1
|
Hornsteiner F, Vierthaler J, Strandt H, Resag A, Fu Z, Ausserhofer M, Tripp CH, Dieckmann S, Kanduth M, Farrand K, Bregar S, Nemati N, Hermann-Kleiter N, Seretis A, Morla S, Mullins D, Finotello F, Trajanoski Z, Wollmann G, Ronchese F, Schmitz M, Hermans IF, Stoitzner P. Tumor-targeted therapy with BRAF-inhibitor recruits activated dendritic cells to promote tumor immunity in melanoma. J Immunother Cancer 2024; 12:e008606. [PMID: 38631706 PMCID: PMC11029477 DOI: 10.1136/jitc-2023-008606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Tumor-targeted therapy causes impressive tumor regression, but the emergence of resistance limits long-term survival benefits in patients. Little information is available on the role of the myeloid cell network, especially dendritic cells (DC) during tumor-targeted therapy. METHODS Here, we investigated therapy-mediated immunological alterations in the tumor microenvironment (TME) and tumor-draining lymph nodes (LN) in the D4M.3A preclinical melanoma mouse model (harboring the V-Raf murine sarcoma viral oncogene homolog B (BRAF)V600E mutation) by using high-dimensional multicolor flow cytometry in combination with multiplex immunohistochemistry. This was complemented with RNA sequencing and cytokine quantification to characterize the immune status of the tumors. The importance of T cells during tumor-targeted therapy was investigated by depleting CD4+ or CD8+ T cells in tumor-bearing mice. Tumor antigen-specific T-cell responses were characterized by performing in vivo T-cell proliferation assays and the contribution of conventional type 1 DC (cDC1) to T-cell immunity during tumor-targeted therapy was assessed using Batf3-/- mice lacking cDC1. RESULTS Our findings reveal that BRAF-inhibitor therapy increased tumor immunogenicity, reflected by an upregulation of genes associated with immune activation. The T cell-inflamed TME contained higher numbers of activated cDC1 and cDC2 but also inflammatory CCR2-expressing monocytes. At the same time, tumor-targeted therapy enhanced the frequency of migratory, activated DC subsets in tumor-draining LN. Even more, we identified a cDC2 population expressing the Fc gamma receptor I (FcγRI)/CD64 in tumors and LN that displayed high levels of CD40 and CCR7 indicating involvement in T cell-mediated tumor immunity. The importance of cDC2 is underlined by just a partial loss of therapy response in a cDC1-deficient mouse model. Both CD4+ and CD8+ T cells were essential for therapy response as their respective depletion impaired therapy success. On resistance development, the tumors reverted to an immunologically inert state with a loss of DC and inflammatory monocytes together with the accumulation of regulatory T cells. Moreover, tumor antigen-specific CD8+ T cells were compromised in proliferation and interferon-γ-production. CONCLUSION Our results give novel insights into the remodeling of the myeloid landscape by tumor-targeted therapy. We demonstrate that the transient immunogenic tumor milieu contains more activated DC. This knowledge has important implications for the development of future combinatorial therapies.
Collapse
Affiliation(s)
- Florian Hornsteiner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Vierthaler
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helen Strandt
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Antonia Resag
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Zhe Fu
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Markus Ausserhofer
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sophie Dieckmann
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Kanduth
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kathryn Farrand
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sarah Bregar
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Niloofar Nemati
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Athanasios Seretis
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Sudhir Morla
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - David Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Francesca Finotello
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, Innsbruck, Austria
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Guido Wollmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Miyagawa F, Ozato K, Tagaya Y, Asada H. Type I IFN Derived from Ly6C hi Monocytes Suppresses Type 2 Inflammation in a Murine Model of Atopic Dermatitis. J Invest Dermatol 2024; 144:520-530.e2. [PMID: 37739337 DOI: 10.1016/j.jid.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/26/2023] [Accepted: 08/31/2023] [Indexed: 09/24/2023]
Abstract
The roles of innate immune cells, including eosinophils, basophils, and group 2 innate lymphoid cells, in atopic dermatitis (AD) have been well-documented, whereas that of monocytes, another component of the innate immunity, remains rather poorly understood, thus necessitating the topic of this study. In addition, cytokines and cellular pathways needed for the resolution of type 2 inflammation in AD need further investigation. Using a murine AD model, we report here that (i) Ly6Chi monocytes were rapidly recruited to the AD lesion in a CCR2-dependent manner, blockade of which exacerbated AD; (ii) type I IFN production is profoundly involved in this suppression because the blockade of it by genetic depletion or antibody neutralization exacerbated AD; and (iii) Ly6Chi monocytes operate through the production of type I IFN because Ly6Chi monocytes from Irf7-null mice, which lack type I IFN production, failed to rescue Ccr2-/- mice from severe AD upon adoptive transfer. In addition, in vitro studies demonstrated type I IFN suppressed basophil expansion from bone marrow progenitor cells and survival of mature basophils. Collectively, our work suggests that Ly6Chi monocytes are the first and dominant inflammatory cells reaching AD lesions that negatively regulate type 2 inflammation through the production of type I IFN.
Collapse
Affiliation(s)
- Fumi Miyagawa
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan.
| | - Keiko Ozato
- Laboratory of Molecular Growth Regulation, Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Yutaka Tagaya
- Cell Biology Lab, Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan
| |
Collapse
|
3
|
Musigk N, Suwalski P, Golpour A, Fairweather D, Klingel K, Martin P, Frustaci A, Cooper LT, Lüscher TF, Landmesser U, Heidecker B. The inflammatory spectrum of cardiomyopathies. Front Cardiovasc Med 2024; 11:1251780. [PMID: 38464847 PMCID: PMC10921946 DOI: 10.3389/fcvm.2024.1251780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Infiltration of the myocardium with various cell types, cytokines and chemokines plays a crucial role in the pathogenesis of cardiomyopathies including inflammatory cardiomyopathies and myocarditis. A more comprehensive understanding of the precise immune mechanisms involved in acute and chronic myocarditis is essential to develop novel therapeutic approaches. This review offers a comprehensive overview of the current knowledge of the immune landscape in cardiomyopathies based on etiology. It identifies gaps in our knowledge about cardiac inflammation and emphasizes the need for new translational approaches to improve our understanding thus enabling development of novel early detection methods and more effective treatments.
Collapse
Affiliation(s)
- Nicolas Musigk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Phillip Suwalski
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Ainoosh Golpour
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Karin Klingel
- Cardiopathology Institute for Pathology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Pilar Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Thomas F. Lüscher
- GZO-Zurich Regional Health Centre, Wetzikon & Cardioimmunology, Centre for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Royal Brompton & Harefield Hospitals and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Bettina Heidecker
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
4
|
Estaleen RA, Reilly CM, Luo XM. A double-edged sword: interactions of CX 3CL1/CX 3CR1 and gut microbiota in systemic lupus erythematosus. Front Immunol 2024; 14:1330500. [PMID: 38299151 PMCID: PMC10828040 DOI: 10.3389/fimmu.2023.1330500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/28/2023] [Indexed: 02/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic chronic disease initiated by an abnormal immune response to self and can affect multiple organs. SLE is characterized by the production of autoantibodies and the deposition of immune complexes. In regard to the clinical observations assessed by rheumatologists, several chemokines and cytokines also contribute to disease progression. One such chemokine and adhesion molecule is CX3CL1 (otherwise known as fractalkine). CX3CL1 is involved in cell trafficking and inflammation through recognition by its receptor, CX3CR1. The CX3CL1 protein consists of a chemokine domain and a mucin-like stalk that allows it to function both as a chemoattractant and as an adhesion molecule. In inflammation and specifically lupus, the literature displays contradictory evidence for the functions of CX3CL1/CX3CR1 interactions. In addition, the gut microbiota has been shown to play an important role in the pathogenesis of SLE. This review highlights current studies that illustrate the interactions of the gut microbiota and CX3CR1 in SLE.
Collapse
Affiliation(s)
- Rana A. Estaleen
- Department of Biomedical Sciences and Pathobiology, Virgnia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Christopher M. Reilly
- Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virgnia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
5
|
Zhao H, Huang M, Jiang L. Potential Roles and Future Perspectives of Chitinase 3-like 1 in Macrophage Polarization and the Development of Diseases. Int J Mol Sci 2023; 24:16149. [PMID: 38003338 PMCID: PMC10671302 DOI: 10.3390/ijms242216149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1), a chitinase-like protein family member, is a secreted glycoprotein that mediates macrophage polarization, inflammation, apoptosis, angiogenesis, and carcinogenesis. Abnormal CHI3L1 expression has been associated with multiple metabolic and neurological disorders, including diabetes, atherosclerosis, and Alzheimer's disease. Aberrant CHI3L1 expression is also reportedly associated with tumor migration and metastasis, as well as contributions to immune escape, playing important roles in tumor progression. However, the physiological and pathophysiological roles of CHI3L1 in the development of metabolic and neurodegenerative diseases and cancer remain unclear. Understanding the polarization relationship between CHI3L1 and macrophages is crucial for disease progression. Recent research has uncovered the complex mechanisms of CHI3L1 in different diseases, highlighting its close association with macrophage functional polarization. In this article, we review recent findings regarding the various disease types and summarize the relationship between macrophages and CHI3L1. Furthermore, this article also provides a brief overview of the various mechanisms and inhibitors employed to inhibit CHI3L1 and disrupt its interaction with receptors. These endeavors highlight the pivotal roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of metabolic diseases, neurodegenerative diseases, and cancers.
Collapse
Affiliation(s)
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| |
Collapse
|
6
|
Wang C, Cheng Y, Li B, Qiu X, Hu H, Zhang X, Lu Z, Zheng F. Transcriptional characteristics and functional validation of three monocyte subsets during aging. Immun Ageing 2023; 20:50. [PMID: 37759225 PMCID: PMC10523626 DOI: 10.1186/s12979-023-00377-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Age-associated changes in immunity are inextricably linked to chronic inflammation and age-related diseases, the impact of aging on monocyte subsets is poorly understood. METHODS Flow cytometry was applied to distinguish three monocyte subsets between 120 young and 103 aged individuals. We then analyzed the expression profiles of three monocyte subsets from 9 young and 9 older donors and CD14+ monocytes from 1202 individuals between 44 and 83 years old. Flow cytometry was used to measure β-galactosidase activities, ROS levels, mitochondrial contents, mitochondrial membrane potentials (MMPs) and intracellular IL-6 levels in three monocyte subsets of young and elderly individuals, and plasma IL-6 levels were detected by electrochemiluminescence immunoassay. Mitochondrial stress and glycolytic rate of CD14+ monocytes from young and aged individuals were measured by Seahorse XFe24 Analyzer. RESULTS Compared with young individuals, the percentage of classical subset in aged persons significantly decreased, while the proportion of nonclassical subset increased. Age-related differential genes were obviously enriched in cellular senescence, ROS, oxidative phosphorylation, mitochondrial respiratory chain, IL-6 and ribosome-related pathways. Compared with young individuals, the β-galactosidase activities, ROS contents, intracellular IL-6 levels of three monocyte subsets, and plasma IL-6 levels in aged individuals were significantly elevated, while the MMPs apparently declined with age and the mitochondrial contents were only increased in intermediate and nonclassical subsets. CD14+ monocytes from elderly adults had conspicuously lower basal and spare respiratory capacity and higher basal glycolysis than those from young individuals. CONCLUSIONS During aging, monocytes exhibited senescence-associated secretory phenotype, mitochondrial dysfunction, decreased oxidative phosphorylation and increased glycolysis and the nonclassical subset displayed the clearest features of aging. Our study comprehensively investigated age-related transcriptional alterations of three monocyte subsets and identified the pivotal pathways of monocyte senescence, which may have significant implications for tactics to alleviate age-related conditions.
