1
|
Zhang Y, Sun H, Gandhi A, Du Y, Ebrahimi S, Jiang Y, Xu S, Uwase H, Seidel A, Bingaman SS, Arnold AC, Nguyen C, Ding W, Woolard MD, Hobbs R, Bagchi P, He P. Role of shear stress-induced red blood cell released ATP in atherosclerosis. Am J Physiol Heart Circ Physiol 2025; 328:H774-H791. [PMID: 39982440 DOI: 10.1152/ajpheart.00875.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/31/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Altered hemodynamics is a key factor for atherosclerosis. For decades, endothelial cell (EC) responses to fluid-generated wall shear stress have been the central focus for atherogenesis. However, circulating blood is not a cell-free fluid, it contains mechanosensitive red blood cells (RBCs) that are also subjected to altered hemodynamics and release a large amount of ATP, but their impact on atherosclerosis has been overlooked. The focus of this study is the role of shear stress (SS)-induced RBC-released ATP in atherosclerosis. Hypercholesterolemic mouse models with and without RBC-Pannexin 1 deletion were used for the study. Results showed that SS-induced release of ATP from RBCs was at µM concentrations, three-orders of magnitude higher than that from other cell types. Suppression of RBC-released ATP via deletion of Pannexin 1, a mechanosensitive ATP-permeable channel, reduced high-fat diet-induced aortic plaque burden by 40%-60%. Importantly, the location and the extent of aortic atherosclerotic lesions spatially matched with the ATP deposition profile at aortic wall predicted by a computational fluid dynamic (CFD) model. Furthermore, hypercholesterolemia increases EC susceptibility to ATP with potentiated increase in [Ca2+]i, an initial signaling for aortic EC barrier dysfunction, and an essential cause for lipid accumulation and inflammatory cell infiltration. The computational prediction also provides a physics-based explanation for RBC-released ATP-induced sex disparities in atherosclerosis. Our study reveals an important role of RBC-released ATP in the initiation and progression of atherosclerosis. These novel findings provide a more comprehensive view of how altered hemodynamics and systemic risk factors synergistically contribute to atherosclerosis.NEW & NOTEWORTHY This study reveals that, in addition to fluid-derived wall shear stress, the disturbed blood flow-induced release of ATP from mechanosensitive red blood cells (RBCs), the major cellular components of blood, along with hypercholesterolemia-induced increases in endothelial cell susceptibility to ATP contribute significantly to the initiation and progression of atherosclerosis. These novel findings advance our current understanding of how altered hemodynamics and hypercholesterolemia synergistically contribute to atherosclerosis for the first time with the inclusion of RBCs.
Collapse
Affiliation(s)
- Yunpei Zhang
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Haoyu Sun
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Aayush Gandhi
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States
| | - Yong Du
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Saman Ebrahimi
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States
| | - Yanyan Jiang
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Sulei Xu
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Hope Uwase
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Alane Seidel
- Transgenic core, Department of Comparative Medicine, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Sarah S Bingaman
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Christian Nguyen
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Wei Ding
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States
| | - Ryan Hobbs
- Department of Dermatology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| | - Prosenjit Bagchi
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania, United States
| |
Collapse
|
2
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
3
|
Maaniitty E, Jalkanen J, Sinisilta S, Gunn J, Vasankari T, Biancari F, Jalkanen S, Airaksinen KEJ, Hollmen M, Kiviniemi T. Differential circulating cytokine profiles in acute coronary syndrome versus stable coronary artery disease. Sci Rep 2024; 14:17269. [PMID: 39068298 PMCID: PMC11283453 DOI: 10.1038/s41598-024-68333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Chronic inflammation plays a crucial role in coronary artery disease (CAD), but differences in specific cytokine profiles between acute coronary syndrome (ACS) and stable CAD remain unknown. We investigated cytokine differences between these two manifestations of CAD. The study included 308 patients with angiographically detected, hemodynamically significant CAD: 150 patients undergone angiography for ACS, 158 patients undergone angiography for stable CAD. To assess dynamic changes, 116 patients had index angiogram at least 3 months earlier. We measured the serum concentrations of 48 circulating cytokines. The ACS group had decreased interleukin (IL) 4 (p = 0.005), and increased IL-8 (p = 0.008), hepatocyte growth factor (HGF) (p < 0.001) and macrophage colony-stimulating factor (M-CSF) (p = 0.002) levels compared with the stable CAD group. Multivariable logistic regression revealed increased levels of HGF (OR 18.050 [95% CI 4.372-74.517], p < 0.001), M-CSF (OR 2.257 [1.375-3.705], p = 0.001) and IL-6 (OR 1.586 [1.131-2.224], p = 0.007), independently associated with ACS. In the post-angiography group, only diminished platelet-derived growth factor-BB levels in ACS-manifested patients were observed (OR 0.478, [0.279-0.818], p = 0.007). Cytokine profiles differ between ACS and stable CAD. Such differences seem to be mainly reversible within 3 months after ACS. Thus, targeting one or two cytokines only might not offer one-size fits all-therapeutic approach for CAD-associated inflammation.Trial registration: NCT03444259.
Collapse
Affiliation(s)
- Eveliina Maaniitty
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland.
| | - Juho Jalkanen
- Vascular Surgery, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Sami Sinisilta
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Jarmo Gunn
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Tuija Vasankari
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Fausto Biancari
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
- Department of Medicine, South Karelia Central Hospital, University of Helsinki, Valto Käkelän Katu 1, 53130, Lappeenranta, Finland
| | - Sirpa Jalkanen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - K E Juhani Airaksinen
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Maija Hollmen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Tuomas Kiviniemi
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland.
| |
Collapse
|
4
|
Ou Z, Cheng Y, Ma H, Chen K, Lin Q, Chen J, Guo R, Huang Z, Cheng Q, Alaeiilkhchi N, Zhu Q, Huang Z, Jiang H. miR-223 accelerates lipid droplets clearance in microglia following spinal cord injury by upregulating ABCA1. J Transl Med 2024; 22:659. [PMID: 39010173 PMCID: PMC11247820 DOI: 10.1186/s12967-024-05480-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is characterized by extensive demyelination and inflammatory responses. Facilitating the clearance of lipid droplets (LDs) within microglia contributes to creating a microenvironment that favors neural recovery and provides essential materials for subsequent remyelination. Therefore, investigating MicroRNAs (miRNAs) that regulate lipid homeostasis after SCI and elucidating their potential mechanisms in promoting LDs clearance in microglia have become focal points of SCI research. METHODS We established a subacute C5 hemicontusion SCI model in mice and performed transcriptomic sequencing on the injury epicenter to identify differentially expressed genes and associated pathways. Confocal imaging was employed to observe LDs accumulation. Multi-omics analyses were conducted to identify differentially expressed mRNA and miRNA post-SCI. Pathway enrichment analysis and protein-protein interaction network construction were performed using bioinformatics methods, revealing miR-223-Abca1 as a crucial miRNA-mRNA pair in lipid metabolism regulation. BV2 microglia cell lines overexpressing miR-223 were engineered, and immunofluorescence staining, western blot, and other techniques were employed to assess LDs accumulation, relevant targets, and inflammatory factor expression, confirming its role in regulating lipid homeostasis in microglia. RESULTS Histopathological results of our hemicontusion SCI model confirmed LDs aggregation at the injury epicenter, predominantly within microglia. Our transcriptomic analysis during the subacute phase of SCI in mice implicated ATP-binding cassette transporter A1 (Abca1) as a pivotal gene in lipid homeostasis, cholesterol efflux and microglial activation. Integrative mRNA-miRNA multi-omics analysis highlighted the crucial role of miR-223 in the neuroinflammation process following SCI, potentially through the regulation of lipid metabolism via Abca1. In vitro experiments using BV2 cells overexpressing miR-223 demonstrated that elevated levels of miR-223 enhance ABCA1 expression in myelin debris and LPS-induced BV2 cells. This promotes myelin debris degradation and LDs clearance, and induces a shift toward an anti-inflammatory M2 phenotype. CONCLUSIONS In summary, our study unveils the critical regulatory role of miR-223 in lipid homeostasis following SCI. The mechanism by which this occurs involves the upregulation of ABCA1 expression, which facilitates LDs clearance and myelin debris degradation, consequently alleviating the lipid burden, and inhibiting inflammatory polarization of microglia. These findings suggest that strategies to enhance miR-223 expression and target ABCA1, thereby augmenting LDs clearance, may emerge as appealing new clinical targets for SCI treatment.
Collapse
Affiliation(s)
- Zhilin Ou
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yongquan Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hao Ma
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qiong Lin
- School of Anesthesiology, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jiayu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ruqin Guo
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiping Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qixian Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Nima Alaeiilkhchi
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, Vancouver, Canada
| | - Qingan Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zucheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Hui Jiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
5
|
Banerjee A, Singla DK. MSC exosomes attenuate sterile inflammation and necroptosis associated with TAK1-pJNK-NFKB mediated cardiomyopathy in diabetic ApoE KO mice. Front Immunol 2024; 15:1348043. [PMID: 38390337 PMCID: PMC10881775 DOI: 10.3389/fimmu.2024.1348043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Introduction Diabetes is a debilitating disease that leads to complications like cardiac dysfunction and heart failure. In this study, we investigated the pathophysiology of diabetes-induced cardiac dysfunction in mice with dyslipidemia. We hypothesize diabetes in ApoE knockout (ApoE-/-) mice induces cardiac dysfunction by increasing inflammation and necroptosis. Methods ApoE-/- mice were divided into experimental groups: Control, Streptozotocin (STZ), STZ + MSC-Exo (mesenchymal stem cell-derived exosomes), and STZ+MEF-Exo (Mouse embryonic fibroblast derived exosomes). At Day 42, we assessed cardiac function, collected blood and heart tissues. Heart tissue samples were analyzed for inflammation, necroptosis, signaling mechanism, hypertrophy and adverse structural remodeling using histology, immunohistochemistry, western blotting, RT-PCR, cytokine array and TF array. Results and Discussion STZ treated ApoE-/- mice developed diabetes, with significantly (p<0.05) increased blood glucose and body weight loss. These mice developed cardiac dysfunction with significantly (p<0.05) increased left ventricular internal diameter end diastole and end systole, and decreased ejection fraction, and fractional shortening. We found significant (p<0.05) increased expression of inflammatory cytokines TNF- a, IL-6, IL-1a, IL-33 and decreased IL-10 expression. Diabetic mice also exhibited significantly (p<0.05) increased necroptosis marker expression and infiltration of inflammatory monocytes and macrophages. MSC-Exos treated mice showed recovery of diabetes associated pathologies with significantly reduced blood glucose, recovered body weight, increased IL-10 secretion and M2 polarized macrophages in the heart. These mice showed reduced TAK1-pJNK-NFKB inflammation associated expression and improved cardiac function with significantly reduced cardiac hypertrophy and fibrosis compared to diabetic mice. Treatment with MEF-Exos did not play a significant role in attenuating diabetes-induced cardiomyopathy as these treatment mice presented with cardiac dysfunction and underlying pathologies observed in STZ mice. Conclusion Thus, we conclude that cardiac dysfunction develops in diabetic ApoE-/- mice, arising from inflammation, necroptosis, and adverse tissue remodeling, which is ameliorated by MSC-Exos, a potential therapeutic for diabetes-induced cardiomyopathy.
