1
|
Fedotova EI, Berezhnov AV, Popov DY, Shitikova EY, Vinokurov AY. The Role of mtDNA Mutations in Atherosclerosis: The Influence of Mitochondrial Dysfunction on Macrophage Polarization. Int J Mol Sci 2025; 26:1019. [PMID: 39940788 PMCID: PMC11817597 DOI: 10.3390/ijms26031019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/27/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Atherosclerosis is a complex inflammatory process associated with high-mortality cardiovascular diseases. Today, there is a growing body of evidence linking atherosclerosis to mutations of mitochondrial DNA (mtDNA). But the mechanism of this link is insufficiently studied. Atherosclerosis progression involves different cell types and macrophages are one of the most important. Due to their high plasticity, macrophages can demonstrate pro-inflammatory and pro-atherogenic (macrophage type M1) or anti-inflammatory and anti-atherogenic (macrophage type M2) effects. These two cell types, formed as a result of external stimuli, differ significantly in their metabolic profile, which suggests the central role of mitochondria in the implementation of the macrophage polarization route. According to this, we assume that mtDNA mutations causing mitochondrial disturbances can play the role of an internal trigger, leading to the formation of macrophage M1 or M2. This review provides a comparative analysis of the characteristics of mitochondrial function in different types of macrophages and their possible associations with mtDNA mutations linked with inflammation-based pathologies including atherosclerosis.
Collapse
Affiliation(s)
- Evgeniya I. Fedotova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Alexey V. Berezhnov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Daniil Y. Popov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Elena Y. Shitikova
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Andrey Y. Vinokurov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| |
Collapse
|
2
|
Ji YF, Wang YJ, Chen SH, Cheng Y. Interleukin-4 promotes the clearance of amyloid-β by monocyte through enhancing the recognition and intracellular processing of amyloid-β. J Alzheimers Dis 2024; 102:1017-1026. [PMID: 39660534 DOI: 10.1177/13872877241295361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2024]
Abstract
BACKGROUND Impaired clearance of amyloid-β protein (Aβ) in the peripheral system is a crucial event in the pathogenesis of sporadic Alzheimer's disease (AD). Dysfunctional monocytes with deficient clearance of Aβ and increased secretion of pro-inflammatory factors in the periphery are considered to contribute to AD development. Multiple studies suggest that IL-4 can inhibit the inflammatory response and enhance the expression and activity of cathepsin protease associated with intracellular clearance of Aβ by monocytes. OBJECTIVE To investigate the effects of interleukin-4 (IL-4) on Aβ clearance and inflammatory response of monocytes in vitro. METHODS In this study, flow cytometry, confocal microscopy and ELISA techniques were used for measurement of Aβ clearance and its related mechanisms of monocytes. RESULTS We found that the mean intracellular content and uptake ratios of Aβ42 in total monocytes, as well as in the CD14 + CD16- subset, were enhanced by IL-4, concomitant with the degradation of Aβ42 by monocytes. And IL-4 treatment also increased expression of scavenger receptor CD36 and Macrophage scavenger receptor 1 (MSR1) on monocytes. It was shown that IL-4 increased the Aβ immunoreactive area within lysosomal markers, including early endosome antigen 1 (EEA1), lysosome-associated membrane glycoprotein 1 and 2 (LAMP1 and 2) and Aβ degradative enzymes cathepsin B and S in monocytes, but reduced secretion of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interferon-γ (IFN-γ) by monocytes. CONCLUSIONS Our study supports the role of IL-4 in regulating Aβ clearance and inflammatory response by monocytes, suggesting that IL-4 may have therapeutic potential for AD.
Collapse
Affiliation(s)
- Yi-Fei Ji
- Department of Neurology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical University, Nanchong, Sichuan, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Si-Han Chen
- Department of Neurology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical University, Nanchong, Sichuan, China
| | - Yuan Cheng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Deochand DK, Dacic M, Bale MJ, Daman AW, Chaudhary V, Josefowicz SZ, Oliver D, Chinenov Y, Rogatsky I. Mechanisms of epigenomic and functional convergence between glucocorticoid- and IL4-driven macrophage programming. Nat Commun 2024; 15:9000. [PMID: 39424780 PMCID: PMC11489752 DOI: 10.1038/s41467-024-52942-x] [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] [Academic Contribution Register] [Received: 01/10/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024] Open
Abstract
Macrophages adopt distinct phenotypes in response to environmental cues, with type-2 cytokine interleukin-4 promoting a tissue-repair homeostatic state (M2IL4). Glucocorticoids (GC), widely used anti-inflammatory therapeutics, reportedly impart a similar phenotype (M2GC), but how such disparate pathways may functionally converge is unknown. We show using integrative functional genomics that M2IL4 and M2GC transcriptomes share a striking overlap mirrored by a shift in chromatin landscape in both common and signal-specific gene subsets. This core homeostatic program is enacted by transcriptional effectors KLF4 and the glucocorticoid receptor, whose genome-wide occupancy and actions are integrated in a stimulus-specific manner by the nuclear receptor cofactor GRIP1. Indeed, many of the M2IL4:M2GC-shared transcriptomic changes were GRIP1-dependent. Consistently, GRIP1 loss attenuated phagocytic activity of both populations in vitro and macrophage tissue-repair properties in the murine colitis model in vivo. These findings provide a mechanistic framework for homeostatic macrophage programming by distinct signals, to better inform anti-inflammatory drug design.
Collapse
Affiliation(s)
- Dinesh K Deochand
- Hospital for Special Surgery Research Institute, David Z. Rosensweig Genomics Center, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Marija Dacic
- Hospital for Special Surgery Research Institute, David Z. Rosensweig Genomics Center, New York, NY, USA
- Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Michael J Bale
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Andrew W Daman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Vidyanath Chaudhary
- Hospital for Special Surgery Research Institute, David Z. Rosensweig Genomics Center, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Steven Z Josefowicz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - David Oliver
- Hospital for Special Surgery Research Institute, David Z. Rosensweig Genomics Center, New York, NY, USA
| | - Yurii Chinenov
- Hospital for Special Surgery Research Institute, David Z. Rosensweig Genomics Center, New York, NY, USA
| | - Inez Rogatsky
- Hospital for Special Surgery Research Institute, David Z. Rosensweig Genomics Center, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
4
|
Han XQ, Cui ZW, Ma ZY, Wang J, Hu YZ, Li J, Ye JM, Tafalla C, Zhang YA, Zhang XJ. Phagocytic Plasma Cells in Teleost Fish Provide Insights into the Origin and Evolution of B Cells in Vertebrates. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:730-742. [PMID: 38984862 DOI: 10.4049/jimmunol.2400182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/02/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Teleost IgM+ B cells can phagocytose, like mammalian B1 cells, and secrete Ag-specific IgM, like mammalian B2 cells. Therefore, teleost IgM+ B cells may have the functions of both mammalian B1 and B2 cells. To support this view, we initially found that grass carp (Ctenopharyngodon idella) IgM+ plasma cells (PCs) exhibit robust phagocytic ability, akin to IgM+ naive B cells. Subsequently, we sorted grass carp IgM+ PCs into two subpopulations: nonphagocytic (Pha-IgM+ PCs) and phagocytic IgM+ PCs (Pha+IgM+ PCs), both of which demonstrated the capacity to secrete natural IgM with LPS and peptidoglycan binding capacity. Remarkably, following immunization of grass carp with an Ag, we observed that both Pha-IgM+ PCs and Pha+IgM+ PCs could secrete Ag-specific IgM. Furthermore, in vitro concatenated phagocytosis experiments in which Pha-IgM+ PCs from an initial phagocytosis experiment were sorted and exposed again to beads confirmed that these cells also have phagocytic capabilities, thereby suggesting that all teleost IgM+ B cells have phagocytic potential. Additionally, we found that grass carp IgM+ PCs display classical phenotypic features of macrophages, providing support for the hypothesis that vertebrate B cells evolved from ancient phagocytes. These findings together reveal that teleost B cells are a primitive B cell type with functions reminiscent of both mammalian B1 and B2 cells, providing insights into the origin and evolution of B cells in vertebrates.
Collapse
Affiliation(s)
- Xue-Qing Han
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Zheng-Wei Cui
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zi-You Ma
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jie Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Ya-Zhen Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jun Li
- School of Science and Medicine, Lake Superior State University, Sault Sainte Marie, MI
| | - Jian-Min Ye
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Carolina Tafalla
- Animal Health Research Center (CISA), National Institute for Agricultural and Food Research and Technology (INIA), National Research Council (CSIC), Madrid, Spain
| | - Yong-An Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xu-Jie Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
5
|
Dahdah N, Tercero-Alcázar C, Malagón MM, Garcia-Roves PM, Guzmán-Ruiz R. Interrelation of adipose tissue macrophages and fibrosis in obesity. Biochem Pharmacol 2024; 225:116324. [PMID: 38815633 DOI: 10.1016/j.bcp.2024.116324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carmen Tercero-Alcázar
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María M Malagón
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Pablo Miguel Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Rocío Guzmán-Ruiz
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain.
| |
Collapse
|
6
|
Tran N, Mills EL. Redox regulation of macrophages. Redox Biol 2024; 72:103123. [PMID: 38615489 PMCID: PMC11026845 DOI: 10.1016/j.redox.2024.103123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/22/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024] Open
Abstract
Redox signaling, a mode of signal transduction that involves the transfer of electrons from a nucleophilic to electrophilic molecule, has emerged as an essential regulator of inflammatory macrophages. Redox reactions are driven by reactive oxygen/nitrogen species (ROS and RNS) and redox-sensitive metabolites such as fumarate and itaconate, which can post-translationally modify specific cysteine residues in target proteins. In the past decade our understanding of how ROS, RNS, and redox-sensitive metabolites control macrophage function has expanded dramatically. In this review, we discuss the latest evidence of how ROS, RNS, and metabolites regulate macrophage function and how this is dysregulated with disease. We highlight the key tools to assess redox signaling and important questions that remain.
Collapse
Affiliation(s)
- Nhien Tran
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Bi YW, Li LS, Ru N, Zhang B, Lei X. Nicotinamide adenine dinucleotide phosphate oxidase in pancreatic diseases: Mechanisms and future perspectives. World J Gastroenterol 2024; 30:429-439. [PMID: 38414585 PMCID: PMC10895600 DOI: 10.3748/wjg.v30.i5.429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/26/2023] [Revised: 12/17/2023] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Abstract
Pancreatitis and pancreatic cancer (PC) stand as the most worrisome ailments affecting the pancreas. Researchers have dedicated efforts to unraveling the mechanisms underlying these diseases, yet their true nature continues to elude their grasp. Within this realm, oxidative stress is often believed to play a causal and contributory role in the development of pancreatitis and PC. Excessive accumulation of reactive oxygen species (ROS) can cause oxidative stress, and the key enzyme responsible for inducing ROS production in cells is nicotinamide adenine dinucleotide phosphate hydrogen oxides (NOX). NOX contribute to pancreatic fibrosis and inflammation by generating ROS that injure acinar cells, activate pancreatic stellate cells, and mediate macrophage polarization. Excessive ROS production occurs during malignant transformation and pancreatic carcinogenesis, creating an oxidative microenvironment that can cause abnormal apoptosis, epithelial to mesenchymal transition and genomic instability. Therefore, understanding the role of NOX in pancreatic diseases contributes to a more in-depth exploration of the exact pathogenesis of these diseases. In this review, we aim to summarize the potential roles of NOX and its mechanism in pancreatic disorders, aiming to provide novel insights into understanding the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Ya-Wei Bi
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Long-Song Li
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Nan Ru
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Zhang
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiao Lei
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
8
|
Suleimanov SK, Efremov YM, Klyucherev TO, Salimov EL, Ragimov AA, Timashev PS, Vlasova II. Radical-Generating Activity, Phagocytosis, and Mechanical Properties of Four Phenotypes of Human Macrophages. Int J Mol Sci 2024; 25:1860. [PMID: 38339139 PMCID: PMC10855323 DOI: 10.3390/ijms25031860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/05/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Macrophages are the major players and orchestrators of inflammatory response. Expressed proteins and secreted cytokines have been well studied for two polar macrophage phenotypes-pro-inflammatory M1 and anti-inflammatory regenerative M2, but little is known about how the polarization modulates macrophage functions. In this study, we used biochemical and biophysical methods to compare the functional activity and mechanical properties of activated human macrophages differentiated from monocyte with GM-CSF (M0_GM) and M-CSF (M0_M) and polarized into M1 and M2 phenotypes, respectively. Unlike GM-CSF, which generates dormant cells with low activity, M-CSF confers functional activity on macrophages. M0_M and M2 macrophages had very similar functional characteristics-high reactive oxygen species (ROS) production level, and higher phagocytosis and survival compared to M1, while M1 macrophages showed the highest radical-generating activity but the lowest phagocytosis and survival among all phenotypes. All phenotypes decreased their height upon activation, but only M1 and M2 cells increased in stiffness, which can indicate a decrease in the migration ability of these cells and changes in their interactions with other cells. Our results demonstrated that while mechanical properties differ between M0 and polarized cells, all four phenotypes of monocyte-derived macrophages differ in their functional activities, namely in cytokine secretion, ROS production, and phagocytosis. Within the broad continuum of human macrophages obtained in experimental models and existing in vivo, there is a diversity of phenotypes with varying combinations of both markers and functional activities.
