151
|
Zimmermann A, Hänsel R, Gemünden K, Kegel-Hübner V, Babel J, Bläker H, Matz-Soja M, Seehofer D, Damm G. In Vivo and In Vitro Characterization of Primary Human Liver Macrophages and Their Inflammatory State. Biomedicines 2021; 9:biomedicines9040406. [PMID: 33918803 PMCID: PMC8070551 DOI: 10.3390/biomedicines9040406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 11/16/2022] Open
Abstract
Liver macrophages (LMs) play a central role in acute and chronic liver pathologies. Investigation of these processes in humans as well as the development of diagnostic tools and new therapeutic strategies require in vitro models that closely resemble the in vivo situation. In our study, we sought to gain further insight into the role of LMs in different liver pathologies and into their characteristics after isolation from liver tissue. For this purpose, LMs were characterized in human liver tissue sections using immunohistochemistry and bioinformatic image analysis. Isolated cells were characterized in suspension using FACS analyses and in culture using immunofluorescence staining and laser scanning microscopy as well as functional assays. The majority of our investigated liver tissues were characterized by anti-inflammatory LMs which showed a homogeneous distribution and increased cell numbers in correlation with chronic liver injuries. In contrast, pro-inflammatory LMs appeared as temporary and locally restricted reactions. Detailed characterization of isolated macrophages revealed a complex disease dependent pattern of LMs consisting of pro- and anti-inflammatory macrophages of different origins, regulatory macrophages and monocytes. Our study showed that in most cases the macrophage pattern can be transferred in adherent cultures. The observed exceptions were restricted to LMs with pro-inflammatory characteristics.
Collapse
Affiliation(s)
- Andrea Zimmermann
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - René Hänsel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Leipzig University, 04107 Leipzig, Germany
| | - Kilian Gemünden
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Victoria Kegel-Hübner
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
| | - Jonas Babel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
| | - Hendrik Bläker
- Institute for Pathology, University Hospital, Leipzig University, 04103 Leipzig, Germany;
| | - Madlen Matz-Soja
- Rudolf-Schönheimer-Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany;
- Department for Hepatology, University Hospital, Leipzig University, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
- Correspondence: ; Tel.: +49-341-9739656
| |
Collapse
|
152
|
Hao X, Guo Y, Wang R, Yu X, He L, Shu M. Exosomes from adipose-derived mesenchymal stem cells promote survival of fat grafts by regulating macrophage polarization via let-7c. Acta Biochim Biophys Sin (Shanghai) 2021; 53:501-510. [PMID: 33704368 DOI: 10.1093/abbs/gmab018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 12/22/2022] Open
Abstract
The rate of fat graft survival is a critical aspect of successful surgery and has been a matter of concern for over 20 years. Owing to their anti-inflammatory effects and regenerative property, adipose-derived mesenchymal stem cells (AD-MSCs) have been adapted for clinical application in fat grafting, although the mechanism underlying their action remains unclear. Recently, exosomes derived from MSCs were suggested as a better alternative, and these exosomes have also been applied in diverse clinical therapies. Accumulating evidence suggests that MSCs modulate macrophage differentiation via exosome secretion, and the connection between macrophage regulation and the rate of fat graft survival has been established. Here, we identified that let-7c, the key factor in the regulatory process, is shuttled by AD-MSC-derived exosomes to downregulate the transcription factor CCAAT/enhancer-binding protein (C/EBP)-δ. The downregulation of C/EBP-δ resulted in the attenuation of pro-inflammatory M1 macrophages and elevation of anti-inflammatory M2 macrophages. These results suggest that AD-MSC-derived exosomes promote the survival of fat grafts by regulating macrophage polarization via let-7c. This is the first study to elucidate the mechanism underlying the promotion of the fat graft survival rate by AD-MSCs and to evaluate the immunotherapeutic potential of AD-MSC-derived exosomes in fat grafting.
Collapse
Affiliation(s)
- Xiaoyan Hao
- Department of Plastic, Aesthetic and Maxillofacial Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yuan Guo
- Department of Plastic, Aesthetic and Maxillofacial Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Rui Wang
- Department of Plastic, Aesthetic and Maxillofacial Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xueyuan Yu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Lin He
- Department of Plastic, Aesthetic and Maxillofacial Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Maoguo Shu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
153
|
Padberg F, Höper T, Henkel S, Driesch D, Luch A, Zellmer S. Novel indirect co-culture of immortalised hepatocytes with monocyte derived macrophages is characterised by pro-inflammatory cytokine networks. Toxicol In Vitro 2021; 73:105134. [PMID: 33662514 DOI: 10.1016/j.tiv.2021.105134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/28/2021] [Accepted: 02/25/2021] [Indexed: 10/22/2022]
Abstract
The liver is composed of different cell populations. Interactions of different cell populations can be investigated by a newly established indirect co-culture system consisting of immortalised primary human hepatocytes and human monocyte derived macrophages (MDMs). Using the time-dependent cytokine secretion of the co-cultures and single cultures, correlation networks (including the cytokines G-CSF, CCL3, MCP-1, CCL20, FGF, TGF-β1, GM-CSF, IL-8 IL-6, IL-1β, and IL-18) were generated and the correlations were validated by application of IL-8 and TNF-α-neutralising antibodies. The data reveal that IL-8 is crucial for the interaction between hepatocytes and macrophages in vitro. In addition, transcriptome analyses showed that a change in the ratio between macrophages and hepatocytes may trigger pro-inflammatory signalling pathways of the acute phase response and the complement system (release of, e.g., certain cyto- and chemokines). Using diclofenac and LPS showed that the release of cytokines is increasing with higher ratios of MDMs. Altogether, we could demonstrate that the current co-culture system is better suited to mirror the in vivo situation when compared to previously established co-culture systems composed of HepG2 and differentiated THP-1 cells. Further, our data reveal that the cytokine IL-8 is crucial for the interaction between hepatocytes and macrophages in vitro.
Collapse
Affiliation(s)
- Florian Padberg
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Max-Dohrn Straße 8-10, 10589 Berlin, Germany; Department of Biology, Chemistry, Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany.
| | - Tessa Höper
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Max-Dohrn Straße 8-10, 10589 Berlin, Germany
| | | | - Dominik Driesch
- BioControl Jena GmbH, Hans-Knöll-Straße 6, 07745 Jena, Germany
| | - Andreas Luch
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Max-Dohrn Straße 8-10, 10589 Berlin, Germany; Department of Biology, Chemistry, Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Sebastian Zellmer
- German Federal Institute for Risk Assessment (BfR), Department of Chemical and Product Safety, Max-Dohrn Straße 8-10, 10589 Berlin, Germany
| |
Collapse
|
154
|
Houston S. Siamon Gordon: A half-century fascination with macrophages. J Exp Med 2021; 217:151889. [PMID: 32579151 PMCID: PMC7336303 DOI: 10.1084/jem.20191069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Siamon Gordon is a Glaxo Wellcome Professor Emeritus of Cellular Pathology at the University of Oxford and a fellow of the Royal Society. Throughout his career, Siamon has focused on macrophages, and his work led to the identification of the pan-macrophage marker F4/80 and the description of a role for Dectin-1 in the innate recognition of β-glucans. I caught up with Siamon to discuss his career path and his thoughts on macrophages.
Collapse
|
155
|
Béraud E, Collignon A, Franceschi C, Olive D, Lombardo D, Mas E. Investigation of a new tumor-associated glycosylated antigen as target for dendritic cell vaccination in pancreatic cancer. Oncoimmunology 2021; 1:56-61. [PMID: 22720212 PMCID: PMC3376954 DOI: 10.4161/onci.1.1.18459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glycoproteins, as valuable targets for dendritic cell (DC)-vaccination in cancers, remain an open question. Glycosylated structures, which are aberrantly modified during cancerisation, impact positively or negatively on glycoprotein immunogenicity. Here is presented an oncofetal glycovariant of bile-salt-dependent-lipase, expressed on human tumoral pancreas and efficiently processed by DC's, inducing T-lymphocyte activation.
Collapse
Affiliation(s)
- Evelyne Béraud
- INSERM; Marseille, France; Aix-Marseille Univ ; Centre de Recherche en Oncologie biologique et Oncopharmacologie; Marseille, France
| | | | | | | | | | | |
Collapse
|
156
|
Mycobacterium tuberculosis Rv1515c antigen enhances survival of M. smegmatis within macrophages by disrupting the host defence. Microb Pathog 2021; 153:104778. [PMID: 33548483 DOI: 10.1016/j.micpath.2021.104778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/08/2023]
Abstract
Mycobacterium tuberculosis (Mtb) infection is the major cause of tuberculosis. Mtb regions of difference (RD) genes are vital for survival of the pathogen within hosts and for the attenuation of the bacillus Calmette-Guérin vaccine. However, the function of most RD proteins largely remains unexplored. In the present study, we focused on Rv1515c, an RD6 member from M. tuberculosis, and characterised it as a cell surface-associated protein that functions in disrupting the cytokine profile and promoting endoplasmic reticulum stress-mediated apoptosis. Rv1515c expression in M. smegmatis, a nonpathogenic species, resulted in enhanced resistance of the bacterium to various in vitro stressors (such as low pH, sodium dodecyl sulfate, oxidative pressure, and nitrogen intermediate) and its cellular survival within macrophages. Our study is the first to identify the role of Rv1515c in the physiology and pathogenesis of mycobacterium.
Collapse
|
157
|
Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2021; 1:1323-1343. [PMID: 23243596 PMCID: PMC3518505 DOI: 10.4161/onci.22009] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are constituted of a variety of cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells. On one hand, the tumor stroma exerts major pro-tumorigenic and immunosuppressive functions, reflecting the capacity of cancer cells to shape the microenvironment to satisfy their own metabolic and immunological needs. On the other hand, there is a component of tumor-infiltrating leucocytes (TILs) that has been specifically recruited in the attempt to control tumor growth. Along with the recognition of the critical role played by the immune system in oncogenesis, tumor progression and response to therapy, increasing attention has been attracted by the potential prognostic and/or predictive role of the immune infiltrate in this setting. Data from large clinical studies demonstrate indeed that a robust infiltration of neoplastic lesions by specific immune cell populations, including (but not limited to) CD8+ cytotoxic T lymphocytes, Th1 and Th17 CD4+ T cells, natural killer cells, dendritic cells, and M1 macrophages constitutes an independent prognostic indicator in several types of cancer. Conversely, high levels of intratumoral CD4+CD25+FOXP3+ regulatory T cells, Th2 CD4+ T cells, myeloid-derived suppressor cells, M2 macrophages and neutrophils have frequently been associated with dismal prognosis. So far, only a few studies have addressed the true predictive potential of TILs in cancer patients, generally comforting the notion that—at least in some clinical settings—the immune infiltrate can reliably predict if a specific patient will respond to therapy or not. In this Trial Watch, we will summarize the results of clinical trials that have evaluated/are evaluating the prognostic and predictive value of the immune infiltrate in the context of solid malignancies.