Collapse
Affiliation(s)
- Chen Wang
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yating Cheng
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Boyu Li
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xueping Qiu
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Hu
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiaokang Zhang
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Fang Zheng
- Center for Gene Diagnosis, Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Rodriguez BL, Chen L, Li Y, Miao S, Peng DH, Fradette JJ, Diao L, Konen JM, Alvarez FRR, Solis LM, Yi X, Padhye A, Gibson LA, Ochieng JK, Zhou X, Wang J, Gibbons DL. Targeting immunosuppressive Ly6C+ classical monocytes reverses anti-PD-1/CTLA-4 immunotherapy resistance. Front Immunol 2023; 14:1161869. [PMID: 37449205 PMCID: PMC10336223 DOI: 10.3389/fimmu.2023.1161869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Despite significant clinical advancement with the use of immune checkpoint blockade (ICB) in non-small cell lung cancer (NSCLC) there are still a major subset of patients that develop adaptive/acquired resistance. Understanding resistance mechanisms to ICB is critical to developing new therapeutic strategies and improving patient survival. The dynamic nature of the tumor microenvironment and the mutational load driving tumor immunogenicity limit the efficacy to ICB. Recent studies indicate that myeloid cells are drivers of ICB resistance. In this study we sought to understand which immune cells were contributing to resistance and if we could modify them in a way to improve response to ICB therapy. Results Our results show that combination anti-PD-1/CTLA-4 produces an initial antitumor effect with evidence of an activated immune response. Upon extended treatment with anti-PD-1/CTLA-4 acquired resistance developed with an increase of the immunosuppressive populations, including T-regulatory cells, neutrophils and monocytes. Addition of anti-Ly6C blocking antibody to anti-PD-1/CTLA-4 was capable of completely reversing treatment resistance and restoring CD8 T cell activity in multiple KP lung cancer models and in the autochthonous lung cancer KrasLSL-G12D/p53fl/fl model. We found that there were higher classical Ly6C+ monocytes in anti-PD-1/CTLA-4 combination resistant tumors. B7 blockade illustrated the importance of dendritic cells for treatment efficacy of anti-Ly6C/PD-1/CTLA-4. We further determined that classical Ly6C+ monocytes in anti-PD-1/CTLA-4 resistant tumors are trafficked into the tumor via IFN-γ and the CCL2-CCR2 axis. Mechanistically we found that classical monocytes from ICB resistant tumors were unable to differentiate into antigen presenting cells and instead differentiated into immunosuppressive M2 macrophages or myeloid-derived suppressor cells (MDSC). Classical Ly6C+ monocytes from ICB resistant tumors had a decrease in both Flt3 and PU.1 expression that prevented differentiation into dendritic cells/macrophages. Conclusions Therapeutically we found that addition of anti-Ly6C to the combination of anti-PD-1/CTLA-4 was capable of complete tumor eradication. Classical Ly6C+ monocytes differentiate into immunosuppressive cells, while blockade of classical monocytes drives dendritic cell differentiation/maturation to reinvigorate the anti-tumor T cell response. These findings support that immunotherapy resistance is associated with infiltrating monocytes and that controlling the differentiation process of monocytes can enhance the therapeutic potential of ICB.
Collapse
Affiliation(s)
- B. Leticia Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Limo Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yanli Li
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shucheng Miao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- United of Texas (UT) Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - David H. Peng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jared J. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lixia Diao
- Department Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jessica M. Konen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Frank R. Rojas Alvarez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M. Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaohui Yi
- Bellicum Pharmaceuticals, Inc., Houston, TX, United States
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aparna Padhye
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- United of Texas (UT) Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Laura A. Gibson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joshua K. Ochieng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaofei Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jing Wang
- Department Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
8
|
Pang B, Hu C, Li H, Nie X, Wang K, Zhou C, Yi H. Myeloidderived suppressor cells: Escorts at the maternal-fetal interface. Front Immunol 2023; 14:1080391. [PMID: 36817414 PMCID: PMC9932974 DOI: 10.3389/fimmu.2023.1080391] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a novel heterogenous group of immunosuppressive cells derived from myeloid progenitors. Their role is well known in tumors and autoimmune diseases. In recent years, the role and function of MDSCs during reproduction have attracted increasing attention. Improving the understanding of their strong association with recurrent implantation failure, pathological pregnancy, and neonatal health has become a focus area in research. In this review, we focus on the interaction between MDSCs and other cell types (immune and non-immune cells) from embryo implantation to postpartum. Furthermore, we discuss the molecular mechanisms that could facilitate the therapeutic targeting of MDSCs. Therefore, this review intends to encourage further research in the field of maternal-fetal interface immunity in order to identify probable pathways driving the accumulation of MDSCs and to effectively target their ability to promote embryo implantation, reduce pathological pregnancy, and increase neonatal health.
Collapse
Affiliation(s)
- Bo Pang
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Cardiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Cong Hu
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huimin Li
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Xinyu Nie
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Keqi Wang
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Cardiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chen Zhou
- General Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huanfa Yi
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
9
|
Connors J, Taramangalam B, Cusimano G, Bell MR, Matt SM, Runner K, Gaskill PJ, DeFilippis V, Nikolich-Žugich J, Kutzler MA, Haddad EK. Aging alters antiviral signaling pathways resulting in functional impairment in innate immunity in response to pattern recognition receptor agonists. GeroScience 2022; 44:2555-2572. [PMID: 35849213 PMCID: PMC9289086 DOI: 10.1007/s11357-022-00612-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/16/2022] [Indexed: 01/06/2023] Open
Abstract
The progressive impairment of immunity to pathogens and vaccines with aging is a significant public health problem as the world population shifts to an increased percentage of older adults (> 65). We have previously demonstrated that cells obtained from older volunteers have delayed and defective induction of type I interferons and T cell and B cell helper cytokines in response to TLR ligands when compared to those from adult subjects. However, the underlying intracellular mechanisms are not well described. Herein, we studied two critical pathways important in the production of type I interferon (IFN), the interferon response factor 7 (pIRF7), and TANK-binding kinase (pTBK-1). We show a decrease in pIRF7 and pTBK-1 in cross-priming dendritic cells (cDC1s), CD4+ T cell priming DCs (cDC2s), and CD14dimCD16+ vascular patrolling monocytes from older adults (n = 11) following stimulation with pathway-specific agonists in comparison with young individuals (n = 11). The decrease in these key antiviral pathway proteins correlates with decreased phagocytosis, suggesting impaired function in Overall, our findings describe molecular mechanisms which explain the innate functional impairment in older adults and thus could inform us of novel approaches to restore these defects.
Collapse
Affiliation(s)
- Jennifer Connors
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Bhavani Taramangalam
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Gina Cusimano
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Matthew R. Bell
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Stephanie M. Matt
- grid.166341.70000 0001 2181 3113Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Kaitlyn Runner
- grid.166341.70000 0001 2181 3113Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Peter J. Gaskill
- grid.166341.70000 0001 2181 3113Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Victor DeFilippis
- grid.5288.70000 0000 9758 5690Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR USA
| | - Janko Nikolich-Žugich
- grid.134563.60000 0001 2168 186XDepartment of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ USA ,grid.134563.60000 0001 2168 186XArizona Center On Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ USA
| | - Michele A. Kutzler
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| | - Elias K. Haddad
- grid.166341.70000 0001 2181 3113Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, PA USA ,grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA USA
| |
Collapse
|
10
|
Yahara Y, Nguyen T, Ishikawa K, Kamei K, Alman BA. The origins and roles of osteoclasts in bone development, homeostasis and repair. Development 2022; 149:275249. [PMID: 35502779 PMCID: PMC9124578 DOI: 10.1242/dev.199908] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying bone development, repair and regeneration are reliant on the interplay and communication between osteoclasts and other surrounding cells. Osteoclasts are multinucleated monocyte lineage cells with resorptive abilities, forming the bone marrow cavity during development. This marrow cavity, essential to hematopoiesis and osteoclast-osteoblast interactions, provides a setting to investigate the origin of osteoclasts and their multi-faceted roles. This Review examines recent developments in the embryonic understanding of osteoclast origin, as well as interactions within the immune environment to regulate normal and pathological bone development, homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Tuyet Nguyen
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Koji Ishikawa
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Orthopaedic Surgery, Showa University School of Medicine, Tokyo, 142-8666, Japan
| | - Katsuhiko Kamei
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
11
|
Hardonnière K, Szely N, El Ali Z, Pallardy M, Kerdine-Römer S. Models of Dendritic Cells to Assess Skin Sensitization. FRONTIERS IN TOXICOLOGY 2022; 4:851017. [PMID: 35373185 PMCID: PMC8971372 DOI: 10.3389/ftox.2022.851017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
Allergic contact dermatitis (ACD) is a complex skin pathology occurring in reaction against environmental substances found in the workplace (cement, hair dyes, textile dyes), in the private environment (e.g., household products, cosmetic ingredients), or following skin exposure to drugs. Many cells are involved in the initiation of ACD during the sensitization phase. The four key events (KE) of skin sensitization AOP are covalent binding to skin proteins (KE1), keratinocyte activation (KE2), activation of DCs (KE3), and T-cell activation and proliferation (KE4), leading to the adverse outcome of ACD. Dendritic cells (DCs) are thus playing a key role in ACD pathophysiology. Indeed, in the presence of chemical sensitizers, DCs migrate from the skin to the draining lymph nodes and present peptide-chemical conjugates to T cells, leading to their activation and proliferation. In vitro methods have been actively developed to assess the activation of DCs by chemicals to establish a reliable in vitro sensitization test. Therefore, this review will detail the most used methods and protocols to develop DC models in vitro. Three different models of DCs will be addressed: 1) DCs derived from Cord Blood (CD34-DCs), 2) DCs derived from Monocytes (Mo-DCs), and 3) DCs derived from mice Bone-Marrow (BM-DCs). In addition, a model of exposition to contact sensitizers to assess KE3 of skin sensitization will be detailed for each of the models presented.
Collapse
|
12
|
Al-Moussawy M, Abdelsamed HA, Lakkis FG. Immunoglobulin-like receptors and the generation of innate immune memory. Immunogenetics 2022; 74:179-195. [PMID: 35034136 PMCID: PMC10074160 DOI: 10.1007/s00251-021-01240-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022]
Abstract
Host immunity is classically divided into "innate" and "adaptive." While the former has always been regarded as the first, rapid, and antigen-nonspecific reaction to invading pathogens, the latter represents the more sophisticated and antigen-specific response that has the potential to persist and generate memory. Recent work however has challenged this dogma, where murine studies have successfully demonstrated the ability of innate immune cells (monocytes and macrophages) to acquire antigen-specific memory to allogeneic major histocompatibility complex (MHC) molecules. The immunoreceptors so far identified that mediate innate immune memory are the paired immunoglobulin-like receptors (PIRs) in mice, which are orthologous to human leukocyte immunoglobulin-like receptors (LILRs). These receptor families are mainly expressed by the myelomonocytic cell lineage, suggesting an important role in the innate immune response. In this review, we will discuss the role of immunoglobulin-like receptors in the development of innate immune memory across species.
Collapse
Affiliation(s)
- Mouhamad Al-Moussawy
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA.
| | - Hossam A Abdelsamed
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA. .,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, USA.
| | - Fadi G Lakkis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA. .,Department of Immunology, University of Pittsburgh, Pittsburgh, USA. .,Department of Medicine, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
13
|
Cutolo M, Soldano S, Gotelli E, Montagna P, Campitiello R, Paolino S, Pizzorni C, Sulli A, Smith V, Tardito S. CTLA4-Ig treatment induces M1-M2 shift in cultured monocyte-derived macrophages from healthy subjects and rheumatoid arthritis patients. Arthritis Res Ther 2021; 23:306. [PMID: 34952630 PMCID: PMC8709961 DOI: 10.1186/s13075-021-02691-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/03/2021] [Indexed: 12/29/2022] Open
Abstract
Background In rheumatoid arthritis (RA), macrophages play an important role in modulating the immunoinflammatory response through their polarisation into “classically” (M1) or “alternatively activated” (M2) phenotypes. In RA, CTLA4-Ig (abatacept) reduces the inflammatory activity of macrophages by interacting with the costimulatory molecule CD86. The study aimed to investigate the efficacy of CTLA4-Ig treatment to induce an M2 phenotype both in M1-polarised monocyte-derived macrophages (MDMs) obtained from healthy subjects (HS) and in cultured MDMs obtained from active RA patients. Methods Cultured MDMs were obtained from peripheral blood mononuclear cells of 7 active RA patients and from 10 HS after stimulation with phorbol myristate acetate (5 ng/mL) for 24 h. HS-MDMs were then stimulated with lipopolysaccharide (LPS, 1 mg/mL) for 4 h to induce M1-MDMs. M1-MDMs and RA-MDMs were treated with CTLA4-Ig (100 μM and 500 μM) for 3, 12, 24, and 48 h. The gene expression of CD80, CD86, and TLR4 (M1 markers); CD163, CD204, and CD206 (surface M2 markers); and MerTK (functional M2 marker) was evaluated by qRT-PCR. The protein synthesis of surface M2 markers was investigated by Western blotting. The statistical analysis was performed by the Wilcoxon t-test. Results In LPS-induced HS-M1-MDMs, CTLA4-Ig 100 μM and 500 μM significantly downregulated the gene expression of M1 markers (3 h p<0.01 for all molecules; 12 h p<0.05 for TLR4 and CD86) and significantly upregulated that of M2 markers, primarily after 12 h of treatment (CD163: p < 0.01 and p < 0.05; CD206: p < 0.05 and p < 0.01; CD204: p < 0.05 by 100 mg/mL). Moreover, in these cells, CTLA4-Ig 500 μM increased the protein synthesis of surface M2 markers (p < 0.05). Similarly, in RA-MDMs, the CTLA4-Ig treatment significantly downregulated the gene expression of M1 markers at both concentrations primarily after 12 h (p < 0.05). Furthermore, both concentrations of CTLA4-Ig significantly upregulated the gene expression of CD206 (after 3 h of treatment; p < 0.05), CD163, and MerTK (after 12 h of treatment, p < 0.05), whereas CD204 gene expression was significantly upregulated by the high concentration of CTLA4-Ig (p < 0.05). The protein synthesis of all surface markers was increased primarily by CTLA4-Ig 500 μM, significantly for CD204 and CD206 after 24 h of treatment (p < 0.05). Conclusions CTLA4-Ig treatment seems to induce the in vitro shift from M1 to M2 macrophages, of both HS-M1-MDMs and RA-MDMs, as observed by the significant downregulation exerted on selected M1 markers and the upregulation of selected M2 markers suggesting an additional mechanism for its modulation of the RA inflammatory process. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02691-9.