Collapse
Affiliation(s)
- Shakir K. Suleimanov
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (S.K.S.); (Y.M.E.); (T.O.K.); (P.S.T.)
- Laboratory of Clinical Smart Nanotechnologies, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Yuri M. Efremov
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (S.K.S.); (Y.M.E.); (T.O.K.); (P.S.T.)
| | - Timofey O. Klyucherev
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (S.K.S.); (Y.M.E.); (T.O.K.); (P.S.T.)
- Laboratory of Clinical Smart Nanotechnologies, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Emin L. Salimov
- Laboratory Blood Transfusion Complex, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.L.S.); (A.A.R.)
| | - Aligeydar A. Ragimov
- Laboratory Blood Transfusion Complex, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.L.S.); (A.A.R.)
| | - Peter S. Timashev
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (S.K.S.); (Y.M.E.); (T.O.K.); (P.S.T.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Irina I. Vlasova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (S.K.S.); (Y.M.E.); (T.O.K.); (P.S.T.)
| |
Collapse
|
9
|
Oshima E, Hayashi Y, Xie Z, Sato H, Hitomi S, Shibuta I, Urata K, Ni J, Iwata K, Shirota T, Shinoda M. M2 macrophage-derived cathepsin S promotes peripheral nerve regeneration via fibroblast-Schwann cell-signaling relay. J Neuroinflammation 2023; 20:258. [PMID: 37946211 PMCID: PMC10636844 DOI: 10.1186/s12974-023-02943-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/18/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Although peripheral nerves have an intrinsic self-repair capacity following damage, functional recovery is limited in patients. It is a well-established fact that macrophages accumulate at the site of injury. Numerous studies indicate that the phenotypic shift from M1 macrophage to M2 macrophage plays a crucial role in the process of axon regeneration. This polarity change is observed exclusively in peripheral macrophages but not in microglia and CNS macrophages. However, the molecular basis of axonal regeneration by M2 macrophage is not yet fully understood. Herein, we aimed to identify the M2 macrophage-derived axon regeneration factor. METHODS We established a peripheral nerve injury model by transection of the inferior alveolar nerve (IANX) in Sprague-Dawley rats. Transcriptome analysis was performed on the injured nerve. Recovery from sensory deficits in the mandibular region and histological reconnection of IAN after IANX were assessed in rats with macrophage depletion by clodronate. We investigated the effects of adoptive transfer of M2 macrophages or M2-derived cathepsin S (CTSS) on the sensory deficit. CTSS initiating signaling was explored by western blot analysis in IANX rats and immunohistochemistry in co-culture of primary fibroblasts and Schwann cells (SCs). RESULTS Transcriptome analysis revealed that CTSS, a macrophage-selective lysosomal protease, was upregulated in the IAN after its injury. Spontaneous but partial recovery from a sensory deficit in the mandibular region after IANX was abrogated by macrophage ablation at the injured site. In addition, a robust induction of c-Jun, a marker of the repair-supportive phenotype of SCs, after IANX was abolished by macrophage ablation. As in transcriptome analysis, CTSS was upregulated at the injured IAN than in the intact IAN. Endogenous recovery from hypoesthesia was facilitated by supplementation of CTSS but delayed by pharmacological inhibition or genetic silencing of CTSS at the injured site. Adoptive transfer of M2-polarized macrophages at this site facilitated sensory recovery dependent on CTSS in macrophages. Post-IANX, CTSS caused the cleavage of Ephrin-B2 in fibroblasts, which, in turn, bound EphB2 in SCs. CTSS-induced Ephrin-B2 cleavage was also observed in human sensory nerves. Inhibition of CTSS-induced Ephrin-B2 signaling suppressed c-Jun induction in SCs and sensory recovery. CONCLUSIONS These results suggest that M2 macrophage-derived CTSS contributes to axon regeneration by activating SCs via Ephrin-B2 shedding from fibroblasts.
Collapse
Affiliation(s)
- Eri Oshima
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 142-8515, Japan
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan.
| | - Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hitoshi Sato
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 142-8515, Japan
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan
| | - Ikuko Shibuta
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan
| | - Kentaro Urata
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 142-8515, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-Ku, Tokyo, 101-8310, Japan
| |
Collapse
|
10
|
Kosyreva A, Vishnyakova P, Tsvetkov I, Kiseleva V, Dzhalilova DS, Miroshnichenko E, Lokhonina A, Makarova O, Fatkhudinov T. Advantages and disadvantages of treatment of experimental ARDS by M2-polarized RAW 264.7 macrophages. Heliyon 2023; 9:e21880. [PMID: 38027880 PMCID: PMC10658332 DOI: 10.1016/j.heliyon.2023.e21880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/14/2023] [Revised: 09/20/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Innate immunity reactions are core to any immunological process, including systemic inflammation and such extremes as acute respiratory distress syndrome (ARDS) and cytokine storm. Macrophages, the key cells of innate immunity, show high phenotypic plasticity: depending on microenvironmental cues, they can polarize into M1 (classically activated, pro-inflammatory) or M2 (alternatively activated, anti-inflammatory). The anti-inflammatory M2 macrophage polarization-based cell therapies constitute a novel prospective modality. Systemic administration of 'educated' macrophages is intended at their homing in lungs in order to mitigate the pro-inflammatory cytokine production and reduce the risks of 'cytokine storm' and related severe complications. Acute respiratory distress syndrome (ARDS) is the main mortality factor in pneumonia including SARS-CoV-associated cases. This study aimed to evaluate the influence of infusions of RAW 264.7 murine macrophage cell line polarized towards M2 phenotype on the development of LPS-induced ARDS in mouse model. The results indicate that the M2-polarized RAW 264.7 macrophage infusions in the studied model of ARDS promote relocation of lymphocytes from their depots in immune organs to the lungs. In addition, the treatment facilitates expression of M2-polarization markers Arg1, Vegfa and Tgfb and decreases of M1-polarization marker Cd38 in lung tissues, which can indicate the anti-inflammatory response activation. However, treatment of ARDS with M2-polarized macrophages didn't change the neutrophil numbers in the lungs. Moreover, the level of the Arg1 protein in lungs decreased throughtout the treatment with M2 macrophages, which is probably because of the pro-inflammatory microenvironment influence on the polarization of macrophages towards M1. Thus, the chemical polarization of macrophages is unstable and depends on the microenvironment. This adverse effect can be reduced through the use of primary autologous macrophages or some alternative methods of M2 polarization, notably siRNA-mediated.
Collapse
Affiliation(s)
- A.M. Kosyreva
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418, Moscow, Russia
| | - P.A. Vishnyakova
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russia
| | - I.S. Tsvetkov
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418, Moscow, Russia
| | - V.V. Kiseleva
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russia
| | - D. Sh. Dzhalilova
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418, Moscow, Russia
| | - E.A. Miroshnichenko
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418, Moscow, Russia
| | - A.V. Lokhonina
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russia
| | - O.V. Makarova
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418, Moscow, Russia
| | - T.H. Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN), 6 Miklukho-Maklaya Street, 117198, Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418, Moscow, Russia
| |
Collapse
|
11
|
Hu X, Li Y, Chen Q, Wang T, Ma L, Zhang W, Yu R, Zhang J, Wan J, Yu C, Yuan Z. Sialic acids promote macrophage M1 polarization and atherosclerosis by upregulating ROS and autophagy blockage. Int Immunopharmacol 2023; 120:110410. [PMID: 37270929 DOI: 10.1016/j.intimp.2023.110410] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/10/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/06/2023]
Abstract
Accumulating evidence suggests that sialic acids is closely related to atherosclerosis. However, the effects and underlying mechanisms of sialic acids in atherosclerosis have been not defined. Macrophages are one of the most important cells during plaque progression. In this study, we investigated the role of sialic acids in the M1 macrophage polarization and pathogenesis of atherosclerosis. Here we found that sialic acids can promote the polarization of RAW264.7 cells to the M1 phenotype, thereby promoting the expression of proinflammatory cytokines in vitro. The proinflammatory effect of sialic acids may result from the inhibition of LKB1-AMPK-Sirt3 signaling pathway to upregulate intracellular ROS and impairing autophagy-lysosome system to block autophagic flux. In the APOE-/- mice, sialic acids in plasma increased during the development of atherosclerosis. Moreover, exogenous supplement of sialic acids can promote plaque progression in aortic arch and aortic sinus being accompanied by the differentiation of macrophages into M1 type in peripheral tissues. These studies demonstrated that sialic acids can promote macrophage polarization toward the M1 phenotype to accentuate atherosclerosis via inducing mitochondrial ROS and blocking autophagy, thus providing clue to a novel therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Xuemei Hu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Yueyue Li
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qingyang Chen
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Ruihong Yu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Jun Zhang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jingyuan Wan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
12
|
Giri PS, Dwivedi M. Meta-analysis for Association of Interleukin 4 VNTR Polymorphism with Rheumatoid Arthritis Risk and Severity. Biochem Genet 2023; 61:823-846. [PMID: 36258103 DOI: 10.1007/s10528-022-10288-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2022] [Accepted: 10/02/2022] [Indexed: 11/02/2022]
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune disease characterized by severe joint pain. There are conflicting results for the association of Interleukin 4 (IL4) variable number tandem repeats (VNTR; rs8179190) polymorphism with RA. Therefore, we performed a meta-analysis of the available studies to investigate the association of IL4 VNTR polymorphism with RA risk and severity in the overall populations and Asian, Egyptian, European, and Turkish ethnicities by sub-group analyses. Eight studies involving 1993 RA patients and 1732 controls were included in this meta-analysis. We found increased RA risk for the susceptible "R2R2" genotype and "R2" allele under heterozygous, recessive, and allelic models in the Asian populations (p < 0.00001, p < 0.0001, p = 0.001). We observed a significant association between "R2R2" genotype and "R2" allele for RA protection in the Turkish population under heterozygous, recessive, and allelic models (p = 0.01, p = 0.004, p = 0.002). Disease severity-based analysis revealed significant association for "R2R2" genotype and "R2" allele with RA severity under homozygous, heterozygous, recessive, dominant, and allelic models(p = 0.0004, p = 0.03, p = 0.02, p = 0.003, p = 0.01), specifically in Asian populations (p = 0.009, p = 0.02, p = 0.003, p = 0.03, p = 0.01) and under heterozygous, dominant, and allelic genetic models in Egyptian (p = 0.0001, p < 0.0001, p < 0.0001) and European (p = 0.002, p = 0.0007, p = 0.0006) populations. In silico analysis suggested that the susceptible "R2" allele changes the RNA secondary structure to a stable form by changing minimum free energy(ΔG) from - 115.20 to - 136.40 kcal/mol, which might lead to increased stability of IL-4 in RA patients. Overall, the meta-analysis suggests for the involvement of susceptible "R2" allele with RA risk in the Asian populations, RA severity in the overall populations (specifically in Asian, Egyptian, & European populations), and RA protection in the Turkish population.