Collapse
Affiliation(s)
- Laura Senovilla
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Orsay, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Omori S, Tsugita M, Hoshikawa Y, Morita M, Ito F, Yamaguchi SI, Xie Q, Noyori O, Yamaguchi T, Takada A, Saitoh T, Toyokuni S, Akiba H, Nagata S, Kinoshita K, Nakayama M. Tim4 recognizes carbon nanotubes and mediates phagocytosis leading to granuloma formation. Cell Rep 2021; 34:108734. [PMID: 33567275 DOI: 10.1016/j.celrep.2021.108734] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/02/2020] [Accepted: 01/19/2021] [Indexed: 12/22/2022] Open
Abstract
Macrophage recognition and phagocytosis of crystals is critical for the associated fibrosis and cancer. Of note, multi-walled carbon nanotubes (MWCNTs), the highly representative products of nanotechnology, induce macrophage NLRP3 inflammasome activation and cause asbestosis-like pathogenesis. However, it remains largely unknown how macrophages efficiently recognize MWCNTs on their cell surfaces. Here, we identify by a targeted screening of phagocyte receptors the phosphatidylserine receptors T cell immunoglobulin mucin 4 (Tim4) and Tim1 as the pattern-recognition receptors for carbon crystals. Docking simulation studies reveal spatiotemporally stable interfaces between aromatic residues in the extracellular IgV domain of Tim4 and one-dimensional carbon crystals. Further, CRISPR-Cas9-mediated deletion of Tim4 and Tim1 reveals that Tim4, but not Tim1, critically contributes to the recognition of MWCNTs by peritoneal macrophages and to granuloma development in a mouse model of direct mesothelium exposure to MWCNTs. These results suggest that Tim4 recognizes MWCNTs through aromatic interactions and mediates phagocytosis leading to granulomas.
Collapse
Affiliation(s)
- Satoshi Omori
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Misato Tsugita
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Yasuto Hoshikawa
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Sendai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumiya Ito
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; CREST, Japan Science and Technology Agency (JST), Kawaguchi, Japan
| | - Shin-Ichiro Yamaguchi
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Qilin Xie
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Osamu Noyori
- Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Tomoya Yamaguchi
- Department of Cancer Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; PRESTO, JST, Kawaguchi, Japan
| | - Ayato Takada
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tatsuya Saitoh
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; CREST, Japan Science and Technology Agency (JST), Kawaguchi, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Hisaya Akiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Kengo Kinoshita
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.
| | - Masafumi Nakayama
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan; Laboratory of Immunology and Microbiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan; PRESTO, JST, Kawaguchi, Japan.
| |
Collapse
|
159
|
Sun X, Wang Z, Shao C, Yu J, Liu H, Chen H, Li L, Wang X, Ren Y, Huang X, Zhang R, Li G. Analysis of chicken macrophage functions and gene expressions following infectious bronchitis virus M41 infection. Vet Res 2021; 52:14. [PMID: 33509253 PMCID: PMC7841764 DOI: 10.1186/s13567-021-00896-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/06/2021] [Indexed: 12/29/2022] Open
Abstract
Infectious bronchitis virus (IBV) is a pathogenic coronavirus with high morbidity and mortality in chicken breeding. Macrophages with normal biofunctions are essential for host immune responses. In this study, the HD11 chicken macrophage cell line and chicken peripheral blood mononuclear cell-derived macrophages (PBMCs-Mφ) were infected with IBV at multiplicity of infection (MOI) of 10. The dynamic changes of their biofunctions, including cell viability, pathogen elimination function, phagocytic ability, and gene expressions of related proteins/mediators in innate and acquired immunity, inflammation, autophagy and apoptosis were analyzed. Results showed that IBV infection decreased chicken macrophage viability and phagocytic ability, and increased pathogen elimination function. Moreover, IBV augmented the gene expressions of most related proteins in macrophages involved in multiple host bioprocesses, and the dynamic changes of gene expressions had a close relationship with virus replication. Among them, MHCII, Fc receptor, TLR3, IFN-α, CCL4, MIF, IL-1β, IL-6, and iNOS showed significantly higher expressions in IBV-infected cells. However, TLR7, MyD88, MDA5, IFN-γ, MHCII, Fc receptor, MARCO, CD36, MIF, XCL1, CXCL12, TNF-α, iNOS, and IL-10 showed early decreased expressions. Overall, chicken macrophages play an important role in host innate and acquired immune responses to resist IBV infection, despite early damage or suppression. Moreover, the IBV-induced autophagy and apoptosis might participate in the virus-host cell interaction which is attributed to the biological process.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zheng Wang
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Changhao Shao
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jia Yu
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Haoyun Liu
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Huijie Chen
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Lu Li
- Large Scale Instrument and Equipment Sharing Service Platform, Northeast Agricultural University, Harbin, 150030, China
| | - Xiurong Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Science, Harbin, 150069, China
| | - Yudong Ren
- Department of Computer Science and Technology, College of Electrical and Information Technology, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaodan Huang
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ruili Zhang
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Guangxing Li
- Department of Basic Veterinary Science, College of Veterinary Medicine, Heilongjiang Key Laboratory for Animal and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
160
|
Zhou X, Liu X, Huang L. Macrophage-Mediated Tumor Cell Phagocytosis: Opportunity for Nanomedicine Intervention. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2006220. [PMID: 33692665 PMCID: PMC7939128 DOI: 10.1002/adfm.202006220] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Indexed: 05/05/2023]
Abstract
Macrophages are one of the most abundant non-malignant cells in the tumor microenvironment, playing critical roles in mediating tumor immunity. As important innate immune cells, macrophages possess the potential to engulf tumor cells and present tumor-specific antigens for adaptive antitumor immunity induction, leading to growing interest in targeting macrophage phagocytosis for cancer immunotherapy. Nevertheless, live tumor cells have evolved to evade phagocytosis by macrophages via the extensive expression of anti-phagocytic molecules, such as CD47. In addition, macrophages also rapidly recognize and engulf apoptotic cells (efferocytosis) in the tumor microenvironment, which inhibits inflammatory responses and facilitates immune escape of tumor cells. Thus, intervention of macrophage phagocytosis by blocking anti-phagocytic signals on live tumor cells or inhibiting tumor efferocytosis presents a promising strategy for the development of cancer immunotherapies. Here, the regulation of macrophage-mediated tumor cell phagocytosis is first summarized, followed by an overview of strategies targeting macrophage phagocytosis for the development of antitumor therapies. Given the potential off-target effects associated with the administration of traditional therapeutics (for example, monoclonal antibodies, small molecule inhibitors), we highlight the opportunity for nanomedicine in macrophage phagocytosis intervention.
Collapse
Affiliation(s)
- Xuefei Zhou
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiangrui Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
161
|
Xu N, Bai X, Liu Y, Yang Y, Tang B, Shi HN, Vallee I, Boireau P, Liu X, Liu M. The Anti-Inflammatory Immune Response in Early Trichinella spiralis Intestinal Infection Depends on Serine Protease Inhibitor-Mediated Alternative Activation of Macrophages. THE JOURNAL OF IMMUNOLOGY 2021; 206:963-977. [PMID: 33495238 DOI: 10.4049/jimmunol.2000290] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022]
Abstract
Trichinella spiralis is recognized for its ability to regulate host immune responses via excretory/secretory (ES) products. Serine protease inhibitors (serpins) play an important role in ES product-mediated immunoregulatory effects during T. spiralis infection. In this study, the immunoregulatory properties of a serpin derived from T. spiralis (Ts-serpin) were explored in BALB/c mice. The results showed that naturally occurring Ts-serpin was detected in the stichosomes of muscle larvae and adult worms. Moreover, enhancing (by injection of a soluble-expressed recombinant Ts-serpin [rTs-serpin]) or blocking (by passive immunization with anti-rTs-serpin serum) the effects of Ts-serpin changed the levels of cytokines related to inflammation induced by T. spiralis infection in the serum, mesenteric lymph nodes, and peritoneal cavity, which then led to a change in the adult worm burden in early T. spiralis infection. Moreover, the phenotypic changes in peritoneal macrophages were found to be related to Ts-serpin-mediated immunoregulation. Furthermore, a STAT6 activation mechanism independent of IL-4Rα has been found to regulate protein-mediated alternative activation of bone marrow-derived macrophages and mimic the immunoregulatory role of Ts-serpin in T. spiralis infection. Finally, the anti-inflammatory properties of rTs-serpin and bone marrow-derived macrophage alternative activation by rTs-serpin were demonstrated using a trinitrobenzene sulfonic acid-induced inflammatory bowel disease model. In summary, a protein-triggered anti-inflammatory mechanism was found to favor the survival of T. spiralis in the early stage of infection and help to elucidate the immunoregulatory effects of T. spiralis on the host immune response.
Collapse
Affiliation(s)
- Ning Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yaming Yang
- Yunnan Institute of Parasitic Diseases, Pu'er City, Yunnan 665000, China
| | - Bin Tang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hai Ning Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129; and
| | - Isabelle Vallee
- UMR de Biologie Moléculaire et d'Immunologie Parasitaires, Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail, École Nationale Vétérinaire d'Alfort, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université Paris-Est, Laboratoire de Santé Animale, 94706 Maisons-Alfort, France
| | - Pascal Boireau
- UMR de Biologie Moléculaire et d'Immunologie Parasitaires, Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail, École Nationale Vétérinaire d'Alfort, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université Paris-Est, Laboratoire de Santé Animale, 94706 Maisons-Alfort, France
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| |
Collapse
|
162
|
Gonçalves JP, de Oliveira CC, da Silva Trindade E, Riegel-Vidotti IC, Vidotti M, Simas FF. In vitro biocompatibility screening of a colloidal gum Arabic-polyaniline conducting nanocomposite. Int J Biol Macromol 2021; 173:109-117. [PMID: 33476624 DOI: 10.1016/j.ijbiomac.2021.01.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 02/05/2023]
Abstract
Although polyaniline (PANI) is a widely investigated conductive polymer for biological applications, studies addressing the biocompatibility of colloidal PANI dispersions are scarcely found in the literature of the area. Therefore, PANI nanoparticles stabilized by the natural polysaccharide gum Arabic (GA) were screened for their biocompatibility. The GA successfully stabilized the colloidal PANI-GA dispersions when exposed to a protein-rich medium, showing compatibility with the biological environment. The results obtained from a series of in vitro assays showed that, after up to 48 h of exposure to a range of PANI-GA concentrations (1-50 μg/mL), both mouse BALB/3T3 fibroblasts and RAW 264.7 macrophages showed no evidence of change in cellular proliferation, viability and metabolic activity. An increase in macrophage granularity poses as evidence of phagocytic uptake of PANI-GA, without resulting activation of this cell type. Additionally, the PANI-GA nanoparticles modulated the cell morphology changes induced on fibroblasts by GA in a concentration-dependent manner. Thus, this unprecedented biocompatibility study of PANI nanoparticles stabilized by a plant gum exudate polysaccharide showed promising results. This simple biomaterial might be further developed into colloidal formulations for biological and biomedical applications, taking advantage of its versatility, biocompatibility, and conductive properties.
Collapse
Affiliation(s)
- Jenifer Pendiuk Gonçalves
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Carolina Camargo de Oliveira
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Edvaldo da Silva Trindade
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Izabel Cristina Riegel-Vidotti
- Macromolecules and Interfaces Research Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Marcio Vidotti
- Macromolecules and Interfaces Research Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil
| | - Fernanda Fogagnoli Simas
- Laboratory of Inflammatory and Neoplastic Cells/Laboratory of Sulfated Polysaccharides Investigation, Cell Biology Department, Section of Biological Sciences, Universidade Federal do Paraná (UFPR), Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil; Macromolecules and Interfaces Research Group, Department of Chemistry, UFPR, Av Cel Francisco H dos Santos, s/n, CEP 81530-980 Curitiba, PR, Brazil.
| |
Collapse
|
163
|
Rombauts A, Abelenda-Alonso G, Cuervo G, Gudiol C, Carratalà J. Role of the inflammatory response in community-acquired pneumonia: clinical implications. Expert Rev Anti Infect Ther 2021; 20:1261-1274. [PMID: 33034228 DOI: 10.1080/14787210.2021.1834848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Despite adequate antibiotic coverage, community-acquired pneumonia (CAP) remains a leading cause of hospitalization and mortality worldwide. It induces both a local pulmonary and a systemic inflammatory response, particularly significant in severe cases. The intensity of the dysregulated host response varies from patient to patient and has a negative impact on survival and other outcomes. AREAS COVERED This comprehensive review summarizes the pathophysiological aspects of the inflammatory response in CAP, briefly discusses the usefulness of biomarkers, and assesses the clinical evidence for modulating the inflammatory pathways. We searched PubMed for the most relevant studies, reviews, and meta-analysis until August 2020. EXPERT OPINION Notable efforts have been made to identify biomarkers that can accurately differentiate between viral and bacterial etiology, and indeed, to enhance risk stratification in CAP. However, none has proven ideal and no recommended biomarker-guided algorithms exist. Biomarker signatures from proteomic and metabolomic studies could be more useful for such assessments. To date, most studies have produced contradictory results concerning the role of immunomodulatory agents (e.g. corticosteroids, macrolides, and statins) in CAP. Adequately identifying the population who may benefit most from effective modulation of the inflammatory response remains a challenge.