Collapse
Affiliation(s)
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
6
|
Maas SL, Donners MMPC, van der Vorst EPC. ADAM10 and ADAM17, Major Regulators of Chronic Kidney Disease Induced Atherosclerosis? Int J Mol Sci 2023; 24:ijms24087309. [PMID: 37108478 PMCID: PMC10139114 DOI: 10.3390/ijms24087309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a major health problem, affecting millions of people worldwide, in particular hypertensive and diabetic patients. CKD patients suffer from significantly increased cardiovascular disease (CVD) morbidity and mortality, mainly due to accelerated atherosclerosis development. Indeed, CKD not only affects the kidneys, in which injury and maladaptive repair processes lead to local inflammation and fibrosis, but also causes systemic inflammation and altered mineral bone metabolism leading to vascular dysfunction, calcification, and thus, accelerated atherosclerosis. Although CKD and CVD individually have been extensively studied, relatively little research has studied the link between both diseases. This narrative review focuses on the role of a disintegrin and metalloproteases (ADAM) 10 and ADAM17 in CKD and CVD and will for the first time shed light on their role in CKD-induced CVD. By cleaving cell surface molecules, these enzymes regulate not only cellular sensitivity to their micro-environment (in case of receptor cleavage), but also release soluble ectodomains that can exert agonistic or antagonistic functions, both locally and systemically. Although the cell-specific roles of ADAM10 and ADAM17 in CVD, and to a lesser extent in CKD, have been explored, their impact on CKD-induced CVD is likely, yet remains to be elucidated.
Collapse
Affiliation(s)
- Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), 80336 Munich, Germany
| |
Collapse
|
7
|
Baradaran A, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Shadbad MA, Khosravi N, Derakhshani A, Alemohammad H, Afrashteh Nour M, Safarpour H, Silvestris N, Brunetti O, Baradaran B. The cross-talk between tumor-associated macrophages and tumor endothelium: Recent advances in macrophage-based cancer immunotherapy. Biomed Pharmacother 2022; 146:112588. [PMID: 35062062 DOI: 10.1016/j.biopha.2021.112588] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/02/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are among the abundant cell populations of the tumor microenvironment (TME), which have pivotal roles in tumor development, chemoresistance, immune evasion, and metastasis. Growing evidence indicates that TAMs and the cross-talk between TAMs and tumoral endothelial cells can substantially contribute to tumor angiogenesis, which is considered a vital process for cancer development. Besides, tumoral endothelial cells can regulate the leukocyte infiltration to the TME in solid cancers and contribute to immune evasion. Therefore, targeting the immunosuppressive TAMs and the cross-talk between them can be a promising strategy for improving anti-tumoral immune responses. This review aims to summarize the biology of TAMs, their recently identified roles in tumor development/angiogenesis, and recent advances in macrophage-based cancer immunotherapy approaches for treating cancers.
Collapse
Affiliation(s)
- Ali Baradaran
- Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia; Research & Development, BSD Robotics, Queensland, Australia
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Hajar Alemohammad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nicola Silvestris
- Medical Oncology Unit-IRCCS IstitutoTumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit-IRCCS IstitutoTumori "Giovanni Paolo II" of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Razeghian-Jahromi I, Karimi Akhormeh A, Razmkhah M, Zibaeenezhad MJ. Immune system and atherosclerosis: Hostile or friendly relationship. Int J Immunopathol Pharmacol 2022; 36:3946320221092188. [PMID: 35410514 PMCID: PMC9009140 DOI: 10.1177/03946320221092188] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
Coronary artery disease has remained a major health challenge despite enormous progress in prevention, diagnosis, and treatment strategies. Formation of atherosclerotic plaque is a chronic process that is developmentally influenced by intrinsic and extrinsic determinants. Inflammation triggers atherosclerosis, and the fundamental element of inflammation is the immune system. The immune system involves in the atherosclerosis process by a variety of immune cells and a cocktail of mediators. It is believed that almost all main components of this system possess a profound contribution to the atherosclerosis. However, they play contradictory roles, either protective or progressive, in different stages of atherosclerosis progression. It is evident that monocytes are the first immune cells appeared in the atherosclerotic lesion. With the plaque growth, other types of the immune cells such as mast cells, and T lymphocytes are gradually involved. Each cell releases several cytokines which cause the recruitment of other immune cells to the lesion site. This is followed by affecting the expression of other cytokines as well as altering certain signaling pathways. All in all, a mix of intertwined interactions determine the final outcome in terms of mild or severe manifestations, either clinical or subclinical. Therefore, it is of utmost importance to precisely understand the kind and degree of contribution which is made by each immune component in order to stop the growing burden of cardiovascular morbidity and mortality. In this review, we present a comprehensive appraisal on the role of immune cells in the atherosclerosis initiation and development.
Collapse
Affiliation(s)
| | - Ali Karimi Akhormeh
- Cardiovascular Research Center, Shiraz University of Medical
Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer
Research, Shiraz University of Medical
Sciences, Shiraz, Iran
| | | |
Collapse
|
9
|
Chan YH, Ramji DP. Key Roles of Inflammation in Atherosclerosis: Mediators Involved in Orchestrating the Inflammatory Response and Its Resolution in the Disease Along with Therapeutic Avenues Targeting Inflammation. Methods Mol Biol 2022; 2419:21-37. [PMID: 35237956 DOI: 10.1007/978-1-0716-1924-7_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Inflammation is a critical driver of all stages of atherosclerosis, from lesion development to plaque rupture. Cytokines are mediators of the immune response and in atherosclerosis, the balance of anti- and pro-inflammatory cytokines is tipped in favor of the latter, resulting in persistent and unresolved inflammation. Although reducing plasma cholesterol levels mainly via the use of statins has positively impacted patient outcomes and reduced mortality rates, the presence of significant residual inflammation and cardiovascular risk posttherapy emphasizes the prevailing risk of primary and secondary events driven by inflammation independently of hyperlipidemia. Given the dominant role of inflammation in driving pathogenesis, alternative therapeutic avenues beyond targeting lowering of plasma lipids are required. This chapter will discuss the role of inflammation and pro-inflammatory cytokines in driving atherogenesis and disease progression, the therapeutic potential of targeting cytokines for atherosclerosis and promising avenues in this area.
Collapse
Affiliation(s)
- Yee-Hung Chan
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK.
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
10
|
van Ingen E, Foks AC, Woudenberg T, van der Bent ML, de Jong A, Hohensinner PJ, Wojta J, Bot I, Quax PHA, Nossent AY. Inhibition of microRNA-494-3p activates Wnt signaling and reduces proinflammatory macrophage polarization in atherosclerosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1228-1239. [PMID: 34853722 PMCID: PMC8607137 DOI: 10.1016/j.omtn.2021.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/24/2021] [Accepted: 10/28/2021] [Indexed: 12/31/2022]
Abstract
We have previously shown that treatment with third-generation antisense oligonucleotides against miR-494-3p (3GA-494) reduces atherosclerotic plaque progression and stabilizes lesions, both in early and established plaques, with reduced macrophage content in established plaques. Within the plaque, different subtypes of macrophages are present. Here, we aimed to investigate whether miR-494-3p directly influences macrophage polarization and activation. Human macrophages were polarized into either proinflammatory M1 or anti-inflammatory M2 macrophages and simultaneously treated with 3GA-494 or a control antisense (3GA-ctrl). We show that 3GA-494 treatment inhibited miR-494-3p in M1 macrophages and dampened M1 polarization, while in M2 macrophages miR-494-3p expression was induced and M2 polarization enhanced. The proinflammatory marker CCR2 was reduced in 3GA-494-treated atherosclerosis-prone mice. Pathway enrichment analysis predicted an overlap between miR-494-3p target genes in macrophage polarization and Wnt signaling. We demonstrate that miR-494-3p regulates expression levels of multiple Wnt signaling components, such as LRP6 and TBL1X. Wnt signaling appears activated upon treatment with 3GA-494, both in cultured M1 macrophages and in plaques of hypercholesterolemic mice. Taken together, 3GA-494 treatment dampened M1 polarization, at least in part via activated Wnt signaling, while M2 polarization was enhanced, which is both favorable in reducing atherosclerotic plaque formation and increasing plaque stability.
Collapse
Affiliation(s)
- Eva van Ingen
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Division of BioTherapeutics, LACDR, Leiden University, 2333 CC Leiden, The Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, LACDR, Leiden University, 2333 CC Leiden, The Netherlands
| | - Tamar Woudenberg
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - M Leontien van der Bent
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Alwin de Jong
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Philipp J Hohensinner
- Department of Internal Medicine II, Medical, University of Vienna, 1090 Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical, University of Vienna, 1090 Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
| | - Ilze Bot
- Division of BioTherapeutics, LACDR, Leiden University, 2333 CC Leiden, The Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Anne Yaël Nossent
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Department of Internal Medicine II, Medical, University of Vienna, 1090 Vienna, Austria.,Department of Laboratory Medicine, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
11
|
Zhang W, Xu L, Zhu L, Liu Y, Yang S, Zhao M. Lipid Droplets, the Central Hub Integrating Cell Metabolism and the Immune System. Front Physiol 2021; 12:746749. [PMID: 34925055 PMCID: PMC8678573 DOI: 10.3389/fphys.2021.746749] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Lipid droplets (LDs) are commonly found in various biological cells and are organelles related to cell metabolism. LDs, the number and size of which are heterogeneous across cell type, are primarily composed of polar lipids and proteins on the surface with neutral lipids in the core. Neutral lipids stored in LDs can be degraded by lipolysis and lipophagocytosis, which are regulated by various proteins. The process of LD formation can be summarized in four steps. In addition to energy production, LDs play an extremely pivotal role in a variety of physiological and pathological processes, such as endoplasmic reticulum stress, lipid toxicity, storage of fat-soluble vitamins, regulation of oxidative stress, and reprogramming of cell metabolism. Interestingly, LDs, the hub of integration between metabolism and the immune system, are involved in antitumor immunity, anti-infective immunity (viruses, bacteria, parasites, etc.) and some metabolic immune diseases. Herein, we summarize the role of LDs in several major immune cells as elucidated in recent years, including T cells, dendritic cells, macrophages, mast cells, and neutrophils. Additionally, we analyze the role of the interaction between LDs and immune cells in two typical metabolic immune diseases: atherosclerosis and Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya Hospital, Central South University, Changsha, China
| | - Linyong Xu
- School of Life Sciences, Central South University, Changsha, China
| | - Ling Zhu
- School of Life Sciences, Central South University, Changsha, China
| | - Yifan Liu
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Siwei Yang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Dang D, Liu Y, Zhou Q, Li H, Wang Y, Wu H. Identification of a novel IRF8 homozygous mutation causing neutrophilia, monocytopenia and fatal infection in a female neonate. INFECTION GENETICS AND EVOLUTION 2021; 96:105121. [PMID: 34666172 DOI: 10.1016/j.meegid.2021.105121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/29/2022]
Abstract
Inborn errors of immunity (IEIs) result from mutations in genes involved in host immune defense and immune regulation. Herein, we report the identification of a novel IRF8 mutation in a neonate with an IEI. DNA samples from both the neonate and her parents were subjected to DNA sequencing, and the immune status of the patient was assessed. We identified a mutation (c.331C > T, p. Arg111*) in the interferon regulatory factor 8 (IRF8) gene that manifested as sever dysfunctional neutrophilia (96.53 × 109/l) and monocytopenia (0.02 × 109/l). The patient's CD3+ T cell and CD8+ T cell counts were decreased. Her levels of IFN-γ were low even during severe infection. The mRNA expression levels of IRF8 were lower than normal. Her clinical manifestations included a recurrent and progressively fatal infection. Since IRF8 plays a key role in the differentiation and development of immune cells, we suspected that the novel mutation (c.331C > T, p. Arg111*) may be consistent with a severe loss of IRF8 function and result in a failure of immune cells to differentiate and maturation, and lead to a severe infection with early onset.
