 as revealed by generalized structural equation modeling. Leukocyte-specific ablation of miR-34a/b/c ameliorates atherosclerotic plaque development and increases Sirt1 and Jag1 expression in an atherosclerosis mouse model confirming the human findings. CONCLUSIONS The present study reveals the clinical significance of the additive role of miR-34a/b/c in vascular ageing and atherosclerotic vascular disease.
Collapse
|
10
|
Lin P, Ji HH, Li YJ, Guo SD. Macrophage Plasticity and Atherosclerosis Therapy. Front Mol Biosci 2021; 8:679797. [PMID: 34026849 PMCID: PMC8138136 DOI: 10.3389/fmolb.2021.679797] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a chronic disease starting with the entry of monocytes into the subendothelium and the subsequent differentiation into macrophages. Macrophages are the major immune cells in atherosclerotic plaques and are involved in the dynamic progression of atherosclerotic plaques. The biological properties of atherosclerotic plaque macrophages determine lesion size, composition, and stability. The heterogenicity and plasticity of atherosclerotic macrophages have been a hotspot in recent years. Studies demonstrated that lipids, cytokines, chemokines, and other molecules in the atherosclerotic plaque microenvironment regulate macrophage phenotype, contributing to the switch of macrophages toward a pro- or anti-atherosclerosis state. Of note, M1/M2 classification is oversimplified and only represent two extreme states of macrophages. Moreover, M2 macrophages in atherosclerosis are not always protective. Understanding the phenotypic diversity and functions of macrophages can disclose their roles in atherosclerotic plaques. Given that lipid-lowering therapy cannot completely retard the progression of atherosclerosis, macrophages with high heterogeneity and plasticity raise the hope for atherosclerosis regression. This review will focus on the macrophage phenotypic diversity, its role in the progression of the dynamic atherosclerotic plaque, and finally discuss the possibility of treating atherosclerosis by targeting macrophage microenvironment.
Collapse
Affiliation(s)
- Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Hong-Hai Ji
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yan-Jie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
11
|
Seneviratne A, Cave L, Hyde G, Moestrup SK, Carling D, Mason JC, Haskard DO, Boyle JJ. Metformin directly suppresses atherosclerosis in normoglycaemic mice via haematopoietic adenosine monophosphate-activated protein kinase. Cardiovasc Res 2021; 117:1295-1308. [PMID: 32667970 PMCID: PMC8064441 DOI: 10.1093/cvr/cvaa171] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 06/03/2018] [Accepted: 06/22/2020] [Indexed: 12/31/2022] Open
Abstract
AIMS Atherosclerotic vascular disease has an inflammatory pathogenesis. Heme from intraplaque haemorrhage may drive a protective and pro-resolving macrophage M2-like phenotype, Mhem, via AMPK and activating transcription factor 1 (ATF1). The antidiabetic drug metformin may also activate AMPK-dependent signalling. Hypothesis: Metformin systematically induces atheroprotective genes in macrophages via AMPK and ATF1, thereby suppresses atherogenesis. METHODS AND RESULTS Normoglycaemic Ldlr-/- hyperlipidaemic mice were treated with oral metformin, which profoundly suppressed atherosclerotic lesion development (P < 5 × 10-11). Bone marrow transplantation from AMPK-deficient mice demonstrated that metformin-related atheroprotection required haematopoietic AMPK [analysis of variance (ANOVA), P < 0.03]. Metformin at a clinically relevant concentration (10 μM) evoked AMPK-dependent and ATF1-dependent increases in Hmox1, Nr1h2 (Lxrb), Abca1, Apoe, Igf1, and Pdgf, increases in several M2-markers and decreases in Nos2, in murine bone marrow macrophages. Similar effects were seen in human blood-derived macrophages, in which metformin-induced protective genes and M2-like genes, suppressible by si-ATF1-mediated knockdown. Microarray analysis comparing metformin with heme in human macrophages indicated that the transcriptomic effects of metformin were related to those of heme, but not identical. Metformin-induced lesional macrophage expression of p-AMPK, p-ATF1, and downstream M2-like protective effects. CONCLUSION Metformin activates a conserved AMPK-ATF1-M2-like pathway in mouse and human macrophages, and results in highly suppressed atherogenesis in hyperlipidaemic mice via haematopoietic AMPK.
Collapse
Affiliation(s)
| | - Luke Cave
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gareth Hyde
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, UK
| | - Justin C Mason
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Dorian O Haskard
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
12
|
Salt IP, Nunes JRC, Fullerton MD. Metformin again? Atheroprotection mediated by macrophage AMPK and ATF1. Cardiovasc Res 2021; 117:1233-1234. [PMID: 33681999 DOI: 10.1093/cvr/cvab065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Julia R C Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.,Centre for Infection, Immunity and Inflammation, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.,Centre for Infection, Immunity and Inflammation, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
13
|
Seneviratne A, Han Y, Wong E, Walter ERH, Jiang L, Cave L, Long NJ, Carling D, Mason JC, Haskard DO, Boyle JJ. Hematoma Resolution In Vivo Is Directed by Activating Transcription Factor 1. Circ Res 2020; 127:928-944. [PMID: 32611235 PMCID: PMC7478221 DOI: 10.1161/circresaha.119.315528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE The efficient resolution of tissue hemorrhage is an important homeostatic function. In human macrophages in vitro, heme activates an AMPK (AMP-activated protein kinase)/ATF1 (activating transcription factor-1) pathway that directs Mhem macrophages through coregulation of HO-1 (heme oxygenase-1; HMOX1) and lipid homeostasis genes. OBJECTIVE We asked whether this pathway had an in vivo role in mice. METHODS AND RESULTS Perifemoral hematomas were used as a model of hematoma resolution. In mouse bone marrow-derived macrophages, heme induced HO-1, lipid regulatory genes including LXR (lipid X receptor), the growth factor IGF1 (insulin-like growth factor-1), and the splenic red pulp macrophage gene Spic. This response was lost in bone marrow-derived macrophages from mice deficient in AMPK (Prkab1-/-) or ATF1 (Atf1-/-). In vivo, femoral hematomas resolved completely between days 8 and 9 in littermate control mice (n=12), but were still present at day 9 in mice deficient in either AMPK (Prkab1-/-) or ATF1 (Atf1-/-; n=6 each). Residual hematomas were accompanied by increased macrophage infiltration, inflammatory activation and oxidative stress. We also found that fluorescent lipids and a fluorescent iron-analog were trafficked to lipid-laden and iron-laden macrophages respectively. Moreover erythrocyte iron and lipid abnormally colocalized in the same macrophages in Atf1-/- mice. Therefore, iron-lipid separation was Atf1-dependent. CONCLUSIONS Taken together, these data demonstrate that both AMPK and ATF1 are required for normal hematoma resolution. Graphic Abstract: An online graphic abstract is available for this article.