Collapse
Affiliation(s)
- Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy.
| | - Stefano Soldano
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Paola Montagna
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Alberto Sulli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Vanessa Smith
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium.,Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Center (IRC), Ghent, Belgium
| | - Samuele Tardito
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| |
Collapse
|
14
|
Guo K, Xiao N, Liu Y, Wang Z, Tóth J, Gyenis J, Thakur VK, Oyane A, Shubhra QT. Engineering polymer nanoparticles using cell membrane coating technology and their application in cancer treatments: Opportunities and challenges. NANO MATERIALS SCIENCE 2021. [DOI: 10.1016/j.nanoms.2021.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol 2021; 18:824-837. [PMID: 34253911 DOI: 10.1038/s41569-021-00582-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
T cells are among the most common cell types present in atherosclerotic plaques and are increasingly being recognized as a central mediator in atherosclerosis development and progression. At the same time, triglycerides and fatty acids have re-emerged as crucial risk factors for atherosclerosis. Triglycerides and fatty acids are important components of the milieu to which the T cell is exposed from the circulation to the plaque, and increasing evidence shows that fatty acids influence T cell function. In this Review, we discuss the effects of fatty acids on four components of the T cell response - metabolism, activation, proliferation and polarization - and the influence of these changes on the pathogenesis of atherosclerosis. We also discuss how quiescent T cells can undergo a type of metabolic reprogramming induced by exposure to fatty acids in the circulation that influences the subsequent functions of T cells after activation, such as in atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands.
- Netherlands Heart Institute, Utrecht, Netherlands.
| |
Collapse
|
16
|
Woodburn SC, Bollinger JL, Wohleb ES. The semantics of microglia activation: neuroinflammation, homeostasis, and stress. J Neuroinflammation 2021; 18:258. [PMID: 34742308 PMCID: PMC8571840 DOI: 10.1186/s12974-021-02309-6] [Citation(s) in RCA: 244] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia are emerging as critical regulators of neuronal function and behavior in nearly every area of neuroscience. Initial reports focused on classical immune functions of microglia in pathological contexts, however, immunological concepts from these studies have been applied to describe neuro-immune interactions in the absence of disease, injury, or infection. Indeed, terms such as 'microglia activation' or 'neuroinflammation' are used ubiquitously to describe changes in neuro-immune function in disparate contexts; particularly in stress research, where these terms prompt undue comparisons to pathological conditions. This creates a barrier for investigators new to neuro-immunology and ultimately hinders our understanding of stress effects on microglia. As more studies seek to understand the role of microglia in neurobiology and behavior, it is increasingly important to develop standard methods to study and define microglial phenotype and function. In this review, we summarize primary research on the role of microglia in pathological and physiological contexts. Further, we propose a framework to better describe changes in microglia1 phenotype and function in chronic stress. This approach will enable more precise characterization of microglia in different contexts, which should facilitate development of microglia-directed therapeutics in psychiatric and neurological disease.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
17
|
Ceprian N, Valera G, Caro J, Yuste C, Serroukh N, González de Pablos I, Oliva C, Figuer A, Praga M, Alique M, Ramirez R, Morales E, Carracedo J. Effect of Kidney Transplantation on Accelerated Immunosenescence and Vascular Changes Induced by Chronic Kidney Disease. Front Med (Lausanne) 2021; 8:705159. [PMID: 34646838 PMCID: PMC8502880 DOI: 10.3389/fmed.2021.705159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Kidney transplantation is the best option for patients with end-stage renal disease. Despite the improvement in cardiovascular burden (leading cause of mortality among patients with chronic kidney disease), cardiovascular adverse outcomes related to the inflammatory process remain a problem. Thus, the aim of the present study was to characterize the immune profile and microvesicles of patients who underwent transplantation. We investigated the lymphocyte phenotype (CD3, CD4, CD8, CD19, and CD56) and monocyte phenotype (CD14, CD16, CD86, and CD54) in peripheral blood, and endothelium-derived microvesicles (annexin V+CD31+CD41–) in plasma of patients with advanced chronic kidney disease (n = 40), patients with transplantation (n = 40), and healthy subjects (n = 18) recruited from the University Hospital “12 de Octubre” (Madrid, Spain). Patients with kidney transplantation had B-cell lymphopenia, an impairment in co-stimulatory (CD86) and adhesion (CD54) molecules in monocytes, and a reduction in endothelium-derived microvesicles in plasma. The correlations between those parameters explained the modifications in the expression of co-stimulatory and adhesion molecules in monocytes caused by changes in lymphocyte populations, as well as the increase in the levels of endothelial-derived microvesicles in plasma caused by changes in lymphocyte and monocytes populations. Immunosuppressive treatment could directly or indirectly induce those changes. Nevertheless, the particular characteristics of these cells may partly explain the persistence of cardiovascular and renal alterations in patients who underwent transplantation, along with the decrease in arteriosclerotic events compared with advanced chronic kidney disease. In conclusion, the expression of adhesion molecules by monocytes and endothelial-derived microvesicles is related to lymphocyte alterations in patients with kidney transplantation.
Collapse
Affiliation(s)
- Noemi Ceprian
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense de Madrid, Instituto de Investigacin Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Gemma Valera
- Departamento Biología de Sistemas (Unidad Fisiología), Facultad de Medicina, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá de Henares, Madrid, Spain
| | - Jara Caro
- Departamento de Nefrología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria 12 de Octubre, Madrid, Spain
| | - Claudia Yuste
- Departamento de Nefrología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria 12 de Octubre, Madrid, Spain
| | - Nadia Serroukh
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense de Madrid, Instituto de Investigacin Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Carlos Oliva
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense de Madrid, Madrid, Spain
| | - Andrea Figuer
- Departamento Biología de Sistemas (Unidad Fisiología), Facultad de Medicina, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá de Henares, Madrid, Spain
| | - Manuel Praga
- Departamento de Nefrología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria 12 de Octubre, Madrid, Spain
| | - Matilde Alique
- Departamento Biología de Sistemas (Unidad Fisiología), Facultad de Medicina, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá de Henares, Madrid, Spain
| | - Rafael Ramirez
- Departamento Biología de Sistemas (Unidad Fisiología), Facultad de Medicina, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá de Henares, Madrid, Spain
| | - Enrique Morales
- Departamento de Nefrología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria 12 de Octubre, Madrid, Spain
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense de Madrid, Instituto de Investigacin Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| |
Collapse
|
18
|
Khan N, Kaur S, Knuth CM, Jeschke MG. CNS-Spleen Axis - a Close Interplay in Mediating Inflammatory Responses in Burn Patients and a Key to Novel Burn Therapeutics. Front Immunol 2021; 12:720221. [PMID: 34539655 PMCID: PMC8448279 DOI: 10.3389/fimmu.2021.720221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
Severe burn-induced inflammation and subsequent hypermetabolic response can lead to profound infection and sepsis, resulting in multiple organ failure and high mortality risk in patients. This represents an extremely challenging issue for clinicians as sepsis is the leading cause of mortality in burn patients. Since hyperinflammation and immune dysfunction are a result of an immune imbalance, restoring these conditions seem to have promising benefits for burn patients. A key network that modulates the immune balance is the central nervous system (CNS)-spleen axis, which coordinates multiple signaling pathways, including sympathetic and parasympathetic pathways. Modulating inflammation is a key strategy that researchers use to understand neuroimmunomodulation in other hyperinflammatory disease models and modulating the CNS-spleen axis has led to improved clinical outcomes in patients. As the immune balance is paramount for recovery in burn-induced sepsis and patients with hyperinflammatory conditions, it appears that severe burn injuries substantially alter this CNS-spleen axis. Therefore, it is essential to address and discuss the potential therapeutic techniques that target the CNS-spleen axis that aim to restore homeostasis in burn patients. To understand this in detail, we have conducted a systematic review to explore the role of the CNS-spleen axis and its impact on immunomodulation concerning the burn-induced hypermetabolic response and associated sepsis complications. Furthermore, this thorough review explores the role of the spleen, CNS-spleen axis in the ebb and flow phases following a severe burn, how this axis induces metabolic factors and immune dysfunction, and therapeutic techniques and chemical interventions that restore the immune balance via neuroimmunomodulation.
Collapse
Affiliation(s)
- Noorisah Khan
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Supreet Kaur
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Carly M Knuth
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Martí I Líndez AA, Reith W. Arginine-dependent immune responses. Cell Mol Life Sci 2021; 78:5303-5324. [PMID: 34037806 PMCID: PMC8257534 DOI: 10.1007/s00018-021-03828-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence indicates that, over the course of evolution of the immune system, arginine has been selected as a node for the regulation of immune responses. An appropriate supply of arginine has long been associated with the improvement of immune responses. In addition to being a building block for protein synthesis, arginine serves as a substrate for distinct metabolic pathways that profoundly affect immune cell biology; especially macrophage, dendritic cell and T cell immunobiology. Arginine availability, synthesis, and catabolism are highly interrelated aspects of immune responses and their fine-tuning can dictate divergent pro-inflammatory or anti-inflammatory immune outcomes. Here, we review the organismal pathways of arginine metabolism in humans and rodents, as essential modulators of the availability of this semi-essential amino acid for immune cells. We subsequently review well-established and novel findings on the functional impact of arginine biosynthetic and catabolic pathways on the main immune cell lineages. Finally, as arginine has emerged as a molecule impacting on a plethora of immune functions, we integrate key notions on how the disruption or perversion of arginine metabolism is implicated in pathologies ranging from infectious diseases to autoimmunity and cancer.
Collapse
Affiliation(s)
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
20
|
Nagashimada M, Sawamoto K, Ni Y, Kitade H, Nagata N, Xu L, Kobori M, Mukaida N, Yamashita T, Kaneko S, Ota T. CX3CL1-CX3CR1 Signaling Deficiency Exacerbates Obesity-induced Inflammation and Insulin Resistance in Male Mice. Endocrinology 2021; 162:6188411. [PMID: 33765141 DOI: 10.1210/endocr/bqab064] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Indexed: 12/21/2022]
Abstract
The CX3CL1-CX3CR1 system plays an important role in disease progression by regulating inflammation both positively and negatively. We reported previously that C-C chemokine receptors 2 and 5 promote obesity-associated adipose tissue inflammation and insulin resistance. Here, we demonstrate that CX3CL1-CX3CR1 signaling is involved in adipose tissue inflammation and insulin resistance in obese mice via adipose tissue macrophage recruitment and M1/M2 polarization. Cx3cl1 expression was persistently decreased in the epididymal white adipose tissue (eWAT) of high-fat diet-induced obese (DIO) mice, despite increased expression of other chemokines. Interestingly, in Cx3cr1-/- mice, glucose tolerance, insulin resistance, and hepatic steatosis induced by DIO or leptin deficiency were exacerbated. CX3CL1-CX3CR1 signaling deficiency resulted in reduced M2-polarized macrophage migration and an M1-dominant shift of macrophages within eWAT. Furthermore, transplantation of Cx3cr1-/- bone marrow was sufficient to impair glucose tolerance, insulin sensitivity, and regulation of M1/M2 status. Moreover, Cx3cl1 administration in vivo led to the attenuation of glucose intolerance and insulin resistance. Thus, therapy targeting the CX3CL1-CX3CR1 system may be beneficial in the treatment of type 2 diabetes by regulating M1/M2 macrophages.
