1
|
Ingelfinger F, De Feo D, Becher B. GM-CSF: Master regulator of the T cell-phagocyte interface during inflammation. Semin Immunol 2021; 54:101518. [PMID: 34763973 DOI: 10.1016/j.smim.2021.101518] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/23/2021] [Indexed: 12/21/2022]
Abstract
The role of granulocyte-macrophage colony-stimulating factor (GM-CSF) was sequentially redefined during the past decades. Originally described as a hematopoietic growth factor for myelopoiesis, GM-CSF was recognized as a central mediator of inflammation bridging the innate and adaptive arms of the immune system. Phagocytes sensing GM-CSF adapt an inflammatory phenotype and facilitate pathogen clearance. However, in the context of chronic tissue inflammation, GM-CSF secreted by tissue-invading lymphocytes has detrimental effects by licensing tissue damage and hyperinflammation. Accordingly, therapeutic intervention at the T cell-phagocyte interface represents an attractive target to ameliorate disease progression and immunopathology. Although GM-CSF is largely dispensable for steady state myelopoiesis, dysregulation, as seen in chronic inflammatory diseases, may however lead to disrupted haematopoiesis and long-term effects on bone marrow output. Here, we will survey the role of GM-CSF during inflammation, discuss the extent to which GM-CSF-secreting T cells, debate their introduction as a separate T cell lineage and explore current and future clinical implications of GM-CSF in human disease settings.
Collapse
Affiliation(s)
- Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland; Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Tuzlak S, Dejean AS, Iannacone M, Quintana FJ, Waisman A, Ginhoux F, Korn T, Becher B. Repositioning T H cell polarization from single cytokines to complex help. Nat Immunol 2021; 22:1210-1217. [PMID: 34545250 DOI: 10.1038/s41590-021-01009-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022]
Abstract
When helper T (TH) cell polarization was initially described three decades ago, the TH cell universe grew dramatically. New subsets were described based on their expression of few specific cytokines. Beyond TH1 and TH2 cells, this led to the coining of various TH17 and regulatory (Treg) cell subsets as well as TH22, TH25, follicular helper (TFH), TH3, TH5 and TH9 cells. High-dimensional single-cell analysis revealed that a categorization of TH cells into a single-cytokine-based nomenclature fails to capture the complexity and diversity of TH cells. Similar to the simple nomenclature used to describe innate lymphoid cells (ILCs), we propose that TH cell polarization should be categorized in terms of the help they provide to phagocytes (type 1), to B cells, eosinophils and mast cells (type 2) and to non-immune tissue cells, including the stroma and epithelium (type 3). Studying TH cells based on their helper function and the cells they help, rather than phenotypic features such as individual analyzed cytokines or transcription factors, better captures TH cell plasticity and conversion as well as the breadth of immune responses in vivo.
Collapse
Affiliation(s)
- Selma Tuzlak
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Anne S Dejean
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITy), INSERM UMR1291-CNRS UMR5051-Université Toulouse III, Toulouse, France
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, the Academia, Singapore, Singapore.,Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Maser IP, Hoves S, Bayer C, Heidkamp G, Nimmerjahn F, Eckmann J, Ries CH. The Tumor Milieu Promotes Functional Human Tumor-Resident Plasmacytoid Dendritic Cells in Humanized Mouse Models. Front Immunol 2020; 11:2082. [PMID: 33013879 PMCID: PMC7507800 DOI: 10.3389/fimmu.2020.02082] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Particular interest to harness the innate immune system for cancer immunotherapy is fueled by limitations of immune checkpoint blockade. Plasmacytoid dendritic cells (pDC) are detected in a variety of solid tumors and correlate with poor clinical outcome. Release of type I interferons in response to toll-like-receptor (TLR)7 and TLR9 activation is the pDC hallmark. Mouse and human pDC differ substantially in their biology concerning surface marker expression and cytokine production. Here, we employed humanized mouse models (HIS) to study pDC function. We performed a comprehensive characterization of transgenic, myeloid-enhanced mouse strains (NOG-EXL and NSG-SGM3) expressing human interleukin-3 (hIL-3) and granulocyte-macrophage colony stimulating factor (GM-CSF) using identical humanization protocols. Only in HIS-NOG-EXL mice sufficient pDC infiltration was detectable. Therefore, we selected this strain for subsequent tumor studies. We analyzed pDC frequency in peripheral blood and tumors by comparing HIS-NOG-EXL with HIS-NOG mice bearing three different ovarian and breast tumors. Despite the substantially increased pDC numbers in peripheral blood of HIS-NOG-EXL mice, we detected TLR7/8 agonist responsive and thus functional pDCs only in certain tumor models independent of the mouse strain employed. However, HIS-NOG-EXL mice showed in general a superior humanization phenotype characterized by reconstitution of different myeloid subsets, NK cells and B cells producing physiologic IgG levels. Hence, we provide first evidence that the tumor milieu but not genetically introduced cytokines defines intratumoral (i.t.) frequencies of the rare pDC subset. This study provides model systems to investigate in vivo pro- and anti-tumoral human pDC functions.
Collapse
Affiliation(s)
- Ilona-Petra Maser
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Sabine Hoves
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Christa Bayer
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Gordon Heidkamp
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Falk Nimmerjahn
- FAU Erlangen, Division of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jan Eckmann
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Carola H Ries
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany.,Dr. Carola Ries Consulting, Penzberg, Germany
| |
Collapse
|
4
|
Ramos MI, Garcia S, Helder B, Aarrass S, Reedquist KA, Jacobsen SE, Tak PP, Lebre MC. cDC1 are required for the initiation of collagen-induced arthritis. J Transl Autoimmun 2020; 3:100066. [PMID: 33015599 PMCID: PMC7522802 DOI: 10.1016/j.jtauto.2020.100066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is chronic autoimmune disease which etiology remains unknown. Several cell types have been described to potentiate/aggravate the arthritic process however the initiating event in synovial inflammation is still elusive. Dendritic cells (DCs) are essential for the initiation of primary immune responses and thus we hypothesized that these cells might be crucial for RA induction. DCs are a heterogeneous population of cells comprising different subsets with distinct phenotype and function. Here we investigated which DC subset(s) is/are crucial for the initiation of the arthritic process. We have previously demonstrated that Flt3−/− mice, with reduced DCs, were protected from collagen induced arthritis (CIA). Here we have shown that GM-CSF derived DCs in Flt3L−/− mice are functional but not sufficient to induce arthritis. Batf3−/− mice lacking both CD103+ and CD8α+ cDC1 were resistant to collagen induced arthritis (CIA), demonstrating that this DC subset is crucial for arthritis development. CEP-701 (a Flt3L inhibitor) treatment prevented CIA induction, and reduced dramatically the numbers CD103+ cDC1s present in the lymph nodes and synovium. Hence this study identified cDC1 as the main subset orchestrating the initiation of cell-mediated immunity in arthritis. Flt3L independent DCs present in Flt3L−/− mice are functional but are not sufficient to induce arthritis. BATF3−/− mice lacking cDC1 are protected from arthritis development indicating that cDC1 are necessary for disease induction. Treatment with a Flt3L inhibitor, CEP701, reduced cDC1 populations and prevented arthritis induction.