Collapse
Affiliation(s)
- Anusha Seneviratne
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus
| | - Yumeng Han
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus.,Molecular Sciences Research Hub, Imperial College London White City Campus (Y.H., E.W., E.R.H.W., L.J., N.J.L.)
| | - Eunice Wong
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus.,Molecular Sciences Research Hub, Imperial College London White City Campus (Y.H., E.W., E.R.H.W., L.J., N.J.L.)
| | - Edward R H Walter
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus.,Molecular Sciences Research Hub, Imperial College London White City Campus (Y.H., E.W., E.R.H.W., L.J., N.J.L.)
| | - Lijun Jiang
- Molecular Sciences Research Hub, Imperial College London White City Campus (Y.H., E.W., E.R.H.W., L.J., N.J.L.)
| | - Luke Cave
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus
| | - Nicholas J Long
- Molecular Sciences Research Hub, Imperial College London White City Campus (Y.H., E.W., E.R.H.W., L.J., N.J.L.)
| | - David Carling
- MRC London Institute of Medical Sciences (D.C.), Imperial College London Hammersmith Campus
| | - Justin C Mason
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus
| | - Dorian O Haskard
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus
| | - Joseph J Boyle
- From the National Heart and Lung Institute (A.S., Y.H., E.W., E.R.H.W., L.C., J.C.M., D.O.H., J.J.B.), Imperial College London Hammersmith Campus
| |
Collapse
|
14
|
Castegna A, Gissi R, Menga A, Montopoli M, Favia M, Viola A, Canton M. Pharmacological targets of metabolism in disease: Opportunities from macrophages. Pharmacol Ther 2020; 210:107521. [PMID: 32151665 DOI: 10.1016/j.pharmthera.2020.107521] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
From advances in the knowledge of the immune system, it is emerging that the specialized functions displayed by macrophages during the course of an immune response are supported by specific and dynamically-connected metabolic programs. The study of immunometabolism is demonstrating that metabolic adaptations play a critical role in modulating inflammation and, conversely, inflammation deeply influences the acquisition of specific metabolic settings.This strict connection has been proven to be crucial for the execution of defined immune functional programs and it is now under investigation with respect to several human disorders, such as diabetes, sepsis, cancer, and autoimmunity. The abnormal remodelling of the metabolic pathways in macrophages is now emerging as both marker of disease and potential target of therapeutic intervention. By focusing on key pathological conditions, namely obesity and diabetes, rheumatoid arthritis, atherosclerosis and cancer, we will review the metabolic targets suitable for therapeutic intervention in macrophages. In addition, we will discuss the major obstacles and challenges related to the development of therapeutic strategies for a pharmacological targeting of macrophage's metabolism.
Collapse
Affiliation(s)
- Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Alessio Menga
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| |
Collapse
|
15
|
Yandrapalli S, Jolly G, Horblitt A, Pemmasani G, Sanaani A, Aronow WS, Frishman WH. Cardiovascular Safety and Benefits of Noninsulin Antihyperglycemic Drugs for the Treatment of Type 2 Diabetes Mellitus-Part 1. Cardiol Rev 2020; 28:177-189. [PMID: 32282393 DOI: 10.1097/crd.0000000000000308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is a major contributor to the morbidity and mortality associated with type 2 diabetes mellitus (T2DM). With T2DM growing in pandemic proportions, there will be profound healthcare implications of CVD in person with diabetes. The ideal drugs to improve outcomes in T2DM are those having antiglycemic efficacy in addition to cardiovascular (CV) safety, which has to be determined in appropriately designed CV outcome trials as mandated by regulatory agencies. Available evidence is largely supportive of metformin's CV safety and potential CVD risk reduction effects, whereas sulfonylureas are either CV risk neutral or are associated with variable CVD risk. Pioglitazone was also associated with improved CVD risk in patients with diabetes. The more recent antihyperglycemic medications have shown promise with regards to CVD risk reduction in T2DM patients at a high CV risk. Glucagon-like peptide-1 receptor agonists, a type of incretin-based therapy, were associated with better CV outcomes and mortality in T2DM patients, leading to the Food and Drug Administration approval of liraglutide to reduce CVD risk in high-risk T2DM patients. Ongoing and planned randomized controlled trials of the newer drugs should clarify the possibility of class effects, and of CVD risk reduction benefits in low-moderate CV risk patients. While metformin remains the first-line antiglycemic therapy in T2DM, glucagon-like peptide-1 receptor agonists should be appropriately prescribed in T2DM patients with baseline CVD or in those at a high CVD risk to improve CV outcomes. Dipeptidyl peptidase-4 inhibitors and sodium-glucose cotransporter-2 inhibitors are discussed in the second part of this review.