Collapse
Affiliation(s)
- Mayumi Nagashimada
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Kazuki Sawamoto
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| | - Yinhua Ni
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
- College of Biological Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Hironori Kitade
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| | - Naoto Nagata
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| | - Liang Xu
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| | - Masuko Kobori
- Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Tatsuya Yamashita
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| | - Shuichi Kaneko
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| | - Tsuguhito Ota
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
21
|
Cordes F, Lenker E, Weinhage T, Spille LJ, Bettenworth D, Varga G, Schmidt HH, Foell D. Impaired IFN-γ-dependent STAT3 Activation Is Associated With Dysregulation of Regulatory and Inflammatory Signaling in Monocytes of Ulcerative Colitis Patients. Inflamm Bowel Dis 2021; 27:887-901. [PMID: 33165509 DOI: 10.1093/ibd/izaa280] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The Janus kinase/signal transducer and activator of transcription (JAK/STAT) inhibitor tofacitinib has been recently approved for the treatment of ulcerative colitis (UC) but not Crohn's disease (CD). Systematic analysis of the JAK/STAT pathway in inflammatory bowel disease is still missing. The aim of this study was to investigate JAK/STAT activation and adjacent signaling in monocytes of patients with inflammatory bowel diseases, which are key players in inflammatory responses. METHODS Blood samples of active UC (n = 28) and CD patients (n = 28) and healthy controls (n = 22) were collected for primary monocyte investigation. STAT phosphorylation (pSTAT), cytokine secretion, and surface marker expression ± prior tofacitinib blockade in addition to Th-17 and regulatory T cell induction in cocultures were analyzed upon interferon (IFN)-γ timulation. RESULTS Baseline frequencies of pSTAT1+ and pSTAT3+ monocytes were significantly higher in UC, whereas IFN-γ-associated crosstalk induction of pSTAT3+ monocytes was missing in UC-derived monocytes compared with controls and CD. This coincided with decreased interleukin (IL)-10 and cluster of differentiation (CD)39 levels, diminished regulatory T cell (Treg) induction, and increased IL-12 and IL-23 secretion compared with controls, which was not observed in CD monocytes. Tofacitinib induced stronger inhibition of inflammatory cytokine release (IL-6, TNFα, IL-12, IL-23) in UC compared with CD monocytes. CONCLUSIONS In UC monocytes, IFN-γ-associated activation of the JAK/STAT pathway is impaired with an imbalance between STAT1 and STAT3, coinciding with stronger induction of inflammatory monocytes by IFN-γ compared with controls or CD. The fact that tofacitinib had stronger regulatory impact on UC than on CD monocytes further underlines a stronger inflammatory involvement of the JAK/STAT pathway in UC pathogenesis, which might result from missing STAT3 activation to counteract STAT1-induced inflammation.
Collapse
Affiliation(s)
- Friederike Cordes
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Eva Lenker
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Toni Weinhage
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Lea J Spille
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Hartmut H Schmidt
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
22
|
Zhang C, Xu C, Gao L, Li X, Zhao C. Porphyromonas gingivalis lipopolysaccharide promotes T-hel per17 cell differentiation by upregulating Delta-like ligand 4 expression on CD14 + monocytes. PeerJ 2021; 9:e11094. [PMID: 33981487 PMCID: PMC8074840 DOI: 10.7717/peerj.11094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/21/2021] [Indexed: 12/17/2022] Open
Abstract
Backgroud To investigate the effect and mechanism of Porphyromonas gingivalis (P. gingivalis) lipopolysaccharide (LPS) on Th17 cell differentiation mediated by CD14+ monocytes. Methods P. gingivalis LPS-activated CD14+ monocytes were co-cultured with CD4+T cells in different cell ratios. An indirect co-culture system was also established using transwell chambers. Furthermore, anti- Delta-like ligand 4 (Dll-4) antibody was used to investigate the role of Dll-4 in Th17 cell response. The mRNA expression was analyzed using quantitative reverse transcription-polymerase chain reaction, and secreted cytokines in culture supernatant were detected using enzyme-linked immunosorbent assay. Flow cytometry was used to determine the frequencies of Th17 cells. IL-17 protein expression levels were determined using western blotting assay. Results P. gingivalis LPS increased the expressions of interleukin (IL)-1β, IL-6, IL-23 and transforming growth factor (TGF)-β in CD14+ monocytes. Th17 cell frequency upregulated, which is not solely cytokine-dependent but rather requires cell-cell contact with activated monocytes, particularly in the 1:10 cell ratio. Furthermore, P. gingivalis LPS increased t he expression of Dll-4 on CD14+ monocytes, whereas the anti- Dll-4 a ntibody decreased the response of Th17 cells. The results suggest that P. gingivalis LPS enhances Th17 cell response via Dll-4 upregulation on CD14+ monocytes.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chenrong Xu
- Department of Periodontology, Guangdong Provincial Hospital of Stomatology, Stomatological Hospital of Southern Medical University, Guangzhou, China
| | - Li Gao
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiting Li
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chuanjiang Zhao
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Lee SJ, Yoon BR, Kim HY, Yoo SJ, Kang SW, Lee WW. Activated Platelets Convert CD14 +CD16 - Into CD14 +CD16 + Monocytes With Enhanced FcγR-Mediated Phagocytosis and Skewed M2 Polarization. Front Immunol 2021; 11:611133. [PMID: 33488616 PMCID: PMC7817612 DOI: 10.3389/fimmu.2020.611133] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Monocytes are important cellular effectors of innate immune defense. Human monocytes are heterogeneous and can be classified into three distinct subsets based on CD14 and CD16 expression. The expansion of intermediate CD14+CD16+ monocytes has been reported in chronic inflammatory diseases including rheumatoid arthritis (RA). However, the mechanism underlying induction of CD16 and its role in monocytes remains poorly understood. Here, we demonstrate that activated platelets are important for induction of CD16 on classical CD14+CD16- monocytes by soluble factors such as cytokines. Cytokine neutralization and signaling inhibition assays reveal that sequential involvement of platelet-derived TGF-β and monocyte-derived IL-6 contribute to CD16 induction on CD14+CD16- monocytes. Activated platelet-induced CD16 on monocytes participates in antibody-dependent cellular phagocytosis (ADCP) and its level is positively correlated with phagocytic activity. CD14+CD16- monocytes treated with activated platelets preferentially differentiate into M2 macrophages, likely the M2c subset expressing CD163 and MerTK. Lastly, the amount of sCD62P, a marker of activated platelets, is significantly elevated in plasma of RA patients and positively correlates with clinical parameters of RA. Our findings suggest an important role of activated platelets in modulating phenotypical and functional features of human monocytes. This knowledge increases understanding of the immunological role of CD14+CD16+ cells in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Su Jeong Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Bo Ruem Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea
| | - Su-Jin Yoo
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea.,Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
24
|
Conte FL, Santiago KB, Conti BJ, Cardoso EDO, Oliveira LPG, Feltran GDS, Zambuzzi WF, Golim MDA, Cruz MT, Sforcin JM. Propolis from southeastern Brazil produced by Apis mellifera affects innate immunity by modulating cell marker expression, cytokine production and intracellular pathways in human monocytes. J Pharm Pharmacol 2020; 73:135-144. [PMID: 33793799 DOI: 10.1093/jpp/rgaa023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/15/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Propolis is a bee-made product used for centuries due to its diverse biological properties, including its immunomodulatory action. This work aimed at investigating whether propolis may affect monocyte functions challenged with retinoic acid (RA), B subunit of Escherichia coli heat-labile enterotoxin (EtxB), human melanoma-associated antigen-1 (MAGE-1) and lipopolysaccharide (LPS). METHODS Monocytes from healthy donors were treated with the stimuli separately or in the presence of propolis. Cell viability was evaluated by MTT assay, cell marker expression was assessed by flow cytometry, cytokine production by ELISA, gene expression by RT-qPCR. KEY FINDINGS Propolis alone maintained TLR-2, TLR-4, HLA-DR, CD40 and CD80 expression in the monocytes; however, its combination with either MAGE-1 or LPS decreased CD40 expression triggered by the stimuli. Propolis maintained RA action on cell marker expression. Propolis inhibited TNF-α (with either EtxB or MAGE-1) and IL-6 (with either RA or MAGE-1), and increased IL-10 (with MAGE-1) production. Propolis downmodulated LC3 expression induced by LPS. It also induced a lower NF-kB expression than control cells and its combination with RA induced a higher expression than the stimulus alone. CONCLUSIONS Propolis potentially affected innate immunity by downmodulating the monocytes pro-inflammatory activity.
Collapse
Affiliation(s)
- Fernanda Lopes Conte
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Karina Basso Santiago
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Bruno José Conti
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Eliza de Oliveira Cardoso
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Lucas Pires Garcia Oliveira
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Geórgia da Silva Feltran
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Willian Fernando Zambuzzi
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Marjorie de Assis Golim
- Botucatu Blood Center, School of Medicine, São Paulo State University (UNESP), São Paulo, Brazil
| | - Maria Teresa Cruz
- Center for Neurosciences and Cellular Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - José Maurício Sforcin
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
25
|
Huang W, Lan Q, Jiang L, Yan W, Tang F, Shen C, Huang H, Zhong H, Lv J, Zeng S, Li M, Mo Z, Hu B, Liang N, Chen Q, Zhang M, Xu F, Cui L. Fasudil attenuates glial cell-mediated neuroinflammation via ERK1/2 and AKT signaling pathways after optic nerve crush. Mol Biol Rep 2020; 47:8963-8973. [PMID: 33161529 DOI: 10.1007/s11033-020-05953-y] [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: 07/05/2020] [Accepted: 10/28/2020] [Indexed: 11/28/2022]
Abstract
To investigate the functional role of fasudil in optic nerve crush (ONC), and further explore its possible molecular mechanism. After ONC injury, the rats were injected intraperitoneally either with fasudil or normal saline once a day until euthanized. RGCs survival was assessed by retrograde labeling with FluoroGold. Retinal glial cells activation and population changes (GFAP, iba-1) were measured by immunofluorescence. The expressions of cleaved caspase 3 and 9, p-ERK1/2 and p-AKT were detected by western blot. The levels of the pro-inflammatory cytokines were determined using real-time polymerase chain reaction. Fasudil treatment inhibited RGCs apoptosis and reduced RGCs loss demonstrated by the decreased apoptosis-associated proteins expression and the increased fluorogold labeling of RGCs after ONC, respectively. In addition, the ONC + fasudil group compared had a significantly lower expression of GFAP and iba1 compared with the ONC group. The levels of pro-inflammatory cytokines were significantly reduced in the ONC + fasudil group than in the ONC group. Furthermore, the phosphorylation levels of ERK1/2 and AKT (p-ERK1/2 and p-AKT) were obviously elevated by the fasudil treatment. Our study demonstrated that fasudil attenuated glial cell-mediated neuroinflammation by up-regulating the ERK1/2 and AKT signaling pathways in rats ONC models. We conclude that fasudil may be a novel treatment for traumatic optic neuropathy.
Collapse
Affiliation(s)
- Wei Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China.,Guangxi Medical University, Nanning, 530021, China
| | - Qianqian Lan
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Li Jiang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Wenya Yan
- Guangzhou Medical University, Guangzhou, 511436, China
| | - Fen Tang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Chaolan Shen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Hui Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Haibin Zhong
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Jian Lv
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Siming Zeng
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Min Li
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Zhongxiang Mo
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Bing Hu
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Ning Liang
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Qi Chen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Mingyuan Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Fan Xu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China.
| | - Ling Cui
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China.
| |
Collapse
|
26
|
The role of peripheral monocytes and macrophages in ischemic stroke. Neurol Sci 2020; 41:3589-3607. [PMID: 33009963 DOI: 10.1007/s10072-020-04777-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
Abstract
After acute ischemic stroke (AIS), peripheral monocytes infiltrate into the lesion site within 24 h, peak at 3 to 7 days, and then differentiate into macrophages. Traditionally, monocytes/macrophages (MMs) are thought to play a deleterious role in AIS. Depletion of MMs in the acute phase can alleviate brain injury induced by ischemia. However, several studies have shown that MMs have anti-inflammatory functions, participate in angiogenesis, phagocytose necrotic neurons, and promote neurovascular repair. Therefore, MMs play dual roles in ischemic stroke, depending mainly upon the MM microenvironment and the window of time post-stroke. Because activated microglia and MMs are similar in morphology and function, previous studies have often investigated them together. However, recent studies have used special methods to distinguish MMs from microglia and have found that MMs have properties which differ from microglia. Here, we review the unique role of MMs and the interaction between MMs and neurovascular units, including neurons, astrocytes, microglia, and microvessels. Future therapeutics targeting MMs should regulate the polarization and subset transformation of the MMs at different stages of AIS rather than comprehensively suppressing MM infiltration and differentiation. In addition, more studies are needed to elucidate the cellular and molecular mechanisms of MM subsets and polarization during ischemic stroke.