Collapse
Affiliation(s)
- Dan Dang
- Department of Neonatology, First Hospital of Jilin University, Changchun 130021, China
| | - Ying Liu
- Department of Neonatology, First Hospital of Jilin University, Changchun 130021, China
| | - Qi Zhou
- Department of Neonatology, First Hospital of Jilin University, Changchun 130021, China
| | - Heng Li
- Department of Neonatology, First Hospital of Jilin University, Changchun 130021, China
| | - Ying Wang
- Research Institute of pediatrics, First Hospital of Jilin University, Changchun 130021, China
| | - Hui Wu
- Department of Neonatology, First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
13
|
Sharifiaghdam M, Shaabani E, Sharifiaghdam Z, De Keersmaecker H, Lucas B, Lammens J, Ghanbari H, Teimoori-Toolabi L, Vervaet C, De Beer T, Faridi-Majidi R, De Smedt SC, Braeckmans K, Fraire JC. Macrophage reprogramming into a pro-healing phenotype by siRNA delivered with LBL assembled nanocomplexes for wound healing applications. NANOSCALE 2021; 13:15445-15463. [PMID: 34505619 DOI: 10.1039/d1nr03830c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Excessive inflammatory responses in wounds are characterized by the presence of high levels of pro-inflammatory M1 macrophages rather than pro-healing M2 macrophages, which leads to delayed wound healing. Macrophage reprogramming from the M1 to M2 phenotype through knockdown of interferon regulatory factor 5 (irf5) has emerged as a possible therapeutic strategy. While downregulation of irf5 could be achieved by siRNA, it very much depends on successful intracellular delivery by suitable siRNA carriers. Here, we report on highly stable selenium-based layer-by-layer (LBL) nanocomplexes (NCs) for siRNA delivery with polyethyleneimine (PEI-LBL-NCs) as the final polymer layer. PEI-LBL-NCs showed good protection of siRNA with only 40% siRNA release in a buffer of pH = 8.5 after 72 h or in simulated wound fluid after 4 h. PEI-LBL-NCs also proved to be able to transfect RAW 264.7 cells with irf5-siRNA, resulting in successful reprogramming to the M2 phenotype as evidenced by a 3.4 and 2.6 times decrease in NOS-2 and TNF-α mRNA expression levels, respectively. Moreover, irf5-siRNA transfected cells exhibited a 2.5 times increase of the healing mediator Arg-1 and a 64% increase in expression of the M2 cell surface marker CD206+. Incubation of fibroblast cells with conditioned medium isolated from irf5-siRNA transfected RAW 264.7 cells resulted in accelerated wound healing in an in vitro scratch assay. These results show that irf5-siRNA loaded PEI-LBL-NCs are a promising therapeutic approach to tune macrophage polarization for improved wound healing.
Collapse
Affiliation(s)
- Maryam Sharifiaghdam
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Elnaz Shaabani
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zeynab Sharifiaghdam
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Herlinde De Keersmaecker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Bart Lucas
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
| | - Joris Lammens
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | | | - Chris Vervaet
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Thomas De Beer
- Laboratory of Pharmaceutical Process Analytical Technology (LPPAT), Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium.
| |
Collapse
|
14
|
Williams H, Mack CD, Li SCH, Fletcher JP, Medbury HJ. Nature versus Number: Monocytes in Cardiovascular Disease. Int J Mol Sci 2021; 22:ijms22179119. [PMID: 34502027 PMCID: PMC8430468 DOI: 10.3390/ijms22179119] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 01/01/2023] Open
Abstract
Monocytes play a key role in cardiovascular disease (CVD) as their influx into the vessel wall is necessary for the development of an atherosclerotic plaque. Monocytes are, however, heterogeneous differentiating from classical monocytes through the intermediate subset to the nonclassical subset. While it is recognized that the percentage of intermediate and nonclassical monocytes are higher in individuals with CVD, accompanying changes in inflammatory markers suggest a functional impact on disease development that goes beyond the increased proportion of these ‘inflammatory’ monocyte subsets. Furthermore, emerging evidence indicates that changes in monocyte proportion and function arise in dyslipidemia, with lipid lowering medication having some effect on reversing these changes. This review explores the nature and number of monocyte subsets in CVD addressing what they are, when they arise, the effect of lipid lowering treatment, and the possible implications for plaque development. Understanding these associations will deepen our understanding of the clinical significance of monocytes in CVD.
Collapse
Affiliation(s)
- Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Corinne D. Mack
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Stephen C. H. Li
- Chemical Pathology, NSW Health Pathology, Westmead Hospital and Institute of Clinical Pathology and Medical Research, Westmead, Sydney, NSW 2145, Australia;
- Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Western Sydney University, Blacktown, NSW 2148, Australia
| | - John P. Fletcher
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Heather J. Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
15
|
Li BY, Tan W, Zou JL, He Y, Yoshida S, Jiang B, Zhou YD. Role of interferons in diabetic retinopathy. World J Diabetes 2021; 12:939-953. [PMID: 34326947 PMCID: PMC8311473 DOI: 10.4239/wjd.v12.i7.939] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/15/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the major causes of visual impairment and irreversible blindness in developed regions. Aside from abnormal angiogenesis, inflammation is the most specific and might be the initiating factor of DR. As a key participant in inflammation, interferon-gamma (IFN-γ) can be detected in different parts of the eye and is responsible for the breakdown of the blood-retina barrier and activation of inflammatory cells and other cytokines, which accelerate neovascularization and neuroglial degeneration. In addition, IFN-γ is involved in other vascular complications of diabetes mellitus and angiogenesis-dependent diseases, such as diabetic nephropathy, cerebral microbleeds, and age-related macular degeneration. Traditional treatments, such as anti-vascular endothelial growth factor agents, vitrectomy, and laser photocoagulation therapy, are more effective for angiogenesis and not tolerable for every patient. Many ongoing clinical trials are exploring effective drugs that target inflammation. For instance, IFN-α acts against viruses and angiogenesis and is commonly used to treat malignant tumors. Moreover, IFN-α has been shown to contribute to alleviating the progression of DR and other ocular diseases. In this review, we emphasize the roles that IFNs play in the pathogenesis of DR and discuss potential clinical applications of IFNs in DR, such as diagnosis, prognosis, and therapeutic treatment.
Collapse
Affiliation(s)
- Bing-Yan Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
- Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
- Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Jing-Ling Zou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
- Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yan He
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
- Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume 830-0011, Fukuoka, Japan
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
- Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Ye-Di Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
- Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
16
|
Su X, Rakshit M, Das P, Gupta I, Das D, Pramanik M, Ng KW, Kwan J. Ultrasonic Implantation and Imaging of Sound-Sensitive Theranostic Agents for the Treatment of Arterial Inflammation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:24422-24430. [PMID: 34019376 DOI: 10.1021/acsami.1c01161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
For site-specific diseases such as atherosclerosis, it is desirable to noninvasively and locally deliver therapeutics for extended periods of time. High-intensity focused ultrasound (HIFU) provides targeted drug delivery, yet remains unable to sustain delivery beyond the HIFU treatment time. Furthermore, methods to validate HIFU-enhanced drug delivery remain limited. In this study, we report on HIFU-targeted implantation of degradable drug-loaded sound-sensitive multicavity PLGA microparticles (mcPLGA MPs) as a theranostic agent for the treatment of arterial lesions. Once implanted into the targeted tissue, mcPLGA MPs eluted dexamethasone for several days, thereby reducing inflammatory markers linked to oxidized lipid uptake in a foam cell spheroid model. Furthermore, implanted mcPLGA MPs created hyperechoic regions on diagnostic ultrasound images, and thus noninvasively verified that the target region was treated with the theranostic agents. This novel and innovative multifunctional theranostic platform may serve as a promising candidate for noninvasive imaging and treatment for site-specific diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Xiaoqian Su
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Moumita Rakshit
- School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore
| | - Prativa Das
- School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore
| | - Ipshita Gupta
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Dhiman Das
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Manojit Pramanik
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment and Water Research Institute, 1 Cleantech Loop, CleanTech One, 637141, Singapore
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, 665 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - James Kwan
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, United Kingdom
| |
Collapse
|
17
|
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
|
18
|
Suzuki H, Mikami T, Iwahara N, Akiyama Y, Wanibuchi M, Komatsu K, Yokoyama R, Hirano T, Hosoda R, Horio Y, Kuno A, Mikuni N. Aging-associated inflammation and fibrosis in arachnoid membrane. BMC Neurol 2021; 21:169. [PMID: 33882882 PMCID: PMC8058966 DOI: 10.1186/s12883-021-02202-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/13/2021] [Indexed: 11/16/2024] Open
Abstract
Background The physiological and pathological significance of the arachnoid membrane (AM) is still unknown. In this study, we investigated various characteristics of the AM, focusing on the influence of inflammation and fibrosis. Methods Small pieces of AM sample were obtained during neurosurgical procedures from 74 cases. The clinical and pathological characteristics of the hyperplastic AM group (≥ 50 μm) and the non-hyperplastic AM group (< 50 μm) were compared. Then, potential correlations between AM thickness and clinical characteristics were analyzed. Moreover, VEGFα, TGFβ, and TGFα levels were quantitated by real time PCR. Then, the potential correlations between AM thickness and these inflammatory or anti-inflammatory markers, and the influence of the original disease were calculated. Results The median age of the patients in hyperplastic AM group was significantly older than that of the non-hyperplastic AM group. Moreover, the number of fibroblasts, CD68+ cells, CD86+ cells, and CD206+ cells in the hyperplastic AM group was significantly higher than that in the non-hyperplastic AM group. The AM thickness was significantly correlated to age and number of fibroblasts, CD68+ cells, CD86+ cells, and CD206+ cells. The thickness of the AM was significantly correlated to the messenger RNA expression levels of VEGFα (ρ = 0.337), and the VEGFα expression levels were significantly correlated with TGFβ and TNFα. Conclusions The AM hyperplasia was influenced by aging and could be a result of inflammation and fibrosis through cytokine secretion from the inflammatory cells and fibroblasts in the AM.
Collapse
Affiliation(s)
- Hime Suzuki
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takeshi Mikami
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan.
| | - Naotoshi Iwahara
- Department of Pharmacology, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yukinori Akiyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Katsuya Komatsu
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Rintaro Yokoyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Tsukasa Hirano
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Ryusuke Hosoda
- Department of Pharmacology, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Nobuhiro Mikuni
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| |
Collapse
|
19
|
Mohd Idrus FN, Ahmad NS, Hoe CH, Azlan M, Norfuad FA, Yusof Z, Wan Isa WYH, Mohamed Ali AA, Yvonne-Tee GB. Differential polarization and the expression of efferocytosis receptor MerTK on M1 and M2 macrophages isolated from coronary artery disease patients. BMC Immunol 2021; 22:21. [PMID: 33761885 PMCID: PMC7992933 DOI: 10.1186/s12865-021-00410-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 01/04/2021] [Indexed: 01/11/2023] Open
Abstract
Background Differential polarization of macrophage into M1 and M2 mediates atherosclerotic plaque clearance through efferocytosis. Higher expression of Mer proto-oncogene tyrosine kinase (MerTK) on M2 macrophage helps in maintaining macrophage efferocytic efficiency. In healthy individuals, macrophage polarization into M1 and M2 occurs in tissues in concomitance with the acquisition of functional phenotypes depending on specific microenvironment stimuli. However, whether the macrophage differential polarization and MerTK expression vary in coronary artery disease (CAD) patients remain unknown. Objective This study aimed to elucidate the polarization of M1 and M2 macrophage from CAD patients as well as to investigate the expression of MerTK in these macrophage phenotypes. Methods A total of 14 (n) CAD patients were recruited and subsequently grouped into “no apparent CAD”, “non-obstructive CAD” and “obstructive CAD” according to the degree of stenosis. Thirty ml of venous blood was withdrawn to obtain monocyte from the patients. The M1 macrophage was generated by treating the monocyte with GMCSF, LPS and IFN-γ while MCSF, IL-4 and IL-13 were employed to differentiate monocyte into M2 macrophage. After 7 days of polarization, analysis of cell surface differentiation markers (CD86+/CD80+ for M1 and CD206+/CD200R+ for M2) and measurement of MerTK expression were performed using flow cytometry. Results Both M1 and M2 macrophage expressed similar level of CD86, CD80 and CD206 in all groups of CAD patients. MerTK expression in no apparent CAD patients was significantly higher in M2 macrophage compared to M1 macrophage [12.58 ± 4.40 vs. 6.58 ± 1.37, p = 0.040]. Conclusion Differential polarization of macrophage into M1 and M2 was highly dynamic and can be varied due to the microenvironment stimuli in atherosclerotic plaque. Besides, higher expression of MerTK in patients with the least coronary obstructive suggest its vital involvement in efferocytosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00410-2.