Collapse
Affiliation(s)
- Alexander Rombauts
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Guillermo Cuervo
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Spanish Network for Research in Infectious Disease (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain.,Institut Català d'Oncologia (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Spanish Network for Research in Infectious Disease (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
164
|
Huo J, Jia Q, Huang H, Zhang J, Li P, Dong X, Huang W. Emerging photothermal-derived multimodal synergistic therapy in combating bacterial infections. Chem Soc Rev 2021; 50:8762-8789. [PMID: 34159993 DOI: 10.1039/d1cs00074h] [Citation(s) in RCA: 305] [Impact Index Per Article: 101.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Due to the emerging bacterial resistance and the protection of tenacious biofilms, it is hard for the single antibacterial modality to achieve satisfactory therapeutic effects nowadays. In recent years, photothermal therapy (PTT)-derived multimodal synergistic treatments have received wide attention and exhibited cooperatively enhanced bactericidal activity. PTT features spatiotemporally controllable generation of hyperthermia that could eradicate bacteria without inducing resistance. The synergy of it with other treatments, such as chemotherapy, photo-dynamic/catalytic therapy (PDT/PCT), immunotherapy, and sonodynamic therapy (SDT), could lower the introduced laser density in PTT and avoid undesired overheating injury of normal tissues. Simultaneously, by heat-induced improvement of the bacterial membrane permeability, PTT is conducive for accelerated intracellular permeation of chemotherapeutic drugs as well as reactive oxygen species (ROS) generated by photosensitizers/sonosensitizers, and could promote infiltration of immune cells. Thereby, it could solve the currently existing sterilization deficiencies of other combined therapeutic modes, for example, bacterial resistance for chemotherapy, low drug permeability for PDT/PCT/SDT, adverse immunoreactions for immunotherapy, etc. Admittedly, PTT-derived synergistic treatments are becoming essential in fighting bacterial infection, especially those caused by antibiotic-resistant strains. This review firstly presents the classical and newly reported photothermal agents (PTAs) in brief. Profoundly, through the introduction of delicately designed nanocomposite platforms, we systematically discuss the versatile photothermal-derived multimodal synergistic therapy with the purpose of sterilization application. At the end, challenges to PTT-derived combinational therapy are presented and promising synergistic bactericidal prospects are anticipated.
Collapse
Affiliation(s)
- Jingjing Huo
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Qingyan Jia
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and State Key Laboratory of Organic Electronics and Information Displays (SKLOEID), Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Han Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Jing Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China and School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, China.
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and State Key Laboratory of Organic Electronics and Information Displays (SKLOEID), Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China and Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| |
Collapse
|
165
|
Shi M, Shen K, Yang B, Zhang P, Lv K, Qi H, Wang Y, Li M, Yuan Q, Zhang Y. An electroporation strategy to synthesize the membrane-coated nanoparticles for enhanced anti-inflammation therapy in bone infection. Theranostics 2021; 11:2349-2363. [PMID: 33500729 PMCID: PMC7797679 DOI: 10.7150/thno.48407] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
The cell membrane-coated nanoparticles (MNPs) showed great potential in treating infectious disease due to their superior biofunctions in improving biocompatibility of nanoparticles and neutralization of pathogen or toxins. However, bone infection is accompanied with severe inflammation and bone loss, which also requires anti-inflammatory and osteoconductive treatment. The conventional membrane coating method has to undergo ultrasonication and extrusion procedures, which reduces the functionality of cell membrane and limits the choice of nanoparticles. In this study, we proposed an electroporation-based membrane coating strategy to facilitate the synthesis of MNPs to tackle those problems. Methods: Magnetic composite nanoparticles with osteoconductive Ca3(PO4)2 and bactericidal TiO2 were assembled into macrophages through phagocytosis and then collected to expose in electric field for obtaining macrophage membrane-coating nanoparticles. By using molecular dynamics simulation and materials characterizations, the cell membrane coating efficiency was confirmed. The in vitro anti-bacterial and anti-inflammatory abilities were tested by bacteria culturing and immune cells activation. Then drug-resistant bacteria induced bone infection model was established to verify its in vivo therapeutic effects. Results: The coated membrane prepared through electroporation reserved the integrality of membrane structure and right-sidedness, with more functional proteins. Those led to the superior properties of recognition and adsorption with bacteria, toxins and inflammatory cytokines. Owing to the benefits of electroporation, the MNPs exhibited significant better antibacterial and anti-inflammatory abilities for enhancing the tissue repair process. Conclusion: This study provides a novel self-assembly cell membrane coating strategy by electroporation to construct multifunctional membrane-coating nanoparticles for bone infection treatment. This strategy not only improves the functions of coated membrane, but is also proved to be universal for varies nanoparticles or cells, indicating a great potential for future applications in the bioengineering field.
Collapse
Affiliation(s)
- Miusi Shi
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kailun Shen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bin Yang
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Key Laboratory of Catalysis and Materials Science, College of Resources and Environmental Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Peng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kangle Lv
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Key Laboratory of Catalysis and Materials Science, College of Resources and Environmental Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Haoning Qi
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yunxiao Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Mei Li
- Key Laboratory of Catalysis and Energy Materials Chemistry of Ministry of Education & Hubei Key Laboratory of Catalysis and Materials Science, College of Resources and Environmental Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Quan Yuan
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yufeng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
166
|
Rehman A, Rehman L, Ullah R, Beg MA, Khan MAH, Abidi SMA. Oxidative status and changes in the adenosine deaminase activity in experimental host infected with tropical liver fluke, Fasciola gigantica. Acta Trop 2021; 213:105753. [PMID: 33166513 DOI: 10.1016/j.actatropica.2020.105753] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/30/2022]
Abstract
Fine tuning of the metabolic, physiological and immunological cues along with interplay between the biomolecules of the host and the parasite could be responsible for the successful establishment of parasitic infections. The present investigation was aimed at evaluating the oxidative status and the level of adenosine deaminase (ADA) in the serum and liver of rabbits experimentally infected with Fasciola gigantica. A significant increase in level of ROS, MDA and 4-HNE along with a decline in the SOD, CAT, GR and GST activity was evident in rabbits experimentally infected with Fasciola gigantica. However, there was an increase in the GPX activity in the sera of infected rabbits. The increased GPX activity and decreased GR activity would have resulted in the depletion of GSH, a key non-enzymatic antioxidant, in the infected animals. The level of GSSG was also found to be higher in the sera and liver tissues of the infected rabbits along with a decline in the GSH/GSSG ratio, indicating a high level of oxidative stress in the infected animals, which also showed a significant increase in the activity of the marker enzymes of liver pathology, AST and ALT. Further, a significant inhibition of the adenosine deaminase (ADA) activity in the infected rabbits was accompanied with the reduction in the level of pro-inflammatory cytokine, IL-6 while the anti-inflammatory cytokine, IL-4 level was significantly elevated. In conclusion, the F. gigantica induced significant oxidative stress as evident from the increased levels of ROS and lipid peroxidation along with the disruption of antioxidant and detoxification cascade ultimately lead to pathogenic and inflammatory responses in the experimental host. Whereas, the altered ADA activity could modulate the host's immune responses toward Th-2 type and would facilitate the successful establishment of flukes within their host, thus indicating that ADA could be exploited as a target for the development of novel anthelmintic drugs against fasciolosis.
Collapse
Affiliation(s)
- Abdur Rehman
- Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, India.
| | - Lubna Rehman
- Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Rizwan Ullah
- Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Mirza A Beg
- Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - M A Hannan Khan
- Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, India; Department of Zoology, School of Biosciences and Biotechnology, BGSB University, Jammu and Kashmir, India
| | - S M A Abidi
- Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, India.
| |
Collapse
|
167
|
Yue Z, Nie L, Zhang P, Chen Q, Lv Q, Wang Q. Tissue-resident macrophage inflammaging aggravates homeostasis dysregulation in age-related diseases. Cell Immunol 2020; 361:104278. [PMID: 33445052 DOI: 10.1016/j.cellimm.2020.104278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/11/2020] [Accepted: 12/26/2020] [Indexed: 02/08/2023]
Abstract
Organs and tissues contain a large number of tissue-resident macrophages (MΦ-Ts), which are essential for regulating homeostasis and ensuring a rapid response to injury. However, the environmental signals shaping MΦ-Ts phenotypes and the contribution of MΦ-Ts to pathological processes are just starting to be identified. MΦ-Ts isolated from aged animals or patients show alterations in morphology and distribution, defects in phagocytosis and autophagy, and loss of tissue-repair capacity. These variations are closely associated with age-associated disorders, such as inflammaging, which is characterized by cell senescence and a senescence-associated secretory phenotype (SASP) and is frequently observed in patients afflicted with chronic diseases. It seems that the role of these resident populations cannot be avoided in the treatment of aging-related diseases. This review will describe the mechanism by which MΦ-Ts support immune homeostasis and will then discuss how MΦ-Ts facilitate inflammaging and age-related diseases, which will be helpful in the development of new interventions and treatments for chronic diseases of the elderly.
Collapse
Affiliation(s)
- Ziqi Yue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Luningxiao Nie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Peng Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qin Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qingguo Lv
- Department of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
168
|
Gong P, Wang Y, Zhang P, Yang Z, Deng W, Sun Z, Yang M, Li X, Ma G, Deng G, Dong S, Cai L, Jiang W. Immunocyte Membrane-Coated Nanoparticles for Cancer Immunotherapy. Cancers (Basel) 2020; 13:E77. [PMID: 33396603 PMCID: PMC7794746 DOI: 10.3390/cancers13010077] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022] Open
Abstract
Despite the advances in surface bioconjugation of synthetic nanoparticles for targeted drug delivery, simple biological functionalization is still insufficient to replicate complex intercellular interactions naturally. Therefore, these foreign nanoparticles are inevitably exposed to the immune system, which results in phagocytosis by the reticuloendothelial system and thus, loss of their biological significance. Immunocyte membranes play a key role in intercellular interactions, and can protect foreign nanomaterials as a natural barrier. Therefore, biomimetic nanotechnology based on cell membranes has developed rapidly in recent years. This paper summarizes the development of immunocyte membrane-coated nanoparticles in the immunotherapy of tumors. We will introduce several immunocyte membrane-coated nanocarriers and review the challenges to their large-scale preparation and application.
Collapse
Affiliation(s)
- Ping Gong
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (P.Z.); (Z.S.); (G.M.); (G.D.); (L.C.)
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Yifan Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (P.Z.); (Z.S.); (G.M.); (G.D.); (L.C.)
| | - Zhaogang Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Weiye Deng
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Zhihong Sun
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (P.Z.); (Z.S.); (G.M.); (G.D.); (L.C.)
- Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Mingming Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Xuefeng Li
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Gongcheng Ma
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (P.Z.); (Z.S.); (G.M.); (G.D.); (L.C.)
| | - Guanjun Deng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (P.Z.); (Z.S.); (G.M.); (G.D.); (L.C.)
| | - Shiyan Dong
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (P.Z.); (Z.S.); (G.M.); (G.D.); (L.C.)
| | - Wen Jiang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2280 Inwood Road, Dallas, TX 75235, USA; (Y.W.); (Z.Y.); (W.D.); (M.Y.); (X.L.); (S.D.)
| |
Collapse
|
169
|
Wang T, Zhao N, Peng L, Li Y, Huang X, Zhu J, Chen Y, Yu S, Zhao Y. DJ-1 Regulates Microglial Polarization Through P62-Mediated TRAF6/IRF5 Signaling in Cerebral Ischemia-Reperfusion. Front Cell Dev Biol 2020; 8:593890. [PMID: 33392187 PMCID: PMC7773790 DOI: 10.3389/fcell.2020.593890] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/25/2020] [Indexed: 12/28/2022] Open
Abstract
The polarization of microglia/macrophage, the resident immune cells in the brain, plays an important role in the injury and repair associated with ischemia-reperfusion (I/R). Previous studies have shown that DJ-1 has a protective effect in cerebral I/R. We found that DJ-1 regulates the polarization of microglial cells/macrophages after cerebral I/R and explored the mechanism by which DJ-1 mediates microglial/macrophage polarization in cerebral I/R. Middle cerebral artery occlusion/reperfusion (MCAO/R) and oxygen and glucose deprivation/reoxygenation (OGD/R) models were used to simulate cerebral I/R in vivo and in vitro, respectively. DJ-1 siRNA and the DJ-1-based polypeptide ND13 were used to produce an effect on DJ-1, and the P62-specific inhibitor XRK3F2 was used to block the effect of P62. Enhancing the expression of DJ-1 induced anti-inflammatory (M2) polarization of microglia/macrophage, and the expression of the anti-inflammatory factors IL-10 and IL-4 increased. Interference with DJ-1 expression induced pro-inflammatory (M1) polarization of microglia/macrophage, and the expression of the proinflammatory factors TNF-α and IL-1β increased. DJ-1 inhibited the expression of P62, impeded the interaction between P62 and TRAF6, and blocked nuclear entry of IRF5. In subsequent experiments, XRK3F2 synergistically promoted the effect of DJ-1 on microglial/macrophage polarization, further attenuating the interaction between P62 and TRAF6.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Na Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Li Peng
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yumei Li
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Xiaohuan Huang
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Jin Zhu
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yanlin Chen
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
170
|
Torretta S, Scagliola A, Ricci L, Mainini F, Di Marco S, Cuccovillo I, Kajaste-Rudnitski A, Sumpton D, Ryan KM, Cardaci S. D-mannose suppresses macrophage IL-1β production. Nat Commun 2020; 11:6343. [PMID: 33311467 PMCID: PMC7733482 DOI: 10.1038/s41467-020-20164-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 11/13/2020] [Indexed: 01/01/2023] Open
Abstract
D-mannose is a monosaccharide approximately a hundred times less abundant than glucose in human blood. Previous studies demonstrated that supraphysiological levels of D-mannose inhibit tumour growth and stimulate regulatory T cell differentiation. It is not known whether D-mannose metabolism affects the function of non-proliferative cells, such as inflammatory macrophages. Here, we show that D-mannose suppresses LPS-induced macrophage activation by impairing IL-1β production. In vivo, mannose administration improves survival in a mouse model of LPS-induced endotoxemia as well as decreases progression in a mouse model of DSS-induced colitis. Phosphomannose isomerase controls response of LPS-activated macrophages to D-mannose, which impairs glucose metabolism by raising intracellular mannose-6-phosphate levels. Such alterations result in the suppression of succinate-mediated HIF-1α activation, imposing a consequent reduction of LPS-induced Il1b expression. Disclosing an unrecognized metabolic hijack of macrophage activation, our study points towards safe D-mannose utilization as an effective intervention against inflammatory conditions.
Collapse
Affiliation(s)
- Simone Torretta
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Alessandra Scagliola
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Luisa Ricci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Francesco Mainini
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Sabrina Di Marco
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | | | | | - Simone Cardaci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
171
|
siRNA delivery to macrophages using aspherical, nanostructured microparticles as delivery system for pulmonary administration. Eur J Pharm Biopharm 2020; 158:284-293. [PMID: 33285246 DOI: 10.1016/j.ejpb.2020.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/16/2020] [Accepted: 11/28/2020] [Indexed: 12/13/2022]
Abstract
The delivery of oligonucleotides such as siRNA to the lung is a major challenge, as this group of drugs has difficulties to overcome biological barriers due to its polyanionic character and the associated hydrophilic properties, resulting in inefficient delivery. Especially in diseases such as asthma, chronic obstructive pulmonary disease and cystic fibrosis, where increased proinflammation is present, a targeted RNA therapy is desirable due to the high potency of these oligonucleotides. To address these problems and to ensure efficient uptake of siRNA in macrophages, a microparticulate, cylindrical delivery system was developed. In the first step, this particle system was tested for its aerodynamic characteristics to evaluate the aerodynamic properties to optimize lung deposition. The mass median aerodynamic diameter of 2.52 ± 0.23 µm, indicates that the desired target should be reached. The inhibition of TNF-α release, as one of the main mediators of proinflammatory reactions, was investigated. We could show that our carrier system can be loaded with siRNA against TNF-α. Gel electrophoreses allowed to demonstrate that the load can be incorporated and released without being degraded. The delivery system was found to transport a mass fraction of 0.371% [%w/w] as determined by inductively coupled plasma mass spectroscopy. When investigating the release kinetics, the results showed that several days are necessary to release a major amount of the siRNA indicating a sustained release. The cylindrical microparticles with an aspect ratio of 3.3 (ratio of length divided by width) were then tested in vitro successfully reducing TNF-α release from human macrophages significantly by more than 30%. The developed formulation presents a possible oligonucleotide delivery system allowing due to its internal structure to load and protect siRNA.
Collapse
|
172
|
Irie H, Morita K, Koizumi M, Mochizuki S. Immune Responses and Antitumor Effect through Delivering to Antigen Presenting Cells by Optimized Conjugates Consisting of CpG-DNA and Antigenic Peptide. Bioconjug Chem 2020; 31:2585-2595. [PMID: 33151667 DOI: 10.1021/acs.bioconjchem.0c00523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunotherapy using antigen-specific cytotoxic T lymphocytes (CTLs) has become one of the most attractive strategies for cancer treatment. For the induction of antigen-specific CTLs in vivo, the co-delivery of CpG-DNAs and antigens to the same antigen-presenting cells (APCs) is a promising strategy. In this study, we prepared conjugates consisting of 40mer of CpG-DNA (CpG40) and antigenic peptide (OVA257-264), which have the following distinctive features: (1) multiple CpG motifs in a molecule; (2) cleavage in the cytosol because of the disulfide bonding via cysteine residue between peptide and CpG-DNA; (3) conjugation designed to induce antigen presentation on MHC class I molecules. Immunization with the conjugate CpG40-C-OVA257-264 at the mouse tail base induced strong CTL activity at a very low peptide dose of 20 ng/head. It was found that the conjugates were internalized into C-type mannose receptor 1 (MRC1)-expressing cells in inguinal lymph nodes, indicating that the CpG portion in the conjugate acts as not only an adjuvant for the activation of TLR9 but also a carrier to APCs expressing MRC1. In a tumor-bearing mice model, mice immunized with CpG40-C-OVA257-264 conjugates exhibited long delays in tumor growth compared with those treated with PBS, OVA257-264 alone, or a mixture of CpG40 and OVA257-264. Therefore, CpG-C-peptide conjugates could be a new and effective platform for peptide vaccine for the treatment of cancers and infectious diseases.
Collapse
Affiliation(s)
- Hitomi Irie
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| | - Koji Morita
- Modality Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Makoto Koizumi
- Modality Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shinichi Mochizuki
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| |
Collapse
|
173
|
Rogeri PS, Gasparini SO, Martins GL, Costa LKF, Araujo CC, Lugaresi R, Kopfler M, Lancha AH. Crosstalk Between Skeletal Muscle and Immune System: Which Roles Do IL-6 and Glutamine Play? Front Physiol 2020; 11:582258. [PMID: 33178046 PMCID: PMC7596683 DOI: 10.3389/fphys.2020.582258] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
The skeletal muscle was always seen from biomechanical and biochemical views. It is well-established that an active muscle brings many benefits for different body organs and tissues, including the immune system. Since the 1970s, many studies have shown the importance of regular exercise and physical activity in increasing the body's ability to fight opportunist infections, as well as a strategy to fight established diseases. This interaction was mainly attributed to the glutamine, a non-essential amino acid produced by the active skeletal muscle and primarily consumed by rapidly dividing cells, including lymphocytes and monocytes/macrophages, as their main source of energy. Therefore, these cells' function would be significantly improved by the presence of a bigger glutamine pool, facilitating phagocytosis, antigen-presentation, proliferative capacity, cytokine synthesis and release, among other functions. Despite its importance, glutamine is not the only molecule to connect these two tissues. The presence of cytokines is crucial for a proper immune system function. Many of them have well-established pro-inflammatory properties, while others are known for their anti-inflammatory role. Interleukin-6 (IL-6), however, has been in the center of many scientific discussions since it can act as pro- and anti-inflammatory cytokine depending on the tissue that releases it. Skeletal muscle is an essential source of IL-6 with anti-inflammatory properties, regulating the function of the immune cells after tissue injury and the healing process. Therefore, this review aims to discuss further the role of these four components (glutamine, and interleukin-6, and its interface with monocytes/macrophages, and lymphocytes) on the communication between the skeletal muscle and the immune system.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Antonio H. Lancha
- Laboratório de Nutrição e Metabolismo, Escola de Educação Física e Esporte da Universidade de São Paulo, EEFE-USP, São Paulo, Brazil
| |
Collapse
|
174
|
Sanches RCO, Souza C, Oliveira SC. Schistosoma antigens as activators of inflammasome pathway: from an unexpected stimulus to an intriguing role. Microbes Infect 2020; 22:534-539. [PMID: 32841730 DOI: 10.1016/j.micinf.2020.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 01/14/2023]
Abstract
Parasites of the genus Schistosoma are organisms capable of living for decades within the definitive host. They interfere with the immune response by interacting with host's receptors. In this review, we discuss from the first reports to the most recent discoveries regarding the ability of Schistosoma antigens in triggering intracellular receptors and inducing inflammasome activation.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cláudia Souza
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270-901, Salvador, Brazil.
| |
Collapse
|
175
|
Belhadj Z, He B, Deng H, Song S, Zhang H, Wang X, Dai W, Zhang Q. A combined "eat me/don't eat me" strategy based on extracellular vesicles for anticancer nanomedicine. J Extracell Vesicles 2020; 9:1806444. [PMID: 32944191 PMCID: PMC7480498 DOI: 10.1080/20013078.2020.1806444] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/07/2020] [Accepted: 07/05/2020] [Indexed: 12/19/2022] Open
Abstract
A long-term and huge challenge in nanomedicine is the substantial uptake and rapid clearance mediated by the mononuclear phagocyte system (MPS), which enormously hinders the development of nanodrugs. Inspired by the natural merits of extracellular vesicles, we therefore developed a combined "eat me/don't eat me" strategy in an effort to achieve MPS escape and efficient drug delivery. Methodologically, cationized mannan-modified extracellular vesicles derived from DC2.4 cells were administered to saturate the MPS (eat me strategy). Then, nanocarriers fused to CD47-enriched exosomes originated from human serum were administered to evade phagocytosis by MPS (don't eat me strategy). The nanocarriers were also loaded with antitumor drugs and functionalized with a novel homing peptide to promote the tumour tissue accumulation and cancer cell uptake (eat me strategy). The concept was proven in vitro as evidenced by the reduced endocytosis of macrophages and enhanced uptake by tumour cells, whereas prolonged circulation time and increased tumour accumulation were demonstrated in vivo. Specially, the strategy induced a 123.53% increase in tumour distribution compared to conventional nanocarrier. The study both shed light on the challenge overcoming of phagocytic evasion and provided a strategy for significantly improving therapeutic outcomes, potentially permitting active drug delivery via targeted nanomedicines.