Collapse
|
27
|
Béland LC, Markovinovic A, Jakovac H, De Marchi F, Bilic E, Mazzini L, Kriz J, Munitic I. Immunity in amyotrophic lateral sclerosis: blurred lines between excessive inflammation and inefficient immune responses. Brain Commun 2020; 2:fcaa124. [PMID: 33134918 PMCID: PMC7585698 DOI: 10.1093/braincomms/fcaa124] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Despite wide genetic, environmental and clinical heterogeneity in amyotrophic lateral sclerosis, a rapidly fatal neurodegenerative disease targeting motoneurons, neuroinflammation is a common finding. It is marked by local glial activation, T cell infiltration and systemic immune system activation. The immune system has a prominent role in the pathogenesis of various chronic diseases, hence some of them, including some types of cancer, are successfully targeted by immunotherapeutic approaches. However, various anti-inflammatory or immunosuppressive therapies in amyotrophic lateral sclerosis have failed. This prompted increased scrutiny over the immune-mediated processes underlying amyotrophic lateral sclerosis. Perhaps the biggest conundrum is that amyotrophic lateral sclerosis pathogenesis exhibits features of three otherwise distinct immune dysfunctions-excessive inflammation, autoimmunity and inefficient immune responses. Epidemiological and genome-wide association studies show only minimal overlap between amyotrophic lateral sclerosis and autoimmune diseases, so excessive inflammation is usually thought to be secondary to protein aggregation, mitochondrial damage or other stresses. In contrast, several recently characterized amyotrophic lateral sclerosis-linked mutations, including those in TBK1, OPTN, CYLD and C9orf72, could lead to inefficient immune responses and/or damage pile-up, suggesting that an innate immunodeficiency may also be a trigger and/or modifier of this disease. In such cases, non-selective immunosuppression would further restrict neuroprotective immune responses. Here we discuss multiple layers of immune-mediated neuroprotection and neurotoxicity in amyotrophic lateral sclerosis. Particular focus is placed on individual patient mutations that directly or indirectly affect the immune system, and the mechanisms by which these mutations influence disease progression. The topic of immunity in amyotrophic lateral sclerosis is timely and relevant, because it is one of the few common and potentially malleable denominators in this heterogenous disease. Importantly, amyotrophic lateral sclerosis progression has recently been intricately linked to patient T cell and monocyte profiles, as well as polymorphisms in cytokine and chemokine receptors. For this reason, precise patient stratification based on immunophenotyping will be crucial for efficient therapies.
Collapse
Affiliation(s)
| | - Andrea Markovinovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Ervina Bilic
- Department of Neurology, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Letizia Mazzini
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, Quebec G1J 2G3, Canada
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
28
|
Cordes F, Foell D, Ding JN, Varga G, Bettenworth D. Differential regulation of JAK/STAT-signaling in patients with ulcerative colitis and Crohn’s disease. World J Gastroenterol 2020; 26:4055-4075. [PMID: 32821070 PMCID: PMC7403801 DOI: 10.3748/wjg.v26.i28.4055] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/24/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
In 2018, the pan-Janus kinase (JAK) inhibitor tofacitinib was launched for the treatment of ulcerative colitis (UC). Although tofacitinib has proven efficacious in patients with active UC, it failed in patients with Crohn’s disease (CD). This finding strongly hints at a different contribution of JAK signaling in both entities. Here, we review the current knowledge on the interplay between the JAK/signal transducer and activator of transcription (STAT) pathway and inflammatory bowel diseases (IBD). In particular, we provide a detailed overview of the differences and similarities of JAK/STAT-signaling in UC and CD, highlight the impact of the JAK/STAT pathway in experimental colitis models and summarize the published evidence on JAK/STAT-signaling in immune cells of IBD as well as the genetic association between the JAK/STAT pathway and IBD. Finally, we describe novel treatment strategies targeting JAK/STAT inhibition in UC and CD and comment on the limitations and challenges of the new drug class.
Collapse
Affiliation(s)
- Friederike Cordes
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - John Nik Ding
- Department of Gastroenterology, St. Vincent’s Hospital, Melbourne 3002, Australia
- Department of Medicine, University of Melbourne, East Melbourne 3002, Australia
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| |
Collapse
|
29
|
Miyagawa F, Tagaya Y, Ozato K, Horie K, Asada H. Inflammatory monocyte-derived dendritic cells mediate autoimmunity in murine model of systemic lupus erythematosus. J Transl Autoimmun 2020; 3:100060. [PMID: 32743540 PMCID: PMC7388367 DOI: 10.1016/j.jtauto.2020.100060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Using a mouse model of systemic lupus erythematosus (SLE), we recently demonstrated that the two major manifestations of SLE are mechanistically independent because the type I IFN pathway leads to the autoantibody production whereas the NF-κB activation is sufficient for the development of glomerulonephritis. To further advance our understandings on the molecular pathways regulating the development of SLE, we studied the role of IRF8 because it controls both type I IFN and NF-κB pathways and saw that IRF8-deficient mice failed to develop either glomerulonephritis or the autoantibody production. Furthermore, these genetically engineered mice prompted us to realize the important role of Ly6Chigh inflammatory monocytes in the development of SLE. These monocytes migrate to the peritoneal cavity in WT and IRF7-deficient mice but not in IRF8-deficient mice, and there they produce both type I IFN and proinflammatory cytokines in WT mice, while in IRF7-deficient mice they only produce proinflammatory cytokines. Upon migration to the spleen, Ly6Chigh inflammatory monocytes differentiate into dendritic cells (DCs) which are capable of producing proinflammatory cytokines in response to dsDNA autoantigen. Collectively, type I IFN produced from inflammatory monocytes/monocyte-derived DCs might be essential for autoantibody production whereas proinflammatory cytokines produced from them might mediate tissue damages in this model. Our study reveals a specialized role for monocyte-derived antigen presenting cells in autoimmunity. Plasticity of monocyte might play an important role not only in the pathogenesis of the disease but also in flare-ups of the disease.
Collapse
Affiliation(s)
- Fumi Miyagawa
- Department of Dermatology, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Yutaka Tagaya
- Cell Biology Lab, Division of Infectious Agents and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Keiko Ozato
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyoji Horie
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|
30
|
Qi Y, Sun D, Yang W, Xu B, Lv D, Han Y, Sun M, Jiang S, Hu W, Yang Y. Mammalian Sterile 20-Like Kinase (MST) 1/2: Crucial Players in Nervous and Immune System and Neurological Disorders. J Mol Biol 2020; 432:3177-3190. [PMID: 32198112 DOI: 10.1016/j.jmb.2020.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/25/2020] [Accepted: 03/09/2020] [Indexed: 12/28/2022]
Abstract
As central components of the Hippo signaling pathway in mammals, the mammalian sterile 20-like kinase 1 (MST1) and MST2 protein kinases regulate cell proliferation, survival, and death and are involved in the homeostasis of many tissues. Recent studies have elucidated the roles of MST1 and MST2 in the nervous system and immune system, particularly in neurological disorders, which are influenced by aging. In this review, we provide a comprehensive overview of these research areas. First, the activation mechanisms and roles of MST1 and MST2 in neurons, non-neuronal cells, and immune cells are introduced. The roles of MST1 and MST2 in neurological disorders, including brain tumors, cerebrovascular diseases, neurodegenerative disorders, and neuromuscular disorders, are then presented. Finally, the existing obstacles for further research are discussed. Collectively, the information compiled herein provides a common framework for the function of MST1 and MST2 in the nervous system, should contribute to the design of further experiments, and sheds light on potential treatments for aging associated neurological disorders.
Collapse
Affiliation(s)
- Yating Qi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Baoping Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Dewen Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yuehu Han
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Meng Sun
- Department of Cardiology, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Wei Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| |
Collapse
|
31
|
Cordes F, Lenker E, Spille LJ, Weinhage T, Bettenworth D, Kessel C, Schmidt HH, Foell D, Varga G. Tofacitinib Reprograms Human Monocytes of IBD Patients and Healthy Controls Toward a More Regulatory Phenotype. Inflamm Bowel Dis 2020; 26:391-406. [PMID: 31560043 DOI: 10.1093/ibd/izz213] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The inhibition of Janus kinases (JAKs) and subsequent signal transducers and activators of transcription (STATs) by tofacitinib represents a new therapeutic strategy in inflammatory bowel diseases (IBD) as clinical trials have led to approval of tofacitinib for ulcerative colitis (UC) and hint at a possible efficacy for Crohn`s disease (CD). However, the impact of tofacitinib on cellular response of monocytes, which are key players in inflammatory responses, has not been investigated so far. We aimed to analyze JAK/STAT-inhibition by tofacitinib in monocytes of IBD patients and healthy controls. METHODS Primary monocytes of IBD patients with active disease and healthy controls (n = 18) were analyzed for cytokine expression and phenotype after granulocyte macrophage colony-stimulating factor (GM-CSF)/interferon (IFN)γ-stimulation and tofacitinib pretreatment (1-1000 nM) and capacity to induce Foxp3+-regulatory T cells (Tregs) in cocultures. In total, 20 UC patients and 21 CD patients were included. Additionally, dose-dependent inhibition of JAK/STAT-phosphorylation was analyzed in controls. RESULTS Pro-inflammatory costimulation with GM-CSF/IFNγ resulted in significant tumor necrosis factor (TNFα) and interleukin (IL)-6 increase, whereas IL-10 expression decreased in monocytes. Tofacitinib modulated the responses of activated monocytes toward a regulatory phenotype through reduced TNFα and IL-6 secretion and enhanced Treg induction in cocultures. However, in monocytes from active IBD patients, higher tofacitinib dosages were needed for blockade of pro-inflammatory cytokines. Tofacitinib induced stronger regulatory phenotypes in monocytes of UC patients, including more effective inhibition of pro-inflammatory pathways and better restoration of anti-inflammatory mechanisms as compared with CD-derived monocytes. CONCLUSION Tofacitinib dose-dependently reprograms monocytes toward a more regulatory cell type. This beneficial effect possibly results from selective JAK/STAT-blockade by adequate tofacitinib dosage with inhibition of pro-inflammatory responses and permission of a balance-shift toward regulatory pathways.
Collapse
Affiliation(s)
- Friederike Cordes
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Eva Lenker
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Lea J Spille
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Toni Weinhage
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Christoph Kessel
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Hartmut H Schmidt
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
32
|
Hurdayal R, Nieuwenhuizen NE, Khutlang R, Brombacher F. Inflammatory Dendritic Cells, Regulated by IL-4 Receptor Alpha Signaling, Control Replication, and Dissemination of Leishmania major in Mice. Front Cell Infect Microbiol 2020; 9:479. [PMID: 32039054 PMCID: PMC6992597 DOI: 10.3389/fcimb.2019.00479] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/27/2019] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis is a vector-borne disease caused by Leishmania parasites. Macrophages are considered the primary parasite host cell, but dendritic cells (DCs) play a critical role in initiating adaptive immunity and controlling Leishmania infection. Accordingly, our previous study in CD11ccreIL-4Rα−/lox mice, which have impaired IL-4 receptor alpha (IL-4Rα) expression on CD11c+ cells including DCs, confirmed a protective role for IL-4/IL-13-responsive DCs in replication and dissemination of parasites during cutaneous leishmaniasis. However, it was unclear which DC subset/s was executing this function. To investigate this, we infected CD11ccreIL-4Rα−/lox and control mice with L. major GFP+ parasites and identified subsets of infected DCs by flow cytometry. Three days after infection, CD11b+ DCs and CD103+ DCs were the main infected DC subsets in the footpad and draining lymph node, respectively and by 4 weeks post-infection, Ly6C+ and Ly6C− CD11b+ DCs were the main infected DC populations in both the lymph nodes and footpads. Interestingly, Ly6C+CD11b+ inflammatory monocyte-derived DCs but not Ly6C−CD11b+ DCs hosted parasites in the spleen. Importantly, intracellular parasitism was significantly higher in IL-4Rα-deficient DCs. In terms of DC effector function, we found no change in the expression of pattern-recognition receptors (TLR4 and TLR9) nor in expression of the co-stimulatory marker, CD80, but MHCII expression was lower in CD11ccreIL-4Rα−/lox mice at later time-points compared to the controls. Interestingly, in CD11ccreIL-4Rα−/lox mice, which have reduced Th1 responses, CD11b+ DCs had impaired iNOS production, suggesting that DC IL-4Rα expression and NO production is important for controlling parasite numbers and preventing dissemination. Expression of the alternative activation marker arginase was unchanged in CD11b+ DCs in CD11creIL-4Rα−/lox mice compared to littermate controls, but RELM-α was upregulated, suggesting IL-4Rα-independent alternative activation. In summary, L. major parasites may use Ly6C+CD11b+ inflammatory DCs derived from monocytes recruited to infection as “Trojan horses” to migrate to secondary lymphoid organs and peripheral sites, and DC IL-4Rα expression is important for controlling infection.