Collapse
Affiliation(s)
- Fatin Najiah Mohd Idrus
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nurul Shuhadah Ahmad
- Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, 16100, Pengkalan Chepa, Kelantan, Malaysia
| | - Chee Hock Hoe
- Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, 16100, Pengkalan Chepa, Kelantan, Malaysia
| | - Maryam Azlan
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Farisha Alia Norfuad
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Zurkurnai Yusof
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Wan Yus Haniff Wan Isa
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Akbar Ali Mohamed Ali
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Get Bee Yvonne-Tee
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
20
|
Tang RZ, Li ZZ, Hu D, Kanwal F, Yuan CB, Mustaqeem M, Batool AI, Rehman MFU. Sanjie Yiliu Formula Inhibits Colorectal Cancer Growth by Suppression of Proliferation and Induction of Apoptosis. ACS OMEGA 2021; 6:7761-7770. [PMID: 33778287 PMCID: PMC7992181 DOI: 10.1021/acsomega.0c05565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/24/2021] [Indexed: 05/04/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide. As current therapies toward CRC, including chemotherapy and radiotherapy, pose limitations, such as multidrug resistance (MDR) as well as the intrinsic and potential cytotoxic effects, necessitating to find more effective treatment options with fewer side effects, traditional Chinese medicine (TCM) has an advantage in complementary therapies. In the present study, 3-(4,5-dimethylthiozol-2-yl)-2,5-diphenyltetrazolium bromide (MTT assays), trypan blue staining, colony formation, 4,6-diamidino-2-phenylindole dihydrochloride (DAPI) staining, cell cycle determination, and Annexin V-FITC/PI staining were used to examine the efficacy of Sanjie Yiliu Formula (SJYLF) against CRC proliferation and to investigate its underlying molecular mechanisms through protein expression of various proapoptotic factors by quantitative polymerase chain reaction (q-PCR) and Western blotting. This four-herb-TCM SJYLF can be suggested as one of the decoctions clinically effective in late-stage cancer treatment. Our results suggest that SJYLF robustly decreased the viability of only CRC cell lines (HCT-8, SW-480, HT-29, and DLD-1) and not the normal human kidney cells (HK-2). Moreover, SJYLF significantly suppressed proliferation and induced apoptosis in HCT-8 and downregulated cyclin D1, CDK4, and BCL-2, while Bax expression was upregulated at both mRNA and protein expression levels.
Collapse
Affiliation(s)
- Rong Zhu Tang
- Department
of Gastroenterology, Seventh People’s
Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New
Area, Shanghai 200137, P. R. China
| | - Zhang Zhi Li
- Department
of Hematology, Taihe Hospital Affiliated
to the Hubei University of Medicine, Shiyan, China
| | - Dan Hu
- Department
of Neurology, The Central Hospital of Xiaogan, Xiaogan, Hubei 432100, P. R. China
| | - Fariha Kanwal
- Med-X
Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 201620, P. R.
China
| | - Cheng Bin Yuan
- Department
of Critical Care Medicine, Shanghai General
Hospital, Shanghai 200080, P. R. China
- School
of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P. R. China
| | - Muhammad Mustaqeem
- Department
of Chemistry, University of Sargodha, Sub-Campus Bhakkar, Bhakkar 30000, Pakistan
| | - Aima Iram Batool
- Department
of Zoology, University of Sargodha, Sargodha 40100, Pakistan
| | | |
Collapse
|
21
|
Javadifar A, Rastgoo S, Banach M, Jamialahmadi T, Johnston TP, Sahebkar A. Foam Cells as Therapeutic Targets in Atherosclerosis with a Focus on the Regulatory Roles of Non-Coding RNAs. Int J Mol Sci 2021; 22:2529. [PMID: 33802600 PMCID: PMC7961492 DOI: 10.3390/ijms22052529] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a major cause of human cardiovascular disease, which is the leading cause of mortality around the world. Various physiological and pathological processes are involved, including chronic inflammation, dysregulation of lipid metabolism, development of an environment characterized by oxidative stress and improper immune responses. Accordingly, the expansion of novel targets for the treatment of atherosclerosis is necessary. In this study, we focus on the role of foam cells in the development of atherosclerosis. The specific therapeutic goals associated with each stage in the formation of foam cells and the development of atherosclerosis will be considered. Processing and metabolism of cholesterol in the macrophage is one of the main steps in foam cell formation. Cholesterol processing involves lipid uptake, cholesterol esterification and cholesterol efflux, which ultimately leads to cholesterol equilibrium in the macrophage. Recently, many preclinical studies have appeared concerning the role of non-encoding RNAs in the formation of atherosclerotic lesions. Non-encoding RNAs, especially microRNAs, are considered regulators of lipid metabolism by affecting the expression of genes involved in the uptake (e.g., CD36 and LOX1) esterification (ACAT1) and efflux (ABCA1, ABCG1) of cholesterol. They are also able to regulate inflammatory pathways, produce cytokines and mediate foam cell apoptosis. We have reviewed important preclinical evidence of their therapeutic targeting in atherosclerosis, with a special focus on foam cell formation.
Collapse
Affiliation(s)
- Amin Javadifar
- Department of Allergy and Immunology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; (A.J.); (S.R.)
| | - Sahar Rastgoo
- Department of Allergy and Immunology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; (A.J.); (S.R.)
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, 93338 Lodz, Poland
- Polish Mother’s Memorial Hospital Research Institute (PMMHRI), 93338 Lodz, Poland
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan 9479176135, Iran;
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Thomas P. Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO 64108-2718, USA;
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| |
Collapse
|
22
|
Hu XC, Lu YB, Yang YN, Kang XW, Wang YG, Ma B, Xing S. Progress in clinical trials of cell transplantation for the treatment of spinal cord injury: how many questions remain unanswered? Neural Regen Res 2021; 16:405-413. [PMID: 32985458 PMCID: PMC7996007 DOI: 10.4103/1673-5374.293130] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 03/17/2020] [Accepted: 04/26/2020] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury can lead to severe motor, sensory and autonomic nervous dysfunctions. However, there is currently no effective treatment for spinal cord injury. Neural stem cells and progenitor cells, bone marrow mesenchymal stem cells, olfactory ensheathing cells, umbilical cord blood stem cells, adipose stem cells, hematopoietic stem cells, oligodendrocyte precursor cells, macrophages and Schwann cells have been studied as potential treatments for spinal cord injury. These treatments were mainly performed in animals. However, subtle changes in sensory function, nerve root movement and pain cannot be fully investigated with animal studies. Although these cell types have shown excellent safety and effectiveness in various animal models, sufficient evidence of efficacy for clinical translation is still lacking. Cell transplantation should be combined with tissue engineering scaffolds, local drug delivery systems, postoperative adjuvant therapy and physical rehabilitation training as part of a comprehensive treatment plan to provide the possibility for patients with SCI to return to normal life. This review summarizes and analyzes the clinical trials of cell transplantation therapy in spinal cord injury, with the aim of providing a rational foundation for the development of clinical treatments for spinal cord injury.
Collapse
Affiliation(s)
- Xu-Chang Hu
- Key Laboratory of Bone and Joint Diseases Research of Gansu Province, Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Yu-Bao Lu
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yong-Na Yang
- Department of Neurology, The First People's Hospital of Lanzhou City, Lanzhou, Gansu Province, China
| | - Xue-Wen Kang
- Key Laboratory of Bone and Joint Diseases Research of Gansu Province, Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Yong-Gang Wang
- Key Laboratory of Bone and Joint Diseases Research of Gansu Province, Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Bing Ma
- Key Laboratory of Bone and Joint Diseases Research of Gansu Province, Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Shuai Xing
- Key Laboratory of Bone and Joint Diseases Research of Gansu Province, Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| |
Collapse
|
23
|
Lin S, Li X, Zhang J, Zhang Y. Omentin-1: Protective impact on ischemic stroke via ameliorating atherosclerosis. Clin Chim Acta 2021; 517:31-40. [PMID: 33607071 DOI: 10.1016/j.cca.2021.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/26/2022]
Abstract
Omentin-1, a newly identified adipokine, has recently been revealed as a novel biomarker for ischemic stroke (IS). Low circulating omentin-1 levels could indicate a high risk of IS, and elevated omentin-1 levels exert a favorable impact on cerebral ischemia. Furthermore, omentin-1 has anti-atherosclerotic, anti-inflammatory, and cardiovascular protective capabilities through the intracellular Akt/AMP-activated protein kinase (AMPK)/ nuclear factor-κB (NF-κB) and certain protein kinase (ERK, JNK, and p38) signaling pathways. Omentin-1 also alleviates endothelial cell dysfunction, improves revascularization via the Akt-endothelial nitric-oxide synthase (eNOS) regulatory axis, promotes endothelium-dependent vasodilation through endothelium-derived NO in an eNOS fashion, and inhibits VSMC proliferation by means of AMPK/ERK signaling pathways, VSMC migration via inactivation of the NADPH oxidase (NOX)/ROS/p38/HSP27 pathways and artery calcification via the PI3K-Akt pathway. These findings indicate that omentin-1 may be a negative mediator of IS. Pharmacologically, several lines of clinical evidence indicate that metformin and statins could elevate omentin-1 levels, although the specific mechanism has not been precisely delineated until now. This study is the first to summarize the comprehensive mechanisms between omentin-1 and atherosclerosis and to review the shielding effect of omentin-1 on IS. We shed light on omentin-1 as a novel therapeutic target for combating IS.
Collapse
Affiliation(s)
- Shiyi Lin
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China; School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiabei Zhang
- School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Yuyang Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
24
|
Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther 2021; 12:102. [PMID: 33536064 PMCID: PMC7860030 DOI: 10.1186/s13287-021-02153-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) often leads to serious motor and sensory dysfunction of the limbs below the injured segment. SCI not only results in physical and psychological harm to patients but can also cause a huge economic burden on their families and society. As there is no effective treatment method, the prevention, treatment, and rehabilitation of patients with SCI have become urgent problems to be solved. In recent years, mesenchymal stem cells (MSCs) have attracted more attention in the treatment of SCI. Although MSC therapy can reduce injured volume and promote axonal regeneration, its application is limited by tumorigenicity, a low survival rate, and immune rejection. Accumulating literature shows that exosomes have great potential in the treatment of SCI. In this review, we summarize the existing MSC-derived exosome studies on SCI and discuss the advantages and challenges of treating SCI based on exosomes derived from MSCs.
Collapse
Affiliation(s)
- Wen-Zhao Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Zhan-Jun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Jie-Ru Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Xue-Wen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China.
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China.
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
25
|
Zhang MW, Shen YJ, Shi J, Yu JG. MiR-223-3p in Cardiovascular Diseases: A Biomarker and Potential Therapeutic Target. Front Cardiovasc Med 2021; 7:610561. [PMID: 33553260 PMCID: PMC7854547 DOI: 10.3389/fcvm.2020.610561] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, involving vasculopathy, cardiac dysfunction, or circulatory disturbance, have become the major cause of death globally and brought heavy social burdens. The complexity and diversity of the pathogenic factors add difficulties to diagnosis and treatment, as well as lead to poor prognosis of these diseases. MicroRNAs are short non-coding RNAs to modulate gene expression through directly binding to the 3′-untranslated regions of mRNAs of target genes and thereby to downregulate the protein levels post-transcriptionally. The multiple regulatory effects of microRNAs have been investigated extensively in cardiovascular diseases. MiR-223-3p, expressed in multiple cells such as macrophages, platelets, hepatocytes, and cardiomyocytes to modulate their cellular activities through targeting a variety of genes, is involved in the pathological progression of many cardiovascular diseases. It participates in regulation of several crucial signaling pathways such as phosphatidylinositol 3-kinase/protein kinase B, insulin-like growth factor 1, nuclear factor kappa B, mitogen-activated protein kinase, NOD-like receptor family pyrin domain containing 3 inflammasome, and ribosomal protein S6 kinase B1/hypoxia inducible factor 1 α pathways to affect cell proliferation, migration, apoptosis, hypertrophy, and polarization, as well as electrophysiology, resulting in dysfunction of cardiovascular system. Here, in this review, we will discuss the role of miR-223-3p in cardiovascular diseases, involving its verified targets, influenced signaling pathways, and regulation of cell function. In addition, the potential of miR-223-3p as therapeutic target and biomarker for diagnosis and prediction of cardiovascular diseases will be further discussed, providing clues for clinicians.