Collapse
Affiliation(s)
- Srikanth Yandrapalli
- From the Department of Medicine, Division of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - George Jolly
- Division of Cardiology, Loma Linda University Medical Center, Loma Linda, CA
| | - Adam Horblitt
- Division of Cardiology, Tulane Medical Center, New Orleans, LA
| | - Gayatri Pemmasani
- From the Department of Medicine, Division of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Abdallah Sanaani
- From the Department of Medicine, Division of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- From the Department of Medicine, Division of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - William H Frishman
- From the Department of Medicine, Division of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
16
|
Yang S, Yuan HQ, Hao YM, Ren Z, Qu SL, Liu LS, Wei DH, Tang ZH, Zhang JF, Jiang ZS. Macrophage polarization in atherosclerosis. Clin Chim Acta 2019; 501:142-146. [PMID: 31730809 DOI: 10.1016/j.cca.2019.10.034] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is a chronic inflammatory response that increases the risk of cardiovascular diseases. An in-depth study of the pathogenesis of atherosclerosis is critical for the treatment of atherosclerotic cardiovascular disease. The development of atherosclerosis involves many cells, such as endothelial cells, vascular smooth muscle cells, macrophages, and others. The considerable effects of macrophages in atherosclerosis are inextricably linked to macrophage polarization and the resulting phenotype. Moreover, the significant impact of macrophages on atherosclerosis depend not only on the function of the different macrophage phenotypes but also on the relative ratio of different phenotypes in the plaque. Research on atherosclerosis therapy indicates that the reduced plaque size and enhanced stability are partly due to modulating macrophage polarization. Therefore, regulating macrophage polarization and changing the proportion of macrophage phenotypes in plaques is a new therapeutic approach for atherosclerosis. This review provides a new perspective for atherosclerosis therapy by summarizing the relationship between macrophage polarization and atherosclerosis, as well as treatment targeting macrophage polarization.
Collapse
Affiliation(s)
- Sai Yang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Hou-Qin Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Ya-Meng Hao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Dang-Heng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Ji-Feng Zhang
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Rd, NCRC Bldg26-357S, Ann Arbor, MI 48109, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South China, Hengyang City, Hunan Province, 421001, PR China.
| |
Collapse
|
17
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
18
|
Abdolmaleki F, Gheibi Hayat SM, Bianconi V, Johnston TP, Sahebkar A. Atherosclerosis and immunity: A perspective. Trends Cardiovasc Med 2019; 29:363-371. [DOI: 10.1016/j.tcm.2018.09.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/09/2018] [Accepted: 09/25/2018] [Indexed: 01/11/2023]
|
19
|
Saenz J, Santa-María C, Reyes-Quiroz ME, Geniz I, Jiménez J, Sobrino F, Alba G. Grapefruit Flavonoid Naringenin Regulates the Expression of LXRα in THP-1 Macrophages by Modulating AMP-Activated Protein Kinase. Mol Pharm 2018; 15:1735-1745. [PMID: 29140707 DOI: 10.1021/acs.molpharmaceut.7b00797] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The present work investigates the modulation of grapefruit flavonoid naringenin over liver X receptor alpha (LXRα) and its target genes in THP-1 macrophages, focusing on AMP-activated protein kinase (AMPK) implication. Naringenin induced LXRα at mRNA and protein levels besides influencing the expression of LXRα target genes ABCA1, ABCG1 (ATP-binding cassette A1 and G1), and SREBP1c (sterol response element binding protein 1c) in THP-1 macrophages. The increased LXRα mRNA and protein expression was reverted when AMPK was inhibited by its chemical inhibitor, compound C or by transfection with AMPK α1 and α2 siRNA. Naringenin treatments were also able to promote reverse cholesterol transport in THP-1 cells, which is in line with the increase in the ABCA1 and ABCG1 expression found. Treatments with this flavonoid also inhibited cell migration in THP-1 cells. In conclusion, LXRα and its target genes are up-regulated by naringenin in an AMPK dependent manner in human macrophages. The enhancement in the expression of genes involved in cholesterol efflux may reveal a new mechanism by which this polyphenol can prevent atherosclerosis and foam cell progression.