Collapse
Affiliation(s)
- Prashant S Giri
- C. G. Bhakta Institute of Biotechnology, Faculty of Science, Uka Tarsadia University, Bardoli, Surat, Gujarat, 394 350, India
| | - Mitesh Dwivedi
- C. G. Bhakta Institute of Biotechnology, Faculty of Science, Uka Tarsadia University, Bardoli, Surat, Gujarat, 394 350, India.
| |
Collapse
|
13
|
Lail SS, Balce DR, Canton J, Yates RM. Approaches to Measuring Reductive and Oxidative Events in Phagosomes. Methods Mol Biol 2023; 2692:139-152. [PMID: 37365466 DOI: 10.1007/978-1-0716-3338-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 06/28/2023]
Abstract
The phagosome is a redox-active organelle. Numerous reductive and oxidative systems play both direct and indirect roles in phagosomal function. With the advent of newer methodologies to study these redox events in live cells, the details of how redox conditions change within the maturing phagosome, how they are regulated, and how they influence other phagosomal functions can be investigated. In this chapter, we detail phagosome-specific, fluorescence-based assays that measure disulfide reduction and the production of reactive oxygen species in live phagocytes such as macrophages and dendritic cells, in real time.
Collapse
Affiliation(s)
- Shranjit S Lail
- Department of Medical Science, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dale R Balce
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Johnathan Canton
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robin M Yates
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.
- Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
14
|
Lopez T, Wendremaire M, Lagarde J, Duquet O, Alibert L, Paquette B, Garrido C, Lirussi F. Wound Healing versus Metastasis: Role of Oxidative Stress. Biomedicines 2022; 10:2784. [PMID: 36359304 PMCID: PMC9687595 DOI: 10.3390/biomedicines10112784] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/20/2022] [Revised: 10/20/2022] [Accepted: 10/29/2022] [Indexed: 10/24/2023] Open
Abstract
Many signaling pathways, molecular and cellular actors which are critical for wound healing have been implicated in cancer metastasis. These two conditions are a complex succession of cellular biological events and accurate regulation of these events is essential. Apart from inflammation, macrophages-released ROS arise as major regulators of these processes. But, whatever the pathology concerned, oxidative stress is a complicated phenomenon to control and requires a finely tuned balance over the different stages and responding cells. This review provides an overview of the pivotal role of oxidative stress in both wound healing and metastasis, encompassing the contribution of macrophages. Indeed, macrophages are major ROS producers but also appear as their targets since ROS interfere with their differentiation and function. Elucidating ROS functions in wound healing and metastatic spread may allow the development of innovative therapeutic strategies involving redox modulators.
Collapse
Affiliation(s)
- Tatiana Lopez
- UMR 1231, Lipides Nutrition Cancer, INSERM, 21000 Dijon, France
- UFR des Sciences de Santé, Université Bourgogne Franche-Comté, 25000 Besançon, France
| | - Maeva Wendremaire
- UMR 1231, Lipides Nutrition Cancer, INSERM, 21000 Dijon, France
- UFR des Sciences de Santé, Université Bourgogne Franche-Comté, 25000 Besançon, France
| | - Jimmy Lagarde
- UMR 1231, Lipides Nutrition Cancer, INSERM, 21000 Dijon, France
- UFR des Sciences de Santé, Université Bourgogne Franche-Comté, 25000 Besançon, France
| | - Oriane Duquet
- UFR des Sciences de Santé, Université Bourgogne Franche-Comté, 25000 Besançon, France
- Plateforme PACE, Laboratoire de Pharmacologie-Toxicologie, Centre Hospitalo-Universitaire Besançon, 25000 Besançon, France
| | - Line Alibert
- Service de Chirurgie, Centre Hospitalo-Universitaire Besançon, 25000 Besançon, France
| | - Brice Paquette
- Service de Chirurgie, Centre Hospitalo-Universitaire Besançon, 25000 Besançon, France
| | - Carmen Garrido
- UMR 1231, Lipides Nutrition Cancer, INSERM, 21000 Dijon, France
- UFR des Sciences de Santé, Université Bourgogne Franche-Comté, 25000 Besançon, France
- Centre Georges François Leclerc, 21000 Dijon, France
| | - Frédéric Lirussi
- UMR 1231, Lipides Nutrition Cancer, INSERM, 21000 Dijon, France
- UFR des Sciences de Santé, Université Bourgogne Franche-Comté, 25000 Besançon, France
- Plateforme PACE, Laboratoire de Pharmacologie-Toxicologie, Centre Hospitalo-Universitaire Besançon, 25000 Besançon, France
| |
Collapse
|
15
|
Zhao X, Chen J, Sun H, Zhang Y, Zou D. New insights into fibrosis from the ECM degradation perspective: the macrophage-MMP-ECM interaction. Cell Biosci 2022; 12:117. [PMID: 35897082 PMCID: PMC9327238 DOI: 10.1186/s13578-022-00856-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/14/2022] [Accepted: 07/16/2022] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is a pathological feature of a variety of chronic inflammatory diseases that can affect almost all organs, which can cause severe consequences and even lead to death. Fibrosis is characterized by the excessive accumulation of extracellular matrix (ECM) due to disruption of the balance between ECM production and degradation. Although overabundance of ECM proteins has long been the focus of studies on fibrosis, another facet of the problem-impaired degradation of the ECM-is gaining increasing attention. Matrix metalloproteinase (MMP) and the tissue inhibitor of metalloproteinase (TIMP) system is the main molecular system contributing to ECM degradation, and macrophages are the major regulators of ECM. However, the relationship among macrophages, the MMP/TIMP system and the ECM is not fully understood in the context of fibrosis. Here, we discuss in detail the role played by the ECM in the development of fibrosis and highlight the macrophage-MMP-ECM interaction that is involved in fibrogenesis and may be a potential therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Xiangyu Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayin Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxiang Sun
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Pérez S, Rius-Pérez S. Macrophage Polarization and Reprogramming in Acute Inflammation: A Redox Perspective. Antioxidants (Basel) 2022; 11:antiox11071394. [PMID: 35883885 PMCID: PMC9311967 DOI: 10.3390/antiox11071394] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/14/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage polarization refers to the process by which macrophages can produce two distinct functional phenotypes: M1 or M2. The balance between both strongly affects the progression of inflammatory disorders. Here, we review how redox signals regulate macrophage polarization and reprogramming during acute inflammation. In M1, macrophages augment NADPH oxidase isoform 2 (NOX2), inducible nitric oxide synthase (iNOS), synaptotagmin-binding cytoplasmic RNA interacting protein (SYNCRIP), and tumor necrosis factor receptor-associated factor 6 increase oxygen and nitrogen reactive species, which triggers inflammatory response, phagocytosis, and cytotoxicity. In M2, macrophages down-regulate NOX2, iNOS, SYNCRIP, and/or up-regulate arginase and superoxide dismutase type 1, counteract oxidative and nitrosative stress, and favor anti-inflammatory and tissue repair responses. M1 and M2 macrophages exhibit different metabolic profiles, which are tightly regulated by redox mechanisms. Oxidative and nitrosative stress sustain the M1 phenotype by activating glycolysis and lipid biosynthesis, but by inhibiting tricarboxylic acid cycle and oxidative phosphorylation. This metabolic profile is reversed in M2 macrophages because of changes in the redox state. Therefore, new therapies based on redox mechanisms have emerged to treat acute inflammation with positive results, which highlights the relevance of redox signaling as a master regulator of macrophage reprogramming.
Collapse
|
17
|
Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022; 11:2091. [PMID: 35805174 PMCID: PMC9266143 DOI: 10.3390/cells11132091] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.
Collapse
Affiliation(s)
| | - Shelley B. Hooks
- Hooks Lab, Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
18
|
Espinosa Gonzalez M, Volk-Draper L, Bhattarai N, Wilber A, Ran S. Th2 cytokines IL-4, IL-13, and IL-10 promote differentiation of pro-lymphatic progenitors derived from bone marrow myeloid precursors. Stem Cells Dev 2022; 31:322-333. [PMID: 35442077 PMCID: PMC9232236 DOI: 10.1089/scd.2022.0004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
Abstract
Myeloid-lymphatic endothelial cell progenitors (M-LECP) are a subset of bone marrow (BM)-derived cells characterized by expression of M2-type macrophage markers. We previously showed significant contribution of M-LECP to tumor lymphatic formation and metastasis in human clinical breast tumors and corresponding mouse models. Since M2-type is induced in macrophages by immunosuppressive Th2 cytokines IL-4, IL-13, and IL-10, we hypothesized that these factors might promote pro-lymphatic specification of M-LECP during their differentiation from BM myeloid precursors. To test this hypothesis, we analyzed expression of Th2 cytokines and their receptors in mouse BM cells under conditions leading to M-LECP differentiation, namely, CSF-1 treatment followed by activation of TLR4. We found that under these conditions, all three Th2 receptors were strongly upregulated in >95% of the cells that also secrete endogenous IL-10 but not IL-4 or IL-13 ligands. However, addition of any of the Th2 factors to CSF-1 primed cells significantly increased generation of myeloid-lymphatic progenitors as indicated by co-induction of lymphatic-specific (e.g., Lyve-1, integrin-a9, collectin-12, and stabilin-1) and M2-type markers (e.g., CD163, CD204, CD206, and PD-L1). Antibody-mediated blockade of either IL-10 receptor (IL-10R) or IL-10 ligand significantly reduced both immunosuppressive and lymphatic phenotypes. Moreover, tumor-recruited Lyve-1+ lymphatic progenitors in vivo expressed all Th2 receptors as well as corresponding ligands including IL-4 and IL-13 that were absent in BM cells. This study presents original evidence for the significant role of Th2 cytokines in co-development of immunosuppressive and lymphatic phenotypes in tumor-recruited M2-type myeloid cells. Progenitor-mediated increase in lymphatic vessels can enhance immunosuppression by physical removal of stimulatory immune cells. Thus, targeting Th2 pathways might simultaneously relieve immunosuppression and inhibit differentiation of pro-lymphatic progenitors that ultimately promote tumor spread.