Collapse
Affiliation(s)
- Meng-Wan Zhang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yun-Jie Shen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Shi
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Guang Yu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
26
|
Pireaux V, Delporte C, Rousseau A, Desmet JM, Van Antwerpen P, Raes M, Zouaoui Boudjeltia K. M2 Monocyte Polarization in Dialyzed Patients Is Associated with Increased Levels of M-CSF and Myeloperoxidase-Associated Oxidative Stress: Preliminary Results. Biomedicines 2021; 9:biomedicines9010084. [PMID: 33467199 PMCID: PMC7830480 DOI: 10.3390/biomedicines9010084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular diseases represent a major issue in terms of morbidity and mortality for dialysis patients. This morbidity is due to the accelerated atherosclerosis observed in these patients. Atherosclerosis is a chronic inflammatory disease characterized by key players such as monocytes, macrophages, or oxidized LDLs. Monocytes-macrophages are classified into subsets of polarized cells, with M1 and M2 macrophages considered, respectively, as pro- and anti-inflammatory. (1) Methods: The monocyte subsets and phenotypes were analyzed by flow cytometry. These data were completed by the quantification of plasma M-CSF, IL-8, CRP, Mox-LDLs, Apo-B, Apo-AI, chloro-tyrosine, and homocitrulline concentrations. The statistical differences and associations between two continuous variables were assessed using the Mann-Whitney U test and Spearman's correlation coefficient, respectively. (2) Results: Hemodialyzed patients showed a significant increase in their concentrations of CRP, M-CSF, and IL-8 (inflammation biomarkers), as well as chloro-tyrosine and homocitrulline (myeloperoxidase-associated oxidative stress biomarkers). Moreover, we observed a higher percentage of M2 monocytes in the plasma of hemodialysis patients as compared to the controls. (3) Conclusions: Our data suggest that oxidative stress and an inflammatory environment, which is amplified in hemodialysis patients, seems to favor an increase in the concentration of circulating M-CSF, therefore leading to an increase in M2 polarization among circulating monocytes.
Collapse
Affiliation(s)
- Valérie Pireaux
- URBC-Narilis, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium; (V.P.); (M.R.)
| | - Cédric Delporte
- Laboratory of Pharmaceutical Chemistry and Analytical Platform, Faculty of Pharmacy, Université libre de Bruxelles (Campus de la Plaine) CP205/05, Boulevard du Triomphe, 1050 Brussels, Belgium; (C.D.); (P.V.A.)
| | - Alexandre Rousseau
- Laboratory of Experimental Medicine (ULB 222 Unit), CHU-Charleroi, ISPPC Hôpital Vésale, Université libre de Bruxelles, 6110 Montigny-Le-Tilleul, Belgium;
| | - Jean-Marc Desmet
- Nephrology-Hemodialysis Unit, CHU-Charleroi, ISPPC Hôpital Vésale, 6110 Montigny-Le-Tilleul, Belgium;
| | - Pierre Van Antwerpen
- Laboratory of Pharmaceutical Chemistry and Analytical Platform, Faculty of Pharmacy, Université libre de Bruxelles (Campus de la Plaine) CP205/05, Boulevard du Triomphe, 1050 Brussels, Belgium; (C.D.); (P.V.A.)
| | - Martine Raes
- URBC-Narilis, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium; (V.P.); (M.R.)
| | - Karim Zouaoui Boudjeltia
- Laboratory of Experimental Medicine (ULB 222 Unit), CHU-Charleroi, ISPPC Hôpital Vésale, Université libre de Bruxelles, 6110 Montigny-Le-Tilleul, Belgium;
- Correspondence: ; Tel.: +32-71-92-47-05; Fax: +32-71-92-47-10
| |
Collapse
|
27
|
Kulshrestha R, Dhanda H, Pandey A, Singh A, Kumar R. Immunopathogenesis and therapeutic potential of macrophage influx in diffuse parenchymal lung diseases. Expert Rev Respir Med 2020; 14:917-928. [PMID: 32600077 DOI: 10.1080/17476348.2020.1776117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION The diffuse parenchymal lung diseases (DPLD)/interstitial lung diseases (ILD) are progressive lung disorders with usually unclear etiology, poor long-term survival and no effective treatment. Their pathogenesis is characterized by alveolar epithelial cell injury, inflammation, epithelial-mesenchymal transition, and parenchymal fibrosis. Macrophages play diverse roles in their development, both in the acute phase and in tissue repair. AREAS COVERED In this review, we summarize the current state of knowledge regarding the role of macrophages and their phenotypes in the immunopathogenesis of DPLDs; CVD-ILD, UIP, NSIP, DIP, RB-ILD, AIP, HP, Sarcoidosis, etc. Our goal is to update the understanding of the immune mechanisms underlying the initiation and progression of fibrosis in DPLDs. This will help in identification of biomarkers and in developing novel therapeutic strategies for DPLDs. A thorough literature search of the published studies in PubMed (from 1975 to 2020) was done. EXPERT OPINION The macrophage associated inflammatory markers needs to be explored for their potential as biomarkers of disease activity and progression. Pharmacological targeting of macrophage activation may reduce the risk of macrophage activation syndrome (MAS) and help improving the survival and prognosis of these patients.
Collapse
Affiliation(s)
| | - Himanshu Dhanda
- Department of Pathology, V.P.Chest Institute , New Delhi, India
| | - Apoorva Pandey
- Department of Pathology, V.P.Chest Institute , New Delhi, India
| | - Amit Singh
- Department of Pathology, V.P.Chest Institute , New Delhi, India
| | - Raj Kumar
- Department of Pulmonary Medicine, V.P.Chest Institute , New Delhi, India
| |
Collapse
|
28
|
Kuznetsova T, Prange KHM, Glass CK, de Winther MPJ. Transcriptional and epigenetic regulation of macrophages in atherosclerosis. Nat Rev Cardiol 2020; 17:216-228. [PMID: 31578516 PMCID: PMC7770754 DOI: 10.1038/s41569-019-0265-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2019] [Indexed: 12/11/2022]
Abstract
Monocytes and macrophages provide defence against pathogens and danger signals. These cells respond to stimulation in a fast and stimulus-specific manner by utilizing complex cascaded activation by lineage-determining and signal-dependent transcription factors. The complexity of the functional response is determined by interactions between triggered transcription factors and depends on the microenvironment and interdependent signalling cascades. Dysregulation of macrophage phenotypes is a major driver of various diseases such as atherosclerosis, rheumatoid arthritis and type 2 diabetes mellitus. Furthermore, exposure of monocytes, which are macrophage precursor cells, to certain stimuli can lead to a hypo-inflammatory tolerized phenotype or a hyper-inflammatory trained phenotype in a macrophage. In atherosclerosis, macrophages and monocytes are exposed to inflammatory cytokines, oxidized lipids, cholesterol crystals and other factors. All these stimuli induce not only a specific transcriptional response but also interact extensively, leading to transcriptional and epigenetic heterogeneity of macrophages in atherosclerotic plaques. Targeting the epigenetic landscape of plaque macrophages can be a powerful therapeutic tool to modulate pro-atherogenic phenotypes and reduce the rate of plaque formation. In this Review, we discuss the emerging role of transcription factors and epigenetic remodelling in macrophages in the context of atherosclerosis and inflammation, and provide a comprehensive overview of epigenetic enzymes and transcription factors that are involved in macrophage activation.
Collapse
Affiliation(s)
- Tatyana Kuznetsova
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers - location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Koen H M Prange
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers - location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers - location AMC, University of Amsterdam, Amsterdam, Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany.
| |
Collapse
|
29
|
Jiang L, Wang J, Jiang J, Zhang C, Zhao M, Chen Z, Wang N, Hu D, Liu X, Peng H, Lian M. Sonodynamic therapy in atherosclerosis by curcumin nanosuspensions: Preparation design, efficacy evaluation, and mechanisms analysis. Eur J Pharm Biopharm 2019; 146:101-110. [PMID: 31841689 DOI: 10.1016/j.ejpb.2019.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/11/2019] [Accepted: 12/08/2019] [Indexed: 12/14/2022]
Abstract
Previous studies have shown that curcumin (Cur) induced by ultrasound has protective effects on atherosclerosis even if low bioavailability of the Cur. The enhancement of bioavailability of the Cur further improved the curative effect of sonodynamic therapy (SDT) on atherosclerosis through nanotechnology. Nanosuspensions as a good drug delivery system had obvious advantages in increasing the solubility and improving the effectiveness of insoluble drugs. The aim of this study was to develop curcumin nanosuspensions (Cur-ns) which used polyvinylpyrrolidone (PVPK30) and sodium dodecyl sulfate (SDS) as stabilizers to improve poor water solubility and bioavailability of the Cur. And then the therapeutic effects of Cur-ns-SDT on atherosclerotic plaques and its possible mechanisms would be investigated and elucidated. Cur-ns with a small particle size has been successfully prepared and the data have confirmed that Cur-ns could be more easily engulfed into RAW264.7 cells than free Cur and accumulated more under the stimulation of the ultrasound. Reactive oxygen species (ROS) inside RAW264.7 cells after SDT led to the decrease of mitochondrial membrane potential (MMP) and the higher expression of cleaved caspase-9/3. The results of in vivo experiments showed that Cur-ns-SDT reduced the level of total cholesterol (TC) and low density lipoprotein (LDL) and promoted the transformation from M1 to M2 macrophages, relieved atherosclerosis syndrome. Therefore, Cur-ns-SDT was a potential treatment of anti-atherosclerosis by enhancing macrophages apoptosis through mitochondrial pathway and inhibiting the progression of plaques by interfering with macrophages polarization.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Jiahe Wang
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Jiaqi Jiang
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Changmei Zhang
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Man Zhao
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Zhong Chen
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Na Wang
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Dandan Hu
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Xiaoying Liu
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China.
| | - Mingming Lian
- Department of Pharmaceutics, Daqing Campus, Harbin Medical University, Key Laboratory of Research and Development of Natural Products at Harbin Medical University, Xin Yang Road, Daqing 163319, China
| |
Collapse
|
30
|
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]
|
31
|
Abstract
Three decades of research in hematopoietic stem cell transplantation and HIV/AIDS fields have shaped a picture of immune restoration disorders. This manuscript overviews the molecular biology of interferon networks, the molecular pathogenesis of immune reconstitution inflammatory syndrome, and post-hematopoietic stem cell transplantation immune restoration disorders (IRD). It also summarizes the effects of thymic involution on T cell diversity, and the results of the assessment of diagnostic biomarkers of IRD, and tested targeted immunomodulatory treatments.