Collapse
Affiliation(s)
- Javier Saenz
- Departamento de Bioquímica Médica y Biología Molecular , Universidad de Sevilla , 41004 Sevilla , Spain
| | - Consuelo Santa-María
- Departamento de Bioquímica y Biología Molecular , Universidad de Sevilla , 41004 Sevilla , Spain
| | - María Edith Reyes-Quiroz
- Departamento de Bioquímica Médica y Biología Molecular , Universidad de Sevilla , 41004 Sevilla , Spain
| | - Isabel Geniz
- Hospital Nuestra Señora de Valme , Servicio Andaluz de Salud , 41001 Sevilla , Spain
| | - Juan Jiménez
- Departamento de Bioquímica Médica y Biología Molecular , Universidad de Sevilla , 41004 Sevilla , Spain
| | - Francisco Sobrino
- Departamento de Bioquímica Médica y Biología Molecular , Universidad de Sevilla , 41004 Sevilla , Spain
| | - Gonzalo Alba
- Departamento de Bioquímica Médica y Biología Molecular , Universidad de Sevilla , 41004 Sevilla , Spain
| |
Collapse
|
20
|
Sáenz J, Alba G, Reyes-Quiroz ME, Geniz I, Jiménez J, Sobrino F, Santa-María C. Curcumin enhances LXRα in an AMP-activated protein kinase-dependent manner in human macrophages. J Nutr Biochem 2018; 54:48-56. [DOI: 10.1016/j.jnutbio.2017.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/11/2017] [Accepted: 11/11/2017] [Indexed: 12/19/2022]
|
21
|
Hao B, Xiao Y, Song F, Long X, Huang J, Tian M, Deng S, Wu Q. Metformin-induced activation of AMPK inhibits the proliferation and migration of human aortic smooth muscle cells through upregulation of p53 and IFI16. Int J Mol Med 2017; 41:1365-1376. [PMID: 29286156 PMCID: PMC5819901 DOI: 10.3892/ijmm.2017.3346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/07/2017] [Indexed: 11/28/2022] Open
Abstract
The proliferation and migration of vascular smooth muscle cells are significant in the development and progression of atherosclerosis and plaque rupture. Metformin is a widely used antidiabetic drug, which has been reported to inhibit cell growth and migration. The antiproliferative and antimigratory effects of metformin have been attributed to 5′ adenosine monophosphate-activated protein kinase (AMPK) activation. The purpose of the present study was to investigate the effects of metformin on primary human aortic muscle cells (HASMCs) in vitro and to clarify the underlying mechanism. We investigated the effectiveness of metformin in inhibiting the proliferation and migration of HASMCs in vitro using RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), cell number counting, cell viability assay, cell cycle assay and cell migration assay. Through transfection with small interfering (si)RNA targeting p53 and interferon-inducible protein 16 (IFI16), the roles of p53 and IFI16 in these processes were evaluated. The present study demonstrated that p53, IFI16 and AMPK were upregulated in senescent primary HASMCs, which exhibited a decrease in proliferation and migration. In addition, metformin was able to activate p53, IFI16 and AMPK, in order to inhibit proliferation and migration of HASMCs. Furthermore, siRNA-mediated knockdown of p53 and IFI16 attenuated AMPK activation and reversed the suppressive effects of metformin. Notably, in response to metformin, the activation of AMPK was not observed in p53- and IFI16-silenced HASMCs. These results indicated that metformin-induced activation of AMPK suppresses the proliferation and migration of HASMCs by upregulating p53 and IFI16. These findings suggested that metformin may have potential use in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Biao Hao
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Yan Xiao
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Fang Song
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Xiangshu Long
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Jing Huang
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Maobo Tian
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Shiyan Deng
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Qiang Wu
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
22
|
Bories GFP, Leitinger N. Macrophage metabolism in atherosclerosis. FEBS Lett 2017; 591:3042-3060. [PMID: 28796886 DOI: 10.1002/1873-3468.12786] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/04/2017] [Accepted: 08/04/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Gael F. P. Bories
- Department of Pharmacology and Robert M. Berne Cardiovascular Research Center; University of Virginia; Charlottsville VA USA
| | - Norbert Leitinger
- Department of Pharmacology and Robert M. Berne Cardiovascular Research Center; University of Virginia; Charlottsville VA USA
| |
Collapse
|
23
|
Wang YW, He SJ, Feng X, Cheng J, Luo YT, Tian L, Huang Q. Metformin: a review of its potential indications. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2421-2429. [PMID: 28860713 PMCID: PMC5574599 DOI: 10.2147/dddt.s141675] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metformin is the most commonly prescribed drug for type 2 diabetes mellitus. In recent years, in addition to glucose lowering, several studies have presented evidence suggesting some potential role for metformin, such as antitumor effect, antiaging effect, cardiovascular protective effect, neuroprotective effect or an optional treatment for polycystic ovary syndrome. This paper will critically review the role of metformin to provide reference for doctors and researchers.
Collapse
Affiliation(s)
- Yi-Wei Wang
- The Comprehensive Cancer Center and Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Si-Jia He
- The Comprehensive Cancer Center and Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiao Feng
- The Comprehensive Cancer Center and Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jin Cheng
- The Comprehensive Cancer Center and Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yun-Tao Luo
- The Comprehensive Cancer Center and Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ling Tian
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qian Huang
- The Comprehensive Cancer Center and Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
24
|
Abstract
The AMP-activated protein kinase (AMPK) is a central regulator of multiple metabolic pathways and may have therapeutic importance for treating obesity, insulin resistance, type 2 diabetes (T2D), non-alcoholic fatty liver disease (NAFLD), and cardiovascular disease (CVD). Given the ubiquitous expression of AMPK, it has been a challenge to evaluate which tissue types may be most beneficially poised for mediating the positive metabolic effects of AMPK-centered treatments. In this review we evaluate the metabolic phenotypes of transgenic mouse models in which AMPK expression and function have been manipulated, and the impact this has on controlling lipid metabolism, glucose homeostasis, and inflammation. This information may be useful for guiding the development of AMPK-targeted therapeutics to treat chronic metabolic diseases.