Collapse
Affiliation(s)
- Maria Espinosa Gonzalez
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Lisa Volk-Draper
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Nihit Bhattarai
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Andrew Wilber
- Southern Illinois University School of Medicine, Medical Microbiology, Immunology and Cell Biology, Springfield, Illinois, United States;
| | - Sophia Ran
- Southern Illinois University School of Medicine, 12249, Medical Microbiology, Immunology and Cell Biology, 801 N. Rutledge, P.O. Box 19626, Springfield, Illinois, United States, 62794;
| |
Collapse
|
19
|
O'Brien EM, Spiller KL. Pro-inflammatory polarization primes Macrophages to transition into a distinct M2-like phenotype in response to IL-4. J Leukoc Biol 2021; 111:989-1000. [PMID: 34643290 DOI: 10.1002/jlb.3a0520-338r] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022] Open
Abstract
Tissue repair is largely regulated by diverse Mϕ populations whose functions are timing- and context-dependent. The early phase of healing is dominated by pro-inflammatory Mϕs, also known as M1, followed by the emergence of a distinct and diverse population that is collectively referred to as M2. The extent of the diversity of the M2 population is unknown. M2 Mϕs may originate directly from circulating monocytes or from phenotypic switching of pre-existing M1 Mϕs within the site of injury. The differences between these groups are poorly understood, but have major implications for understanding and treating pathologies characterized by deficient M2 activation, such as chronic wounds, which also exhibit diminished M1 Mϕ behavior. This study investigated the influence of prior M1 activation on human Mϕ polarization to an M2 phenotype in response to IL-4 treatment in vitro. Compared to unactivated (M0) Mϕs, M1 Mϕs up-regulated several receptors that promote the M2 phenotype, including the primary receptor for IL-4. M1 Mϕs also up-regulated M2 markers in response to lower doses of IL-4, including doses as low as 10 pg/mL, and accelerated STAT6 phosphorylation. However, M1 activation appeared to also change the Mϕ response to treatment with IL-4, generating an M2-like phenotype with a distinct gene and protein expression signature compared to M2 Mϕs prepared directly from M0 Mϕs. Functionally, compared to M0-derived M2 Mϕs, M1-derived M2 Mϕs demonstrated increased migratory response to SDF-1α, and conditioned media from these Mϕs promoted increased migration of endothelial cells in transwell assays, although other common Mϕ-associated functions such as phagocytosis were not affected by prior polarization state. In summary, M1 polarization appears to prime Mϕs to transition into a distinct M2 phenotype in response to IL4, which leads to increased expression of some genes and proteins and decreased expression of others, as well as functional differences. Together, these findings indicate the importance of prior M1 activation in regulating subsequent M2 behavior, and suggest that correcting M1 behavior may be a therapeutic target in dysfunctional M2 activation.
Collapse
Affiliation(s)
- Erin M O'Brien
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Fountain A, Inpanathan S, Alves P, Verdawala MB, Botelho RJ. Phagosome maturation in macrophages: Eat, digest, adapt, and repeat. Adv Biol Regul 2021; 82:100832. [PMID: 34717137 DOI: 10.1016/j.jbior.2021.100832] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 11/30/2022]
Abstract
Phagocytosis is a dynamic process that requires an intricate interplay between phagocytic receptors, membrane lipids, and numerous signalling proteins and their effectors, to coordinate the engulfment of a bound particle. These particles are diverse in their physico-chemical properties such as size and shape and include bacteria, fungi, apoptotic cells, living tumour cells, and abiotic particles. Once engulfed, these particles are enclosed within a phagosome, which undergoes a striking transformation referred to as phagosome maturation, which will ultimately lead to the processing and degradation of the enclosed particulate. In this review, we focus on recent advancements in phagosome maturation in macrophages, highlighting new discoveries and emerging themes. Such advancements include identification of new GTPases and their effectors and the intricate spatio-temporal dynamics of phosphoinositides in governing phagosome maturation. We then explore phagosome fission and recycling, the emerging role of membrane contact sites, and delve into mechanisms of phagosome resolution to recycle and reform lysosomes. We further illustrate how phagosome maturation is context-dependent, subject to the type of particle, phagocytic receptors, the phagocytes and their state of activation during phagocytosis. Lastly, we discuss how phagosomes serve as signalling platforms to help phagocytes adapt to their environmental conditions. Overall, this review aims to cover recent findings, identify emerging themes, and highlight current challenges and directions to improve our understanding of phagosome maturation in macrophages.
Collapse
Affiliation(s)
- Aaron Fountain
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Subothan Inpanathan
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Patris Alves
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Munira B Verdawala
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Roberto J Botelho
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada.
| |
Collapse
|
21
|
Levin SG, Pershina EV, Bugaev-Makarovskiy NA, Chernomorets IY, Konakov MV, Arkhipov VI. Why Do Levels Of Anti-inflammatory Cytokines Increase During Memory Acquisition? Neuroscience 2021; 473:159-169. [PMID: 34418518 DOI: 10.1016/j.neuroscience.2021.08.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/20/2021] [Revised: 07/14/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022]
Abstract
The role of anti-inflammatory cytokines in the mechanisms of learning and memory, modulation of synaptic plasticity in the mammalian brain has not received sufficient attention. These issues are discussed in this review, and among the many cytokines, attention is paid to the most studied in this respect IL-10, IL-4, IL-13 and TGF-β. The level of anti-inflammatory cytokines in the brain tends to increase during memory acquisition, but the significance of such an increase is unclear. We hypothesize that anti-inflammatory cytokines primarily protect and optimize the functioning of neuronal circuits involved in information processing. The increased local activity of neurons during memory acquisition activates many signaling molecules, and some of them can trigger unwanted processes (including neuroinflammation), but increased levels of anti-inflammatory cytokines prevent this triggering. Each of the anti-inflammatory cytokines plays a specific role in supporting information processing. For example, the role of IL-4 and IL-13 in recruiting T cells to the meninges during training in healthy animals has been most studied. It has also been shown that TGF-β is able to optimize late stage LTP in the hippocampus and support the consolidation of memory traces in behavioral studies. Cytokines have an effect on learning and memory through their influence on neuroplasticity, neurogenesis in the hippocampus and regulation of the neurovascular unit. Experiments have shown such an effect, and the data obtained create the prerequisites for new therapeutic approaches to the correction of cognitive impairments.
Collapse
Affiliation(s)
- Sergey G Levin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Ekaterina V Pershina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia.
| | - Nickolay A Bugaev-Makarovskiy
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Irina Yu Chernomorets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Maxim V Konakov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Vladimir I Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| |
Collapse
|
22
|
Lukácsi S, Farkas Z, Saskői É, Bajtay Z, Takács-Vellai K. Conserved and Distinct Elements of Phagocytosis in Human and C. elegans. Int J Mol Sci 2021; 22:ijms22168934. [PMID: 34445642 PMCID: PMC8396242 DOI: 10.3390/ijms22168934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Endocytosis provides the cellular nutrition and homeostasis of organisms, but pathogens often take advantage of this entry point to infect host cells. This is counteracted by phagocytosis that plays a key role in the protection against invading microbes both during the initial engulfment of pathogens and in the clearance of infected cells. Phagocytic cells balance two vital functions: preventing the accumulation of cell corpses to avoid pathological inflammation and autoimmunity, whilst maintaining host defence. In this review, we compare elements of phagocytosis in mammals and the nematode Caenorhabditis elegans. Initial recognition of infection requires different mechanisms. In mammals, pattern recognition receptors bind pathogens directly, whereas activation of the innate immune response in the nematode rather relies on the detection of cellular damage. In contrast, molecules involved in efferocytosis—the engulfment and elimination of dying cells and cell debris—are highly conserved between the two species. Therefore, C. elegans is a powerful model to research mechanisms of the phagocytic machinery. Finally, we show that both mammalian and worm studies help to understand how the two phagocytic functions are interconnected: emerging data suggest the activation of innate immunity as a consequence of defective apoptotic cell clearance.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Éva Saskői
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
- Department of Immunology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary
| | - Krisztina Takács-Vellai
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
- Correspondence:
| |
Collapse
|
23
|
Fields L, Ito T, Kobayashi K, Ichihara Y, Podaru MN, Hussain M, Yamashita K, Machado V, Lewis-McDougall F, Suzuki K. Epicardial placement of human MSC-loaded fibrin sealant films for heart failure: Preclinical efficacy and mechanistic data. Mol Ther 2021; 29:2554-2570. [PMID: 33887461 PMCID: PMC8353205 DOI: 10.1016/j.ymthe.2021.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/02/2020] [Revised: 02/28/2021] [Accepted: 04/15/2021] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal cell (MSC) transplantation has been investigated as an advanced treatment of heart failure; however, further improvement of the therapeutic efficacy and mechanistic understanding are needed. Our previous study has reported that epicardial placement of fibrin sealant films incorporating rat amniotic membrane-derived (AM)-MSCs (MSC-dressings) could address limitations of traditional transplantation methods. To progress this finding toward clinical translation, this current study aimed to examine the efficacy of MSC-dressings using human AM-MSCs (hAM-MSCs) and the underpinning mechanism for myocardial repair. Echocardiography demonstrated that cardiac function and structure were improved in a rat ischemic cardiomyopathy model after hAM-MSC-dressing therapy. hAM-MSCs survived well in the rat heart, enhanced myocardial expression of reparative genes, and attenuated adverse remodeling. Copy number analysis by qPCR revealed that upregulated reparative genes originated from endogenous rat cells rather than hAM-MSCs. These results suggest hAM-MSC-dressing therapy stimulates a secondary release of paracrine factors from endogenous cells improving myocardial repair ("secondary paracrine effect"), and cardiac M2-like macrophages were identified as a potential cell source of repair. We demonstrated hAM-MSCs increased M2-like macrophages through not only enhancing M2 polarization but also augmenting their proliferation and migration capabilities via PGE2, CCL2, and TGF-β1, resulting in enhanced cardiac function after injury.
Collapse
Affiliation(s)
- Laura Fields
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tomoya Ito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kazuya Kobayashi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Yuki Ichihara
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mihai-Nicolae Podaru
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mohsin Hussain
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kizuku Yamashita
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vanessa Machado
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Fiona Lewis-McDougall
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ken Suzuki
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
24
|
Gaiser AK, Bauer S, Ruez S, Holzmann K, Fändrich M, Syrovets T, Simmet T. Serum Amyloid A1 Induces Classically Activated Macrophages: A Role for Enhanced Fibril Formation. Front Immunol 2021; 12:691155. [PMID: 34276683 PMCID: PMC8278318 DOI: 10.3389/fimmu.2021.691155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/05/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
AA amyloidosis belongs to the group of amyloid diseases which can follow chronic inflammatory conditions of various origin. The disease is characterized by the deposition of insoluble amyloid fibrils formed by serum amyloid A1 (SAA1) leading eventually to organ failure. Macrophages are intimately involved in the fibrillogenesis as well as in the clearance of amyloid fibrils. In vivo, macrophages may occur as classically (M1) or alternatively activated (M2) macrophages. We investigate here how SAA1 might affect the macrophage phenotype and function. Gene microarray analysis revealed upregulation of 64 M1-associated genes by SAA1. M1-like polarization was further confirmed by the expression of the M1-marker MARCO, activation of the NF-κB transcription factor, and secretion of the M1-cytokines TNF-α, IL-6, and MCP-1. Additionally, we demonstrate here that M1-polarized macrophages exhibit enhanced fibrillogenic activity towards SAA1. Based on our data, we propose reconsideration of the currently used cellular amyloidosis models towards an in vitro model employing M1-polarized macrophages. Furthermore, the data suggest macrophage repolarization as potential intervention strategy in AA amyloidosis.