Collapse
Affiliation(s)
- Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hailiang Deng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Siyang Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
176
|
Kumar V. Pulmonary Innate Immune Response Determines the Outcome of Inflammation During Pneumonia and Sepsis-Associated Acute Lung Injury. Front Immunol 2020; 11:1722. [PMID: 32849610 PMCID: PMC7417316 DOI: 10.3389/fimmu.2020.01722] [Citation(s) in RCA: 318] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
The lung is a primary organ for gas exchange in mammals that represents the largest epithelial surface in direct contact with the external environment. It also serves as a crucial immune organ, which harbors both innate and adaptive immune cells to induce a potent immune response. Due to its direct contact with the outer environment, the lung serves as a primary target organ for many airborne pathogens, toxicants (aerosols), and allergens causing pneumonia, acute respiratory distress syndrome (ARDS), and acute lung injury or inflammation (ALI). The current review describes the immunological mechanisms responsible for bacterial pneumonia and sepsis-induced ALI. It highlights the immunological differences for the severity of bacterial sepsis-induced ALI as compared to the pneumonia-associated ALI. The immune-based differences between the Gram-positive and Gram-negative bacteria-induced pneumonia show different mechanisms to induce ALI. The role of pulmonary epithelial cells (PECs), alveolar macrophages (AMs), innate lymphoid cells (ILCs), and different pattern-recognition receptors (PRRs, including Toll-like receptors (TLRs) and inflammasome proteins) in neutrophil infiltration and ALI induction have been described during pneumonia and sepsis-induced ALI. Also, the resolution of inflammation is frequently observed during ALI associated with pneumonia, whereas sepsis-associated ALI lacks it. Hence, the review mainly describes the different immune mechanisms responsible for pneumonia and sepsis-induced ALI. The differences in immune response depending on the causal pathogen (Gram-positive or Gram-negative bacteria) associated pneumonia or sepsis-induced ALI should be taken in mind specific immune-based therapeutics.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, Faculty of Medicine, School of Clinical Medicine, Mater Research, University of Queensland, Brisbane, QLD, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
177
|
Mondragon-Shem K, Wongtrakul-Kish K, Kozak RP, Yan S, Wilson IBH, Paschinger K, Rogers ME, Spencer DIR, Acosta-Serrano A. Insights into the salivary N-glycome of Lutzomyia longipalpis, vector of visceral leishmaniasis. Sci Rep 2020; 10:12903. [PMID: 32737362 PMCID: PMC7395719 DOI: 10.1038/s41598-020-69753-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/14/2020] [Indexed: 01/08/2023] Open
Abstract
During Leishmania transmission sand flies inoculate parasites and saliva into the skin of vertebrates. Saliva has anti-haemostatic and anti-inflammatory activities that evolved to facilitate bloodfeeding, but also modulate the host's immune responses. Sand fly salivary proteins have been extensively studied, but the nature and biological roles of protein-linked glycans remain overlooked. Here, we characterised the profile of N-glycans from the salivary glycoproteins of Lutzomyia longipalpis, vector of visceral leishmaniasis in the Americas. In silico predictions suggest half of Lu. longipalpis salivary proteins may be N-glycosylated. SDS-PAGE coupled to LC-MS analysis of sand fly saliva, before and after enzymatic deglycosylation, revealed several candidate glycoproteins. To determine the diversity of N-glycan structures in sand fly saliva, enzymatically released sugars were fluorescently tagged and analysed by HPLC, combined with highly sensitive LC-MS/MS, MALDI-TOF-MS, and exoglycosidase treatments. We found that the N-glycan composition of Lu. longipalpis saliva mostly consists of oligomannose sugars, with Man5GlcNAc2 being the most abundant, and a few hybrid-type species. Interestingly, some glycans appear modified with a group of 144 Da, whose identity has yet to be confirmed. Our work presents the first detailed structural analysis of sand fly salivary glycans.
Collapse
Affiliation(s)
- Karina Mondragon-Shem
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Katherine Wongtrakul-Kish
- Ludger Ltd., Culham Science Centre, Oxfordshire, OX14 3EB, UK
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
| | | | - Shi Yan
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine, A-1210, Vienna, Austria
| | - Iain B H Wilson
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Katharina Paschinger
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Matthew E Rogers
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | | | - Alvaro Acosta-Serrano
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK.
| |
Collapse
|
178
|
Lubow J, Collins KL. Vpr Is a VIP: HIV Vpr and Infected Macrophages Promote Viral Pathogenesis. Viruses 2020; 12:E809. [PMID: 32726944 PMCID: PMC7472745 DOI: 10.3390/v12080809] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
HIV infects several cell types in the body, including CD4+ T cells and macrophages. Here we review the role of macrophages in HIV infection and describe complex interactions between viral proteins and host defenses in these cells. Macrophages exist in many forms throughout the body, where they play numerous roles in healthy and diseased states. They express pattern-recognition receptors (PRRs) that bind viral, bacterial, fungal, and parasitic pathogens, making them both a key player in innate immunity and a potential target of infection by pathogens, including HIV. Among these PRRs is mannose receptor, a macrophage-specific protein that binds oligosaccharides, restricts HIV replication, and is downregulated by the HIV accessory protein Vpr. Vpr significantly enhances infection in vivo, but the mechanism by which this occurs is controversial. It is well established that Vpr alters the expression of numerous host proteins by using its co-factor DCAF1, a component of the DCAF1-DDB1-CUL4 ubiquitin ligase complex. The host proteins targeted by Vpr and their role in viral replication are described in detail. We also discuss the structure and function of the viral protein Env, which is stabilized by Vpr in macrophages. Overall, this literature review provides an updated understanding of the contributions of macrophages and Vpr to HIV pathogenesis.
Collapse
Affiliation(s)
- Jay Lubow
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Kathleen L. Collins
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
179
|
Donalisio M, Cirrincione S, Rittà M, Lamberti C, Civra A, Francese R, Tonetto P, Sottemano S, Manfredi M, Lorenzato A, Moro GE, Giribaldi M, Cavallarin L, Giuffrida MG, Bertino E, Coscia A, Lembo D. Extracellular Vesicles in Human Preterm Colostrum Inhibit Infection by Human Cytomegalovirus In Vitro. Microorganisms 2020; 8:microorganisms8071087. [PMID: 32708203 PMCID: PMC7409124 DOI: 10.3390/microorganisms8071087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 07/17/2020] [Indexed: 11/16/2022] Open
Abstract
Breast milk is a complex biofluid that nourishes infants, supports their growth and protects them from diseases. However, at the same time, breastfeeding is a transmission route for human cytomegalovirus (HCMV), with preterm infants being at a great risk of congenital disease. The discrepancy between high HCMV transmission rates and the few reported cases of infants with severe clinical illness is likely due to the protective effect of breast milk. The aim of this study was to investigate the anti-HCMV activity of human preterm colostrum and clarify the role of colostrum-derived extracellular vesicles (EVs). Preterm colostrum samples were collected and the EVs were purified and characterized. The in vitro anti-HCMV activity of both colostrum and EVs was tested against HCMV, and the viral replication step inhibited by colostrum-purified EVs was examined. We investigated the putative role EV surface proteins play in impairing HCMV infection using shaving experiments and proteomic analysis. The obtained results confirmed the antiviral action of colostrum against HCMV and demonstrated a remarkable antiviral activity of colostrum-derived EVs. Furthermore, we demonstrated that EVs impair the attachment of HCMV to cells, with EV surface proteins playing a role in mediating this action. These findings contribute to clarifying the mechanisms that underlie the protective role of human colostrum against HCMV infection.
Collapse
Affiliation(s)
- Manuela Donalisio
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
- Correspondence: (M.D.); (A.C.); Tel.: +39-011-6705427 (M.D.); +39-011-3134437 (A.C.)
| | - Simona Cirrincione
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Massimo Rittà
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| | - Cristina Lamberti
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Andrea Civra
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| | - Rachele Francese
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| | - Paola Tonetto
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
| | - Stefano Sottemano
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy;
| | - Annalisa Lorenzato
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo (TO), Italy;
- Department of Oncology, University of Turin, 10060 Candiolo (TO), Italy
| | - Guido E. Moro
- Italian Association of Human Milk Banks, 20126 Milano, Italy;
| | - Marzia Giribaldi
- Research Centre for Engineering and Agro-food Processing (CREA), 10135 Torino, Italy;
| | - Laura Cavallarin
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Maria Gabriella Giuffrida
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Enrico Bertino
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
| | - Alessandra Coscia
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
- Correspondence: (M.D.); (A.C.); Tel.: +39-011-6705427 (M.D.); +39-011-3134437 (A.C.)
| | - David Lembo
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| |
Collapse
|
180
|
High-resolution and differential analysis of rat microglial markers in traumatic brain injury: conventional flow cytometric and bioinformatics analysis. Sci Rep 2020; 10:11991. [PMID: 32686718 PMCID: PMC7371644 DOI: 10.1038/s41598-020-68770-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 06/30/2020] [Indexed: 12/23/2022] Open
Abstract
Traumatic brain injury (TBI) results in a cascade of cellular responses, which produce neuroinflammation, partly due to microglial activation. Transforming from surveying to primed phenotypes, microglia undergo considerable molecular changes. However, specific microglial profiles in rat remain elusive due to tedious methodology and limited availability of reagents. Here, we present a flow cytometry-based analysis of rat microglia 24 h after TBI using the controlled cortical impact model, validated with a bioinformatics approach. Isolated microglia are analyzed for morphological changes and their expression of activation markers using flow cytometry, traditional gating-based analysis methods and support the data by employing bioinformatics statistical tools. We use CD45, CD11b/c, and p2y12 receptor to identify microglia and evaluate their activation state using CD32, CD86, RT1B, CD200R, and CD163. The results from logic-gated flow cytometry analysis was validated with bioinformatics-based analysis and machine learning algorithms to detect quantitative changes in morphology and marker expression in microglia due to activation following TBI.
Collapse
|
181
|
Modulation of microglial phenotypes improves sepsis-induced hippocampus-dependent cognitive impairments and decreases brain inflammation in an animal model of sepsis. Clin Sci (Lond) 2020; 134:765-776. [PMID: 32219335 DOI: 10.1042/cs20191322] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/17/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND In order to modulate microglial phenotypes in vivo, M1 microglia were depleted by administration of gadolinium chloride and the expression of M2 microglia was induced by IL-4 administration in an animal model of sepsis to better characterize the role of microglial phenotypes in sepsis-induced brain dysfunction. METHODS Wistar rats were submitted to sham or cecal ligation and perforation (CLP) and treated with IL-4 or GdCl3. Animals were submitted to behavioral tests 10 days after surgery. In a separated cohort of animals at 24 h, 3 and 10 days after surgery, hippocampus was removed and cytokine levels, M1/M2 markers and CKIP-1 levels were determined. RESULTS Modulation of microglia by IL-4 and GdCl3 was associated with an improvement in long-term cognitive impairment. When treated with IL-4 and GdCl3, the reduction of pro-inflammatory cytokines was apparent in almost all analyzed time points. Additionally, CD11b and iNOS were increased after CLP at all time points, and both IL-4 and GdCl3 treatments were able to reverse this. There was a significant decrease in CD11b gene expression in the CLP+GdCl3 group. IL-4 treatment was able to decrease iNOS expression after sepsis. Furthermore, there was an increase of CKIP-1 in the hippocampus of GdCl3 and IL-4 treated animals 10 days after CLP induction. CONCLUSIONS GdCl3 and IL-4 are able to manipulate microglial phenotype in an animal models of sepsis, by increasing the polarization toward an M2 phenotype IL-4 and GdCl3 treatment was associated with decreased brain inflammation and functional recovery.