Collapse
Affiliation(s)
- Maria Ines Ramos
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Samuel Garcia
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Boy Helder
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Saida Aarrass
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Kris A Reedquist
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Sten E Jacobsen
- Haematopoietic Stem Cell Laboratory and MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Maria Cristina Lebre
- Department of Clinical Immunology and Rheumatology.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Russo MA, Georgius P, Pires AS, Heng B, Allwright M, Guennewig B, Santarelli DM, Bailey D, Fiore NT, Tan VX, Latini A, Guillemin GJ, Austin PJ. Novel immune biomarkers in complex regional pain syndrome. J Neuroimmunol 2020; 347:577330. [PMID: 32731051 DOI: 10.1016/j.jneuroim.2020.577330] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023]
Abstract
We investigated serum levels of 29 cytokines and immune-activated kynurenine and tetrahydrobiopterin pathway metabolites in 15 complex regional pain syndrome (CRPS) subjects and 14 healthy controls. Significant reductions in interleukin-37 and tryptophan were found in CRPS subjects, along with positive correlations between kynurenine/tryptophan ratio and TNF-α levels with kinesiophobia, tetrahydrobiopterin levels with McGill pain score, sRAGE, and xanthurenic acid and neopterin levels with depression, anxiety and stress scores. Using machine learning, we identified a set of binary variables, including IL-37 and GM-CSF, capable of distinguishing controls from established CRPS subjects. These results suggest possible involvement of various inflammatory markers in CRPS pathogenesis.
Collapse
Affiliation(s)
- Marc A Russo
- Hunter Pain Specialists, 91 Chatham Street, Broadmeadow, NSW, 2292, Australia; Genesis Research Services, 220 Denison St, Broadmeadow, NSW, 2292, Australia
| | - Peter Georgius
- Pain Rehab, Suite 4 Noosa Central, 6 Bottlebrush Avenue, Sunshine Coast, QLD, 4567, Australia
| | - Ananda Staats Pires
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia; Laboratório de Bioenergética e Estresse Oxidativo, LABOX; Departamento de Bioquímica, CCB; Universidade Federal de Santa Catarina; Florianópolis / SC, Brazil
| | - Benjamin Heng
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Michael Allwright
- ForeFront, Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Boris Guennewig
- ForeFront, Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | | | - Dominic Bailey
- Genesis Research Services, 220 Denison St, Broadmeadow, NSW, 2292, Australia
| | - Nathan T Fiore
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Vanessa X Tan
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Alexandra Latini
- Laboratório de Bioenergética e Estresse Oxidativo, LABOX; Departamento de Bioquímica, CCB; Universidade Federal de Santa Catarina; Florianópolis / SC, Brazil
| | - Gilles J Guillemin
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Paul J Austin
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| |
Collapse
|
6
|
Targeting Granulocyte-Monocyte Colony-Stimulating Factor Signaling in Rheumatoid Arthritis: Future Prospects. Drugs 2020; 79:1741-1755. [PMID: 31486005 DOI: 10.1007/s40265-019-01192-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic, autoimmune disease that affects joints and extra-articular structures. In the last decade, the management of this chronic disease has dramatically changed with the introduction of several targeted mechanisms of action, such as tumor necrosis factor-α inhibition, T-cell costimulation inhibition, B-cell depletion, interleukin-6 blockade, and Janus kinase inhibition. Beyond its well-known hematopoietic role on the proliferation and differentiation of myeloid cells, granulocyte-monocyte colony-stimulating factor (GM-CSF) is a proinflammatory mediator acting as a cytokine, with a proven pathogenetic role in autoimmune disorders such as RA. In vitro studies clearly demonstrated the effect of GM-CSF in the communication between resident tissue cells and activated macrophages at chronic inflammation sites, and confirmed the elevation of GM-CSF levels in inflamed synovial tissue of RA subjects compared with healthy controls. Moreover, a pivotal role of GM-CSF in the perception of pain has been clearly confirmed. Therefore, blockade of the GM-CSF pathway by monoclonal antibodies directed against the cytokine itself or its receptor has been investigated in refractory RA patients. Overall, the safety profile of GM-CSF inhibitors seems to be very favorable, with a particularly low incidence of infectious complications. The efficacy of this new mechanism of action is comparable with main competitors, even though the response rates reported in phase II randomized controlled trials (RCTs) appear to be numerically lower than the response rates observed with other biological disease-modifying antirheumatic drugs already licensed for RA. Mainly because of this reason, nowadays the development program of most GM-CSF blockers for RA has been discontinued, with the exception of otilimab, which is under evaluation in two phase III RCTs with a head-to head non-inferiority design against tofacitinib. These studies will likely be useful for better defining the potential role of GM-CSF inhibition in the therapeutic algorithm of RA. On the other hand, the potential role of GM-CSF blockade in the treatment of other rheumatic diseases is now under investigation. Phase II trials are ongoing with the aim of evaluating mavrilimumab for the treatment of giant cell arteritis, and namilumab for the treatment of spondyloarthritis. Moreover, GM-CSF inhibitors have been tested in osteoarthritis and diffuse subtype of systemic sclerosis. This review aims to describe in detail the available evidence on the GM-CSF blocking pathway in RA management, paving the way to a possible alternative treatment for RA patients. Novel insights regarding the potential use of GM-CSF blockers for alternative indications will be also addressed.