Collapse
Affiliation(s)
- Ann-Kathrin Gaiser
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Shanna Bauer
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Stephanie Ruez
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | | | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, Ulm, Germany
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| |
Collapse
|
25
|
Danz JC, Kantarci A, Bornstein MM, Katsaros C, Stavropoulos A. Impact of Orthodontic Forces on Plasma Levels of Markers of Bone Turnover and Inflammation in a Rat Model of Buccal Expansion. Front Physiol 2021; 12:637606. [PMID: 34113259 PMCID: PMC8186951 DOI: 10.3389/fphys.2021.637606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2020] [Accepted: 04/22/2021] [Indexed: 11/21/2022] Open
Abstract
Plasma levels of protein analytes might be markers to predict and monitor the kinetics of bone and tissue remodeling, including maximization of orthodontic treatment stability. They could help predict/prevent and/or diagnose possible adverse effects such as bone dehiscences, gingival recession, or root resorption. The objective of this study was to measure plasma levels of markers of bone turnover and inflammation during orthodontic force application in a rat model of orthodontic expansion. Two different orthodontic forces for bilateral buccal expansion of the maxillary arches around second and third molars were applied in 10 rats equally distributed in low-force (LF) or conventional force (CF) groups. Four rats served as the control group. Blood samples were collected at days 0, 1, 2, 3, 6, 13, 21, and 58. Longitudinal concentrations of osteoprotegerin (OPG), soluble receptor activator of nuclear factor kappaB ligand (sRANKL), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), tumor necrosis factor α (TNF), and parathyroid hormone (PTH) were determined in blood samples by a multiplex immunoassay. CF and LF resulted in a significantly maxillary skeletal expansion while the CF group demonstrated significantly higher expansion than the LF group in the long term. Bone turnover demonstrated a two-phase response. During the “early phase” (up to 6 days of force application), LF resulted in more sRANKL expression and increased sRANKL/OPG ratio than the CF and control animals. There was a parallel increase in PTH levels in the early phase in response to LF. During the “late phase” (6–58 days), the markers of bone turnover were stable in both groups. IL-4, IL-6, and IL-10 levels did not significantly change the test groups throughout the study. These results suggest that maxillary expansion in response to different orthodontic forces follows different phases of bone turnover that may be force specific.
Collapse
Affiliation(s)
- Jan C Danz
- Department of Orthodontics and Dentofacial Orthopedics, School of Dental Medicine ZMK, University of Bern, Bern, Switzerland
| | | | - Michael M Bornstein
- Department of Oral Health and Medicine, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Christos Katsaros
- Department of Orthodontics and Dentofacial Orthopedics, School of Dental Medicine ZMK, University of Bern, Bern, Switzerland
| | - Andreas Stavropoulos
- Division of Regenerative Dental Medicine and Periodontology, University Clinics of Dental Medicine (CUMD), University of Geneva, Geneva, Switzerland.,Department of Periodontology, Faculty of Odontology, Malmö University, Malmö, Sweden
| |
Collapse
|
26
|
Li J, Yu S, Lu X, Cui K, Tang X, Xu Y, Liang X. The phase changes of M1/M2 phenotype of microglia/macrophage following oxygen-induced retinopathy in mice. Inflamm Res 2021; 70:183-192. [PMID: 33386422 DOI: 10.1007/s00011-020-01427-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2020] [Revised: 10/27/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Microglia/macrophage activation is previously reported to be involved in various ocular diseases. However, the separate role of M1/M2 phenotype microglia/macrophage in the pathological process of oxygen-induced retinopathy (OIR) remains unknown. In this research, we explored the role and regulatory mechanism of M1/M2 microglia/macrophage in OIR in C57BL/6J mice. Furthermore, we demonstrated the time phase of M1/M2 shifting of microglia/macrophage during the natural process of OIR, which is very essential for further investigations. MATERIALS AND METHODS C57BL/6j pups were exposed to hyperoxia environment from postnatal 7(P7) to P12 then returned to normoxia. The mice were then euthanized, and the eyes were harvested at a series of time points for further investigation. The M1/M2 phenotype microglia/macrophage activity was presented by immunofluorescent staining and real-time quantitative polymerase chain reaction (qPCR). The NF-κb-STAT3 signaling and IL-4-STAT6-PPAR-γ signaling pathway activity was examined by western blot analysis. RESULTS The microglia/macrophage were activated when the OIR model was set up after P12. The M1 microglia/macrophage activation was found in neovascularization (NV) tufts in both central and peripheral retina, which started from P12 when the mice were returned to normoxia environment and peaked at P17. During this period of time, the NF-κb-STAT3 signaling pathway was activated, resulting in the upregulated M1 phenotype microglia/macrophage polarization, along with the enhanced inflammatory cytokine expression including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1β. Consequently, the NV tufts were observed from P12 and the volume continued to increase until P17. However, the M2 phenotype microglia/macrophage activity took over during the late phase of OIR started from P17. The IL-4-STAT6-PPAR-γ signaling activity was upregulated from P17 and peaked at P20, inducing M2 phenotype microglia polarization, which consequently led to the inhibition of inflammatory cytokines and spontaneous regression of NV tufts. CONCLUSIONS Microglia/macrophage participate actively in the natural process of OIR in mice, and two phenotypes exert different functions. Treatment modulating microglia/macrophage polarize toward M2 phenotype might be a novel and promising method for ocular neovascular diseases such as retinopathy of prematurity (ROP), wet age-related macular degeneration (wAMD), and diabetic retinopathy (DR).
Collapse
Affiliation(s)
- Jia Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China.
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510030, Guangdong, China.
| |
Collapse
|
27
|
Chen M, Tse G, Wong WT. Interleukin-4 increases phagocytosis of necrotic cells by macrophages through scavenger receptor CD36. Clin Exp Pharmacol Physiol 2021; 48:129-136. [PMID: 32852093 DOI: 10.1111/1440-1681.13399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Interleukin-4 (IL-4) signalling pathways regulate the activity of macrophages, enhance their proteolytic capacity and drive resolution of inflammation during tissue repair. The aim of this study was to examine whether IL-4 can enhance phagocytosis of necrotic cells and elucidate the molecular mechanisms. METHODS Phagocytosis of necrotic thymocytes by RAW264.7 cells, a macrophage cell line, with or without IL-4 treatment, was determined by flow cytometry. Protein expression was determined by western blot analysis. RESULTS The phagocytosis index was significantly increased by IL-4 (10 ng/mL). IL-4-enhanced phagocytosis was mediated by upregulation of scavenger receptor CD36. STAT6 activation is required for IL-4-mediated phagocytosis. CONCLUSIONS Interleukin-4 can accelerate cell debris clearance by stimulating expression of CD36, which requires downstream STAT6 activation. Its beneficial effects on driving tissue repair and regeneration should be explored in future studies.
Collapse
Affiliation(s)
- Mingzhuang Chen
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary Tse
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
28
|
The Degree of Helicobacter pylori Infection Affects the State of Macrophage Polarization through Crosstalk between ROS and HIF-1 α. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5281795. [PMID: 33376580 PMCID: PMC7746446 DOI: 10.1155/2020/5281795] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/20/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022]
Abstract
Methods The expression of CD86, CD206, and HIF-1α in the gastric mucosa was evaluated through immunohistochemistry. RAW 264.7 cells were cocultured with H. pylori at various multiplicities of infection (MOIs), and iNOS, CD86, Arg-1, CD206, and HIF-1α expression was detected by Western blot, PCR, and ELISA analyses. ROS expression was detected with the fluorescent probe DCFH-DA. Macrophages were also treated with the ROS inhibitor NAC or HIF-1α inhibitor YC-1. Results Immunohistochemical staining revealed that the macrophage polarization state was associated with the progression of gastric lesions and state of H. pylori infection. The MOI of H. pylori affected macrophage polarization, and H. pylori enhanced the expression of ROS and HIF-1α in macrophages. A low MOI of H. pylori promoted both the M1 and M2 phenotypes, while a high MOI suppressed the M2 phenotype. Furthermore, ROS inhibition attenuated HIF-1α expression and switched macrophage polarization from M1 to M2. However, HIF-1α inhibition suppressed ROS expression and inhibited both the M1 phenotype and the M2 phenotype. Inhibition of ROS or HIF-1α also suppressed the activation of the Akt/mTOR pathway, which was implicated in H. pylori-induced macrophage polarization. Conclusions Macrophage polarization is associated with the progression of gastric lesions and state of H. pylori infection. The MOI of H. pylori influences the macrophage polarization state. Crosstalk between ROS and HIF-1α regulates H. pylori-induced macrophage polarization via the Akt/mTOR pathway.
Collapse
|
29
|
Wang J, Su Y, Xu L, Li D. Micro-patterned surface construction on BCP ceramics and the regulation on inflammation-involved osteogenic differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111220. [DOI: 10.1016/j.msec.2020.111220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/03/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023]
|
30
|
Arnhold J. The Dual Role of Myeloperoxidase in Immune Response. Int J Mol Sci 2020; 21:E8057. [PMID: 33137905 PMCID: PMC7663354 DOI: 10.3390/ijms21218057] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/05/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
The heme protein myeloperoxidase (MPO) is a major constituent of neutrophils. As a key mediator of the innate immune system, neutrophils are rapidly recruited to inflammatory sites, where they recognize, phagocytose, and inactivate foreign microorganisms. In the newly formed phagosomes, MPO is involved in the creation and maintenance of an alkaline milieu, which is optimal in combatting microbes. Myeloperoxidase is also a key component in neutrophil extracellular traps. These helpful properties are contrasted by the release of MPO and other neutrophil constituents from necrotic cells or as a result of frustrated phagocytosis. Although MPO is inactivated by the plasma protein ceruloplasmin, it can interact with negatively charged components of serum and the extracellular matrix. In cardiovascular diseases and many other disease scenarios, active MPO and MPO-modified targets are present in atherosclerotic lesions and other disease-specific locations. This implies an involvement of neutrophils, MPO, and other neutrophil products in pathogenesis mechanisms. This review critically reflects on the beneficial and harmful functions of MPO against the background of immune response.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, 04 107 Leipzig, Germany
| |
Collapse
|
31
|
Allan ERO, Blouin MS. Heat shock increases hydrogen peroxide release from circulating hemocytes of the snail Biomphalaria glabrata. FISH & SHELLFISH IMMUNOLOGY 2020; 105:203-208. [PMID: 32702479 PMCID: PMC7501213 DOI: 10.1016/j.fsi.2020.07.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/12/2020] [Revised: 07/01/2020] [Accepted: 07/14/2020] [Indexed: 06/11/2023]
Abstract
Planorbid freshwater snails are important intermediate hosts for parasitic diseases caused by parasitic worms, most notably schistosomiasis. There are numerous reports of snails, specifically Biomphalaria glabrata, having compromised defences against schistosomes after being exposed to thermal stress. Environmental modifications to the defenses of schistosome transmitting snails could have negative ramifications for human disease risk in the context of climate change. Here the effects of heat shock on the production of hydrogen peroxide, a primary anti-microbial effector in many molluscs, were examined. The present findings show that heat shock increases NADPH oxidase 2 mRNA levels and hydrogen peroxide produced by snail hemocytes, and that both of these phenotypes could be reversed by an HSP-90 inhibitor. These findings indicate that snail defense systems are altered by heat shock at a molecular level in B. glabrata, and that snail immunity to many pathogens may be altered by the rapid variations in temperature that are associated with global climate change.
Collapse
Affiliation(s)
- Euan R O Allan
- Department of Pathobiology, School of Veterinary Medicine, St. George's University, West Indies, Grenada.
| | - Michael S Blouin
- Department of Integrative Biology, College of Science, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
32
|
Quarta A, Berneman Z, Ponsaerts P. Neuroprotective modulation of microglia effector functions following priming with interleukin 4 and 13: current limitations in understanding their mode-of-action. Brain Behav Immun 2020; 88:856-866. [PMID: 32224056 DOI: 10.1016/j.bbi.2020.03.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/27/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years the long-standing theory of microglia's properties for dual polarization towards a pro- or anti-inflammatory phenotype has been deeply challenged. Furthermore, the elucidation of microglia ontogenesis exposed intrinsic differences between microglia and peripheral myeloid cells, thereby further underscoring the need to re-evaluate microglia-specific activation behavior, especially within an inflamed central nervous system (CNS) environment. This review critically summarizes recent literature on the in vitro and in vivo response of murine microglia to the immune-modulatory cytokines interleukin 4 (IL4) and interleukin 13 (IL13), i.e. those driving the so-called anti-inflammatory phenotype. Here we highlight several pivotal factors that may influence experimental outcome and/or interpretation of in vitro and in vivo studies evaluating microglia's phenotypical and functional properties upon IL4/IL13 treatment. Finally, the current therapeutic relevance of IL4/IL13-induced microglia activation in both acute and chronic CNS disorders is discussed.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
33
|
Huo SC, Yue B. Approaches to promoting bone marrow mesenchymal stem cell osteogenesis on orthopedic implant surface. World J Stem Cells 2020; 12:545-561. [PMID: 32843913 PMCID: PMC7415248 DOI: 10.4252/wjsc.v12.i7.545] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/28/2020] [Revised: 05/13/2020] [Accepted: 05/30/2020] [Indexed: 02/06/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) play a critical role in the osseointegration of bone and orthopedic implant. However, osseointegration between the Ti-based implants and the surrounding bone tissue must be improved due to titanium’s inherent defects. Surface modification stands out as a versatile technique to create instructive biomaterials that can actively direct stem cell fate. Here, we summarize the current approaches to promoting BMSC osteogenesis on the surface of titanium and its alloys. We will highlight the utilization of the unique properties of titanium and its alloys in promoting tissue regeneration, and discuss recent advances in understanding their role in regenerative medicine. We aim to provide a systematic and comprehensive review of approaches to promoting BMSC osteogenesis on the orthopedic implant surface.