Collapse
Affiliation(s)
- Emily A Day
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada
| | - Rebecca J Ford
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada
| | - Gregory R Steinberg
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, Canada.
| |
Collapse
|
25
|
Barbati SA, Colussi C, Bacci L, Aiello A, Re A, Stigliano E, Isidori AM, Grassi C, Pontecorvi A, Farsetti A, Gaetano C, Nanni S. Transcription Factor CREM Mediates High Glucose Response in Cardiomyocytes and in a Male Mouse Model of Prolonged Hyperglycemia. Endocrinology 2017; 158:2391-2405. [PMID: 28368536 DOI: 10.1210/en.2016-1960] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/16/2017] [Indexed: 01/31/2023]
Abstract
This study aims at investigating the epigenetic landscape of cardiomyocytes exposed to elevated glucose levels. High glucose (30 mM) for 72 hours determined some epigenetic changes in mouse HL-1 and rat differentiated H9C2 cardiomyocytes including upregulation of class I and III histone deacetylase protein levels and activity, inhibition of histone acetylase p300 activity, increase in histone H3 lysine 27 trimethylation, and reduction in H3 lysine 9 acetylation. Gene expression analysis focused on cardiotoxicity revealed that high glucose induced markers associated with tissue damage, fibrosis, and cardiac remodeling such as Nexilin (NEXN), versican, cyclic adenosine 5'-monophosphate-responsive element modulator (CREM), and adrenoceptor α2A (ADRA2). Notably, the transcription factor CREM was found to be important in the regulation of cardiotoxicity-associated genes as assessed by specific small interfering RNA and chromatin immunoprecipitation experiments. In CD1 mice, made hyperglycemic by streptozotoicin (STZ) injection, cardiac structural alterations were evident at 6 months after STZ treatment and were associated with a significant increase of H3 lysine 27 trimethylation and reduction of H3 lysine 9 acetylation. Consistently, NEXN, CREM, and ADRA2 expression was significantly induced at the RNA and protein levels. Confocal microscopy analysis of NEXN localization showed this protein irregularly distributed along the sarcomeres in the heart of hyperglycemic mice. This evidence suggested a structural alteration of cardiac Z-disk with potential consequences on contractility. In conclusion, high glucose may alter the epigenetic landscape of cardiac cells. Sildenafil, restoring guanosine 3', 5'-cyclic monophosphate levels, counteracted the increase of CREM and NEXN, providing a protective effect in the presence of hyperglycemia.
Collapse
Affiliation(s)
- Saviana A Barbati
- Institute of Human Physiology, Università Cattolica di Roma, 00168 Rome, Italy
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Claudia Colussi
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
| | - Lorenza Bacci
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Aurora Aiello
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
| | - Agnese Re
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
| | - Egidio Stigliano
- Department of Histopathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Alfredo Pontecorvi
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| | - Antonella Farsetti
- Institute of Cell Biology and Neurobiology, National Research Council, 00143 Rome, Italy
- Medicine Clinic III, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Carlo Gaetano
- Medicine Clinic III, Division of Cardiovascular Epigenetics, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Simona Nanni
- Institute of Medical Pathology, Università Cattolica di Roma, 00168 Rome, Italy
| |
Collapse
|
26
|
Epigenetic changes in blood leukocytes following an omega-3 fatty acid supplementation. Clin Epigenetics 2017; 9:43. [PMID: 28450971 PMCID: PMC5405524 DOI: 10.1186/s13148-017-0345-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/14/2017] [Indexed: 12/24/2022] Open
Abstract
Background Omega-3 polyunsaturated fatty acids (n-3 FAs) have several beneficial effects on cardiovascular (CV) disease risk factors. These effects on CV risk profile may be mediated by several factors, including epigenetic modifications. Our objective is to investigate, using genome-wide DNA methylation analyses, methylation changes following an n-3 FA supplementation in overweight and obese subjects and to identify specific biological pathways potentially altered by the supplementation. Results Blood leukocytes genome-wide DNA methylation profiles of 36 overweight and obese subjects before and after a 6-week supplementation with 3 g of n-3 FAs were compared using GenomeStudio software. After supplementation, 308 CpG sites, assigned to 231 genes, were differentially methylated (FDR-corrected Diffscore ≥│13│~ P ≤ 0.05). Using Ingenuity Pathway Analysis system, a total of 55 pathways were significantly overrepresented following supplementation. Among these pathways, 16 were related to inflammatory and immune response, lipid metabolism, type 2 diabetes, and cardiovascular signaling. Changes in methylation levels of CpG sites within AKT3, ATF1, HDAC4, and IGFBP5 were correlated with changes in plasma triglyceride and glucose levels as well as with changes in the ratio of total cholesterol/HDL-cholesterol following the supplementation. Conclusions These data provide key differences in blood leukocytes DNA methylation profiles of subjects following an n-3 FA supplementation, which brings new, potential insights on metabolic pathways underlying the effects of n-3 FAs on CV health. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0345-3) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Guo L, Harari E, Virmani R, Finn AV. Linking Hemorrhage, Angiogenesis, Macrophages, and Iron Metabolism in Atherosclerotic Vascular Diseases. Arterioscler Thromb Vasc Biol 2017; 37:e33-e39. [DOI: 10.1161/atvbaha.117.309045] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Liang Guo