Collapse
Affiliation(s)
- Hesham Mohei
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Usha Kellampalli
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
32
|
Biological profile of monocyte-derived macrophages in coronary heart disease patients: implications for plaque morphology. Sci Rep 2019; 9:8680. [PMID: 31213640 PMCID: PMC6581961 DOI: 10.1038/s41598-019-44847-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
The prevalence of a macrophage phenotype in atherosclerotic plaque may drive its progression and/or instability. Macrophages from coronary plaques are not available, and monocyte-derived macrophages (MDMs) are usually considered as a surrogate. We compared the MDM profile obtained from coronary artery disease (CAD) patients and healthy subjects, and we evaluated the association between CAD MDM profile and in vivo coronary plaque characteristics assessed by optical coherence tomography (OCT). At morphological analysis, MDMs of CAD patients had a higher prevalence of round than spindle cells, whereas in healthy subjects the prevalence of the two morphotypes was similar. Compared to healthy subjects, MDMs of CAD patients had reduced efferocytosis, lower transglutaminase-2, CD206 and CD163 receptor levels, and higher tissue factor (TF) levels. At OCT, patients with a higher prevalence of round MDMs showed more frequently a lipid-rich plaque, a thin-cap fibroatheroma, a greater intra-plaque macrophage accumulation, and a ruptured plaque. The MDM efferocytosis correlated with minimal lumen area, and TF levels in MDMs correlated with the presence of ruptured plaque. MDMs obtained from CAD patients are characterized by a morpho-phenotypic heterogeneity with a prevalence of round cells, showing pro-inflammatory and pro-thrombotic properties. The MDM profile allows identifying CAD patients at high risk.
Collapse
|
33
|
Bruen R, Fitzsimons S, Belton O. miR-155 in the Resolution of Atherosclerosis. Front Pharmacol 2019; 10:463. [PMID: 31139076 PMCID: PMC6527595 DOI: 10.3389/fphar.2019.00463] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory disease where advanced lesions can eventually completely obstruct blood flow resulting in clinical events, such as a myocardial infarction or stroke. Monocytes and macrophages are the dominant biologically active immune cells involved in atherosclerosis disease and play a pivotal role during initiation, progression, and regression of disease. Altering macrophage inflammation is critical to induce regression of atherosclerosis and microRNAs (miRs) have emerged as key regulators of the macrophage phenotype. MiRs are small noncoding RNAs that regulate gene expression. They are dysregulated during atherosclerosis development and are key regulators of macrophage function and polarization. MiRs are short nucleotide transcripts that are very stable in circulation and thus have potential as therapeutics and/or biomarkers in the context of atherosclerosis. Of relevance to this review is that inhibition of macrophage-specific miR-155 may be a viable therapeutic strategy to decrease inflammation associated with atherosclerosis. However, further studies on these miRs and advancements in miR therapeutic delivery are required for these therapeutics to advance to the clinical setting. Conjugated linoleic acid (CLA), a pro-resolving lipid mediator, is an agonist of the peroxisome proliferator-activated receptor (PPAR)-γ. The biological activities of CLA have been documented to have anti-atherogenic effects in experimental models of atherosclerosis, inducing regression and impacting on monocyte and macrophage cells. Our work and that of others on PPAR-γ agonists and polyunsaturated fatty acids have shown that these mediators regulate candidate miRNAs and promote pro-resolving atherosclerotic plaque microenvironments.
Collapse
Affiliation(s)
- Robyn Bruen
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Orina Belton
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Curtale G, Rubino M, Locati M. MicroRNAs as Molecular Switches in Macrophage Activation. Front Immunol 2019; 10:799. [PMID: 31057539 PMCID: PMC6478758 DOI: 10.3389/fimmu.2019.00799] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/26/2019] [Indexed: 12/25/2022] Open
Abstract
The efficacy of macrophage- mediated inflammatory response relies on the coordinated expression of key factors, which expression is finely regulated at both transcriptional and post-transcriptional level. Several studies have provided compelling evidence that microRNAs play pivotal roles in modulating macrophage activation, polarization, tissue infiltration, and resolution of inflammation. In this review, we highlight the essential molecular mechanisms underlying the different phases of inflammation that are targeted by microRNAs to inhibit or accelerate restoration to tissue integrity and homeostasis. We further review the impact of microRNA-dependent regulation of tumor-associated macrophages and the relative implication for tumor biology.
Collapse
Affiliation(s)
- Graziella Curtale
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Marcello Rubino
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| |
Collapse
|
35
|
Mehrpouri M, Bashash D, Mohammadi MH, Gheydari ME, Satlsar ES, Hamidpour M. Co-culture of Platelets with Monocytes Induced M2 Macrophage Polarization and Formation of Foam Cells: Shedding Light on the Crucial Role of Platelets in Monocyte Differentiation. Turk J Haematol 2019; 36:97-105. [PMID: 30859801 PMCID: PMC6516100 DOI: 10.4274/tjh.galenos.2019.2018.0449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective Far beyond hemostasis and thrombosis, significant evidence has indicated the critical role of platelets in atherosclerosis. SDF-1 is among the pro-inflammatory chemokines that are increased in platelets of patients with coronary artery disease (CAD). The goal of the current work is to identify the in vitro effect of platelets from either CAD patients or healthy volunteers on the induction of macrophages and foam cells. Materials and Methods The expression of SDF-1 on platelet surfaces in CAD patients and healthy volunteers was investigated using flow cytometry. We also evaluated the CXCR4/CXCR7 expression on monocytes from buffy coats of healthy volunteers. The effect of platelets from CAD patients and healthy volunteers on differentiation of monocytes and foam cell formation was evaluated using Oil Red O (ORO) staining. Flow cytometry and real-time PCR were also employed to evaluate surface markers and mRNA expression of genes involved in this process after co-culture of platelets with monocytes. Results Monocytes in co-culture with platelets acquired a spindleshape appearance and ORO-positive lipid droplets. In addition, platelets could induce CD163 expression, as an important marker of M2 macrophage, and upregulate the mRNA expression of the SRB, CD36, ACAT, LXR-α, and ABCA1 genes in monocytes. Notably, platelets of CAD patients with higher expression of SDF-1, increased the expression of genes encoding SRB and CD36 as compared to platelets of healthy volunteers. Conclusion Our results indicate that platelets from CAD patients could provoke monocyte differentiation into macrophages with an M2 phenotype, which in turn may participate in an atheroprotective process.
Collapse
Affiliation(s)
- Mahdieh Mehrpouri
- Shahid Beheshti University of Medical Sciences, Faculty of Allied Medical Sciences, Department of Hematology and Blood Banking, Tehran, Iran
| | - Davood Bashash
- Shahid Beheshti University of Medical Sciences, Faculty of Allied Medical Sciences, Department of Hematology and Blood Banking, Tehran, Iran
| | - Mohammad Hossien Mohammadi
- HSCT Research Centre, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Esmail Gheydari
- Shahid Beheshti University of Medical Sciences Faculty of Medicine, Taleghani General Hospital, Department of Cardiology, Tehran, Iran
| | - Esmail Shahabi Satlsar
- Shahid Beheshti University of Medical Sciences, Faculty of Allied Medical Sciences, Department of Hematology and Blood Banking, Tehran, Iran
| | - Mohsen Hamidpour
- HSCT Research Centre, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
M2 Macrophages as a Potential Target for Antiatherosclerosis Treatment. Neural Plast 2019; 2019:6724903. [PMID: 30923552 PMCID: PMC6409015 DOI: 10.1155/2019/6724903] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammation course, which could induce life-threatening diseases such as stroke and myocardial infarction. Optimal medical treatments for atherosclerotic risk factors with current antihypertensive and lipid-lowering drugs (for example, statins) are widely used in clinical practice. However, many patients with established disease still continue to have recurrent cardiovascular events in spite of treatment with a state-of-the-art therapy. Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality worldwide. Hence, current treatment of atherosclerosis is still far from being satisfactory. Recently, M2 macrophages have been found associated with atherosclerosis regression. The M2 phenotype can secrete anti-inflammatory factors such as IL-10 and TGF-β, promote tissue remodeling and repairing through collagen formation, and clear dying cells and debris by efferocytosis. Therefore, modulators targeting macrophages' polarization to the M2 phenotype could be another promising treatment strategy for atherosclerosis. Two main signaling pathways, the Akt/mTORC/LXR pathway and the JAK/STAT6 pathway, are found playing important roles in M2 polarization. In addition, researchers have reported several potential approaches to modulate M2 polarization. Inhibiting or activating some kinds of enzymes, affecting transcription factors, or acting on several membrane receptors could regulate the polarization of the M2 phenotype. Besides, biomolecules, for example vitamin D, were found to affect the process of M2 polarization. Pomegranate juice could promote M2 polarization via unclear mechanism. In this review, we will discuss how M2 macrophages affect atherosclerosis regression, signal transduction in M2 polarization, and outline potential targets and compounds that affect M2 polarization, thus controlling the progress of atherosclerosis.
Collapse
|
37
|
Aberrant DNA methylation of M1-macrophage genes in coronary artery disease. Sci Rep 2019; 9:1429. [PMID: 30723273 PMCID: PMC6363807 DOI: 10.1038/s41598-018-38040-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/19/2018] [Indexed: 01/22/2023] Open
Abstract
M1 and M2 macrophage balance in atherosclerosis has attracted much interest. Though, it remains unknown how macrophage heterogeneity is regulated. Moreover, the regulation of macrophage polarization and activation also involve DNA methylation. However, it remains ambiguous which genes are under direct regulation by DNA methylation. Our aim was to evaluate the gene-specific promoter DNA methylation status of M1/M2 polarization markers in PBMCs of CAD patients. A case-control study was performed with 25 CAD patients and 25 controls to study the promoter DNA methylation status of STAT1, STAT6, MHC2, IL12b, iNOS, JAK1, JAK2 and SOCS5 using MS-HRM analysis. Our data indicates that there was a clear-cut difference in the pattern of gene-specific promoter DNA methylation of CAD patients in comparison to controls. A significant difference was observed between the percentage methylation of STAT1, IL12b, MHC2, iNOS, JAK1 and JAK2 in CAD patients and control subjects. In conclusion, our data show that MS-HRM assay is a rapid and inexpensive method for qualitatively identifying aberrant gene-specific promoter DNA methylation changes in CAD. Furthermore, we propose that gene-specific promoter DNA methylation based on monocyte/macrophage might aid as diagnostic marker for clinical application or DNA methylation-related drug interventions may offer novel possibilities for atherosclerotic disease management.
Collapse
|
38
|
Momtazi-Borojeni AA, Abdollahi E, Nikfar B, Chaichian S, Ekhlasi-Hundrieser M. Curcumin as a potential modulator of M1 and M2 macrophages: new insights in atherosclerosis therapy. Heart Fail Rev 2019; 24:399-409. [DOI: 10.1007/s10741-018-09764-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
39
|
Splenectomy Promotes Macrophage Polarization in a Mouse Model of Concanavalin A- (ConA-) Induced Liver Fibrosis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5756189. [PMID: 30723740 PMCID: PMC6339718 DOI: 10.1155/2019/5756189] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/24/2018] [Indexed: 12/11/2022]
Abstract
Background Splenectomy can improve liver function and survival in patients with autoimmune hepatitis (AIH) and liver cirrhosis. We investigated the underlying mechanism in a mouse model of concanavalin A- (ConA-) induced liver fibrosis. Methods We used ConA to induce immune liver fibrosis in BALB/c mice. Splenectomy was performed alone or with the administration of dexamethasone (DEX). Changes in blood and liver tissues were evaluated. Results Mice treated with ConA for 7 weeks developed advanced liver fibrosis, while splenectomy suppressed liver fibrosis. Although the populations of macrophages/monocytes and M1 macrophages decreased after splenectomy, the inflammatory factors associated with M2 macrophages increased after splenectomy. Furthermore, the population of circulating CD11b+Ly6Chigh myeloid-derived suppressor cells (MDSCs) increased after splenectomy. After ConA treatment, elevated levels of activated and total NF-kBp65/p50 combined with DNA were observed in hepatic tissues. In contrast, the levels of NF-κB p65/p50 decreased after splenectomy. Conclusions Splenectomy may promote the polarization of CD11b+Ly6Chigh MDSCs and the differentiation of M2 macrophages while restricting the level of NF-κB p65-p50 heterodimers. These factors may suppress the progression of liver fibrosis.