Collapse
Affiliation(s)
- Shi-Cheng Huo
- Department of Bone and Joint Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
34
|
Redox Regulation of PPAR γ in Polarized Macrophages. PPAR Res 2020; 2020:8253831. [PMID: 32695149 PMCID: PMC7350077 DOI: 10.1155/2020/8253831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/22/2019] [Revised: 04/28/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
The peroxisome proliferator-activated receptor (PPARγ) is a central mediator of cellular lipid metabolism and immune cell responses during inflammation. This is facilitated by its role as a transcription factor as well as a DNA-independent protein interaction partner. We addressed how the cellular redox milieu in the cytosol and the nucleus of lipopolysaccharide (LPS)/interferon-γ- (IFNγ-) and interleukin-4- (IL4-) polarized macrophages (MΦ) initiates posttranslational modifications of PPARγ, that in turn alter its protein function. Using the redox-sensitive GFP2 (roGFP2), we validated oxidizing and reducing conditions following classical and alternative activation of MΦ, while the redox status of PPARγ was determined via mass spectrometry. Cysteine residues located in the zinc finger regions (amino acid fragments AA 90-115, AA 116-130, and AA 160-167) of PPARγ were highly oxidized, accompanied by phosphorylation of serine 82 in response to LPS/IFNγ, whereas IL4-stimulation provoked minor serine 82 phosphorylation and less cysteine oxidation, favoring a reductive milieu. Mutating these cysteines to alanine to mimic a redox modification decreased PPARγ-dependent reporter gene transactivation supporting a functional shift of PPARγ associated with the MΦ phenotype. These data suggest distinct mechanisms for regulating PPARγ function based on the redox state of MΦ.
Collapse
|
35
|
Esser AK, Ross MH, Fontana F, Su X, Gabay A, Fox GC, Xu Y, Xiang J, Schmieder AH, Yang X, Cui G, Scott M, Achilefu S, Chauhan J, Fletcher S, Lanza GM, Weilbaecher KN. Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer. Theranostics 2020; 10:7510-7526. [PMID: 32685002 PMCID: PMC7359087 DOI: 10.7150/thno.44523] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/03/2020] [Accepted: 05/30/2020] [Indexed: 12/27/2022] Open
Abstract
Tumor-associated macrophages (TAMs) enhance tumor growth in mice and are correlated with a worse prognosis for breast cancer patients. While early therapies sought to deplete all macrophages, current therapeutics aim to reprogram pro-tumor macrophages (M2) and preserve those necessary for anti-tumor immune responses (M1). Recent studies have shown that c-MYC (MYC) is induced in M2 macrophages in vitro and in vivo where it regulates the expression of tumor-promoting genes. In a myeloid lineage MYC KO mouse model, MYC had important roles in macrophage maturation and function leading to reduced tumor growth. We therefore hypothesized that targeted delivery of a MYC inhibitor to established M2 TAMs could reduce polarization toward an M2 phenotype in breast cancer models. Methods: In this study, we developed a MYC inhibitor prodrug (MI3-PD) for encapsulation within perfluorocarbon nanoparticles, which can deliver drugs directly to the cytosol of the target cell through a phagocytosis independent mechanism. We have previously shown that M2-like TAMs express significant levels of the vitronectin receptor, integrin β3, and in vivo targeting and therapeutic potential was evaluated using αvβ3 integrin targeted rhodamine-labeled nanoparticles (NP) or integrin αvβ3-MI3-PD nanoparticles. Results: We observed that rhodamine, delivered by αvβ3-rhodamine NP, was incorporated into M2 tumor promoting macrophages through both phagocytosis-independent and dependent mechanisms, while NP uptake in tumor suppressing M1 macrophages was almost exclusively through phagocytosis. In a mouse model of breast cancer (4T1-GFP-FL), M2-like TAMs were significantly reduced with αvβ3-MI3-PD NP treatment. To validate this effect was independent of drug delivery to tumor cells and was specific to the MYC inhibitor, mice with integrin β3 knock out tumors (PyMT-Bo1 β3KO) were treated with αvβ3-NP or αvβ3-MI3-PD NP. M2 macrophages were significantly reduced with αvβ3-MI3-PD nanoparticle therapy but not αvβ3-NP treatment. Conclusion: These data suggest αvβ3-NP-mediated drug delivery of a c-MYC inhibitor can reduce protumor M2-like macrophages while preserving antitumor M1-like macrophages in breast cancer.
Collapse
|
36
|
Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur J Pharmacol 2020; 877:173090. [PMID: 32234529 DOI: 10.1016/j.ejphar.2020.173090] [Citation(s) in RCA: 1201] [Impact Index Per Article: 240.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/04/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
Macrophages can be affected by a variety of factors to change their phenotype and thus affect their function. Activated macrophages are usually divided into two categories, M1-like macrophages and M2-like macrophages. Both M1 macrophages and M2 macrophages are closely related to inflammatory responses, among which M1 macrophages are mainly involved in pro-inflammatory responses and M2 macrophages are mainly involved in anti-inflammatory responses. Improving the inflammatory environment by modulating the activation state of macrophages is an effective method for the treatment of diseases. In this review, we analyzed the mechanism of macrophage polarization from the tumor microenvironment, nanocarriers, nuclear receptor PPARγ, phagocytosis, NF-κB signaling pathways, and other pathways.
Collapse
Affiliation(s)
- Chen Yunna
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Hu Mengru
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Wang Lei
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China; Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, Anhui, 230012, China.
| | - Chen Weidong
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China; Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, Anhui, 230012, China.
| |
Collapse
|
37
|
Regulation of the Proteolytic Activity of Cysteine Cathepsins by Oxidants. Int J Mol Sci 2020; 21:ijms21061944. [PMID: 32178437 PMCID: PMC7139492 DOI: 10.3390/ijms21061944] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/20/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
Besides their primary involvement in the recycling and degradation of proteins in endo-lysosomal compartments and also in specialized biological functions, cysteine cathepsins are pivotal proteolytic contributors of various deleterious diseases. While the molecular mechanisms of regulation via their natural inhibitors have been exhaustively studied, less is currently known about how their enzymatic activity is modulated during the redox imbalance associated with oxidative stress and their exposure resistance to oxidants. More specifically, there is only patchy information on the regulation of lung cysteine cathepsins, while the respiratory system is directly exposed to countless exogenous oxidants contained in dust, tobacco, combustion fumes, and industrial or domestic particles. Papain-like enzymes (clan CA, family C1, subfamily C1A) encompass a conserved catalytic thiolate-imidazolium pair (Cys25-His159) in their active site. Although the sulfhydryl group (with a low acidic pKa) is a potent nucleophile highly susceptible to chemical modifications, some cysteine cathepsins reveal an unanticipated resistance to oxidative stress. Besides an introductory chapter and peculiar attention to lung cysteine cathepsins, the purpose of this review is to afford a concise update of the current knowledge on molecular mechanisms associated with the regulation of cysteine cathepsins by redox balance and by oxidants (e.g., Michael acceptors, reactive oxygen, and nitrogen species).
Collapse
|
38
|
Hoy Z, Wright TW, Elliott M, Malone J, Bhagwat S, Wang J, Gigliotti F. Combination Immunotherapy with Passive Antibody and Sulfasalazine Accelerates Fungal Clearance and Promotes the Resolution of Pneumocystis-Associated Immunopathogenesis. Infect Immun 2020; 88:e00640-19. [PMID: 31611280 PMCID: PMC6977122 DOI: 10.1128/iai.00640-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/14/2019] [Accepted: 10/10/2019] [Indexed: 12/31/2022] Open
Abstract
The pulmonary immune response protects healthy individuals against Pneumocystis pneumonia (PcP). However, the immune response also drives immunopathogenesis in patients who develop severe PcP, and it is generally accepted that optimal treatment requires combination strategies that promote fungal killing and also provide effective immunomodulation. The anti-inflammatory drug sulfasalazine programs macrophages for enhanced Pneumocystis phagocytosis and also suppresses PcP-related immunopathogenesis. Anti-Pneumocystis antibody opsonizes Pneumocystis organisms for greater phagocytosis and may also mask antigens that drive immunopathogenesis. Thus, we hypothesized that combining antibody and sulfasalazine would have the dual benefit of enhancing fungal clearance while dampening immunopathogenesis and allow the rescue of severe PcP. To model a clinically relevant treatment scenario in mice, therapeutic interventions were withheld until clear symptoms of pneumonia were evident. When administered individually, both passive antibody and sulfasalazine improved pulmonary function and enhanced Pneumocystis clearance to similar degrees. However, combination treatment with antibody and sulfasalazine produced a more rapid improvement, with recovery of body weight, a dramatic improvement in pulmonary function, reduced lung inflammation, and the rapid clearance of the Pneumocystis organisms. Accelerated fungal clearance in the combination treatment group was associated with a significant increase in macrophage phagocytosis of Pneumocystis Both passive antibody and sulfasalazine resulted in the suppression of Th1 cytokines and a marked increase in lung macrophages displaying an alternatively activated phenotype, which were enhanced by combination treatment. Our data support the concept that passive antibody and sulfasalazine could be an effective and specific adjunctive therapy for PcP, with the potential to accelerate fungal clearance while attenuating PcP-associated immunopathogenesis.
Collapse
Affiliation(s)
- Zachary Hoy
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, USA
| | - Terry W Wright
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael Elliott
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jane Malone
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, USA
| | - Samir Bhagwat
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, USA
| | - Jing Wang
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, USA
| | - Francis Gigliotti
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
39
|
Schneider A, Wood HN, Geden S, Greene CJ, Yates RM, Masternak MM, Rohde KH. Growth hormone-mediated reprogramming of macrophage transcriptome and effector functions. Sci Rep 2019; 9:19348. [PMID: 31852980 PMCID: PMC6920138 DOI: 10.1038/s41598-019-56017-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2019] [Accepted: 11/29/2019] [Indexed: 01/07/2023] Open
Abstract
Macrophages are an important component of the innate immune response. Priming and activation of macrophages is stimulated by cytokines (i.e IFNγ). However, growth hormone (GH) can also stimulate macrophage activation. Based on these observations, the goal of this work was to 1) to compare the transcriptome profile of macrophages activated in vitro with GH and IFNγ, and 2) to assess the impact of GH on key macrophage functional properties like reactive oxygen species (ROS) production and phagosomal proteolysis. To assess the global transcriptional and functional impact of GH on macrophage programming, bone marrow derived macrophages were treated with GH or IFNγ. Our data strongly support a potential link between GH, which wanes with age, and impaired macrophage function. The notable overlap of GH with IFNγ-induced pathways involved in innate immune sensing of pathogens and antimicrobial responses argue for an important role for GH in macrophage priming and maturation. By using functional assays that report on biochemical activities within the lumen of phagosomes, we have also shown that GH alters physiologically relevant processes such as ROS production and proteolysis. These changes could have far reaching impacts on antimicrobial capacity, signaling, and antigen presentation.