Collapse
|
182
|
Shin JY, Lee PH. Mesenchymal stem cells modulate misfolded α-synuclein in parkinsonian disorders: A multitarget disease-modifying strategy. Stem Cell Res 2020; 47:101908. [PMID: 32683319 DOI: 10.1016/j.scr.2020.101908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/22/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
α-Synuclein (α-Syn) aggregates, the major toxic component of Lewy bodies, are proteinaceous fibrillar cytoplasmic inclusions observed in α-synucleinopathies, such as Parkinson's disease (PD), multiple system atrophy, and dementia with Lewy bodies. Overexpression of α-syn induce neuronal loss and α-syn aggregation in PD animals. Recent studies show that α-syn is released by exocytosis and can be transmitted between brain areas through cell-to-cell propagation. Moreover, aggregates of extracellular α-syn can induce neuroinflammation-mediated neurotoxic signaling through microglial activation and release of pro-inflammatory factors. Thus, modulation of α-syn might be a potential therapeutic strategy for modifying disease progression of α-synucleinopathies. Our previous studies have revealed that MSCs have potent neuroprotective effects in PD animal through modulation of neuroinflammation, inhibition of cell death, and promotion of neurogenesis. Here, we provide further evidence that MSCs have the potential to modulate α-syn-related microenvironments via enhancement of autophagy, proteolysis of α-syn aggregates, inhibition of cell-to-cell transmission of α-syn, stabilization of axonal transport, and phagocytic clearance of α-syn by microglial M2 polarization. With advantages in clinical applications, these data suggests that the use of MSCs as pharmacological modulators of α-syn propagation would be an effective therapeutic approach in PD.
Collapse
Affiliation(s)
- Jin Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University, Seoul, South Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University, Seoul, South Korea.
| |
Collapse
|
183
|
Arnaud-Sampaio VF, Rabelo ILA, Bento CA, Glaser T, Bezerra J, Coutinho-Silva R, Ulrich H, Lameu C. Using Cytometry for Investigation of Purinergic Signaling in Tumor-Associated Macrophages. Cytometry A 2020; 97:1109-1126. [PMID: 32633884 DOI: 10.1002/cyto.a.24035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/25/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023]
Abstract
Tumor-associated macrophages are widely recognized for their importance in guiding pro-tumoral or antitumoral responses. Mediating inflammation or immunosuppression, these cells support many key events in cancer progression: cell growth, chemotaxis, invasiveness, angiogenesis and cell death. The communication between cells in the tumor microenvironment strongly relies on the secretion and recognition of several molecules, including damage-associated molecular patterns (DAMPs), such as adenosine triphosphate (ATP). Extracellular ATP (eATP) and its degradation products act as signaling molecules and have extensively described roles in immune response and inflammation, as well as in cancer biology. These multiple functions highlight the purinergic system as a promising target to investigate the interplay between macrophages and cancer cells. Here, we reviewed purinergic signaling pathways connecting cancer cells and macrophages, a yet poorly investigated field. Finally, we present a new tool for the characterization of macrophage phenotype within the tumor. Image cytometry emerges as a cutting-edge tool, capable of providing a broad set of information on cell morphology, expression of specific markers, and its cellular or subcellular localization, preserving cell-cell interactions within the tumor section and providing high statistical strength in small-sized experiments. Thus, image cytometry allows deeper investigation of tumor heterogeneity and interactions between these cells. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
| | - Izadora L A Rabelo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Carolina A Bento
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Jean Bezerra
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Claudiana Lameu
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
184
|
de la Rosa G, Gómez AI, Baños MC, Pelegrín P. Signaling Through Purinergic Receptor P2Y 2 Enhances Macrophage IL-1β Production. Int J Mol Sci 2020; 21:ijms21134686. [PMID: 32630144 PMCID: PMC7370188 DOI: 10.3390/ijms21134686] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/19/2020] [Accepted: 06/27/2020] [Indexed: 12/14/2022] Open
Abstract
The release of nucleotides during necrosis or apoptosis has been described to have both proinflammatory and anti-inflammatory effect on the surrounding cells. Here we describe how low concentrations of UTP and ATP applied during macrophage priming enhance IL-1β production when subsequently the NLRP3 inflammasome is activated in murine resident peritoneal macrophages. Deficiency or pharmacological inhibition of the purinergic receptor P2Y2 reverted the increase of IL-1β release induced by nucleotides. IL-1β increase was found dependent on the expression of Il1b gene and probably involving JNK activity. On the contrary, nucleotides decreased the production of a different proinflammatory cytokines such as TNF-α. These results suggest that nucleotides could shape the response of macrophages to obtain a unique proinflammatory signature that might be relevant in unrevealing specific inflammatory conditions.
Collapse
|
185
|
Qu Z, Guo Y, Li M, Cao C, Wang J, Gao M. Recombinant ferritin nanoparticles can induce dendritic cell maturation through TLR4/NF-κB pathway. Biotechnol Lett 2020; 42:2489-2500. [PMID: 32567013 DOI: 10.1007/s10529-020-02944-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/14/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Immune response initiation and regulation require activation of dendritic cells (DCs). However, the mechanism by which ferritin, a carrier for immunogen, induces DCs maturation remains unclear. RESULTS Recombinant ferritin nanoparticle (RFNp), were prepared through the baculovirus expression vector system, formed spherical and hollow cage-liked proteins with a diameter of approximately 12.17 ± 0.87 nm. They induced bone marrow-derived DC (BMDC) maturation via surface molecules up-regulation of (MHC II, CD80, CD86 and CD40), increased pro-inflammatory cytokines production (IL-6, IL-12, TNF-α, and IFN-γ), and decreased antigen capturing capacity. They positively regulated IκBα and NF-κB (p65) phosphorylation, and facilitate NF-κB (p65) translocation into mature BMDCs nuclei. Following pre-treatment of RFNp-treated BMDCs with TLR4 and NF-κB (p65) inhibitors, respectively, surface molecule expression, pro-inflammatory cytokines production, and IκBα and NF-κB (p65) activities were suppressed. RFNp-treated BMDCs can also facilitate T-cell proliferation and differentiation into Th1 and Th2. CONCLUSION RFNps induced DCs maturation lends the potential application of RFNps as carrier platforms in DC-based vaccine.
Collapse
Affiliation(s)
- Zhehui Qu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, Heilongjiang, People's Republic of China
- College of Animal Science and Veterinary Medicine, Xinyang Agriculture and Forestry University, Xinyang, 464000, Henan, People's Republic of China
| | - Yongli Guo
- Animal Disease Prevention and Control Center of Heilongjiang Province, Harbin, 150069, Heilongjiang, People's Republic of China
| | - Mingzhu Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, Heilongjiang, People's Republic of China
| | - Chong Cao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, Heilongjiang, People's Republic of China
| | - Junwei Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, Heilongjiang, People's Republic of China.
| | - Mingchun Gao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, Heilongjiang, People's Republic of China.
| |
Collapse
|
186
|
Carrageenan from red algae: an application in the development of inhalable tuberculosis therapy targeting the macrophages. Drug Deliv Transl Res 2020; 10:1675-1687. [DOI: 10.1007/s13346-020-00799-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
187
|
Fraunberger EA, DeJesus P, Zanier ER, Shutt TE, Esser MJ. Acute and Persistent Alterations of Cerebellar Inflammatory Networks and Glial Activation in a Rat Model of Pediatric Mild Traumatic Brain Injury. J Neurotrauma 2020; 37:1315-1330. [DOI: 10.1089/neu.2019.6714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Erik A. Fraunberger
- Hotchkiss Brain Institute, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Pauline DeJesus
- Hotchkiss Brain Institute, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Elisa R. Zanier
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Timothy E. Shutt
- Hotchkiss Brain Institute, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
- Department of Medical Genetics, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Michael J. Esser
- Hotchkiss Brain Institute, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
- Cumming School of Medicine, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
188
|
Zhao K, Dong R, Yu Y, Tu C, Li Y, Cui Y, Bao L, Ling C. Cigarette smoke-induced lung inflammation in COPD mediated via CCR1/JAK/STAT /NF-κB pathway. Aging (Albany NY) 2020; 12:9125-9138. [PMID: 32463796 PMCID: PMC7288948 DOI: 10.18632/aging.103180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/16/2020] [Indexed: 01/24/2023]
Abstract
Inflammation is an important cause of chronic obstructive pulmonary disease (COPD) and its acute exacerbation. However, the critical role of C-C chemokine receptor (CCR)1 in progression of cigarette smoke-induced chronic inflammation remains unclear. We studied CCR1 expression using immunohistochemistry, immunofluorescence, and real-time polymerase chain reaction (RT-PCR) in COPD patients and controls. Cytokine levels in peripheral blood were measured by enzyme-linked immunosorbent assay (ELISA). In vitro, we investigated Janus kinase/signal transducers and activators of transcription (JAK/STAT)/nuclear factor-κB (NF-κB) signaling in cigarette smoke extract-induced or CCR1 deficiency/overexpressed mouse macrophage cell line MH-S by RT-PCR and western blot, and measured the cytokine levels in the supernatant with ELISA. We found that CCR1 expression was upregulated in COPD patients and there was a negative correlation between CCR1 mRNA levels and predicted % forced expiratory volume in 1 min. Inflammatory cytokine levels in the peripheral blood were higher in COPD patients than controls, and these were positively correlated with CCR1 levels. CCR1 was shown to play a critical role in regulating smoke-induced inflammation via JAK/STAT3/NF-κB signaling in vitro. CCR1 may play a critical role in airway inflammation in COPD. Additionally, understanding the molecular mechanism may help develop novel methods for the treatment of COPD.
Collapse
Affiliation(s)
- Kaishun Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215000, China
- Department of Respiratory Medicine, Jiading Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201800, China
| | - Ran Dong
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Yanfang Yu
- Department of Respiratory Medicine, Jiading Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201800, China
| | - Chunlin Tu
- Department of Respiratory Medicine, Jiading Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201800, China
| | - Ying Li
- Department of Respiratory Medicine, Jiading Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201800, China
| | - YuJuan Cui
- Department of Respiratory Medicine, Jiading Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201800, China
| | - Lei Bao
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215000, China
| | - Chunhua Ling
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215000, China
| |
Collapse
|
189
|
Patial S, Saini Y. Lung macrophages: current understanding of their roles in Ozone-induced lung diseases. Crit Rev Toxicol 2020; 50:310-323. [PMID: 32458707 DOI: 10.1080/10408444.2020.1762537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Through the National Ambient Air Quality Standards (NAAQS), the Clean Air Act of the United States outlines acceptable levels of six different air pollutants considered harmful to humans and the environment. Included in this list is ozone (O3), a highly reactive oxidant gas, respiratory health hazard, and common environmental air pollutant at ground level. The respiratory health effects due to O3 exposure are often associated with molecular and cellular perturbations in the respiratory tract. Periodic review of NAAQS requires comprehensive scientific evaluation of the public health effects of these pollutants, which is formulated through integrated science assessment (ISA) of the most policy-relevant scientific literature. This review focuses on the protective and pathogenic effects of macrophages in the O3-exposed respiratory tract, with emphasis on mouse model-based toxicological studies. Critical findings from 39 studies containing the words O3, macrophage, mice, and lung within the full text were assessed. While some of these studies highlight the presence of disease-relevant pathogenic macrophages in the airspaces, others emphasize a protective role for macrophages in O3-induced lung diseases. Moreover, a comprehensive list of currently known macrophage-specific roles in O3-induced lung diseases is included in this review and the significant knowledge gaps that still exist in the field are outlined. In conclusion, there is a vital need in this field for additional policy-relevant scientific information, including mechanistic studies to further define the role of macrophages in response to O3.