- From the CVPath Institute, Inc, Gaithersburg, MD
| | | | - Renu Virmani
- From the CVPath Institute, Inc, Gaithersburg, MD
| | | |
Collapse
|
28
|
Abdel Malik R, Zippel N, Frömel T, Heidler J, Zukunft S, Walzog B, Ansari N, Pampaloni F, Wingert S, Rieger MA, Wittig I, Fisslthaler B, Fleming I. AMP-Activated Protein Kinase α2 in Neutrophils Regulates Vascular Repair via Hypoxia-Inducible Factor-1α and a Network of Proteins Affecting Metabolism and Apoptosis. Circ Res 2016; 120:99-109. [PMID: 27777247 PMCID: PMC5213742 DOI: 10.1161/circresaha.116.309937] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE The AMP-activated protein kinase (AMPK) is stimulated by hypoxia, and although the AMPKα1 catalytic subunit has been implicated in angiogenesis, little is known about the role played by the AMPKα2 subunit in vascular repair. OBJECTIVE To determine the role of the AMPKα2 subunit in vascular repair. METHODS AND RESULTS Recovery of blood flow after femoral artery ligation was impaired (>80%) in AMPKα2-/- versus wild-type mice, a phenotype reproduced in mice lacking AMPKα2 in myeloid cells (AMPKα2ΔMC). Three days after ligation, neutrophil infiltration into ischemic limbs of AMPKα2ΔMC mice was lower than that in wild-type mice despite being higher after 24 hours. Neutrophil survival in ischemic tissue is required to attract monocytes that contribute to the angiogenic response. Indeed, apoptosis was increased in hypoxic neutrophils from AMPKα2ΔMC mice, fewer monocytes were recruited, and gene array analysis revealed attenuated expression of proangiogenic proteins in ischemic AMPKα2ΔMC hindlimbs. Many angiogenic growth factors are regulated by hypoxia-inducible factor, and hypoxia-inducible factor-1α induction was attenuated in AMPKα2-deficient cells and accompanied by its enhanced hydroxylation. Also, fewer proteins were regulated by hypoxia in neutrophils from AMPKα2ΔMC mice. Mechanistically, isocitrate dehydrogenase expression and the production of α-ketoglutarate, which negatively regulate hypoxia-inducible factor-1α stability, were attenuated in neutrophils from wild-type mice but remained elevated in cells from AMPKα2ΔMC mice. CONCLUSIONS AMPKα2 regulates α-ketoglutarate generation, hypoxia-inducible factor-1α stability, and neutrophil survival, which in turn determine further myeloid cell recruitment and repair potential. The activation of AMPKα2 in neutrophils is a decisive event in the initiation of vascular repair after ischemia.
Collapse
Affiliation(s)
- Randa Abdel Malik
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Nina Zippel
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Timo Frömel
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Juliana Heidler
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Sven Zukunft
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Barbara Walzog
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Nariman Ansari
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Francesco Pampaloni
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Susanne Wingert
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Michael A Rieger
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Ilka Wittig
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Beate Fisslthaler
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.)
| | - Ingrid Fleming
- From the Institute for Vascular Signaling, Centre for Molecular Medicine (R.A.M., N.Z., T.F., S.Z., B.F., I.F.), Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine (J.H., I.W.), ECCPS Metabolomics Facility, Institute for Vascular Signaling, Centre for Molecular Medicine (S.Z.), Department of Hematology/Oncology (S.W., M.A.R.), and Buchmann Institute for Molecular Life Sciences (N.A., F.P.), Goethe University, Frankfurt am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany (R.A.M., T.F., J.H., S.Z., B.F., I.F.); and Walter-Brendel-Centre of Experimental Medicine, Department of Cardiovascular Physiology and Pathophysiology, Ludwig Maximilians University, Munich, Germany (B.W.).
| |
Collapse
|
29
|
Kemmerer M, Wittig I, Richter F, Brüne B, Namgaladze D. AMPK activates LXRα and ABCA1 expression in human macrophages. Int J Biochem Cell Biol 2016; 78:1-9. [DOI: 10.1016/j.biocel.2016.06.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/10/2016] [Accepted: 06/21/2016] [Indexed: 11/16/2022]
|
30
|
Zhou Z, Tang Y, Jin X, Chen C, Lu Y, Liu L, Shen C. Metformin Inhibits Advanced Glycation End Products-Induced Inflammatory Response in Murine Macrophages Partly through AMPK Activation and RAGE/NF κB Pathway Suppression. J Diabetes Res 2016; 2016:4847812. [PMID: 27761470 PMCID: PMC5059570 DOI: 10.1155/2016/4847812] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/22/2016] [Indexed: 01/12/2023] Open
Abstract
Advanced glycation end products (AGEs) are major inflammatory mediators in diabetes, affecting atherosclerosis progression via macrophages. Metformin slows diabetic atherosclerosis progression through mechanisms that remain to be fully elucidated. The present study of murine bone marrow derived macrophages showed that (1) AGEs enhanced proinflammatory cytokines (interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α)) mRNA expression, RAGE expression, and NFκB activation; (2) metformin pretreatment inhibited AGEs effects and AGEs-induced cluster designation 86 (CD86) (M1 marker) expression, while promoting CD206 (M2 marker) surface expression and anti-inflammatory cytokine (IL-10) mRNA expression; and (3) the AMPK inhibitor, Compound C, attenuated metformin effects. In conclusion, metformin inhibits AGEs-induced inflammatory response in murine macrophages partly through AMPK activation and RAGE/NFκB pathway suppression.