Collapse
|
7
|
Avila-Calderón ED, Flores-Romo L, Sharon W, Donis-Maturano L, Becerril-García MA, Arreola MGA, Reynoso BA, Güemes FS, Contreras-Rodríguez A. Dendritic cells and Brucella spp. interaction: the sentinel host and the stealthy pathogen. Folia Microbiol (Praha) 2020; 65:1-16. [PMID: 30783994 PMCID: PMC7224029 DOI: 10.1007/s12223-019-00691-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/06/2019] [Indexed: 01/18/2023]
Abstract
As dendritic cells (DCs) are among the first cells to encounter antigens, these cells trigger both innate and T cell responses, and are the most potent antigen-presenting cells. Brucella spp., which is an intracellular facultative and stealthy pathogen, is able to evade the bactericidal activities of professional phagocytes. Several studies have demonstrated that Brucella can survive and replicate intracellularly, thereby provoking impaired maturation of DCs. Therefore, the interaction between DCs and Brucella becomes an interesting model to study the immune response. In this review, we first will describe the most common techniques for DCs differentiation in vitro as well as general features of brucellosis. Then, the interaction of DCs and Brucella, including pathogen recognition, molecular mechanisms of bacterial pathogenesis, and intracellular trafficking of Brucella to subvert innate response, will be reviewed. Finally, we will debate diversity in immunological DC response and the controversial role of DC activation against Brucella infection.
Collapse
Affiliation(s)
- Eric Daniel Avila-Calderón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Leopoldo Flores-Romo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
| | - Witonsky Sharon
- Center for Molecular Medicine and Infectious Diseases/Center for One Health, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
| | - Luis Donis-Maturano
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana 3918, Zona Playitas, 22860, Ensenada, Baja California, Mexico
| | - Miguel Angel Becerril-García
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Avenida Francisco I Madero y Dr. Aguirre Pequeño S/N Mitras Centro, 64460, Monterrey, Nuevo León, Mexico
| | - Ma Guadalupe Aguilera Arreola
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Beatriz Arellano Reynoso
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Francisco Suarez Güemes
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Araceli Contreras-Rodríguez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico.
| |
Collapse
|
8
|
Zhan Y, Lew AM, Chopin M. The Pleiotropic Effects of the GM-CSF Rheostat on Myeloid Cell Differentiation and Function: More Than a Numbers Game. Front Immunol 2019; 10:2679. [PMID: 31803190 PMCID: PMC6873328 DOI: 10.3389/fimmu.2019.02679] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 10/30/2019] [Indexed: 12/27/2022] Open
Abstract
Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) is a myelopoietic growth factor that has pleiotropic effects not only in promoting the differentiation of immature precursors into polymorphonuclear neutrophils (PMNs), monocytes/macrophages (MØs) and dendritic cells (DCs), but also in controlling the function of fully mature myeloid cells. This broad spectrum of GM-CSF action may elicit paradoxical outcomes-both immunostimulation and immunosuppression-in infection, inflammation, and cancer. The complexity of GM-CSF action remains to be fully unraveled. Several aspects of GM-CSF action could contribute to its diverse biological consequences. Firstly, GM-CSF as a single cytokine affects development of most myeloid cells from progenitors to mature immune cells. Secondly, GM-CSF activates JAK2/STAT5 and also activate multiple signaling modules and transcriptional factors that direct different biological processes. Thirdly, GM-CSF can be produced by different cell types including tumor cells in response to different environmental cues; thus, GM-CSF quantity can vary greatly under different pathophysiological settings. Finally, GM-CSF signaling is also fine-tuned by other less defined feedback mechanisms. In this review, we will discuss the role of GM-CSF in orchestrating the differentiation, survival, and proliferation during the generation of multiple lineages of myeloid cells (PMNs, MØs, and DCs). We will also discuss the role of GM-CSF in regulating the function of DCs and the functional polarization of MØs. We highlight how the dose of GM-CSF and corresponding signal strength acts as a rheostat to fine-tune cell fate, and thus the way GM-CSF may best be targeted for immuno-intervention in infection, inflammation and cancer.
Collapse
Affiliation(s)
- Yifan Zhan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Michael Chopin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Abstract
Pre-clinical models and clinical trials demonstrate that targeting the action of the cytokine, granulocyte macrophage-colony stimulating factor (GM-CSF), can be efficacious in inflammation/autoimmunity reinforcing the importance of understanding how GM-CSF functions; a significant GM-CSF-responding cell in this context is likely to be the monocyte. This article summarizes critically the literature on the downstream cellular pathways regulating GM-CSF interaction with monocytes (and macrophages), highlighting some contentious issues, and conclusions surrounding this biology. It also suggests future directions which could be undertaken so as to more fully understand this aspect of GM-CSF biology. Given the focus of this collection of articles on monocytes, the following discussion in general will be limited to this population or to its more mature progeny, the macrophage, even though GM-CSF biology is broader than this.
Collapse
Affiliation(s)
- John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia
| |
Collapse
|
10
|
Calmeiro J, Carrascal M, Gomes C, Falcão A, Cruz MT, Neves BM. Biomaterial-based platforms for in situ dendritic cell programming and their use in antitumor immunotherapy. J Immunother Cancer 2019; 7:238. [PMID: 31484548 PMCID: PMC6727507 DOI: 10.1186/s40425-019-0716-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 08/23/2019] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) are central players in the immune system, with an exquisite capacity to initiate and modulate immune responses. These functional characteristics have led to intense research on the development of DC-based immunotherapies, particularly for oncologic diseases. During recent decades, DC-based vaccines have generated very promising results in animal studies, and more than 300 clinical assays have demonstrated the safety profile of this approach. However, clinical data are inconsistent, and clear evidence of meaningful efficacy is still lacking. One of the reasons for this lack of evidence is the limited functional abilities of the used ex vivo-differentiated DCs. Therefore, alternative approaches for targeting and modulating endogenous DC subpopulations have emerged as an attractive concept. Here, we sought to revise the evolution of several strategies for the in situ mobilization and modulation of DCs. The first approaches using chemokine-secreting irradiated tumor cells are addressed, and special attention is given to the cutting-edge injectable bioengineered platforms, programmed to release chemoattractants, tumor antigens and DC maturating agents. Finally, we discuss how our increasing knowledge of DC biology, the use of neoantigens and their combination with immune checkpoint inhibitors can leverage the refinement of these polymeric vaccines to boost their antitumor efficacy.