Collapse
Affiliation(s)
- Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Hillary N Wood
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Sandra Geden
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Catherine J Greene
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA.
- Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland.
| | - Kyle H Rohde
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA.
| |
Collapse
|
40
|
Rogers KJ, Brunton B, Mallinger L, Bohan D, Sevcik KM, Chen J, Ruggio N, Maury W. IL-4/IL-13 polarization of macrophages enhances Ebola virus glycoprotein-dependent infection. PLoS Negl Trop Dis 2019; 13:e0007819. [PMID: 31825972 PMCID: PMC6905523 DOI: 10.1371/journal.pntd.0007819] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/23/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ebolavirus (EBOV) outbreaks, while sporadic, cause tremendous morbidity and mortality. No therapeutics or vaccines are currently licensed; however, a vaccine has shown promise in clinical trials. A critical step towards development of effective therapeutics is a better understanding of factors that govern host susceptibility to this pathogen. As macrophages are an important cell population targeted during virus replication, we explore the effect of cytokine polarization on macrophage infection. METHODS/MAIN FINDINGS We utilized a BSL2 EBOV model virus, infectious, recombinant vesicular stomatitis virus encoding EBOV glycoprotein (GP) (rVSV/EBOV GP) in place of its native glycoprotein. Macrophages polarized towards a M2-like anti-inflammatory state by combined IL-4 and IL-13 treatment were more susceptible to rVSV/EBOV GP, but not to wild-type VSV (rVSV/G), suggesting that EBOV GP-dependent entry events were enhanced by these cytokines. Examination of RNA expression of known surface receptors that bind and internalize filoviruses demonstrated that IL-4/IL-13 stimulated expression of the C-type lectin receptor DC-SIGN in human macrophages and addition of the competitive inhibitor mannan abrogated IL-4/IL-13 enhanced infection. Two murine DC-SIGN-like family members, SIGNR3 and SIGNR5, were upregulated by IL-4/IL-13 in murine macrophages, but only SIGNR3 enhanced virus infection in a mannan-inhibited manner, suggesting that murine SIGNR3 plays a similar role to human DC-SIGN. In vivo IL-4/IL-13 administration significantly increased virus-mediated mortality in a mouse model and transfer of ex vivo IL-4/IL-13-treated murine peritoneal macrophages into the peritoneal cavity of mice enhanced pathogenesis. SIGNIFICANCE These studies highlight the ability of macrophage polarization to influence EBOV GP-dependent virus replication in vivo and ex vivo, with M2a polarization upregulating cell surface receptor expression and thereby enhancing virus replication. Our findings provide an increased understanding of the host factors in macrophages governing susceptibility to filoviruses and identify novel murine receptors mediating EBOV entry.
Collapse
Affiliation(s)
- Kai J. Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Bethany Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Laura Mallinger
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Dana Bohan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Kristina M. Sevcik
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Jing Chen
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Natalie Ruggio
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
- * E-mail:
| |
Collapse
|
41
|
Cathepsin L secretion by host and neoplastic cells potentiates invasion. Oncotarget 2019; 10:5560-5568. [PMID: 31565189 PMCID: PMC6756864 DOI: 10.18632/oncotarget.27182] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/23/2019] [Accepted: 07/21/2019] [Indexed: 12/25/2022] Open
Abstract
The presence of macrophages within breast tumors correlates with metastatic potential. These tumor-associated macrophages often take on a pro-tumorigenic (M2-like) phenotype resulting in the secretion of growth factors and proteases, including the lysosomal protease cathepsin L. Since cathepsin L also is frequently secreted by breast cancer cells and contributes to tumor invasion, metastasis, and angiogenesis, we hypothesized that secretion of cathepsin L by both tumor-associated macrophages and neoplastic cells would facilitate the metastatic phenotype. Our results showed that the novel cathepsin L/K inhibitors KGP94 and KGP207 could inhibit in vitro M2 macrophage invasion and reduce the macrophage-stimulated invasion of 4T1 murine breast cancer cells. KGP94 and KGP207 treatment also reduced the expression of several M2-associated markers, suggesting that cathepsin L activity may be important for IL-4-driven M0 to M2 differentiation. In addition, cathepsin L shRNA knockdown studies revealed that cathepsin L from both the tumor cell and the macrophage population is important for tumor cell invasion. Thus our data suggest that tumor cells and macrophages may both contribute to the cathepsin L-driven metastatic phenotype of breast cancer. Taken together, these studies highlight the importance of cathepsin L in macrophage functions and suggest that cathepsin inhibition strategies may be therapeutically beneficial by impairing the progression of tumors with high infiltration of M2 macrophages.
Collapse
|
42
|
Establishment of BV2 microglia polarization model and its effect on Toxoplasma gondii proliferation. Res Vet Sci 2019; 125:382-389. [PMID: 31404885 DOI: 10.1016/j.rvsc.2019.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2019] [Revised: 06/08/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023]
Abstract
Toxoplasma gondii is an intracellular opportunistic, parasitic protozoan. Microglia have been classified into two main types: M1 (classically activated macrophages) and M2 (alternatively activated macrophages). BV2 cells were used in this study, together with lipopolysaccharide (LPS) and interferon (IFN)-γ or interleukin (IL)-4, which were used to induce resting microglia. Expression levels of M1/M2 markers were determined at both mRNA and protein levels, using PCR, western blot, and flow cytometry. Furthermore, cells were infected with T. gondii PLK strain, and the dynamic changes in M1/M2 marker expression levels were determined. An in vitro polarization model was successfully established. Expression of Nos2 and M1-associated markers was significantly upregulated at 12 h post-infection in BV2 cells. Further, the JAK/STAT1 and NF-κB signaling pathways were also activated following T. gondii infection. This demonstrated that T. gondii infection induces M1-type microglial polarization in vitro. The present study demonstrated that T. gondii infection affects microglial activation in vitro and elucidated the effects of activated microglia on T. gondii proliferation. This data may serve as a useful reference for more detailed elucidation of interactions between T. gondii and the innate immune system.
Collapse
|
43
|
Wang Y, Qi H, Miron RJ, Zhang Y. Modulating macrophage polarization on titanium implant surface by poly(dopamine)-assisted immobilization of IL4. Clin Implant Dent Relat Res 2019; 21:977-986. [PMID: 31373150 DOI: 10.1111/cid.12819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/05/2019] [Revised: 05/14/2019] [Accepted: 06/29/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND In the past few decades, very little research has been carried out to modify implant surfaces to improve osteointegration through the regulation of immune cells. PURPOSE The aim of this study is to investigate whether the poly(dopamine) (pDA)-assisted immobilization of IL4 on titanium surfaces could modulate the inflammatory profile of macrophages in vitro and search for the possibility of enhancing implant integration in this way. MATERIAL AND METHODS The surface composition, topography, and roughness of SLA, SLA-pDA, and SLA-pDA-IL4 discs were examined by scanning electron microscope (SEM) and energy dispersive spectrometer (EDS). Then the releasing profile of the SLA-pDA-IL4 implants was recorded for 1 week and the bioactivity of released IL4 was investigated by ELISA. Then macrophage polarization was investigated via three methods including: (a) surface marker via immunofluorescence; (b) mRNA levels of M1 and M2 polarization markers via real-time PCR, and (c) cytokine release via ELISA. RESULTS SEM and EDS revealed that pDA and IL4 were coated successfully on SLA surfaces. The ELISA results showed that IL4 remained its bioactivity on SLA surface and were immobilized on the SLA surface. The immobilization of IL4 through pDA has no significant influence on the attachment, morphology, and proliferation of macrophages, while it increased the M2/M1 proportion in human macrophages revealed by immunofluorescence. The real-time PCR and ELISA results demonstrated that SLA-pDA-IL4 surface reduced the pro-inflammatory profile compared with SLA-pDA and SLA surfaces. CONCLUSIONS The SLA-pDA-IL4 surfaces described here is able to activate adherent macrophages into M2 phenotype and reduce the release of pro-inflammatory cytokines. Immobilization of IL4 via pDA is convenient and effective, thus providing an applicable way to control macrophage behavior upon implant insertion and is anticipated to accelerating further bone integration.
Collapse
Affiliation(s)
- Yulan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Haoning Qi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Richard J Miron
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yufeng Zhang
- Department of Oral Implantology, School of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
44
|
Yao K, Zu HB. Microglial polarization: novel therapeutic mechanism against Alzheimer's disease. Inflammopharmacology 2019; 28:95-110. [PMID: 31264132 DOI: 10.1007/s10787-019-00613-5] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/16/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease that results in progressive dementia, and exhibits high disability and fatality rates. Recent evidence has demonstrated that neuroinflammation is critical in the pathophysiological processes of AD, which is characterized by the activation of microglia and astrocytes. Under different stimuli, microglia are usually activated into two polarized states, termed the classical 'M1' phenotype and the alternative 'M2' phenotype. M1 microglia are considered to promote inflammatory injury in AD; in contrast, M2 microglia exert neuroprotective effects. Imbalanced microglial polarization, in the form of excessive activation of M1 microglia and dysfunction of M2 microglia, markedly promotes the development of AD. Furthermore, an increasing number of studies have shown that the transition of microglia from the M1 to M2 phenotype could potently alleviate pathological damage in AD. Hence, this article reviews the current knowledge regarding the role of microglial M1/M2 polarization in the pathophysiology of AD. In addition, we summarize several approaches that protect against AD by altering the polarization states of microglia. This review aims to contribute to a better understanding of the pathogenesis of AD and, moreover, to explore the potential of novel drugs for the treatment of AD in the future.
Collapse
Affiliation(s)
- Kai Yao
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, China
| | - Heng-Bing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
45
|
Li X, Lin S, Chen X, Huang W, Li Q, Zhang H, Chen X, Yang S, Jin K, Shao B. The Prognostic Value of Serum Cytokines in Patients with Acute Ischemic Stroke. Aging Dis 2019; 10:544-556. [PMID: 31164999 PMCID: PMC6538221 DOI: 10.14336/ad.2018.0820] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammatory response is an unavoidable process and contributes to the destruction of cerebral tissue during the acute ischemic stroke (AIS) phase and has not been addressed fully to date. Insightful understanding of correlation of inflammatory mediators and stroke outcome may provide new biomarkers or therapeutic approaches for ischemic stroke. Here, we prospectively recruited 180 first-ever AIS patients within 72 hrs after stroke onset. We used the National Institutes of Health Stroke Scale (NIHSS) to quantify stroke severity and modified Rankin scale (mRS) to assess the 3-month outcome for AIS patients. Initially, we screened 35 cytokines, chemokines, and growth factors in sera from 75 AIS patients and control subjects. Cytokines that were of interest were further investigated in the 180 AIS patients and 14 heathy controls. We found that IL-1RA, IL-1β, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-13, IL-15, EGF, G-CSF, Flt-3L, GM-CSF and Fractalkine levels were significantly decreased in severe stroke patients. In particular, IL-1β, IL-4, IL-5, IL-7, IL-9, IL-10, IL-15, G-CSF and GM-CSF were significantly reduced in AIS patients with poor outcome, compared to those with good prognosis. IL-6 was notably higher in the poor outcome group. Only IL-9 level decreased in the large infarct volume group. After adjusting for confounders, we found that IL-5 was an independent protective factor for prognosis in AIS patients with an adjusted OR of 0.042 (P = 0.007), whereas IL-6 was an independent risk predictor for AIS patients with an adjusted OR of 1.293 (P = 0.003). Our study suggests the levels of serum cytokines are related to stroke severity, short-term prognosis and cerebral infarct volume in AIS patients.