Collapse
Affiliation(s)
- Zhong'e Zhou
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
- Department of Cardiology, Central Hospital of Minhang District, 170 Xinsong Road, Shanghai 201199, China
| | - Yong Tang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Xian Jin
- Department of Cardiology, Central Hospital of Minhang District, 170 Xinsong Road, Shanghai 201199, China
- *Xian Jin: and
| | - Chengjun Chen
- Department of Cardiology, Central Hospital of Minhang District, 170 Xinsong Road, Shanghai 201199, China
| | - Yi Lu
- Department of Cardiology, Central Hospital of Minhang District, 170 Xinsong Road, Shanghai 201199, China
| | - Liang Liu
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Chengxing Shen
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
- *Chengxing Shen:
| |
Collapse
|
31
|
Abstract
Initiation and progression of atherosclerosis depend on local inflammation and accumulation of lipids in the vascular wall. Although many cells are involved in the development and progression of atherosclerosis, macrophages are fundamental contributors. For nearly a decade, the phenotypic heterogeneity and plasticity of macrophages has been studied. In atherosclerotic lesions, macrophages are submitted to a large variety of micro-environmental signals, such as oxidized lipids and cytokines, which influence the phenotypic polarization and activation of macrophages resulting in a dynamic plasticity. The macrophage phenotype spectrum is characterized, at the extremes, by the classical M1 macrophages induced by T-helper 1 (Th-1) cytokines and by the alternative M2 macrophages induced by Th-2 cytokines. M2 macrophages can be further classified into M2a, M2b, M2c, and M2d subtypes. More recently, additional plaque-specific macrophage phenotypes have been identified, termed as Mox, Mhem, and M4. Understanding the mechanisms and functional consequences of the phenotypic heterogeneity of macrophages will contribute to determine their potential role in lesion development and plaque stability. Furthermore, research on macrophage plasticity could lead to novel therapeutic approaches to counteract cardiovascular diseases such as atherosclerosis. The present review summarizes our current knowledge on macrophage subsets in atherosclerotic plaques and mechanism behind the modulation of the macrophage phenotype.
Collapse
Affiliation(s)
- Sophie Colin
- Université Lille 2, Lille, France; Inserm, U1011, Lille, France; Institut Pasteur de Lille, Lille, France; European Genomic Institute for Diabetes (EGID), FR 3508, Lille, France
| | | | | |
Collapse
|
32
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, U970, Paris, France.
| |
Collapse
|
33
|
Busch AW, Montgomery BL. Interdependence of tetrapyrrole metabolism, the generation of oxidative stress and the mitigative oxidative stress response. Redox Biol 2015; 4:260-71. [PMID: 25618582 PMCID: PMC4315935 DOI: 10.1016/j.redox.2015.01.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 01/01/2023] Open
Abstract
Tetrapyrroles are involved in light harvesting and light perception, electron-transfer reactions, and as co-factors for key enzymes and sensory proteins. Under conditions in which cells exhibit stress-induced imbalances of photosynthetic reactions, or light absorption exceeds the ability of the cell to use photoexcitation energy in synthesis reactions, redox imbalance can occur in photosynthetic cells. Such conditions can lead to the generation of reactive oxygen species (ROS) associated with alterations in tetrapyrrole homeostasis. ROS accumulation can result in cellular damage and detrimental effects on organismal fitness, or ROS molecules can serve as signals to induce a protective or damage-mitigating oxidative stress signaling response in cells. Induced oxidative stress responses include tetrapyrrole-dependent and -independent mechanisms for mitigating ROS generation and/or accumulation. Thus, tetrapyrroles can be contributors to oxidative stress, but are also essential in the oxidative stress response to protect cells by contributing to detoxification of ROS. In this review, we highlight the interconnection and interdependence of tetrapyrrole metabolism with the occurrence of oxidative stress and protective oxidative stress signaling responses in photosynthetic organisms. Tetrapyrroles are involved in light sensing and oxidative stress mitigation. Reactive oxygen species (ROS) can form upon light exposure of free tetrapyrroles. Tetrapyrrole homeostasis must be tightly regulated to avoid oxidative stress. ROS can result in cellular damage or oxidative stress signaling in cells.
Collapse
|
34
|
Fullerton MD, Ford RJ, McGregor CP, LeBlond ND, Snider SA, Stypa SA, Day EA, Lhoták Š, Schertzer JD, Austin RC, Kemp BE, Steinberg GR. Salicylate improves macrophage cholesterol homeostasis via activation of Ampk. J Lipid Res 2015; 56:1025-33. [PMID: 25773887 PMCID: PMC4409279 DOI: 10.1194/jlr.m058875] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Indexed: 02/02/2023] Open
Abstract
Atherosclerosis stems from imbalances in lipid metabolism and leads to maladaptive inflammatory responses. The AMP-activated protein kinase (Ampk) is a highly conserved serine/threonine kinase that regulates many aspects of lipid and energy metabolism, although its specific role in controlling macrophage cholesterol homeostasis remains unclear. We sought to address this question by testing the effects of direct Ampk activators in primary bone marrow-derived macrophages from Ampk β1-deficient (β1−/−) mice. Macrophages from Ampk β1−/− mice had enhanced lipogenic capacity and diminished cholesterol efflux, although cholesterol uptake was unaffected. Direct activation of Ampk β1 via salicylate (the unacetylated form of aspirin) or A-769662 (a small molecule activator), decreased the synthesis of FAs and sterols in WT but not Ampk β1−/− macrophages. In lipid-laden macrophages, Ampk activation decreased cholesterol content (foam cell formation) and increased cholesterol efflux to HDL and apoA-I, effects that occurred in an Ampk β1-dependent manner. Increased cholesterol efflux was also associated with increased gene expression of the ATP binding cassette transporters, Abcg1 and Abca1. Moreover, in vivo reverse cholesterol transport was suppressed in mice that received Ampk β1−/− macrophages compared with the WT control. Our data highlight the therapeutic potential of targeting macrophage Ampk with new or existing drugs for the possible reduction in foam cell formation during the early stages of atherosclerosis.