Collapse
Affiliation(s)
- João Calmeiro
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Mylène Carrascal
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Tecnimede Group, Sintra, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Agra do Crasto - Edifício 30, 3810-193, Aveiro, Portugal.
| |
Collapse
|
11
|
Chrisikos TT, Zhou Y, Slone N, Babcock R, Watowich SS, Li HS. Molecular regulation of dendritic cell development and function in homeostasis, inflammation, and cancer. Mol Immunol 2019; 110:24-39. [PMID: 29549977 PMCID: PMC6139080 DOI: 10.1016/j.molimm.2018.01.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 01/04/2018] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are the principal antigen-presenting cells of the immune system and play key roles in controlling immune tolerance and activation. As such, DCs are chief mediators of tumor immunity. DCs can regulate tolerogenic immune responses that facilitate unchecked tumor growth. Importantly, however, DCs also mediate immune-stimulatory activity that restrains tumor progression. For instance, emerging evidence indicates the cDC1 subset has important functions in delivering tumor antigens to lymph nodes and inducing antigen-specific lymphocyte responses to tumors. Moreover, DCs control specific therapeutic responses in cancer including those resulting from immune checkpoint blockade. DC generation and function is influenced profoundly by cytokines, as well as their intracellular signaling proteins including STAT transcription factors. Regardless, our understanding of DC regulation in the cytokine-rich tumor microenvironment is still developing and must be better defined to advance cancer treatment. Here, we review literature focused on the molecular control of DCs, with a particular emphasis on cytokine- and STAT-mediated DC regulation. In addition, we highlight recent studies that delineate the importance of DCs in anti-tumor immunity and immune therapy, with the overall goal of improving knowledge of tumor-associated factors and intrinsic DC signaling cascades that influence DC function in cancer.
Collapse
Affiliation(s)
- Taylor T Chrisikos
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yifan Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natalie Slone
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rachel Babcock
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| | - Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Onai N, Asano J, Kurosaki R, Kuroda S, Ohteki T. Flexible fate commitment of E2-2high common DC progenitors implies tuning in tissue microenvironments. Int Immunol 2018; 29:443-456. [PMID: 29106601 DOI: 10.1093/intimm/dxx058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/26/2017] [Indexed: 12/16/2022] Open
Abstract
The basic helix-loop-helix transcription factor E2-2 is essential for the development of plasmacytoid dendritic cells (pDCs) but not conventional DCs (cDCs). Here, we generated E2-2 reporter mice and demonstrated that an E2-2high fraction among common DC progenitors, which are a major source of pDCs and cDCs in the steady state, strictly gave rise to pDCs in the presence of Flt3 (Fms-like tyrosine kinase receptor-3) ligand ex vivo or in the secondary lymphoid organs when transferred in vivo. However, in the small intestine, some of these E2-2high progenitors differentiated into cDCs that produced retinoic acid. This transdifferentiation was driven by signaling via the common β receptor, a receptor for the cytokines IL-3, IL-5 and GM-CSF, which are abundant in the gut. In the presence of GM-CSF and Flt3 ligand, E2-2high-progenitor-derived cDCs consistently induced Foxp3+ Treg cells ex vivo. Our findings reveal the commitment and flexibility of E2-2high progenitor differentiation and imply that pertinent tuning machinery is present in the gut microenvironment.
Collapse
Affiliation(s)
- Nobuyuki Onai
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan.,Department of Immunology, Kanazawa Medical University, Japan
| | - Jumpei Asano
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Rumiko Kurosaki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Shoko Kuroda
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| |
Collapse
|
13
|
Zhang W, Ding Y, Sun L, Hong Q, Sun Y, Han L, Zi M, Xu Y. Bone marrow-derived inflammatory and steady state DCs are different in both functions and survival. Cell Immunol 2018; 331:100-109. [DOI: 10.1016/j.cellimm.2018.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 05/06/2018] [Accepted: 06/06/2018] [Indexed: 01/15/2023]
|
14
|
Sun L, Rautela J, Delconte RB, Souza-Fonseca-Guimaraes F, Carrington EM, Schenk RL, Herold MJ, Huntington ND, Lew AM, Xu Y, Zhan Y. GM-CSF Quantity Has a Selective Effect on Granulocytic vs. Monocytic Myeloid Development and Function. Front Immunol 2018; 9:1922. [PMID: 30210491 PMCID: PMC6120981 DOI: 10.3389/fimmu.2018.01922] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/06/2018] [Indexed: 01/14/2023] Open
Abstract
GM-CSF promotes myeloid differentiation of cultured bone marrow cells into cells of the granulocytic and monocytic lineage; the latter can further differentiate into monocytes/macrophages and dendritic cells. How GM-CSF selects for these different myeloid fates is unresolved. GM-CSF levels can change either iatrogenically (e.g., augmenting leukopoiesis after radiotherapy) or naturally (e.g., during infection or inflammation) resulting in different immunological outcomes. Therefore, we asked whether the dose of GM-CSF may regulate the development of three types of myeloid cells. Here, we showed that GM-CSF acted as a molecular rheostat where the quantity determined which cell type was favored; moreover, the cellular process by which this was achieved was different for each cell type. Thus, low quantities of GM-CSF promoted the granulocytic lineage, mainly through survival. High quantities promoted the monocytic lineage, mainly through proliferation, whereas moderate quantities promoted moDCs, mainly through differentiation. Finally, we demonstrated that monocytes/macrophages generated with different doses of GM-CSF differed in function. We contend that this selective effect of GM-CSF dose on myeloid differentiation and function should be taken into consideration during pathophysiological states that may alter GM-CSF levels and during GM-CSF agonistic or antagonistic therapy.