Collapse
Affiliation(s)
- Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
190
|
Glia maturation factor-γ regulates murine macrophage iron metabolism and M2 polarization through mitochondrial ROS. Blood Adv 2020; 3:1211-1225. [PMID: 30971398 DOI: 10.1182/bloodadvances.2018026070] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/16/2019] [Indexed: 12/19/2022] Open
Abstract
In macrophages, cellular iron metabolism status is tightly integrated with macrophage phenotype and associated with mitochondrial function. However, how molecular events regulate mitochondrial activity to integrate regulation of iron metabolism and macrophage phenotype remains unclear. Here, we explored the important role of the actin-regulatory protein glia maturation factor-γ (GMFG) in the regulation of cellular iron metabolism and macrophage phenotype. We found that GMFG was downregulated in murine macrophages by exposure to iron and hydrogen peroxide. GMFG knockdown altered the expression of iron metabolism proteins and increased iron levels in murine macrophages and concomitantly promoted their polarization toward an anti-inflammatory M2 phenotype. GMFG-knockdown macrophages exhibited moderately increased levels of mitochondrial reactive oxygen species (mtROS), which were accompanied by decreased expression of some mitochondrial respiration chain components, including the iron-sulfur cluster assembly scaffold protein ISCU as well as the antioxidant enzymes SOD1 and SOD2. Importantly, treatment of GMFG-knockdown macrophages with the antioxidant N-acetylcysteine reversed the altered expression of iron metabolism proteins and significantly inhibited the enhanced gene expression of M2 macrophage markers, suggesting that mtROS is mechanistically linked to cellular iron metabolism and macrophage phenotype. Finally, GMFG interacted with the mitochondrial membrane ATPase ATAD3A, suggesting that GMFG knockdown-induced mtROS production might be attributed to alteration of mitochondrial function in macrophages. Our findings suggest that GMFG is an important regulator in cellular iron metabolism and macrophage phenotype and could be a novel therapeutic target for modulating macrophage function in immune and metabolic disorders.
Collapse
|
191
|
Donninelli G, Saraf-Sinik I, Mazziotti V, Capone A, Grasso MG, Battistini L, Reynolds R, Magliozzi R, Volpe E. Interleukin-9 regulates macrophage activation in the progressive multiple sclerosis brain. J Neuroinflammation 2020; 17:149. [PMID: 32375811 PMCID: PMC7204302 DOI: 10.1186/s12974-020-01770-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/09/2020] [Indexed: 12/22/2022] Open
Abstract
Background Multiple sclerosis (MS) is an immune-mediated, chronic inflammatory, and demyelinating disease of the central nervous system (CNS). Several cytokines are thought to be involved in the regulation of MS pathogenesis. We recently identified interleukin (IL)-9 as a cytokine reducing inflammation and protecting from neurodegeneration in relapsing–remitting MS patients. However, the expression of IL-9 in CNS, and the mechanisms underlying the effect of IL-9 on CNS infiltrating immune cells have never been investigated. Methods To address this question, we first analyzed the expression levels of IL-9 in post-mortem cerebrospinal fluid of MS patients and the in situ expression of IL-9 in post-mortem MS brain samples by immunohistochemistry. A complementary investigation focused on identifying which immune cells express IL-9 receptor (IL-9R) by flow cytometry, western blot, and immunohistochemistry. Finally, we explored the effect of IL-9 on IL-9-responsive cells, analyzing the induced signaling pathways and functional properties. Results We found that macrophages, microglia, and CD4 T lymphocytes were the cells expressing the highest levels of IL-9 in the MS brain. Of the immune cells circulating in the blood, monocytes/macrophages were the most responsive to IL-9. We validated the expression of IL-9R by macrophages/microglia in post-mortem brain sections of MS patients. IL-9 induced activation of signal transducer and activator of transcription (STAT)1, STAT3, and STAT5 and reduced the expression of activation markers, such as CD45, CD14, CD68, and CD11b in inflammatory macrophages stimulated in vitro with lipopolysaccharide and interferon (IFN)-γ. Similarly, in situ the number of activated CD68+ macrophages was significantly reduced in areas with high levels of IL-9. Moreover, in the same conditions, IL-9 increased the secretion of the anti-inflammatory cytokine, transforming growth factor (TGF)-β. Conclusions These results reveal a new cytokine expressed in the CNS, with a role in the context of MS. We have demonstrated that IL-9 and its receptor are both expressed in CNS. Moreover, we found that IL-9 decreases the activation state and promotes the anti-inflammatory properties of human macrophages. This mechanism may contribute to the beneficial effects of IL-9 that are observed in MS, and may be therapeutically potentiated by modulating IL-9 expression in MS.
Collapse
Affiliation(s)
- Gloria Donninelli
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Inbar Saraf-Sinik
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Valentina Mazziotti
- Neurology section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico G.B. Rossi, P.le L.A. Scuro, 10, 37134, Verona, Italy
| | - Alessia Capone
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Roberta Magliozzi
- Neurology section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Policlinico G.B. Rossi, P.le L.A. Scuro, 10, 37134, Verona, Italy. .,Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK.
| | - Elisabetta Volpe
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy.
| |
Collapse
|
192
|
Sadh S, Hajjar S, Ariana A, Phuong MS, Cai D, Thakker P, Sad S. Coating M-CSF on plastic surface results in the generation of increased numbers of macrophages in vitro. J Immunol Methods 2020; 481-482:112788. [PMID: 32304707 DOI: 10.1016/j.jim.2020.112788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/29/2020] [Accepted: 03/31/2020] [Indexed: 11/15/2022]
Abstract
Macrophages are one of the important cell types in the innate immune system that are present in various anatomical regions of the body and promote early control of pathogens. The relative proportion of macrophages in various lymphoid and non-lymphoid regions is small, and as such it is tedious to purify these cells to homogeneity. Culture of bone marrow precursors with macrophage colony-stimulating factor (M-CSF) results in their differentiation to macrophages, however this procedure results in low numbers of differentiated macrophages. Herein we reveal a new approach of generating increased numbers of differentiated macrophages from bone marrow precursors. We show that M-CSF delivered in a plate-bound form results in the differentiation of significantly more macrophages in comparison to soluble M-CSF. Furthermore, the macrophages differentiated with plate-bound M-CSF display increased metabolic activity and cell death following infection with pathogens.
Collapse
Affiliation(s)
- Sanathan Sadh
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Stephanie Hajjar
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Ardeshir Ariana
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Melissa Sen Phuong
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - David Cai
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Parva Thakker
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada; University of Ottawa Centre for Infection, Immunity and Inflammation, Ottawa, K1H 8M5 Ontario, Canada.
| |
Collapse
|
193
|
Li B, Tan TB, Wang L, Zhao XY, Tan GJ. p38MAPK/SGK1 signaling regulates macrophage polarization in experimental autoimmune encephalomyelitis. Aging (Albany NY) 2020; 11:898-907. [PMID: 30716717 PMCID: PMC6382436 DOI: 10.18632/aging.101786] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is characterized with multifocal demyelination resulting from activation and infiltration of inflammatory cells into the central nerve system. Recent reports suggest that p38 mitogen-activated protein kinase (MAPK) / serum- and glucocorticoid-inducible protein kinase 1 (SGK1) signaling pathway contributes to the pathology of MS through regulation of immunity. However, the role of this signaling pathway in MS-related macrophage activation and polarization has not been studied. Here, we used an experimental autoimmune encephalomyelitis (EAE) model for MS to study the role of p38MAPK/SGK1 signaling in the macrophage polarization and its effects on the development and severity of EAE. Here, we found that p38MAPK/SGK1 signaling is required for IL4-induced M2 macrophage polarization in vitro. Chitin-induced M2 macrophage polarization reduces the severity of EAE in mice. Generation of an adeno-associated virus (AAV) carrying sh-p38 or sh-SGK1 under the control of a CD68 promoter successfully knockdown p38 or SGK1 levels in vitro and in vivo. Treatment with AAV-sh-p38 or AAV-sh-SGK1 abolished the effects of Chitin on macrophage polarization and the severity of EAE. Thus, our data suggest that p38MAPK/SGK1 signaling induces M2 macrophage polarization, which reduces the severity of EAE, a model for MS.
Collapse
Affiliation(s)
- Bo Li
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang 050000, China
| | | | - Liang Wang
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.,Key Laboratory of Hebei Neurology, Shijiazhuang 050000, China
| | - Xiao-Yun Zhao
- Laboratory Medicine Center of Qilu Hospital of Shandong University (Qingdao), Qingdao 266035, China
| | - Guo-Jun Tan
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.,Key Laboratory of Hebei Neurology, Shijiazhuang 050000, China
| |
Collapse
|
194
|
Samiea A, Yoon JSJ, Cheung ST, Chamberlain TC, Mui ALF. Interleukin-10 contributes to PGE2 signalling through upregulation of EP4 via SHIP1 and STAT3. PLoS One 2020; 15:e0230427. [PMID: 32240179 PMCID: PMC7117666 DOI: 10.1371/journal.pone.0230427] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/28/2020] [Indexed: 12/15/2022] Open
Abstract
Macrophage cells form part of our first line defense against pathogens. Macrophages become activated by microbial products such as lipopolysaccharide (LPS) to produce inflammatory mediators, such as TNFα and other cytokines, which orchestrate the host defense against the pathogen. Once the pathogen has been eradicated, the activated macrophage must be appropriately deactivated or inflammatory diseases result. Interleukin-10 (IL10) is a key anti-inflammatory cytokine which deactivates the activated macrophage. The IL10 receptor (IL10R) signals through the Jak1/Tyk2 tyrosine kinases, STAT3 transcription factor and the SHIP1 inositol phosphatase. However, IL10 has also been described to induce the activation of the cyclic adenosine monophosphate (cAMP) regulated protein kinase A (PKA). We now report that IL10R signalling leads to STAT3/SHIP1 dependent expression of the EP4 receptor for prostaglandin E2 (PGE2). In macrophages, EP4 is a Gαs-protein coupled receptor that stimulates adenylate cyclase (AC) production of cAMP, leading to downstream activation of protein kinase A (PKA) and phosphorylation of the CREB transcription factor. IL10 induction of phospho-CREB and inhibition of LPS-induced phosphorylation of p85 PI3K and p70 S6 kinase required the presence of EP4. These data suggest that IL10R activation of STAT3/SHIP1 enhances EP4 expression, and that it is EP4 which activates cAMP-dependent signalling. The coordination between IL10R and EP4 signalling also provides an explanation for why cAMP elevating agents synergize with IL10 to elicit anti-inflammatory responses.