Collapse
Affiliation(s)
- Morgan D Fullerton
- Divisions of Endocrinology and Metabolism McMaster University, Hamilton, Canada Department of Medicine, and Departments of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Canada Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Rebecca J Ford
- Divisions of Endocrinology and Metabolism McMaster University, Hamilton, Canada
| | - Chelsea P McGregor
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Nicholas D LeBlond
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Shayne A Snider
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Stephanie A Stypa
- Divisions of Endocrinology and Metabolism McMaster University, Hamilton, Canada
| | - Emily A Day
- Divisions of Endocrinology and Metabolism McMaster University, Hamilton, Canada
| | - Šárka Lhoták
- Hamilton Centre for Kidney Research, St. Joseph's Healthcare Hamilton, Hamilton, Canada Nephrology, McMaster University, Hamilton, Canada
| | - Jonathan D Schertzer
- Department of Medicine, and Departments of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Canada Pediatrics, McMaster University, Hamilton, Canada
| | - Richard C Austin
- Hamilton Centre for Kidney Research, St. Joseph's Healthcare Hamilton, Hamilton, Canada Nephrology, McMaster University, Hamilton, Canada
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, Australia
| | - Gregory R Steinberg
- Divisions of Endocrinology and Metabolism McMaster University, Hamilton, Canada Department of Medicine, and Departments of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Canada
| |
Collapse
|
35
|
Rojas J, Salazar J, Martínez MS, Palmar J, Bautista J, Chávez-Castillo M, Gómez A, Bermúdez V. Macrophage Heterogeneity and Plasticity: Impact of Macrophage Biomarkers on Atherosclerosis. SCIENTIFICA 2015; 2015:851252. [PMID: 26491604 PMCID: PMC4600540 DOI: 10.1155/2015/851252] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/09/2015] [Indexed: 05/15/2023]
Abstract
Cardiovascular disease (CVD) is a global epidemic, currently representing the worldwide leading cause of morbidity and mortality. Atherosclerosis is the fundamental pathophysiologic component of CVD, where the immune system plays an essential role. Monocytes and macrophages are key mediators in this aspect: due to their heterogeneity and plasticity, these cells may act as either pro- or anti-inflammatory mediators. Indeed, monocytes may develop heterogeneous functional phenotypes depending on the predominating pro- or anti-inflammatory microenvironment within the lesion, resulting in classic, intermediate, and non-classic monocytes, each with strikingly differing features. Similarly, macrophages may also adopt heterogeneous profiles being mainly M1 and M2, the former showing a proinflammatory profile while the latter demonstrates anti-inflammatory traits; they are further subdivided in several subtypes with more specialized functions. Furthermore, macrophages may display plasticity by dynamically shifting between phenotypes in response to specific signals. Each of these distinct cell profiles is associated with diverse biomarkers which may be exploited for therapeutic intervention, including IL-10, IL-13, PPAR-γ, LXR, NLRP3 inflammasomes, and microRNAs. Direct modulation of the molecular pathways concerning these potential macrophage-related targets represents a promising field for new therapeutic alternatives in atherosclerosis and CVD.
Collapse
Affiliation(s)
- Joselyn Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
- Endocrinology Department, Maracaibo University Hospital, Maracaibo 4004, Venezuela
- *Joselyn Rojas:
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Jim Palmar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Jordan Bautista
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Mervin Chávez-Castillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Alexis Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Valmore Bermúdez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW In overweight patients with diabetes, treatment with metformin improves cardiovascular outcomes. This observation has fuelled the hypothesis that metformin has direct cardiovascular protective properties over and above glucose lowering. Here, we discuss the various cardiovascular effects of metformin observed in preclinical studies and recent clinical trials in patients, which fail to reproduce these findings. RECENT FINDINGS Laboratory studies suggest that metformin limits atherosclerosis. Also, metformin consistently limits myocardial infarct size and reduces postinfarction remodeling in rodents.Confirmation of these effects in patients, however, appears difficult. In nondiabetic patients, metformin does not reduce carotid intima media thickness. In myocardial infarction patients, the effects of metformin on infarct size are inconclusive, but these studies suffer from methodological shortcomings. Finally, chronic administration of metformin does not affect postinfarction cardiac remodeling in nondiabetic patients. SUMMARY Although recent trials in nondiabetic patients could not confirm direct effects of metformin on atherosclerosis and cardiac remodeling, an acute cardioprotective effect of metformin cannot be excluded yet. We might have to consider, though, that the beneficial effect of metformin on cardiovascular prognosis in patients with diabetes is due to its effects on glucose metabolism and body weight rather than due to pleiotropic direct cardiovascular effects.
Collapse
Affiliation(s)
- Niels P Riksen
- aDepartment of Internal Medicine bPharmacology-Toxicology, Radboud university medical center, Nijmegen, the Netherlands
| | | |
Collapse
|
37
|
Abstract
Macrophage accumulation within the vascular wall is a hallmark of atherosclerosis. In atherosclerotic lesions, macrophages respond to various environmental stimuli, such as modified lipids, cytokines, and senescent erythrocytes, which can modify their functional phenotypes. The results of studies on human atherosclerotic plaques demonstrate that the relative proportions of macrophage subsets within a plaque might be a better indicator of plaque phenotype and stability than the total number of macrophages. Understanding the function of specific macrophage subsets and their contribution to the composition and growth of atherosclerotic plaques would aid the identification of novel strategies to delay or halt the development of the disease and its associated pathophysiological consequences. However, most studies aimed at characterizing the phenotypes of human macrophages are performed in vitro and, therefore, their functional relevance to human pathology remains uncertain. In this Review, the diverse range of macrophage phenotypes in atherosclerotic lesions and their potential roles in both plaque progression and stability are discussed, with an emphasis on human pathology.
Collapse
Affiliation(s)
- Giulia Chinetti-Gbaguidi
- INSERM U1011, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, BP 245, Lille 59019, France
| | - Sophie Colin
- INSERM U1011, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, BP 245, Lille 59019, France
| | - Bart Staels
- INSERM U1011, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, BP 245, Lille 59019, France
| |
Collapse
|