Collapse
Affiliation(s)
- Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Jai Rautela
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Rebecca B Delconte
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Emma M Carrington
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Robyn L Schenk
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Marco J Herold
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Nicholas D Huntington
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Yifan Zhan
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Guangzhou Women and Children's Medical Centre, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Tao K, He M, Tao F, Xu G, Ye M, Zheng Y, Li Y. Development of NKG2D-based chimeric antigen receptor-T cells for gastric cancer treatment. Cancer Chemother Pharmacol 2018; 82:815-827. [PMID: 30132099 DOI: 10.1007/s00280-018-3670-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022]
Abstract
Gastric cancer is the third leading cause of cancer-related mortalities worldwide and mostly incurable. It remains an urgent need for novel strategies in the management of patients with advanced gastric cancer. Chimeric antigen receptor (CAR) T therapy has shown unprecedented clinical success in hematological malignancies and potential utility is going on various solid tumors like gastric cancer. In this study, a broad expression of NKG2D ligands was observed in gastric cancer cell lines, making them suitable targets for gastric cancer therapy. T cells were engineered with an NKG2D-based second-generation CAR and the resulting NKG2D-CAR-T cells showed significantly increased cytolytic activity against gastric cancer compared to untransduced T cells. In vivo, these cells can significantly suppressed the growth of established gastric cancer xenografts. Besides, cisplatin was shown to upregulate NKG2D ligand expression in gastric cancer cells and enhance the susceptibility to NKG2D-CAR-T-cell-mediated cytotoxicity. In conclusion, NKG2D-based CAR-T cells have potent in vivo and in vitro anti-tumor activities against gastric cancer and could be a new paradigm for patients with gastric cancer, either used alone or combined with chemotherapy.
Collapse
Affiliation(s)
- Kelong Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Meng He
- Department of Respiratory Medicine, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, People's Republic of China
| | - Feng Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Guangen Xu
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Minfeng Ye
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, People's Republic of China
| | - Yaoqing Li
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China.
| |
Collapse
|
16
|
Rogers PB, Schwartz EH. Generation of Large Numbers of Myeloid Progenitors and Dendritic Cell Precursors from Murine Bone Marrow Using a Novel Cell Sorting Strategy. J Vis Exp 2018:57365. [PMID: 30148496 PMCID: PMC6126687 DOI: 10.3791/57365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cultures of monocyte-derived dendritic cells (moDC) generated from mouse bone marrow using Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) have recently been recognized to be more heterogeneous than previously appreciated. These cultures routinely contain moDC as well monocyte-derived macrophages (moMac), and even some less developed cells such as monocytes. The goal of this protocol is to provide a consistent method for identification and separation of the many cell types present in these cultures as they develop, so that their specific functions may be further investigated. The sorting strategy presented here separates cells first into four populations based on expression of Ly6C and CD115, both of which are expressed transiently by cells as they develop in GM-CSF-driven culture. These four populations include Common myeloid progenitors or CMP (Ly6C-, CD115-), granulocyte/macrophage progenitors or GMP (Ly6C+, CD115-), monocytes (Ly6C+, CD115+), and monocyte-derived macrophages or moMac (Ly6C-, CD115+). CD11c is also added to the sorting strategy to distinguish two populations within the Ly6C-, CD115- population: CMP (CD11c-) and moDC (CD11c+). Finally, two populations may be further distinguished within the Ly6C-, CD115+ population based on the level of MHC class II expression. MoMacs express lower levels of MHC class II, while a monocyte-derived DC precursor (moDP) expresses higher MHC class II. This method allows for the reliable isolation of several developmentally distinct populations in numbers sufficient for a variety of functional and developmental analyses. We highlight one such functional readout, the differential responses of these cell types to stimulation with Pathogen-Associated Molecular Patterns (PAMPs).
Collapse
|
17
|
Chen S, Li X, Zhang W, Zi M, Xu Y. Inflammatory compound lipopolysaccharide promotes the survival of GM-CSF cultured dendritic cell via PI3 kinase-dependent upregulation of Bcl-x. Immunol Cell Biol 2018; 96:912-921. [PMID: 29624724 DOI: 10.1111/imcb.12051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 12/28/2022]
Abstract
As professional antigen-presenting cells, dendritic cells (DCs) initiate and regulate immune responses against inflammation. The invasion of pathogens into the body, however, can in turn cause the change of DCs in both activity and viability, which ultimately affect immune homeostasis. The exact mechanisms that the bacteria utilize to alter the lifespan of DCs, however, are far from clear. In this study, we found that the bacterial wall compound lipopolysaccharide (LPS) can promote the survival of GM-CSF-differentiated DCs (GM-DCs). At molecular levels, we demonstrated that GM-DCs had distinct pattern of mRNA expression for anti-apoptotic BCL-2 family members, of which, Bcl-x increased significantly following LPS stimulation. Interestingly, specific inhibition of BCL-XL protein alone was sufficient to remove the anti-apoptotic effects of LPS on BM-DCs. Further study of the signaling mechanisms revealed that although LPS can activate both Erk MAP kinase and PI3 kinase pathways, only blocking of PI3K abolished both Bcl-x upregulation and the enhanced survival phenotype, suggesting that the PI3K signaling mediated the upregulation of Bcl-x for the LPS-induced pro-survival in GM-DCs. Collectively, this study unveils a molecular mechanism that DCs adapt to maintain innate immunity against the invasion of pathogens.