Collapse
Affiliation(s)
- Ramona Hurdayal
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Natalie Eva Nieuwenhuizen
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rethabile Khutlang
- Identity Authentication Research Group, Defence and Security, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, South African Medical Research Council on Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
33
|
Liu C, Choi MW, Xue X, Cheung PCK. Immunomodulatory Effect of Structurally Characterized Mushroom Sclerotial Polysaccharides Isolated from Polyporus rhinocerus on Bone Marrow Dendritic Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12137-12143. [PMID: 31566976 DOI: 10.1021/acs.jafc.9b03294] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This study evaluated the immunomodulatory effects of two high-molecular-weight and structurally different mushroom polysaccharides, an alkali-soluble polysaccharide (mPRSon) and a water-soluble polysaccharide-protein complex (PRW), isolated previously from the sclerotia of Pleurotus rhinocerus, on the maturation of murine bone-marrow-derived dendritic cells (BMDCs). The effects of mPRSon and PRW on the expression of morphological change, surface molecules, phagocytic activity, and cytokine release in BMDCs were determined by flow cytometry and a mouse cytokine array. The results showed that both mPRSon and PRW could induce phenotypic and functional maturation of BMDCs. At the same time, mPRSon upregulated the expression of membrane phenotypic marker CD86 and PRW markedly upregulated CD40, CD80, and CD86. In addition, mPRSon could bind to the dectin-1 receptor and stimulate the release of MIP-1α, MIP-2, and IL-2, while PRW could bind to complement receptor 3 and toll-like receptor 2 with an upregulation of the expression of IL-2, IL-6, MIP-1α, MIP-2, RANTES, IL-12p40p70, IL-12p70, TIMP-1, IFN-γ, KC, MCP-1, and GCSF. The study provides additional information on how structural differences in sclerotial polysaccharides influence their immunomodulatory activities on BMDCs involving different PAMP receptors. It is anticipated that more understanding of the interactions between the sclerotial polysaccharides and their receptors in immune cells can facilitate their future application for cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoran Liu
- Medical Research Center , The People's Hospital of Longhua , Shenzhen , 518109 , People's Republic of China
- Food and Nutritional Sciences, School of Life Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong Special Administrative Region of the People's Republic of China
| | - Man Wing Choi
- Food and Nutritional Sciences, School of Life Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong Special Administrative Region of the People's Republic of China
| | - Xingkui Xue
- Medical Research Center , The People's Hospital of Longhua , Shenzhen , 518109 , People's Republic of China
| | - Peter C K Cheung
- Food and Nutritional Sciences, School of Life Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong Special Administrative Region of the People's Republic of China
| |
Collapse
|
34
|
Jian Z, Liu R, Zhu X, Smerin D, Zhong Y, Gu L, Fang W, Xiong X. The Involvement and Therapy Target of Immune Cells After Ischemic Stroke. Front Immunol 2019; 10:2167. [PMID: 31572378 PMCID: PMC6749156 DOI: 10.3389/fimmu.2019.02167] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
After ischemic stroke, the integrity of the blood-brain barrier is compromised. Peripheral immune cells, including neutrophils, T cells, B cells, dendritic cells, and macrophages, infiltrate into the ischemic brain tissue and play an important role in regulating the progression of ischemic brain injury. In this review, we will discuss the role of different immune cells after stroke in the secondary inflammatory reaction and focus on the phenotypes and functions of macrophages in ischemic stroke, as well as briefly introduce the anti-ischemic stroke therapy targeting macrophages.
Collapse
Affiliation(s)
- Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology and Toxicology, Shandong Institute for Food and Drug Control, Jinan, China
| | - Xiqun Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daniel Smerin
- Department of Neurosurgery, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
35
|
Lin W, Xu D, Austin CD, Caplazi P, Senger K, Sun Y, Jeet S, Young J, Delarosa D, Suto E, Huang Z, Zhang J, Yan D, Corzo C, Barck K, Rajan S, Looney C, Gandham V, Lesch J, Liang WC, Mai E, Ngu H, Ratti N, Chen Y, Misner D, Lin T, Danilenko D, Katavolos P, Doudemont E, Uppal H, Eastham J, Mak J, de Almeida PE, Bao K, Hadadianpour A, Keir M, Carano RAD, Diehl L, Xu M, Wu Y, Weimer RM, DeVoss J, Lee WP, Balazs M, Walsh K, Alatsis KR, Martin F, Zarrin AA. Function of CSF1 and IL34 in Macrophage Homeostasis, Inflammation, and Cancer. Front Immunol 2019; 10:2019. [PMID: 31552020 PMCID: PMC6736990 DOI: 10.3389/fimmu.2019.02019] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Colony-stimulating factor 1 (CSF1) and interleukin 34 (IL34) signal via the CSF1 receptor to regulate macrophage differentiation. Studies in IL34- or CSF1-deficient mice have revealed that IL34 function is limited to the central nervous system and skin during development. However, the roles of IL34 and CSF1 at homeostasis or in the context of inflammatory diseases or cancer in wild-type mice have not been clarified in vivo. By neutralizing CSF1 and/or IL34 in adult mice, we identified that they play important roles in macrophage differentiation, specifically in steady-state microglia, Langerhans cells, and kidney macrophages. In several inflammatory models, neutralization of both CSF1 and IL34 contributed to maximal disease protection. However, in a myeloid cell-rich tumor model, CSF1 but not IL34 was required for tumor-associated macrophage accumulation and immune homeostasis. Analysis of human inflammatory conditions reveals IL34 upregulation that may account for the protection requirement of IL34 blockade. Furthermore, evaluation of IL34 and CSF1 blockade treatment during Listeria infection reveals no substantial safety concerns. Thus, IL34 and CSF1 play non-redundant roles in macrophage differentiation, and therapeutic intervention targeting IL34 and/or CSF1 may provide an effective treatment in macrophage-driven immune-pathologies.
Collapse
Affiliation(s)
- WeiYu Lin
- Genentech, South San Francisco, CA, United States
| | - Daqi Xu
- Genentech, South San Francisco, CA, United States
| | | | | | - Kate Senger
- Genentech, South San Francisco, CA, United States
| | - Yonglian Sun
- Genentech, South San Francisco, CA, United States
| | | | - Judy Young
- Genentech, South San Francisco, CA, United States
| | | | - Eric Suto
- Genentech, South San Francisco, CA, United States
| | - Zhiyu Huang
- Genentech, South San Francisco, CA, United States
| | - Juan Zhang
- Genentech, South San Francisco, CA, United States
| | - Donghong Yan
- Genentech, South San Francisco, CA, United States
| | - Cesar Corzo
- Genentech, South San Francisco, CA, United States
| | - Kai Barck
- Genentech, South San Francisco, CA, United States
| | | | | | | | - Justin Lesch
- Genentech, South San Francisco, CA, United States
| | | | - Elaine Mai
- Genentech, South San Francisco, CA, United States
| | - Hai Ngu
- Genentech, South San Francisco, CA, United States
| | | | - Yongmei Chen
- Genentech, South San Francisco, CA, United States
| | - Dinah Misner
- Genentech, South San Francisco, CA, United States
| | - Tori Lin
- Genentech, South San Francisco, CA, United States
| | | | | | | | | | | | - Judy Mak
- Genentech, South San Francisco, CA, United States
| | | | | | | | - Mary Keir
- Genentech, South San Francisco, CA, United States
| | | | - Lauri Diehl
- Genentech, South San Francisco, CA, United States
| | - Min Xu
- Genentech, South San Francisco, CA, United States
| | - Yan Wu
- Genentech, South San Francisco, CA, United States
| | | | - Jason DeVoss
- Genentech, South San Francisco, CA, United States
| | - Wyne P Lee
- Genentech, South San Francisco, CA, United States
| | | | - Kevin Walsh
- Genentech, South San Francisco, CA, United States
| | | | | | - Ali A Zarrin
- Genentech, South San Francisco, CA, United States
| |
Collapse
|
36
|
Hey YY, O'Neill TJ, O'Neill HC. A novel myeloid cell in murine spleen defined through gene profiling. J Cell Mol Med 2019; 23:5128-5143. [PMID: 31210415 PMCID: PMC6653018 DOI: 10.1111/jcmm.14382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
A novel myeloid antigen presenting cell can be generated through in vitro haematopoiesis in long‐term splenic stromal cocultures. The in vivo equivalent subset was recently identified as phenotypically and functionally distinct from the spleen subsets of macrophages, conventional (c) dendritic cells (DC), resident monocytes, inflammatory monocytes and eosinophils. This novel subset which is myeloid on the basis of cell surface phenotype, but dendritic‐like on the basis of cell surface marker expression and antigen presenting function, has been tentatively labelled “L‐DC.” Transcriptome analysis has now been employed to determine the lineage relationship of this cell type with known splenic cDC and monocyte subsets. Principal components analysis showed separation of “L‐DC” and monocytes from cDC subsets in the second principal component. Hierarchical clustering then indicated a close lineage relationship between this novel subset, resident monocytes and inflammatory monocytes. Resident monocytes were the most closely aligned, with no genes specifically expressed by the novel subset. This subset, however, showed upregulation of genes reflecting both dendritic and myeloid lineages, with strong upregulation of several genes, particularly CD300e. While resident monocytes were found to be dependent on Toll‐like receptor signalling for development and were reduced in number in Myd88‐/‐ and Trif‐/‐ mutant mice, both the novel subset and inflammatory monocytes were unaffected. Here, we describe a novel myeloid cell type closely aligned with resident monocytes in terms of lineage but distinct in terms of development and functional capacity.
Collapse
Affiliation(s)
- Ying-Ying Hey
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
| | | | - Helen C O'Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
| |
Collapse
|
37
|
Cespuglio R, Amrouni D, Raymond EF, Bouteille B, Buguet A. Cerebral inducible nitric oxide synthase protein expression in microglia, astrocytes and neurons in Trypanosoma brucei brucei-infected rats. PLoS One 2019; 14:e0215070. [PMID: 30995270 PMCID: PMC6469759 DOI: 10.1371/journal.pone.0215070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/26/2019] [Indexed: 11/18/2022] Open
Abstract
To study the anatomo-biochemical substrates of brain inflammatory processes, Wistar male rats were infected with Trypanosoma brucei brucei. With this reproducible animal model of human African trypanosomiasis, brain cells (astrocytes, microglial cells, neurons) expressing the inducible nitric oxide synthase (iNOS) enzyme were revealed. Immunohistochemistry was achieved for each control and infected animal through eight coronal brain sections taken along the caudorostral axis of the brain (brainstem, cerebellum, diencephalon and telencephalon). Specific markers of astrocytes (anti-glial fibrillary acidic protein), microglial cells (anti-integrin alpha M) or neurons (anti-Neuronal Nuclei) were employed. The iNOS staining was present in neurons, astrocytes and microglial cells, but not in oligodendrocytes. Stained astrocytes and microglial cells resided mainly near the third cavity in the rostral part of brainstem (periaqueductal gray), diencephalon (thalamus and hypothalamus) and basal telencephalon. Stained neurons were scarce in basal telencephalon, contrasting with numerous iNOS-positive neuroglial cells. Contrarily, in dorsal telencephalon (neocortex and hippocampus), iNOS-positive neurons were plentiful, contrasting with the marked paucity of labelled neuroglial (astrocytes and microglial) cells. The dual distribution between iNOS-labelled neuroglial cells and iNOS-labelled neurons is a feature that has never been described before. Functionalities attached to such a divergent distribution are discussed.