Collapse
|
40
|
Shoulders H, Garner KH, Singla DK. Macrophage depletion by clodronate attenuates bone morphogenetic protein-7 induced M2 macrophage differentiation and improved systolic blood velocity in atherosclerosis. Transl Res 2019; 203:1-14. [PMID: 30107156 PMCID: PMC6314201 DOI: 10.1016/j.trsl.2018.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022]
Abstract
Bone morphogenetic protein-7 (BMP-7) affects the presence of macrophage subtypes in vitro and in vivo at an early stage of atherosclerosis (ATH); however, it remains unknown whether BMP-7 treatment affects the development and progression of ATH at a mid-stage of the disease. We therefore performed a Day 28 (D28) study to examine BMP-7's potential to affect monocyte differentiation. Atherosclerosis was developed in ApoE KO mice, and these animals were treated with intravenous injections of BMP-7 and/or liposomal clodronate (LC). BMP-7 significantly (P < 0.05) lowers plaque formation following induction of atherosclerosis. However, upon macrophage depletion, BMP-7 fails to significantly alter plaque progression suggesting a direct role of BMP-7 on macrophages. Immunohistochemical staining of carotid arteries was performed to determine BMP-7's effect on pro-inflammatory M1 inducible nitric oxide synthase and anti-inflammatory M2 (cluster of differentiation [CD]206, Arginase-1) macrophages, and monocytes ( CD14). BMP-7 significantly reduced pro-inflammatory M1 macrophages and increased anti-inflammatory M2 macrophages at D28, while BMP-7 showed no effect on M2 macrophage differentiation in animals treated with LC. Enzyme-linked immunosorbent assay data showed significant reduction in proinflammatory cytokines (Interleukin-6 [IL-6]), monocyte chemoattractant protein-1, and tumor necrosis factor-α) and a significant increase in anti-inflammatory cytokine (IL-10) in BMP-7 treated mice (P < 0.05).Western blot analysis of arterial tissue confirms a significant increase in pro-survival kinases extracellular-signal regulated kinase and SMAD and a reduction in pro-inflammatory kinases p38 and c-Jun N-terminal kinase in BMP-7 treated mice (P < 0.05). Overall, this study suggests that clodronate treatment inhibits BMP-7 induced differentiation of monocytes into M2 macrophages and improved systolic blood velocity.
Collapse
Affiliation(s)
- Heidi Shoulders
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Kaley H Garner
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida.
| |
Collapse
|
41
|
de Gaetano M, McEvoy C, Andrews D, Cacace A, Hunter J, Brennan E, Godson C. Specialized Pro-resolving Lipid Mediators: Modulation of Diabetes-Associated Cardio-, Reno-, and Retino-Vascular Complications. Front Pharmacol 2018; 9:1488. [PMID: 30618774 PMCID: PMC6305798 DOI: 10.3389/fphar.2018.01488] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022] Open
Abstract
Diabetes and its associated chronic complications present a healthcare challenge on a global scale. Despite improvements in the management of chronic complications of the micro-/macro-vasculature, their growing prevalence and incidence highlights the scale of the problem. It is currently estimated that diabetes affects 425 million people globally and it is anticipated that this figure will rise by 2025 to 700 million people. The vascular complications of diabetes including diabetes-associated atherosclerosis and kidney disease present a particular challenge. Diabetes is the leading cause of end stage renal disease, reflecting fibrosis leading to organ failure. Moreover, diabetes associated states of inflammation, neo-vascularization, apoptosis and hypercoagulability contribute to also exacerbate atherosclerosis, from the metabolic syndrome to advanced disease, plaque rupture and coronary thrombosis. Current therapeutic interventions focus on regulating blood glucose, glomerular and peripheral hypertension and can at best slow the progression of diabetes complications. Recently advanced knowledge of the pathogenesis underlying diabetes and associated complications revealed common mechanisms, including the inflammatory response, insulin resistance and hyperglycemia. The major role that inflammation plays in many chronic diseases has led to the development of new strategies aiming to promote the restoration of homeostasis through the "resolution of inflammation." These strategies aim to mimic the spontaneous activities of the 'specialized pro-resolving mediators' (SPMs), including endogenous molecules and their synthetic mimetics. This review aims to discuss the effect of SPMs [with particular attention to lipoxins (LXs) and resolvins (Rvs)] on inflammatory responses in a series of experimental models, as well as evidence from human studies, in the context of cardio- and reno-vascular diabetic complications, with a brief mention to diabetic retinopathy (DR). These data collectively support the hypothesis that endogenously generated SPMs or synthetic mimetics of their activities may represent lead molecules in a new discipline, namely the 'resolution pharmacology,' offering hope for new therapeutic strategies to prevent and treat, specifically, diabetes-associated atherosclerosis, nephropathy and retinopathy.
Collapse
Affiliation(s)
- Monica de Gaetano
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Caitriona McEvoy
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
- Renal Transplant Program, University Health Network, Toronto, ON, Canada
| | - Darrell Andrews
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Antonino Cacace
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Jonathan Hunter
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
42
|
Tomiotto-Pellissier F, Bortoleti BTDS, Assolini JP, Gonçalves MD, Carloto ACM, Miranda-Sapla MM, Conchon-Costa I, Bordignon J, Pavanelli WR. Macrophage Polarization in Leishmaniasis: Broadening Horizons. Front Immunol 2018; 9:2529. [PMID: 30429856 PMCID: PMC6220043 DOI: 10.3389/fimmu.2018.02529] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/15/2018] [Indexed: 01/14/2023] Open
Abstract
Leishmaniasis is a vector-borne neglected tropical disease that affects more than 700,000 people annually. Leishmania parasites cause the disease, and different species trigger a distinct immune response and clinical manifestations. Macrophages are the final host cells for the proliferation of Leishmania parasites, and these cells are the key to a controlled or exacerbated response that culminates in clinical manifestations. M1 and M2 are the two main macrophage phenotypes. M1 is a pro-inflammatory subtype with microbicidal properties, and M2, or alternatively activated, is an anti-inflammatory/regulatory subtype that is related to inflammation resolution and tissue repair. The present review elucidates the roles of M1 and M2 polarization in leishmaniasis and highlights the role of the salivary components of the vector and the action of the parasite in the macrophage plasticity.
Collapse
Affiliation(s)
- Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - João Paulo Assolini
- Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Manoela Daiele Gonçalves
- Laboratory of Biotransformation and Phytochemistry, Department of Chemistry, State University of Londrina, Universitary Hospital, Londrina, Brazil
| | | | | | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Juliano Bordignon
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Molecular Virology, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil
| | - Wander Rogério Pavanelli
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
43
|
TIPE1 suppresses osteosarcoma tumor growth by regulating macrophage infiltration. Clin Transl Oncol 2018; 21:334-341. [PMID: 30062520 DOI: 10.1007/s12094-018-1927-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Osteosarcoma is the most common primary malignancy of the bone, and macrophages play a promotional role during osteosarcoma development and progression. TIPE1 is known to function as a tumor suppressor in diverse cancers by inducing cell arrest and apoptosis. However, the biological function of TIPE1 in osteosarcoma is still unclear. PURPOSE The purpose of this study was to investigate the expression and function of TIPE1 in osteosarcoma. METHODS In the present study, TIPE1 expression in osteosarcoma cancer cells was determined by qPCR and western blotting. A subcutaneous tumor model was established to investigate the potential anti-tumor activity of TIPE1 in osteosarcoma. Further, flow cytometry, western blotting, immunofluorescence staining, and ELISA were performed to clarify the underlying mechanism by which TIPE1 regulates growth of osteosarcoma. RESULTS Our results suggest that TIPE1 is downregulated in osteosarcoma cancer cells, and ectopic expression TIPE1 significantly inhibited osteosarcoma tumor growth in vivo. Furthermore, TIPE1 inhibits the infiltration of macrophages in osteosarcoma tumor by suppressing MCP-1 expression in osteosarcoma cells. Further in vivo study revealed that inhibition of MCP-1/CCR2 axis by Bindarit blocked the inhibitory effect of TIPE1 on osteosarcoma growth. CONCLUSION Collectively, our results demonstrate the anti-tumor role of TIPE1 in osteosarcoma and reveal a novel therapy target for osteosarcoma.
Collapse
|
44
|
Huang YY, Zhang Q, Zhang JN, Zhang YN, Gu L, Yang HM, Xia N, Wang XM, Zhang H. Triptolide up-regulates metabotropic glutamate receptor 5 to inhibit microglia activation in the lipopolysaccharide-induced model of Parkinson's disease. Brain Behav Immun 2018; 71:93-107. [PMID: 29649522 DOI: 10.1016/j.bbi.2018.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/27/2018] [Accepted: 04/07/2018] [Indexed: 12/11/2022] Open
Abstract
Metabotropic glutamate receptor (mGlu)5 regulates microglia activation, which contributes to inflammation. However, the role of mGlu5 in neuroinflammation associated with Parkinson's disease (PD) remains unclear. Triptolide (T10) exerts potent immunosuppressive and anti-inflammatory effects and protects neurons by inhibiting microglia activation. In this study, we investigated the role of mGlu5 in the anti-inflammatory effect of T10 in a lipopolysaccharide (LPS)-induced PD model. In cultured BV2 cells and primary microglia, blocking mGlu5 activity or knocking down its expression abolished T10-inhibited release of proinflammatory cytokines induced by LPS. Moreover, T10 up-regulated mGlu5 expression decreased by LPS through enhancing mRNA expression and protein stability. T10 also reversed the reduction in mGlu5 membrane localization and modulated receptor-mediated mitogen-activated protein kinase activity induced by LPS. Pharmacological inhibition of signaling molecules increased nitric oxide level and inducible nitric oxide synthase (iNOS), tumor necrosis factor-α, and interleukin (IL)-1β and -6 transcript levels that were downregulated by treatment with T10. Consistent with these in vitro findings, blocking mGlu5 attenuated the anti-inflammatory effects of T10 in an LPS-induced PD model and blocked the decreases in the number and morphology of ionized calcium binding adaptor molecule 1-positive microglia and LPS-induced iNOS protein expression caused by T10 treatment. Besides, mGlu5 mediated the effect of T10 on microglia-induced astrocyte activation in vitro and in vivo. The findings provide evidence for a novel mechanism by which mGlu5 regulates T10-inhibited microglia activation via modulating protein expression of the receptor and its intracellular signaling. The study might contribute to the biological effects of Chinese herbs as an approach for protecting against neurotoxicity in PD.
Collapse
Affiliation(s)
- Yi-Ying Huang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Qian Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Ya-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Ning Xia
- Burke Medical Research Institute, Weill Cornell Medicine, White Plains, NY, 10605, USA
| | - Xiao-Min Wang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
45
|
Pseudolaric acid B attenuates atherosclerosis progression and inflammation by suppressing PPARγ-mediated NF-κB activation. Int Immunopharmacol 2018; 59:76-85. [DOI: 10.1016/j.intimp.2018.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/20/2018] [Accepted: 03/31/2018] [Indexed: 12/12/2022]
|
46
|
Guo M, Xiao J, Sheng X, Zhang X, Tie Y, Wang L, Zhao L, Ji X. Ginsenoside Rg3 Mitigates Atherosclerosis Progression in Diabetic apoE-/- Mice by Skewing Macrophages to the M2 Phenotype. Front Pharmacol 2018; 9:464. [PMID: 29867472 PMCID: PMC5954105 DOI: 10.3389/fphar.2018.00464] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/20/2018] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis (AS) in diabetic patients is often associated with low stability, which might be largely attributed to unfavorable macrophage polarization and increased inflammatory response induced by hyperglycaemia. Ginsenoside Rg3 is one of the main active principles of Panax Ginseng, which has been reported to be a natural ligand of peroxisome proliferator-activated receptor-gamma (PPARγ), a key nuclear transcriptional factor involved in inflammation and macrophage differentiation. However, it remains unclear if Rg3 could exert protective effects on plaque stability in diabetes. In this study, we investigated the role of ginsenoside 20(S)-Rg3 in macrophage polarization and AS plaque stability using advanced glycation end products-treated macrophages and diabetic AS mice models. In vitro, advanced glycation end products (AGEs) treatment promoted the expression of proinflammatory molecules and M1 surface markers, whereas 20(S)-Rg3 could reverse the M1 polarization to the M2 phenotype. In vivo, the administration of 20(S)-Rg3 promoted AS lesion stability and reduced the plaque burden, accompanied by increased M2 macrophages and reduced M1 macrophages. In addition, PPARγ antagonist GW9662 co-administration mostly blocked these effects, suggesting the important role of PPARγ pathways in mediating 20(S)-Rg3 effects in macrophage polarization and atherosclerosis progression. Together, these results demonstrated an immunomodulatory role of ginsenoside 20(S)-Rg3 in promoting macrophages to a profile of the M2 type through PPARγ-dependent mechanisms, and indicated a potential role of 20(S)-Rg3 in the prevention and treatment of diabetic atherosclerosis.