Collapse
Affiliation(s)
- Shun Chen
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| | - Xinchen Li
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| | - Mengting Zi
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| |
Collapse
|
18
|
Becher B, Tugues S, Greter M. GM-CSF: From Growth Factor to Central Mediator of Tissue Inflammation. Immunity 2017; 45:963-973. [PMID: 27851925 DOI: 10.1016/j.immuni.2016.10.026] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 10/19/2016] [Accepted: 10/25/2016] [Indexed: 12/21/2022]
Abstract
The granulocyte-macrophage colony-stimulating factor (GM-CSF) was initially classified as a hematopoietic growth factor. However, unlike its close relatives macrophage CSF (M-CSF) and granulocyte CSF (G-CSF), the majority of myeloid cells do not require GM-CSF for steady-state myelopoiesis. Instead, in inflammation, GM-CSF serves as a communication conduit between tissue-invading lymphocytes and myeloid cells. Even though lymphocytes are in all likelihood the instigators of chronic inflammatory disease, GM-CSF-activated phagocytes are well equipped to cause tissue damage. The pivotal role of GM-CSF at the T cell:myeloid cell interface might shift our attention toward studying the function of the myeloid compartment in immunopathology. Targeting specifically the crosstalk between T cells and myeloid cells through GM-CSF holds promise for the development of therapeutics to combat chronic tissue inflammation. Here, we will review some of the major discoveries of the recent past, which indicate that GM-CSF is so much more than its name suggests.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of Experimental Immunology, University of Zurich Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Sonia Tugues
- Institute of Experimental Immunology, University of Zurich Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
19
|
Zhao W, Zhou X, Zhao G, Lin Q, Wang X, Yu X, Wang B. Enrichment of Ly6C hi monocytes by multiple GM-CSF injections with HBV vaccine contributes to viral clearance in a HBV mouse model. Hum Vaccin Immunother 2017; 13:2872-2882. [PMID: 28699816 PMCID: PMC5718782 DOI: 10.1080/21645515.2017.1344797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adjuvants are considered a necessary component for HBV therapeutic vaccines but few are licensed in clinical practice due to concerns about safety or efficiency. In our recent study, we established that a combination protocol of 3-day pretreatments with GM-CSF before a vaccination (3 × GM-CSF+VACCINE) into the same injection site could break immune tolerance and cause over 90% reduction of HBsAg level in the HBsAg transgenic mouse model. Herein, we further investigated the therapeutic potential of the combination in AAV8–1.3HBV-infected mice. After 4 vaccinations, both serum HBeAg and HBsAg were cleared and there was a 95% reduction of HBV-positive hepatocytes, in addition to the presence of large number of infiltrating CD8+ T cells in the livers. Mechanistically, the HBV-specific T-cell responses were elicited via a 3 × GM-CSF+VACCINE-induced conversion of CCR2-dependent CD11b+ Ly6Chi monocytes into CD11b+CD11c+ DCs. Experimental depletion of Ly6Chi monocytes resulted in a defective HBV-specific immune response thereby abrogating HBV eradication. This vaccination strategy could lead to development of an effective therapeutic protocol against chronic HBV in infected patients.
Collapse
Affiliation(s)
- Weidong Zhao
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Xian Zhou
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Gan Zhao
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Qing Lin
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Xianzheng Wang
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| | - Xueping Yu
- b Department of Infectious Diseases, Huashan Hospital , Fudan University , Shanghai , China
| | - Bin Wang
- a Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences , Fudan University , Shanghai , China
| |
Collapse
|
20
|
Crotti C, Raimondo MG, Becciolini A, Biggioggero M, Favalli EG. Spotlight on mavrilimumab for the treatment of rheumatoid arthritis: evidence to date. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:211-223. [PMID: 28144129 PMCID: PMC5245809 DOI: 10.2147/dddt.s104233] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The introduction of biological therapies into clinical practice has dramatically modified the natural history of chronic inflammatory diseases, such as rheumatoid arthritis (RA). RA is a systemic autoimmune disease that causes articular damage and has a great negative impact on patients’ quality of life. Despite the wide spectrum of available biological treatments, ~30% of RA patients are still unresponsive, resulting in high disability and increased morbidity and mortality. In the last few decades, the scientific knowledge on RA pathogenesis vastly improved, leading to the identification of new proinflammatory molecules as potential therapeutic targets. Several in vitro and in vivo studies showed that granulocyte-macrophage colony-stimulating factor (GM-CSF), known to be a hematopoietic factor, is also one of the proinflammatory cytokines involved in macrophage activation, crucial for the pathogenic network of RA. Mavrilimumab, a human monoclonal antibody targeting the subunit α of GM-CSF receptor, was recently developed as a competitive antagonist of GM-CSF pathway and successfully adopted in human trials for mild to moderate RA. Mavrilimumab phase I and phase II studies reported an overall good efficacy and safety profile of the drug, and these encouraging results promoted the initiation of worldwide phase III studies. In particular, 158-week results of phase II trials did not show long-term lung toxicity, addressing the major concern about this target of pulmonary alveolar proteinosis development. However, further clinical studies conducted in larger RA populations are needed to confirm these promising results. This review summarizes the biological role of GM-CSF in RA and the preclinical and clinical data on mavrilimumab and other monoclonal antibodies targeted on this pathway as an alternative therapeutic option in RA patients who are unresponsive to conventional biological drugs.
Collapse
Affiliation(s)
- Chiara Crotti
- Department of Clinical Sciences and Health Community, University of Milan, Division of Rheumatology, Gaetano Pini Institute
| | - Maria Gabriella Raimondo
- Department of Clinical Sciences and Health Community, University of Milan, Division of Rheumatology, Gaetano Pini Institute
| | | | - Martina Biggioggero
- Department of Clinical Sciences and Health Community, University of Milan, Division of Rheumatology, Gaetano Pini Institute
| | | |
Collapse
|
21
|
Cook AD, Louis C, Robinson MJ, Saleh R, Sleeman MA, Hamilton JA. Granulocyte macrophage colony-stimulating factor receptor α expression and its targeting in antigen-induced arthritis and inflammation. Arthritis Res Ther 2016; 18:287. [PMID: 27908288 PMCID: PMC5134062 DOI: 10.1186/s13075-016-1185-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/16/2016] [Indexed: 12/23/2022] Open
Abstract
Background Blockade of granulocyte macrophage colony-stimulating factor (GM-CSF) and its receptor (GM-CSFRα) is being successfully tested in trials in rheumatoid arthritis (RA) with clinical results equivalent to those found with neutralization of the current therapeutic targets, TNF and IL-6. To explore further the role of GM-CSF as a pro-inflammatory cytokine, we examined the effect of anti-GM-CSFRα neutralization on myeloid cell populations in antigen-driven arthritis and inflammation models and also compared its effect with that of anti-TNF and anti-IL-6. Methods Cell population changes upon neutralization by monoclonal antibodies (mAbs) in the antigen-induced arthritis (AIA) and antigen-induced peritonitis (AIP) models were monitored by flow cytometry and microarray. Adoptive transfer of monocytes into the AIP cavity was used to assess the GM-CSF dependence of the development of macrophages and monocyte-derived dendritic cells (Mo-DCs) at a site of inflammation. Results Therapeutic administration of a neutralizing anti-GM-CSF mAb, but not of an anti-colony-stimulating factor (anti-CSF)-1 or an anti-CSF-1R mAb, ameliorated AIA disease. Using the anti-GM-CSFRα mAb, the relative surface expression of different inflammatory myeloid populations was found to be similar in the inflamed tissues in both the AIA and AIP models; however, the GM-CSFRα mAb, but not neutralizing anti-TNF and anti-IL-6 mAbs, preferentially depleted Mo-DCs from these sites. In addition, we were able to show that locally acting GM-CSF upregulated macrophage/Mo-DC numbers via GM-CSFR signalling in donor monocytes. Conclusions Our findings suggest that GM-CSF blockade modulates inflammatory responses differently to TNF and IL-6 blockade and may provide additional insight into how targeting the GM-CSF/GM-CSFRα system is providing efficacy in RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1185-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew D Cook
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia.