Collapse
Affiliation(s)
- Xianmei Li
- 1Department of Rehabilitation, Wenzhou People's Hospital, Wenzhou, China
| | - Siyang Lin
- 2Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoli Chen
- 1Department of Rehabilitation, Wenzhou People's Hospital, Wenzhou, China
| | - Wensi Huang
- 3Department of Neurology, The People's Hospital of Pingyang, Wenzhou, China
| | - Qian Li
- 4Department of Neurology, Jinhua Municipal Central Hospital, Wenzhou, China
| | - Hongxia Zhang
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Xudong Chen
- 2Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shaohua Yang
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Kunlin Jin
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Bei Shao
- 2Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
Guo M, Härtlova A, Gierliński M, Prescott A, Castellvi J, Losa JH, Petersen SK, Wenzel UA, Dill BD, Emmerich CH, Ramon Y Cajal S, Russell DG, Trost M. Triggering MSR1 promotes JNK-mediated inflammation in IL-4-activated macrophages. EMBO J 2019; 38:embj.2018100299. [PMID: 31028084 PMCID: PMC6545745 DOI: 10.15252/embj.2018100299] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alternatively activated M2 macrophages play an important role in maintenance of tissue homeostasis by scavenging dead cells, cell debris and lipoprotein aggregates via phagocytosis. Using proteomics, we investigated how alternative activation, driven by IL‐4, modulated the phagosomal proteome to control macrophage function. Our data indicate that alternative activation enhances homeostatic functions such as proteolysis, lipolysis and nutrient transport. Intriguingly, we identified the enhanced recruitment of the TAK1/MKK7/JNK signalling complex to phagosomes of IL‐4‐activated macrophages. The recruitment of this signalling complex was mediated through K63 polyubiquitylation of the macrophage scavenger receptor 1 (MSR1). Triggering of MSR1 in IL‐4‐activated macrophages leads to enhanced JNK activation, thereby promoting a phenotypic switch from an anti‐inflammatory to a pro‐inflammatory state, which was abolished upon MSR1 deletion or JNK inhibition. Moreover, MSR1 K63 polyubiquitylation correlated with the activation of JNK signalling in ovarian cancer tissue from human patients, suggesting that it may be relevant for macrophage phenotypic shift in vivo. Altogether, we identified that MSR1 signals through JNK via K63 polyubiquitylation and provides evidence for the receptor's involvement in macrophage polarization.
Collapse
Affiliation(s)
- Manman Guo
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Anetta Härtlova
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK .,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marek Gierliński
- Data Analysis Group, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alan Prescott
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Josep Castellvi
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Javier Hernandez Losa
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Sine K Petersen
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf A Wenzel
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Brian D Dill
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Christoph H Emmerich
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Santiago Ramon Y Cajal
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Matthias Trost
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK .,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
47
|
The NADPH Oxidase Nox4 Controls Macrophage Polarization in an NF κB-Dependent Manner. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3264858. [PMID: 31178956 PMCID: PMC6501210 DOI: 10.1155/2019/3264858] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 12/13/2018] [Revised: 03/04/2019] [Accepted: 03/21/2019] [Indexed: 01/21/2023]
Abstract
The family of NADPH oxidases represents an important source of reactive oxygen species (ROS) within the cell. Nox4 is a special member of this family as it constitutively produces H2O2 and its loss promotes inflammation. A major cellular component of inflammation is the macrophage population, which can be divided into several subpopulations depending on their phenotype, with proinflammatory M(LPS+IFNγ) and wound-healing M(IL4+IL13) macrophages being extremes of the functional spectrum. Whether Nox4 is expressed in macrophages is discussed controversially. Here, we show that macrophages besides a high level of Nox2 indeed express Nox4. As Nox4 contributes to differentiation of many cells, we hypothesize that Nox4 plays a role in determining the polarization and the phenotype of macrophages. In bone marrow-derived monocytes, ex vivo treatment with LPS/IFNγ or IL4/IL13 results in polarization of the cells into M(LPS+IFNγ) or M(IL4+IL13) macrophages, respectively. In this ex vivo setting, Nox4 deficiency reduces M(IL4+IL13) polarization and forces M(LPS+IFNγ). Nox4-/- M(LPS+IFNγ)-polarized macrophages express more Nox2 and produce more superoxide anions than wild type M(LPS+IFNγ)-polarized macrophages. Mechanistically, Nox4 deficiency reduces STAT6 activation and promotes NFκB activity, with the latter being responsible for the higher level of Nox2 in Nox4-deficient M(LPS+IFNγ)-polarized macrophages. According to those findings, in vivo, in a murine inflammation-driven fibrosarcoma model, Nox4 deficiency forces the expression of proinflammatory genes and cytokines, accompanied by an increase in the number of proinflammatory Ly6C+ macrophages in the tumors. Collectively, the data obtained in this study suggest an anti-inflammatory role for Nox4 in macrophages. Nox4 deficiency results in less M(IL4+IL13) polarization and suppression of NFκB activity in monocytes.
Collapse
|
48
|
Skjesol A, Yurchenko M, Bösl K, Gravastrand C, Nilsen KE, Grøvdal LM, Agliano F, Patane F, Lentini G, Kim H, Teti G, Kumar Sharma A, Kandasamy RK, Sporsheim B, Starheim KK, Golenbock DT, Stenmark H, McCaffrey M, Espevik T, Husebye H. The TLR4 adaptor TRAM controls the phagocytosis of Gram-negative bacteria by interacting with the Rab11-family interacting protein 2. PLoS Pathog 2019; 15:e1007684. [PMID: 30883606 PMCID: PMC6438586 DOI: 10.1371/journal.ppat.1007684] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2018] [Revised: 03/28/2019] [Accepted: 03/07/2019] [Indexed: 02/06/2023] Open
Abstract
Phagocytosis is a complex process that eliminates microbes and is performed by specialised cells such as macrophages. Toll-like receptor 4 (TLR4) is expressed on the surface of macrophages and recognizes Gram-negative bacteria. Moreover, TLR4 has been suggested to play a role in the phagocytosis of Gram-negative bacteria, but the mechanisms remain unclear. Here we have used primary human macrophages and engineered THP-1 monocytes to show that the TLR4 sorting adapter, TRAM, is instrumental for phagocytosis of Escherichia coli as well as Staphylococcus aureus. We find that TRAM forms a complex with Rab11 family interacting protein 2 (FIP2) that is recruited to the phagocytic cups of E. coli. This promotes activation of the actin-regulatory GTPases Rac1 and Cdc42. Our results show that FIP2 guided TRAM recruitment orchestrates actin remodelling and IRF3 activation, two events that are both required for phagocytosis of Gram-negative bacteria. The Gram-negative bacteria E. coli is the most common cause of severe human pathological conditions like sepsis. Sepsis is a clinical syndrome defined by pathological changes due to systemic inflammation, resulting in paralysis of adaptive T-cell immunity with IFN-β as a critical factor. TLR4 is a key sensing receptor of lipopolysaccharide on Gram-negative bacteria. Inflammatory signalling by TLR4 is initiated by the use of alternative pair of TIR-adapters, MAL-MyD88 or TRAM-TRIF. MAL-MyD88 signaling occurs mainly from the plasma membrane giving pro-inflammatory cytokines like TNF, while TRAM-TRIF signaling occurs from vacuoles like endosomes and phagosomes to give type I interferons like IFN-β. It has previously been shown that TLR4 can control phagocytosis and phagosomal maturation through MAL-MyD88 in mice, however, these data have been disputed and published before the role of TRAM was defined in the induction of IFN-β. A role for TRAM or TRIF in phagocytosis has not previously been reported. Here we describe a novel mechanism where TRAM and its binding partner Rab11-FIP2 control phagocytosis of E. coli and regulate IRF3 dependent production of IFN-β. The significance of these results is that we define Rab11-FIP2 as a potential target for modulation of TLR4-dependent signalling in different pathological states.
Collapse
Affiliation(s)
- Astrid Skjesol
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mariia Yurchenko
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Korbinian Bösl
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Caroline Gravastrand
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kaja Elisabeth Nilsen
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lene Melsæther Grøvdal
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Federica Agliano
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Francesco Patane
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Germana Lentini
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Hera Kim
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Giuseppe Teti
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Aditya Kumar Sharma
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørnar Sporsheim
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian K. Starheim
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Douglas T. Golenbock
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Harald Stenmark
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department for Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo Norway
| | - Mary McCaffrey
- Molecular Cell Biology Laboratory, Biochemistry Department, Biosciences Institute, University College Cork, Cork, Ireland
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- The Central Norway Regional Health Authority, St. Olavs Hospital HF, Trondheim, Norway
| | - Harald Husebye
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- The Central Norway Regional Health Authority, St. Olavs Hospital HF, Trondheim, Norway
- * E-mail:
| |
Collapse
|
49
|
Baranov MV, Bianchi F, Schirmacher A, van Aart MAC, Maassen S, Muntjewerff EM, Dingjan I, Ter Beest M, Verdoes M, Keyser SGL, Bertozzi CR, Diederichsen U, van den Bogaart G. The Phosphoinositide Kinase PIKfyve Promotes Cathepsin-S-Mediated Major Histocompatibility Complex Class II Antigen Presentation. iScience 2018; 11:160-177. [PMID: 30612035 PMCID: PMC6319320 DOI: 10.1016/j.isci.2018.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2018] [Revised: 11/28/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
Antigen presentation to T cells in major histocompatibility complex class II (MHC class II) requires the conversion of early endo/phagosomes into lysosomes by a process called maturation. Maturation is driven by the phosphoinositide kinase PIKfyve. Blocking PIKfyve activity by small molecule inhibitors caused a delay in the conversion of phagosomes into lysosomes and in phagosomal acidification, whereas production of reactive oxygen species (ROS) increased. Elevated ROS resulted in reduced activity of cathepsin S and B, but not X, causing a proteolytic defect of MHC class II chaperone invariant chain Ii processing. We developed a novel universal MHC class II presentation assay based on a bio-orthogonal "clickable" antigen and showed that MHC class II presentation was disrupted by the inhibition of PIKfyve, which in turn resulted in reduced activation of CD4+ T cells. Our results demonstrate a key role of PIKfyve in the processing and presentation of antigens, which should be taken into consideration when targeting PIKfyve in autoimmune disease and cancer.
Collapse
Affiliation(s)
- Maksim V Baranov
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands
| | - Frans Bianchi
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands; Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen 9747 AG, the Netherlands
| | - Anastasiya Schirmacher
- Institute of Organic and Biomolecular Chemistry, Georg-August-University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany
| | - Melissa A C van Aart
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands
| | - Sjors Maassen
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands; Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen 9747 AG, the Netherlands
| | - Elke M Muntjewerff
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands
| | - Ilse Dingjan
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands
| | | | - Carolyn R Bertozzi
- Department of Chemistry and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Ulf Diederichsen
- Institute of Organic and Biomolecular Chemistry, Georg-August-University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525GA Nijmegen, the Netherlands; Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen 9747 AG, the Netherlands.
| |
Collapse
|
50
|
Abstract
Efferocytosis, the clearing of dead or dying cells from living tissues, is a highly programmed, vital process to maintain the healthy functioning of every organism. Disorders of efferocytosis have been linked to several chronic diseases including atherosclerosis and auto-immune diseases. To date several different assays to determine phagocytosis, using microscopy or FACS analysis with labelled targets, have been developed. However, many of these are unable to differentiate between cells that have truly been phagocytosed and those still present on the surface of the macrophages hindering exact assessment of efferocytotic capacity. We herein describe AnxA5-pHrodo and its negative control M1234-pHrodo as new molecular probes to measure in vitro as well as ex-vivo efferocytotic capacity.
Collapse
|