Collapse
Affiliation(s)
- Raymond Cespuglio
- Neuroscience Research Centre of Lyon (CRNL), Neurochem, Faculty of Medicine, Claude-Bernard Lyon-1 University, Lyon, France
- Sechenov 1st Moscow State Medical University, Laboratory of Psychiatric Neurobiology, Moscow, Russia
| | - Donia Amrouni
- Neuroscience Research Centre of Lyon (CRNL), Neurochem, Faculty of Medicine, Claude-Bernard Lyon-1 University, Lyon, France
| | - Elizabeth F. Raymond
- Faculty of Medicine, team EA 4171, Claude-Bernard Lyon-1 University, Lyon, France
| | - Bernard Bouteille
- Department of Parasitology, Dupuytren University Hospital, Limoges, France
| | - Alain Buguet
- Malaria Research Unit, UMR 5246 CNRS, Claude-Bernard Lyon-1 University, Villeurbanne, France
| |
Collapse
|
38
|
Knobloch J, Panek S, Yanik SD, Jamal Jameel K, Bendella Z, Jungck D, Bürger P, Bülthoff E, Struck B, Giannakis N, Rupp J, Kronsbein J, Peters M, Koch A. The monocyte-dependent immune response to bacteria is suppressed in smoking-induced COPD. J Mol Med (Berl) 2019; 97:817-828. [PMID: 30929031 DOI: 10.1007/s00109-019-01778-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 03/04/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022]
Abstract
COPD patients have an increased susceptibility to bacterial airway infections that can induce exacerbations. In response to infections, circulating monocytes become recruited to the infected tissue and secrete cytokines. We hypothesized that this cytokine response is reduced in COPD. Cultured peripheral blood monocytes of never smokers (NS) and smokers without (S) and with COPD (3 study populations, n = 36-37) were stimulated with extracts of Haemophilus influenzae, Staphylococcus aureus, or Streptococcus pneumoniae or with four different pathogen-associated molecular patterns (PAMPs). Four cytokines and 9 PAMP-related signaling molecules were measured and compared between the groups. Granulocyte-macrophage-colony-stimulating-factor responses to all stimulants were reduced in S and COPD compared to NS. Tumor-necrosis-factor-α responses to all bacterial extracts, peptidoglycan, and lipopolysaccharide were reduced in S and/or COPD. Interleukin-10 responses to S. aureus and lipoteichoic acid were increased in COPD. Correlations to pack-years and lung function were found. The peptidoglycan-receptor NOD2 and the mRNA of the lipopolysaccharide-receptor TLR4 were reduced in S and COPD. Cytokine responses of monocytes to bacteria are suppressed by smoking and in COPD possibly due to NOD2 and TLR4 reduction and/or interleukin-10 increase. This might help to explain the increased susceptibility to bacterial infections. These systemic molecular pathologies might be targets for therapeutic strategies to prevent infection-induced exacerbations. KEY MESSAGES: COPD subjects have an increased susceptibility to bacterial infections. This implies defects in the immune response to bacteria and is critical for disease progression. The cytokine response of monocytes to bacteria is reduced in COPD. This might be due to a reduced NOD2 and TLR4 and an increased IL-10 expression. This can explain the increased susceptibility to infections and help to identify drug targets.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany.
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany.
| | - Susanne Panek
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Sarah Derya Yanik
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Kaschin Jamal Jameel
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Zeynep Bendella
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Department of Radiology, University of Bonn Medical Center, Bonn, Germany
| | - David Jungck
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
- Department of Pneumology, Clinic III for Internal Medicine, University of Cologne, Cologne, Germany
- Department of Internal Medicine II, Pneumology, Allergology and Respiratory Medicine, Bethel Teaching Hospital, Berlin, Germany
| | - Paul Bürger
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Eike Bülthoff
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Birte Struck
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Nikolaos Giannakis
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology, Allergology, Sleep- and Respiratory Medicine, Bergmannsheil University Hospital, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Germany
| | - Marcus Peters
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | - Andrea Koch
- Medical Clinic V, Ludwig-Maximilians-University LMU, Munich, Germany
- German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
39
|
Wang J, Li C, Fu J, Wang X, Feng X, Pan X. Tim-3 regulates inflammatory cytokine expression and Th17 cell response induced by monocytes from patients with chronic hepatitis B. Scand J Immunol 2019; 89:e12755. [PMID: 30729555 DOI: 10.1111/sji.12755] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 02/06/2023]
Abstract
Tim-3 is expressed on monocytes/macrophages and is involved in the regulation of inflammatory responses. The aim of this study was to determine the effect of Tim-3 on inflammatory response triggered by peripheral monocytes from patients with chronic hepatitis B (CHB). Tim-3 expression on peripheral monocytes and frequency of Th17 cells in peripheral blood mononuclear cells (PBMCs) derived from CHB patients were detected. Followed by lipopolysaccharides (LPS) activation of circulating monocytes from CHB patients, expression of inflammatory cytokines including TNF-α,IL-1β and IL-6 were examined in the presence and absence of Galectin-9 which is the ligand for Tim-3. Subsequently, after purified CD4+T cells were cocultured with LPS-activated monocytes from CHB patients in the presence of anti-Tim-3 antibody, percentage of Th17 cells and production of IL-17 were measured. Tim-3 expression was significantly upregulated and closely correlated to the frequency of Th17 cells in patients with CHB. Expression of TNF-α,IL-1β and IL-6 increased significantly in monocytes stimulated with LPS and Galectin-9, compared to LPS stimulation alone. LPS-activated monocytes from CHB patients could drive differentiation of memory CD4+T cells to Th17 cells. However, under the blockade of Tim-3 signalling by anti-Tim-3 antibody, percentage of Th17 cells and production of IL-17 decreased significantly. Our results demonstrate that upregulated expression of Tim-3 on circulating monocytes accelerates inflammatory response by promoting production of inflammatory cytokines and Th17 responses in CHB.
Collapse
Affiliation(s)
- Junyan Wang
- Department of Infectious Disease, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chan Li
- Department of Infectious Disease, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Juanjuan Fu
- Department of Infectious Disease, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xia Wang
- Department of Infectious Disease, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xia Feng
- Central Laboratory of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiucheng Pan
- Department of Infectious Disease, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
40
|
Luo L, Han Y, Song X, Zhu T, Zeng Y, Li T. CD16-expressing monocytes correlate with arterial stiffness in HIV-infected ART-naïve men. HIV CLINICAL TRIALS 2019; 19:39-45. [PMID: 29770747 DOI: 10.1080/15284336.2018.1437863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Objectives To determine the association of the markers of monocyte activation and arterial stiffness among HIV-infected antiretroviral therapy (ART)-naïve men. Methods Sixty HIV-infected ART-naïve men and 20 HIV-uninfected male controls without symptoms or history of cardiovascular disease were recruited. Pulse wave velocity (PWV) were used as the marker of arterial stiffness and determined using a pulse pressure analyzer. The percentage of CD16-expressing monocytes was used as a marker of monocyte activation. Plasma neopterin concentration, one of the monocyte/macrophage activation markers and plasma tissue factor (TF), the coagulation marker in response to inflammatory stimuli, were also analyzed. Multivariate analyses were used to explore the association of the percentage of CD16-expressing monocytes with arterial stiffness in HIV-infected men. Results HIV-infected ART-naïve men demonstrated significantly higher PWV (1252.8 ± 161.6 vs.1159.2 ± 108.3 cm/s, p = 0.018). The percentage of CD16-expressing monocytes was significantly higher in HIV-infected men comparing male controls (23.4 ± 6.0% vs. 19.6 ± 4.6%, p = 0.012). Plasma concentrations of neopterin (0.91 vs. 0.64 ng/ml), p < 0.001) and TF (5.29 vs. 4.43 pg/ml, p = 0.04) were higher in HIV-infected men comparing controls. In the multivariate model for PWV among HIV-infected men, the percentage of CD16-expressing monocytes (p = 0.023) and age (p = 0.017) were significantly associated with PWV. HIV viral load, CD4 count, percentage of CD8+CD38+T cells and percentage of CD8+HLA-DR+ T cells were not associated with PWV. Discussion Higher level of monocyte activation marker is associated with higher level of arterial stiffness in ART naïve HIV-infected men. HIV viral load, CD4 count, and the markers of CD8 T cell activation were unrelated to PWV.
Collapse
Affiliation(s)
- Ling Luo
- a Department of Infectious Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing , China
| | - Yang Han
- a Department of Infectious Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing , China
| | - Xiaojing Song
- a Department of Infectious Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing , China
| | - Ting Zhu
- a Department of Infectious Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing , China
| | - Yong Zeng
- b Department of Cardiovascular Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing , China
| | - Taisheng Li
- a Department of Infectious Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing , China
| |
Collapse
|
41
|
Ha JW, You MJ, Park HS, Kim JW, Kwon MS. Differential effect of LPS and paclitaxel on microglial functional phenotypes and circulating cytokines: the possible role of CX3CR1 and IL-4/10 in blocking persistent inflammation. Arch Pharm Res 2019; 42:359-368. [PMID: 30852731 DOI: 10.1007/s12272-019-01137-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/23/2019] [Indexed: 10/27/2022]
Abstract
Neuroinflammation plays a role in cancer chemotherapy-induced chronic pain. Thus far, most studies have focused on neuroinflammation suppression. However, there are limited reports of which factor is involved in the transition from acute inflammation to chronic inflammation, resulting in neuroinflammation and chronic pain. Here, we compared the inflammatory reaction and pain response induced by LPS and paclitaxel. LPS (0.5 mg/kg) or paclitaxel (2 mg/kg/day for 5 days) was administered intraperitoneally to mice, and mechanical allodynia was examined by von Frey test. LPS induced transient mechanical allodynia, whereas paclitaxel induced persistent mechanical allodynia. The CD86/CX3CR1 ratio remained unchanged due to CX3CR1 elevation following LPS injection, whereas the ratio was increased on day 1 after paclitaxel injection. LPS also increased CD45, CCL2, and CCL5 mRNA in the spinal cord and circulating pro- and anti-inflammatory cytokines 1 day after injection; however, the pattern was not consistent. Paclitaxel gradually increased inflammatory cytokines in the spinal cord. CX3CR1 might be involved in blocking the transition from acute pain to persistent pain in the LPS group. In addition, serum IL-4 and IL-10 elevation in the LPS group may be associated with chronic pain prevention. Therefore, targeting CX3CR1, IL-4, and IL-10 might be an alternative therapeutic strategy.
Collapse
Affiliation(s)
- Joong-Won Ha
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.,Department of Orthopedic Surgery, National Health Insurance Service Ilsan Hospital, Ilsan, 10444, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyun-Sun Park
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jong Wan Kim
- Department of Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 40 Sukwoo-Dong, Hwaseong-Si, Gyeonggi-do, 14068, Republic of Korea.
| | - Min-Soo Kwon
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
42
|
Ropert C. How toll-like receptors reveal monocyte plasticity: the cutting edge of antiinflammatory therapy. Cell Mol Life Sci 2019; 76:745-755. [PMID: 30413835 PMCID: PMC11105477 DOI: 10.1007/s00018-018-2959-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/23/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLR)s are central in immune response by recognizing pathogen-associated molecular patterns (PAMP)s. If they are essential to eliminate pathogens in earlier stages of infection, they also might play a role in homeostasis and tissue repair. TLR versatility parallels the plasticity of monocytes, which represent an heterogeneous population of immune cells. They are rapidly recruited to sites of infection and involved in clearance of pathogens and in tissue healing. This review underlines how TLRs have proved to be an interesting tool to study the properties of monocytes and why different therapeutic strategies exploring monocyte plasticity may be relevant in the context of chronic inflammatory disorders.
Collapse
Affiliation(s)
- Catherine Ropert
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Pampulha, Belo Horizonte, MG, 31270-910, Brazil.
| |
Collapse
|
43
|
de Gaetano M, McEvoy C, Andrews D, Cacace A, Hunter J, Brennan E, Godson C. Specialized Pro-resolving Lipid Mediators: Modulation of Diabetes-Associated Cardio-, Reno-, and Retino-Vascular Complications. Front Pharmacol 2018; 9:1488. [PMID: 30618774 PMCID: PMC6305798 DOI: 10.3389/fphar.2018.01488] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022] Open
Abstract
Diabetes and its associated chronic complications present a healthcare challenge on a global scale. Despite improvements in the management of chronic complications of the micro-/macro-vasculature, their growing prevalence and incidence highlights the scale of the problem. It is currently estimated that diabetes affects 425 million people globally and it is anticipated that this figure will rise by 2025 to 700 million people. The vascular complications of diabetes including diabetes-associated atherosclerosis and kidney disease present a particular challenge. Diabetes is the leading cause of end stage renal disease, reflecting fibrosis leading to organ failure. Moreover, diabetes associated states of inflammation, neo-vascularization, apoptosis and hypercoagulability contribute to also exacerbate atherosclerosis, from the metabolic syndrome to advanced disease, plaque rupture and coronary thrombosis. Current therapeutic interventions focus on regulating blood glucose, glomerular and peripheral hypertension and can at best slow the progression of diabetes complications. Recently advanced knowledge of the pathogenesis underlying diabetes and associated complications revealed common mechanisms, including the inflammatory response, insulin resistance and hyperglycemia. The major role that inflammation plays in many chronic diseases has led to the development of new strategies aiming to promote the restoration of homeostasis through the "resolution of inflammation." These strategies aim to mimic the spontaneous activities of the 'specialized pro-resolving mediators' (SPMs), including endogenous molecules and their synthetic mimetics. This review aims to discuss the effect of SPMs [with particular attention to lipoxins (LXs) and resolvins (Rvs)] on inflammatory responses in a series of experimental models, as well as evidence from human studies, in the context of cardio- and reno-vascular diabetic complications, with a brief mention to diabetic retinopathy (DR). These data collectively support the hypothesis that endogenously generated SPMs or synthetic mimetics of their activities may represent lead molecules in a new discipline, namely the 'resolution pharmacology,' offering hope for new therapeutic strategies to prevent and treat, specifically, diabetes-associated atherosclerosis, nephropathy and retinopathy.