| | - Cynthia Louis
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| | - Matthew J Robinson
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK
| | - Reem Saleh
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| | - Matthew A Sleeman
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,, Present Address: Regeneron, 777 Old Saw Mill River Rd, Tarrytown, NY, USA
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, 3050, Australia
| |
Collapse
|
22
|
Plasmacytoid dendritic cells are short-lived: reappraising the influence of migration, genetic factors and activation on estimation of lifespan. Sci Rep 2016; 6:25060. [PMID: 27112985 PMCID: PMC4844974 DOI: 10.1038/srep25060] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/08/2016] [Indexed: 12/21/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) play an important role in immunity to certain pathogens and immunopathology in some autoimmune diseases. They are thought to have a longer lifespan than conventional DCs (cDCs), largely based on a slower rate of BrdU labeling by splenic pDCs. Here we demonstrated that pDC expansion and therefore BrdU labeling by pDCs occurs in bone marrow (BM). The rate of labeling was similar between BM pDCs and spleen cDCs. Therefore, slower BrdU labeling of spleen pDCs likely reflects the "migration time" (∼2 days) for BrdU labeled pDCs to traffic to the spleen, not necessarily reflecting longer life span. Tracking the decay of differentiated DCs showed that splenic pDCs and cDCs decayed at a similar rate. We suggest that spleen pDCs have a shorter in vivo lifespan than estimated utilizing some of the previous approaches. Nevertheless, pDC lifespan varies between mouse strains. pDCs from lupus-prone NZB mice survived longer than C57BL/6 pDCs. We also demonstrated that activation either positively or negatively impacted on the survival of pDCs via different cell-death mechanisms. Thus, pDCs are also short-lived. However, the pDC lifespan is regulated by genetic and environmental factors that may have pathological consequence.
Collapse
|
23
|
Papadopoulos A, Gagnaire A, Degos C, de Chastellier C, Gorvel JP. Brucella discriminates between mouse dendritic cell subsets upon in vitro infection. Virulence 2015; 7:33-44. [PMID: 26606688 PMCID: PMC4871654 DOI: 10.1080/21505594.2015.1108516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Brucella is a Gram-negative bacterium responsible for brucellosis, a worldwide re-emerging zoonosis. Brucella has been shown to infect and replicate within Granulocyte macrophage colony-stimulating factor (GMCSF) in vitro grown bone marrow-derived dendritic cells (BMDC). In this cell model, Brucella can efficiently control BMDC maturation. However, it has been shown that Brucella infection in vivo induces spleen dendritic cells (DC) migration and maturation. As DCs form a complex network composed by several subpopulations, differences observed may be due to different interactions between Brucella and DC subsets. Here, we compare Brucella interaction with several in vitro BMDC models. The present study shows that Brucella is capable of replicating in all the BMDC models tested with a high infection rate at early time points in GMCSF-IL15 DCs and Flt3l DCs. GMCSF-IL15 DCs and Flt3l DCs are more activated than the other studied DC models and consequently intracellular bacteria are not efficiently targeted to the ER replicative niche. Interestingly, GMCSF-DC and GMCSF-Flt3l DC response to infection is comparable. However, the key difference between these 2 models concerns IL10 secretion by GMCSF DCs observed at 48 h post-infection. IL10 secretion can explain the weak secretion of IL12p70 and TNFα in the GMCSF-DC model and the low level of maturation observed when compared to GMCSF-IL15 DCs and Flt3l DCs. These models provide good tools to understand how Brucella induce DC maturation in vivo and may lead to new therapeutic design using DCs as cellular vaccines capable of enhancing immune response against pathogens.
Collapse
Affiliation(s)
- Alexia Papadopoulos
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Aurélie Gagnaire
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Clara Degos
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Chantal de Chastellier
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Jean-Pierre Gorvel
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| |
Collapse
|
24
|
Croxford AL, Spath S, Becher B. GM-CSF in Neuroinflammation: Licensing Myeloid Cells for Tissue Damage. Trends Immunol 2015; 36:651-662. [DOI: 10.1016/j.it.2015.08.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 12/23/2022]
|
25
|
Louis C, Cook AD, Lacey D, Fleetwood AJ, Vlahos R, Anderson GP, Hamilton JA. Specific Contributions of CSF-1 and GM-CSF to the Dynamics of the Mononuclear Phagocyte System. THE JOURNAL OF IMMUNOLOGY 2015; 195:134-44. [DOI: 10.4049/jimmunol.1500369] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/27/2015] [Indexed: 12/23/2022]
|
26
|
Bhattacharya P, Budnick I, Singh M, Thiruppathi M, Alharshawi K, Elshabrawy H, Holterman MJ, Prabhakar BS. Dual Role of GM-CSF as a Pro-Inflammatory and a Regulatory Cytokine: Implications for Immune Therapy. J Interferon Cytokine Res 2015; 35:585-99. [PMID: 25803788 DOI: 10.1089/jir.2014.0149] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Granulocyte macrophage colony stimulating factor (GM-CSF) is generally recognized as an inflammatory cytokine. Its inflammatory activity is primarily due its role as a growth and differentiation factor for granulocyte and macrophage populations. In this capacity, among other clinical applications, it has been used to bolster anti-tumor immune responses. GM-CSF-mediated inflammation has also been implicated in certain types of autoimmune diseases, including rheumatoid arthritis and multiple sclerosis. Thus, agents that can block GM-CSF or its receptor have been used as anti-inflammatory therapies. However, a review of literature reveals that in many situations GM-CSF can act as an anti-inflammatory/regulatory cytokine. We and others have shown that GM-CSF can modulate dendritic cell differentiation to render them "tolerogenic," which, in turn, can increase regulatory T-cell numbers and function. Therefore, the pro-inflammatory and regulatory effects of GM-CSF appear to depend on the dose and the presence of other relevant cytokines in the context of an immune response. A thorough understanding of the various immunomodulatory effects of GM-CSF will facilitate more appropriate use and thus further enhance its clinical utility.