Collapse
MESH Headings
- Animals
- Dinoprostone/metabolism
- Female
- Interleukin-10/pharmacology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Oxytocics/metabolism
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
- RAW 264.7 Cells
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Abrar Samiea
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Jeff S. J. Yoon
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Department of Surgery, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Sylvia T. Cheung
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Department of Surgery, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Thomas C. Chamberlain
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Department of Surgery, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Alice L. -F. Mui
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Department of Surgery, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
195
|
Umasuthan N, Xue X, Caballero-Solares A, Kumar S, Westcott JD, Chen Z, Fast MD, Skugor S, Nowak BF, Taylor RG, Rise ML. Transcriptomic Profiling in Fins of Atlantic Salmon Parasitized with Sea Lice: Evidence for an Early Imbalance Between Chalimus-Induced Immunomodulation and the Host's Defense Response. Int J Mol Sci 2020; 21:E2417. [PMID: 32244468 PMCID: PMC7177938 DOI: 10.3390/ijms21072417] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/27/2020] [Indexed: 01/08/2023] Open
Abstract
Parasitic sea lice (e.g., Lepeophtheirus salmonis) cause costly outbreaks in salmon farming. Molecular insights into parasite-induced host responses will provide the basis for improved management strategies. We investigated the early transcriptomic responses in pelvic fins of Atlantic salmon parasitized with chalimus I stage sea lice. Fin samples collected from non-infected (i.e. pre-infected) control (PRE) and at chalimus-attachment sites (ATT) and adjacent to chalimus-attachment sites (ADJ) from infected fish were used in profiling global gene expression using 44 K microarrays. We identified 6568 differentially expressed probes (DEPs, FDR < 5%) that included 1928 shared DEPs between ATT and ADJ compared to PRE. The ATT versus ADJ comparison revealed 90 DEPs, all of which were upregulated in ATT samples. Gene ontology/pathway term network analyses revealed profound changes in physiological processes, including extracellular matrix (ECM) degradation, tissue repair/remodeling and wound healing, immunity and defense, chemotaxis and signaling, antiviral response, and redox homeostasis in infected fins. The QPCR analysis of 37 microarray-identified transcripts representing these functional themes served to confirm the microarray results with a significant positive correlation (p < 0.0001). Most immune/defense-relevant transcripts were downregulated in both ATT and ADJ sites compared to PRE, suggesting that chalimus exerts immunosuppressive effects in the salmon's fins. The comparison between ATT and ADJ sites demonstrated the upregulation of a suite of immune-relevant transcripts, evidencing the salmon's attempt to mount an anti-lice response. We hypothesize that an imbalance between immunomodulation caused by chalimus during the early phase of infection and weak defense response manifested by Atlantic salmon makes it a susceptible host for L. salmonis.
Collapse
Affiliation(s)
- Navaneethaiyer Umasuthan
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John’s, NL A1C 5S7, Canada; (X.X.); (A.C.-S.); (S.K.)
| | - Xi Xue
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John’s, NL A1C 5S7, Canada; (X.X.); (A.C.-S.); (S.K.)
| | - Albert Caballero-Solares
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John’s, NL A1C 5S7, Canada; (X.X.); (A.C.-S.); (S.K.)
| | - Surendra Kumar
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John’s, NL A1C 5S7, Canada; (X.X.); (A.C.-S.); (S.K.)
| | - Jillian D. Westcott
- Fisheries and Marine Institute, Memorial University of Newfoundland, P.O. Box 4920, St. John’s, NL A1C 5R3, Canada; (J.D.W.); (Z.C.)
| | - Zhiyu Chen
- Fisheries and Marine Institute, Memorial University of Newfoundland, P.O. Box 4920, St. John’s, NL A1C 5R3, Canada; (J.D.W.); (Z.C.)
| | - Mark D. Fast
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada;
| | - Stanko Skugor
- Cargill Aqua Nutrition, Cargill, Sea Lice Research Center (SLRC), Hanaveien 17, 4327 Sandnes, Norway;
| | - Barbara F. Nowak
- Institute of Marine and Antarctic Studies, University of Tasmania, Locked Bag 1370, Launceston 7250, TAS, Australia;
| | - Richard G. Taylor
- Cargill Animal Nutrition, 10383 165th Avenue NW, Elk River, MN 55330, USA;
| | - Matthew L. Rise
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John’s, NL A1C 5S7, Canada; (X.X.); (A.C.-S.); (S.K.)
| |
Collapse
|
196
|
Gordon S, Plüddemann A, Mukhopadhyay S. Plasma membrane receptors of tissue macrophages: functions and role in pathology. J Pathol 2020; 250:656-666. [PMID: 32086805 DOI: 10.1002/path.5404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
The cells of the mononuclear phagocyte system (MPS) constitute a dispersed organ, which is distributed throughout the body. Macrophages in different tissues display distinctive mosaic phenotypes as resident and recruited cells of embryonic and bone marrow origin, respectively. They help to maintain homeostasis during development and throughout adult life, yet contribute to the pathogenesis of many disease processes, including inflammation, innate and adaptive immunity, metabolic disorders, and cancer. Heterogeneous tissue macrophage populations display a wide variety of surface molecules to recognise and respond to host, microbial, and exogenous ligands in their environment; their receptors mediate the uptake and destruction of effete and dying host cells and pathogens, as well as contribute trophic and secretory functions within every organ in the body. Apart from local cellular interactions, macrophage surface molecules and products serve to mobilise and coordinate systemic humoral and cellular responses. Their use as antigen markers in pathogenesis and as potential drug targets has lagged in clinical pathology and human immunotherapy. In this review, we summarise the properties of selected surface molecules expressed on macrophages in different tissues and disease processes, to provide a functional basis for diagnosis, further research, and treatment. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Siamon Gordon
- College of Medicine, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Annette Plüddemann
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Subhankar Mukhopadhyay
- Peter Gorer Department of Immunobiology, Medical Research Council Centre for Transplantation, King's College London, London, UK
| |
Collapse
|
197
|
Abstract
Phagocytosis is a pivotal immunological process, and its discovery by Elia Metchnikoff in 1882 was a step toward the establishment of the innate immune system as a separate branch of immunology. Elia Metchnikoff received the Nobel Prize in physiology and medicine for this discovery in 1908. Since its discovery almost 140 years before, phagocytosis remains the hot topic of research in immunology. The phagocytosis research has seen a great advancement since its first discovery. Functionally, phagocytosis is a simple immunological process required to engulf and remove pathogens, dead cells and tumor cells to maintain the immune homeostasis. However, mechanistically, it is a very complex process involving different mechanisms, induced and regulated by several pattern recognition receptors, soluble pattern recognition molecules, scavenger receptors (SRs) and opsonins. These mechanisms involve the formation of phagosomes, their maturation into phagolysosomes causing pathogen destruction or antigen synthesis to present them to major histocompatibility complex molecules for activating an adaptive immune response. Any defect in this mechanism may predispose the host to certain infections and inflammatory diseases (autoinflammatory and autoimmune diseases) along with immunodeficiency. The article is designed to discuss its mechanistic complexity at each level, varying from phagocytosis induction to phagolysosome resolution.
Collapse
Affiliation(s)
- Vijay Kumar
- Faculty of Medicine, Children's Health Queensland Clinical Unit, School of Clinical Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
198
|
Puthenparampil M, Stropparo E, Zywicki S, Bovis F, Cazzola C, Federle L, Grassivaro F, Rinaldi F, Perini P, Sormani MP, Gallo P. Wide Cytokine Analysis in Cerebrospinal Fluid at Diagnosis Identified CCL-3 as a Possible Prognostic Factor for Multiple Sclerosis. Front Immunol 2020; 11:174. [PMID: 32194540 PMCID: PMC7066207 DOI: 10.3389/fimmu.2020.00174] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Apart from IgG oligoclonal bands, no other biomarker has, to date, been validated for diagnostic and/or prognostic purposes in multiple sclerosis (MS). Aim: To investigate a wide panel of cytokines and chemokines in the cerebrospinal fluid (CSF) of relapsing-remitting MS (RRMS) patients and evaluate their association with clinical and magnetic resonance imaging (MRI) parameters, as well as their predictive clinical value. Methods: Fifty-one RRMS at clinical onset and 17 other not inflammatory neurological disorders (ONINDs) underwent brain MRI (including 3D-T1, 3D-FLAIR, and 3-DIR sequences) and CSF examination. Eighty-seven cytokines and chemokines were analyzed in CSF by Multiplex technology. Results: Compared to ONIND, CXCL-10, CXCL-11, CXCL-13, CCL-1, CCL-2, CCL-3, CCL-22, IL-16, and BAFF were significantly (p < 0.05) increased in RRMS CSF. However, only CCL-3 was associated with both MS diagnosis and IgGOB detection. Based on a 95%CI in ONIND (cut-off value: 0.798 pg/ml) and ROC analysis (cut-off value: 0.495 pg/ml), RRMS patients were stratified in CCL-3high (>0.736 pg/mL), CCL-3medium, and CCL-3low (<0.495 pg/ml). Survival analysis disclosed a strong association between high CCL-3 values and disease reactivation (OR = 4.9, 95%CI: 1.8-13.3, p < 0.005) in the following 2 years. Conclusions: CCL-3 deserves further investigation as a candidate prognostic biomarker for RRMS.
Collapse
Affiliation(s)
- Marco Puthenparampil
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Erica Stropparo
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Sofia Zywicki
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Francesca Bovis
- Biostatistics Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Chiara Cazzola
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Lisa Federle
- Multiple Sclerosis Centre, ULSS8 Berica, Ospedale San Bortolo, Vicenza, Italy
| | - Francesca Grassivaro
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Francesca Rinaldi
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Paola Perini
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| | - Maria Pia Sormani
- Biostatistics Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Paolo Gallo
- Department of Neurosciences DNS, Multiple Sclerosis Centre, Università degli Studi di Padova, Padua, Italy
| |
Collapse
|
199
|
Agarwal M, Biswas P, Bhattacharya A, Sinha DK. Reactive oxygen species-mediated cytoplasmic stiffening impairs the phagocytic ability of the macrophage. J Cell Sci 2020; 133:jcs.236471. [PMID: 32005700 DOI: 10.1242/jcs.236471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
The phagocytic ability of macrophages empowers them to enforce innate immunity. RAW264.7, THP-1 and peripheral blood mononuclear cell-derived macrophages display considerable variability with regards to their phagocytic ability. We identify the underlying causes that attenuate the phagocytic abilities of a macrophage. Deformability of the cytoplasm and cortex influences the macrophage's phagocytic ability, and macrophages use the large cell-to-cell variability of their cytoplasmic stiffness to modulate their phagocytic ability. We find that the more-deformable macrophages have a higher phagocytic ability than those that are less deformable. Further, the subcellular spatial variability of cortex stiffness gives rise to more-deformable subdomains on the membrane for pathogen ingestion. We report a previously unknown negative-feedback loop that is triggered by the phagocytic oxidative burst. Macrophages utilize the excess reactive oxygen species to stiffen the cytoplasm, reducing their phagocytic propensity. In organisms, ageing or pathological conditions impair the phagocytic ability of macrophages. Our findings identify the targets that could potentially be utilized for restoring the phagocytic ability of the defunct macrophages.
Collapse
Affiliation(s)
- Mahesh Agarwal
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 32, India
| | - Parijat Biswas
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 32, India
| | - Anindita Bhattacharya
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 32, India
| | - Deepak Kumar Sinha
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 32, India
| |
Collapse
|
200
|
Behnke V, Wolf A, Langmann T. The role of lymphocytes and phagocytes in age-related macular degeneration (AMD). Cell Mol Life Sci 2020; 77:781-788. [PMID: 31897541 PMCID: PMC11104950 DOI: 10.1007/s00018-019-03419-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment of the elderly population. Since AMD is a multifactorial age-related disease with various genetic risk factors, the understanding of its complex pathophysiology is still limited. However, animal experiments, genome-wide association data and the molecular profiling of AMD patient samples have highlighted a key role of systemic and local immune processes that contribute to this chronic eye disease. In this overview article, we concentrate on the role of lymphocytes and mononuclear phagocytes and their interplay in triggering a persistent immune response in the AMD retina. We preferentially review findings from human immune cell analyses and complement these with related findings in experimental models. We conclude that both immune cell types as their signaling network may be a rich source to identify novel molecular targets for immunomodulation in AMD.
Collapse
Affiliation(s)
- Verena Behnke
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), 50931, Cologne, Germany.
| |
Collapse
|