Collapse
Affiliation(s)
- Mengqi Guo
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Xiao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xi Sheng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinyu Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanyuan Tie
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lang Zhao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoping Ji
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
47
|
Nasr SH, Tonson A, El-Dakdouki MH, Zhu DC, Agnew D, Wiseman R, Qian C, Huang X. Effects of Nanoprobe Morphology on Cellular Binding and Inflammatory Responses: Hyaluronan-Conjugated Magnetic Nanoworms for Magnetic Resonance Imaging of Atherosclerotic Plaques. ACS APPLIED MATERIALS & INTERFACES 2018; 10:11495-11507. [PMID: 29558108 PMCID: PMC5995107 DOI: 10.1021/acsami.7b19708] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Atherosclerosis is an inflammatory disease of arterial walls and the rupturing of atherosclerotic plaques is a major cause of heart attack and stroke. Imaging techniques that can enable the detection of atherosclerotic plaques before clinical manifestation are urgently needed. Magnetic resonance imaging (MRI) is a powerful technique to image the morphology of atherosclerotic plaques. In order to better analyze molecular processes in plaques, contrast agents that can selectively bind to plaque receptors will prove invaluable. CD44 is a cell surface protein overexpressed in plaque tissues, the level of which can be correlated with the risks of plaque rupture. Thus, targeting CD44 is an attractive strategy for detection of atherosclerotic plaques. Herein, we report the synthesis of hyaluronan-conjugated iron oxide nanoworms (HA-NWs). A new purification and gel electrophoresis protocol was developed to ensure the complete removal of free HA from HA-NWs. Compared to the more traditional spherical HA-bearing nanoparticles, HA-NWs had an elongated shape, which interacted much stronger with CD44-expressing cells in CD44- and HA-dependent manners. Furthermore, the HA-NWs did not induce much inflammatory response compared to the spherical HA nanoparticles. When assessed in vivo, HA-NWs enabled successful imaging of atherosclerotic plaques in a clinically relevant model of ApoE knockout transgenic mice for noninvasive plaque detection by MRI. Thus, nanoprobe shape engineering can be a useful strategy to significantly enhance their desired biological properties.
Collapse
Affiliation(s)
| | - Anne Tonson
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Mohammad H. El-Dakdouki
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Chemistry, Beirut Arab University, P.O. Box 11-5020, Riad El Solh 11072809, Beirut, Lebanon
| | - David C. Zhu
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824, United States
| | - Robert Wiseman
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
48
|
Xie Z, Wang X, Liu X, Du H, Sun C, Shao X, Tian J, Gu X, Wang H, Tian J, Yu B. Adipose-Derived Exosomes Exert Proatherogenic Effects by Regulating Macrophage Foam Cell Formation and Polarization. J Am Heart Assoc 2018; 7:JAHA.117.007442. [PMID: 29502100 PMCID: PMC5866320 DOI: 10.1161/jaha.117.007442] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Obesity is causally associated with atherosclerosis, and adipose tissue (AT)–derived exosomes may be implicated in the metabolic complications of obesity. However, the precise role of AT‐exosomes in atherogenesis remains unclear. We herein aimed to assess the effect of AT‐exosomes on macrophage foam cell formation and polarization and subsequent atherosclerosis development. Methods and Results Four types of exosomes isolated from the supernatants of ex vivo subcutaneous AT and visceral AT (VAT) explants that were derived from wild‐type mice and high‐fat diet (HFD)–induced obese mice were effectively taken up by RAW264.7 macrophages. Both treatment with wild‐type VAT exosomes and HFD‐VAT exosomes, but not subcutaneous AT exosomes, markedly facilitated macrophage foam cell generation through the downregulation of ATP‐binding cassette transporter (ABCA1 and ABCG1)–mediated cholesterol efflux. Decreased expression of liver X receptor‐α was also observed. Among the 4 types of exosomes, only HFD‐VAT exosomes significantly induced M1 phenotype transition and proinflammatory cytokine (tumor necrosis factor α and interleukin 6) secretion in RAW264.7 macrophages, which was accompanied by increased phosphorylation of NF‐κB‐p65 but not the cellular expression of NF‐κB‐p65 or IκB‐α. Furthermore, systematic intravenous injection of HFD‐VAT exosomes profoundly exacerbated atherosclerosis in hyperlipidemic apolipoprotein E–deficient mice, as indicated by the M1 marker (CD16/32 and inducible nitric oxide synthase)–positive areas and the Oil Red O/Sudan IV–stained area, without affecting the plasma lipid profile and body weight. Conclusions This study demonstrated a proatherosclerotic role for HFD‐VAT exosomes, which is exerted by regulating macrophage foam cell formation and polarization, indicating a novel link between AT and atherosclerosis in the context of obesity.
Collapse
Affiliation(s)
- Zulong Xie
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuedong Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinxin Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huaan Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changbin Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Shao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiangtian Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xia Gu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Cardiology, Heilongjiang Provincial Hospital, Harbin, China
| | - Hailong Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
49
|
Zhang X, Liu M, Qiao L, Zhang X, Liu X, Dong M, Dai H, Ni M, Luan X, Guan J, Lu H. Ginsenoside Rb1 enhances atherosclerotic plaque stability by skewing macrophages to the M2 phenotype. J Cell Mol Med 2018; 22:409-416. [PMID: 28944992 PMCID: PMC5742675 DOI: 10.1111/jcmm.13329] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 07/03/2017] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis (AS) is characterized as progressive arterial plaque, which is easy to rupture under low stability. Macrophage polarization and inflammation response plays an important role in regulating plaque stability. Ginsenoside Rb1 (Rb1), one of the main active principles of Panax Ginseng, has been found powerful potential in alleviating inflammatory response. However, whether Rb1 could exert protective effects on AS plaque stability remains unclear. This study investigated the role of Rb1 on macrophage polarization and atherosclerotic plaque stability using primary peritoneal macrophages isolated from C57BL/6 mice and AS model in ApoE-/- mice. In vitro, Rb1 treatment promoted the expression of arginase-I (Arg-I) and macrophage mannose receptor (CD206), two classic M2 macrophages markers, while the expression of iNOS (M1 macrophages) was decreased. Rb1 increased interleukin-4 (IL-4) and interleukin-13 (IL-13) secretion in supernatant and promoted STAT6 phosphorylation. IL-4 and/or IL-13 neutralizing antibodies and leflunomide, a STAT6 inhibitor attenuated the up-regulation of M2 markers induced by Rb1. In vivo, the administration of Rb1 promoted atherosclerotic lesion stability, accompanied by increased M2 macrophage phenotype and reduced MMP-9 staining. These data suggested that Rb1 enhanced atherosclerotic plaque stability through promoting anti-inflammatory M2 macrophage polarization, which is achieved partly by increasing the production of IL-4 and/or IL-13 and STAT6 phosphorylation. Our study provides new evidence for possibility of Rb1 in prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xue Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
- Department of CardiologyQingdao Municipal HospitalQingdaoChina
| | - Ming‐hao Liu
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Lei Qiao
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Xin‐yu Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Xiao‐ling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Hong‐yan Dai
- Department of CardiologyQingdao Municipal HospitalQingdaoChina
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Xiao‐rong Luan
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| | - Jun Guan
- Department of CardiologyQingdao Municipal HospitalQingdaoChina
| | - Hui‐xia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine; Department of CardiologyQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
50
|
Yuan XM, Ward LJ, Forssell C, Siraj N, Li W. Carotid Atheroma From Men Has Significantly Higher Levels of Inflammation and Iron Metabolism Enabled by Macrophages. Stroke 2017; 49:419-425. [PMID: 29284736 DOI: 10.1161/strokeaha.117.018724] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/26/2017] [Accepted: 11/29/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Men differ from women in the manifestation of atherosclerosis and iron metabolism. Intraplaque hemorrhage and hemoglobin (Hb) catabolism by macrophages are associated with atherosclerotic lesion instability. The study aims were to investigate sex differences in (1) lesion severity in relation to blood Hb, (2) iron homeostasis in human carotid plaques, and (3) macrophage polarization within atheroma. METHODS The carotid artery samples from 39 men and 23 women were immunostained with cell markers for macrophages, smooth muscle cells, ferritin, and TfR1 (transferrin receptor 1), which were further analyzed according to sex in relation to iron, Hb, and lipids in circulation. Additionally, samples of predefined regions from human carotid atherosclerotic lesions, including internal controls, were used for proteomic analysis by mass spectrometry. RESULTS Male patients, compared with women, had larger necrotic cores and more plaque rupture, which were associated with higher levels of Hb. Atheroma of male patients had significantly higher levels of Hb in circulation and CD68 macrophages, ferritin, and TfR1 in lesions. CD68 macrophages were significantly correlated with ferritin and TfR1. Plaques from male patients comparatively possessed higher levels of inflammatory macrophage subsets, CD86 (M1) and CD163 (M2), but lower levels of STF (serotransferrin) and HPX (hemopexin). CONCLUSIONS Male patients with carotid atheroma had more advanced and ruptured lesions associated with significantly higher levels of inflammatory macrophage infiltration and high iron stores in the blood and in their plaques. These findings help to understand sex differences and iron metabolism in atherosclerosis and factors related to atheroma progression.
Collapse
Affiliation(s)
- Xi-Ming Yuan
- From the Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine (X.-M.Y., L.J.W., N.S.) and Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine (L.J.W., W.L.), Linköping University, Sweden; Vascular Surgery, Linköping University Hospital, Sweden (C.F.); and Department of Internal Medicine, University of Alberta, Edmonton, Canada (N.S.).
| | - Liam J Ward
- From the Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine (X.-M.Y., L.J.W., N.S.) and Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine (L.J.W., W.L.), Linköping University, Sweden; Vascular Surgery, Linköping University Hospital, Sweden (C.F.); and Department of Internal Medicine, University of Alberta, Edmonton, Canada (N.S.)
| | - Claes Forssell
- From the Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine (X.-M.Y., L.J.W., N.S.) and Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine (L.J.W., W.L.), Linköping University, Sweden; Vascular Surgery, Linköping University Hospital, Sweden (C.F.); and Department of Internal Medicine, University of Alberta, Edmonton, Canada (N.S.)
| | - Nabeel Siraj
- From the Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine (X.-M.Y., L.J.W., N.S.) and Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine (L.J.W., W.L.), Linköping University, Sweden; Vascular Surgery, Linköping University Hospital, Sweden (C.F.); and Department of Internal Medicine, University of Alberta, Edmonton, Canada (N.S.)
| | - Wei Li
- From the Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine (X.-M.Y., L.J.W., N.S.) and Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine (L.J.W., W.L.), Linköping University, Sweden; Vascular Surgery, Linköping University Hospital, Sweden (C.F.); and Department of Internal Medicine, University of Alberta, Edmonton, Canada (N.S.)
| |
Collapse
|