Collapse
Affiliation(s)
- Monica de Gaetano
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Caitriona McEvoy
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
- Renal Transplant Program, University Health Network, Toronto, ON, Canada
| | - Darrell Andrews
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Antonino Cacace
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Jonathan Hunter
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute and UCD School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
44
|
Pérez-Mazliah DE, Castro Eiro MD, Álvarez MG, Lococo B, Bertocchi G, César G, Natale MA, Albareda MC, Viotti R, Laucella SA. Distinct monocyte subset phenotypes in patients with different clinical forms of chronic Chagas disease and seronegative dilated cardiomyopathy. PLoS Negl Trop Dis 2018; 12:e0006887. [PMID: 30346948 PMCID: PMC6211766 DOI: 10.1371/journal.pntd.0006887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 11/01/2018] [Accepted: 10/01/2018] [Indexed: 01/16/2023] Open
Abstract
Background Chronic infection with Trypanosoma cruzi leads to a constant stimulation of the host immune system. Monocytes, which are recruited in response to inflammatory signals, are divided into classical CD14hiCD16—, non-classical CD14loCD16+ and intermediate CD14hiCD16+ subsets. In this study, we evaluated the frequencies of monocyte subsets in the different clinical stages of chronic Chagas disease in comparison with the monocyte profile of seronegative heart failure subjects and seronegative healthy controls. The effect of the anti-parasite drug therapy benznidazole on monocyte subsets was also explored. Methodology/Principal findings The frequencies of the different monocyte subsets and their phenotypes were measured by flow cytometry. Trypanosoma cruzi-specific antibodies were quantified by conventional serological tests. T. cruzi-infected subjects with mild or no signs of cardiac disease and patients suffering from dilated cardiomyopathy unrelated to T. cruzi infection showed increased levels of non-classical CD14loCD16+ monocytes compared with healthy controls. In contrast, the monocyte profile in T. cruzi-infected subjects with severe cardiomyopathy was skewed towards the classical and intermediate subsets. After benznidazole treatment, non-classical monocytes CD14loCD16+ decreased while classical monocytes CD14hiCD16—increased. Conclusions/Significance The different clinical stages of chronic Chagas disease display distinct monocyte profiles that are restored after anti-parasite drug therapy. T. cruzi-infected subjects with severe cardiac disease displayed a profile of monocytes subsets suggestive of a more pronounced inflammatory environment compared with subjects suffering from heart failure not related to T. cruzi infection, supporting that parasite persistence might also alter cell components of the innate immune system. Monocytes are key players during infection, and they leave the bloodstream and migrate into tissues in response to inflammatory signals. Although the recruitment of monocytes is essential for the effective control and clearance of microorganisms, they can also be highly damaging to neighboring tissues. Based on the expression of CD14 and CD16, monocytes are classified into classical, non-classical and intermediate subsets, all of which exert different functions. Because chronic T. cruzi infection induces a constant activation of the host immune system, inflammatory signals are exacerbated, possibly leading to alterations in the frequencies of monocyte subsets. In this study, we evaluated the monocyte profile in Trypanosoma cruzi-infected subjects with different degrees of cardiac dysfunction and explored whether this profile was similar between seropositive and seronegative subjects with heart failure. We found that the different clinical stages of chronic Chagas disease displayed distinct monocyte profiles, which are susceptible to being restored by modulating the parasite load with anti-parasite drug therapy. T. cruzi-infected subjects with severe cardiac disease displayed a profile of monocytes subsets suggestive of a more pronounced inflammatory environment compared with subjects suffering from heart failure not related to T. cruzi infection, supporting that parasite persistence might be a detrimental factor in the evolution of the cardiac disease induced by T. cruzi.
Collapse
Affiliation(s)
| | | | | | - Bruno Lococo
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Graciela Bertocchi
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Gonzalo César
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - María A. Natale
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - María C. Albareda
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Rodolfo Viotti
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Susana A. Laucella
- Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
45
|
Yao K, Zhao YF. Aging modulates microglia phenotypes in neuroinflammation of MPTP-PD mice. Exp Gerontol 2018; 111:86-93. [DOI: 10.1016/j.exger.2018.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/14/2018] [Accepted: 07/10/2018] [Indexed: 01/25/2023]
|
46
|
Effects of chronic heat stress on lactational performance and the transcriptomic profile of blood cells in lactating dairy goats. J DAIRY RES 2018; 85:423-430. [PMID: 30236165 DOI: 10.1017/s0022029918000705] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
High temperature is a major stress that negatively affects welfare, health, and productivity of dairy animals. Heat-stressed animals are more prone to disease, suggesting that their immunity is hindered. Although productive and physiologic responses of dairy animals to heat stress are well known, there is still limited information on the response at the transcriptome level. Our objective was to evaluate the changes in performance and blood transcriptomics of dairy goats under heat stress. Eight multiparous Murciano-Granadina dairy goats in mid-lactation were assigned to 1 of 2 climatic treatments for 35 d. Treatments and temperature-humidity index (THI) were: (1) thermal neutral (TN: n = 4; 15-20 °C, 40-45%, THI = 59-65), and (2) heat stress (HS: n = 4; 12 h at 37 °C-40%, THI = 86; 12 h at 30 °C-40%, THI = 77). Rectal temperature, respiratory rate, feed intake and milk yield were recorded daily. Additionally, milk composition was evaluated weekly. Blood samples were collected at d 35 and RNA was extracted for microarray analyses (Affymetrix GeneChip Bovine Genome Array). Differences in rectal temperature and respiratory rate between HS and TN goats were maximal during the first 3 d of the experiment, reduced thereafter, but remained significant throughout the 35-d experimental period. Heat stress reduced feed intake, milk yield, milk protein and milk fat contents by 29, 8, 12, and 13%, respectively. Microarray analysis of blood revealed that 55 genes were up-regulated, whereas 88 were down-regulated by HS. Bioinformatics analysis using the Dynamic Impact Approach revealed that 31 biological pathways were impacted by HS. Pathways associated with leukocyte transendothelial migration, cell adhesion, hematopoietic cell lineage, calcium signaling, and PPAR signaling were negatively impacted by HS, whereas nucleotide metabolism was activated. In conclusion, heat stress not only negatively affected milk production in dairy goats, but also resulted in alterations in the functionality of immune cells, which would make the immune system of heat-stressed goats less capable of fending-off diseases.
Collapse
|
47
|
Merkel SF, Andrews AM, Lutton EM, Razmpour R, Cannella LA, Ramirez SH. Dexamethasone Attenuates the Enhanced Rewarding Effects of Cocaine Following Experimental Traumatic Brain Injury. Cell Transplant 2018; 26:1178-1192. [PMID: 28933216 PMCID: PMC5447499 DOI: 10.1177/0963689717714341] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical studies have identified traumatic brain injury (TBI) as a risk factor for the development of cocaine dependence. This claim is supported by our recent preclinical studies showing enhancement of the rewarding effects of cocaine in mice sustaining moderate controlled cortical impact (CCI) injury during adolescence. Here we test the efficacy of dexamethasone, an anti-inflammatory corticosteroid, to attenuate augmentation of the behavioral response to cocaine observed in CCI-TBI animals using the conditioned place preference (CPP) assay. These studies were performed in order to determine whether proinflammatory activity in the nucleus accumbens (NAc), a key brain nucleus in the reward pathway, mediates enhanced cocaine-induced CPP in adolescent animals sustaining moderate CCI-TBI. Our data reveal robust glial activation in the NAc following CCI-TBI and a significant increase in the cocaine-induced CPP of untreated CCI-TBI mice. Furthermore, our results show that dexamethasone treatment following CCI-TBI can attenuate the cocaine place preference of injured animals without producing aversion in the CPP assay. Our studies also found that dexamethasone treatment significantly reduced the expression of select immune response genes including Monocyte chemoattractant protein-1 (MCP-1/CCL2) and intercellular adhesion molecule-1 ( ICAM-1), returning their expression to control levels, which prompted an investigation of peripheral blood monocytes in dexamethasone-treated animals. Experimental findings showed that no craniectomy/dexamethasone mice had a significant increase, while CCI-TBI/dexamethasone animals had a significant decrease in the percentage of circulating nonclassical patrolling monocytes. These results suggest that a portion of these monocytes may migrate to the brain in response to CCI-TBI, potentially sparing the development of chronic neuroinflammation in regions associated with the reward circuitry such as the NAc. Overall, our findings indicate that anti-inflammatory agents, such as dexamethasone, may be effective in normalizing the rewarding effects of cocaine following CCI-TBI.
Collapse
Affiliation(s)
- Steven F Merkel
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Allison M Andrews
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Evan M Lutton
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Roshanak Razmpour
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Lee Anne Cannella
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Servio H Ramirez
- 1 Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,2 Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,3 Shriners Hospitals Pediatric Research Center, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
48
|
Royal W, Can A, Gould TD, Guo M, Huse J, Jackson M, Davis H, Bryant J. Cigarette smoke and nicotine effects on brain proinflammatory responses and behavioral and motor function in HIV-1 transgenic rats. J Neurovirol 2018; 24:246-253. [PMID: 29644536 DOI: 10.1007/s13365-018-0623-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/21/2018] [Accepted: 02/15/2018] [Indexed: 01/08/2023]
Abstract
Cognitive impairment in HIV-1 infection is associated with the induction of chronic proinflammatory responses in the brains of infected individuals. The risk of HIV-related cognitive impairment is increased by cigarette smoking, which induces brain inflammation in rodent models. To better understand the role of smoking and the associated immune response on behavioral and motor function in HIV infection, wild-type F344 and HIV-1 transgenic (HIV1Tg) rats were exposed to either smoke from nicotine-containing (regular) cigarettes, smoke from nicotine-free cigarettes, or to nicotine alone. The animals were then tested using the rotarod test (RRT), the novel object recognition test (NORT), and the open field test (OFT). Subsequently, brain frontal cortex from the rats was analyzed for levels of TNF-α, IL-1, and IL-6. On the RRT, impairment was noted for F344 rats exposed to either nicotine-free cigarette smoke or nicotine alone and for F344 and HIV1Tg rats exposed to regular cigarette smoke. Effects from the exposures on the OFT were seen only for HIV1Tg rats, for which function was worse following exposure to regular cigarette smoke as compared to exposure to nicotine alone. Expression levels for all three cytokines were overall higher for HIV1Tg than for F344 rats. For HIV1Tg rats, TNF-α, IL-1, and IL-6 gene expression levels for all exposure groups were higher than for control rats. All F344 rat exposure groups also showed significantly increased TNF-α expression levels. However, for F344 rats, IL-1 expression levels were higher only for rats exposed to nicotine-free and nicotine-containing CS, and no increase in IL-6 gene expression was noted with any of the exposures as compared to controls. These studies, therefore, demonstrate that F344 and HIV1Tg rats show differential behavioral and immune effects from these exposures. These effects may potentially reflect differences in the responsiveness of the various brain regions in the two animal species as well as the result of direct toxicity mediated by the proinflammatory cytokines that are produced by HIV proteins and by other factors that are present in regular cigarette smoke.
Collapse
Affiliation(s)
- Walter Royal
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA. .,Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| | - Adem Can
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Todd D Gould
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ming Guo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jared Huse
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Myles Jackson
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Harry Davis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph Bryant
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
49
|
The Biology of Monocytes and Dendritic Cells: Contribution to HIV Pathogenesis. Viruses 2018; 10:v10020065. [PMID: 29415518 PMCID: PMC5850372 DOI: 10.3390/v10020065] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023] Open
Abstract
Myeloid cells such as monocytes, dendritic cells (DC) and macrophages (MΦ) are key components of the innate immune system contributing to the maintenance of tissue homeostasis and the development/resolution of immune responses to pathogens. Monocytes and DC, circulating in the blood or infiltrating various lymphoid and non-lymphoid tissues, are derived from distinct bone marrow precursors and are typically short lived. Conversely, recent studies revealed that subsets of tissue resident MΦ are long-lived as they originate from embryonic/fetal precursors that have the ability to self-renew during the life of an individual. Pathogens such as the human immunodeficiency virus type 1 (HIV-1) highjack the functions of myeloid cells for viral replication (e.g., MΦ) or distal dissemination and cell-to-cell transmission (e.g., DC). Although the long-term persistence of HIV reservoirs in CD4+ T-cells during viral suppressive antiretroviral therapy (ART) is well documented, the ability of myeloid cells to harbor replication competent viral reservoirs is still a matter of debate. This review summarizes the current knowledge on the biology of monocytes and DC during homeostasis and in the context of HIV-1 infection and highlights the importance of future studies on long-lived resident MΦ to HIV persistence in ART-treated patients.
Collapse
|
50
|
Wang J, Li Z, Gao L, Qi Y, Zhu H, Qin X. The regulation effect of AMPK in immune related diseases. SCIENCE CHINA-LIFE SCIENCES 2017; 61:523-533. [DOI: 10.1007/s11427-017-9169-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022]
|