Collapse
Affiliation(s)
- Palash Bhattacharya
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| | - Isadore Budnick
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| | - Medha Singh
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| | - Muthusamy Thiruppathi
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| | - Khaled Alharshawi
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| | - Hatem Elshabrawy
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| | - Mark J Holterman
- 2 Department of Surgery, College of Medicine, University of Illinois , Chicago, Illinois
| | - Bellur S Prabhakar
- 1 Department of Microbiology and Immunology, College of Medicine, University of Illinois , Chicago, Illinois
| |
Collapse
|
27
|
Cystatin C is a disease-associated protein subject to multiple regulation. Immunol Cell Biol 2015; 93:442-51. [PMID: 25643616 PMCID: PMC7165929 DOI: 10.1038/icb.2014.121] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 02/06/2023]
Abstract
A protease inhibitor, cystatin C (Cst C), is a secreted cysteine protease inhibitor abundantly expressed in body fluids. Clinically, it is mostly used to measure glomerular filtration rate as a marker for kidney function due to its relatively small molecular weight and easy detection. However, recent findings suggest that Cst C is regulated at both transcriptional and post‐translational levels, and Cst C production from haematopoietic cell lineages contributes significantly to the systematic pools of Cst C. Furthermore, Cst C is directly linked to many pathologic processes through various mechanisms. Thus fluctuation of Cst C levels might have serious clinical implications rather than a mere reflection of kidney functions. Here, we summarize the pathophysiological roles of Cst C dependent and independent on its inhibition of proteases, outline its change of expression by various stimuli, and elucidate the regulatory mechanisms to control this disease‐related protease inhibitor. Finally, we discuss the clinical implications of these findings for translational gains.
Collapse
|
28
|
Guerrero AD, Dong MB, Zhao Y, Lau-Kilby A, Tarbell KV. Interleukin-2-mediated inhibition of dendritic cell development correlates with decreased CD135 expression and increased monocyte/macrophage precursors. Immunology 2015; 143:640-50. [PMID: 24954893 DOI: 10.1111/imm.12345] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 06/06/2014] [Accepted: 06/18/2014] [Indexed: 11/27/2022] Open
Abstract
We have previously shown that interleukin-2 (IL-2) inhibits dendritic cell (DC) development from mouse bone marrow (BM) precursors stimulated with the ligand for FMS-like tyrosine kinase 3 receptor (Flt3L), and have provided evidence that this inhibition occurs at the monocyte DC precursor stage of DC development. Here, we explored the mechanism of IL-2-mediated inhibition of DC development. First, we showed that these in vitro cultures accurately model DCs that develop in vivo by comparing gene and protein expression of the three main Flt3L-induced DC subsets from the BM, CD11b(+) and CD24(+) conventional DCs (cDCs) and plasmacytoid DCs (pDCs) with their respective ex vivo spleen DC subsets (CD11b(+), CD8(+) and pDCs). Next, gene expression changes were quantified in Flt3L DC subsets that developed in the presence of IL-2. These changes included increased expression of Bcl2l11, which encodes the apoptosis-inducing protein Bim, and decreased expression of Flt3 (CD135), the receptor that initiates DC development. Interleukin-2 also significantly reduced Flt3 protein expression on all three Flt3L DC subsets, and attenuated Flt3L-induced STAT3 phosphorylation in DCs. Based on these data, we hypothesized that decreased Flt3 signalling may divert BM precursors down monocyte and macrophage lineages. Indeed, addition of IL-2 led to increases in Flt3(-) cells, including cKit(+) Ly6C(+) CD11b(-) populations consistent with the recently identified committed monocyte/macrophage progenitor. Therefore, IL-2 can inhibit DC development via decreased signalling through Flt3 and increased monocyte/macrophage development.
Collapse
Affiliation(s)
- Alan D Guerrero
- Immune Tolerance Section, Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes, and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
29
|
Loeffler DA. Should development of Alzheimer's disease-specific intravenous immunoglobulin be considered? J Neuroinflammation 2014; 11:198. [PMID: 25476011 PMCID: PMC4265363 DOI: 10.1186/s12974-014-0198-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 11/08/2014] [Indexed: 12/15/2022] Open
Abstract
Recent phase II and III studies with intravenous immunoglobulin (IVIG) in patients with Alzheimer's disease (AD) did not find evidence for the slowing of AD progression compared to placebo-treated patients, in contrast to encouraging results in pilot studies. An additional phase III trial is ongoing. If negative results are found, then further AD studies with IVIG are unlikely unless a manufacturer opts for a trial with high-dose IVIG, which would increase its anti-inflammatory effects but also the risk for adverse events. An alternative approach could be an AD-specific IVIG, supplementing IVIG with higher concentrations of selected antibodies purified from it or produced via recombinant polyclonal antibody technology. These antibodies could include those to amyloid-beta (Aβ, tau protein, inflammatory cytokines, complement activation proteins, and the receptor for advanced glycation end products. IgG fragment crystallizable (Fc) fragments containing terminal sialic acid could be added to increase anti-inflammatory effects. While this product might be more effective in slowing AD clinical progression than current IVIG, there are difficulties with this approach. Preclinical studies would be required to determine which of the antibodies of interest for supplementing current IVIG (for example, antibodies to phosphorylated or oligomeric tau) are actually present (and, therefore, available for purification) in IVIG, and the effects of the product in mouse models of AD. An Investigational New Drug application for an AD-specific IVIG would require United States Food and Drug Administration approval. If the drug would be found to benefit AD patients, meeting the increased demand for IVIG would be challenging.
Collapse
Affiliation(s)
- David A Loeffler
- Department of Internal Medicine, Division of Neurology, Beaumont Health System, 3601 West Thirteen Mile Road, Royal Oak, MI, 48073, USA.
| |
Collapse
|
30
|
Martínez-López M, Iborra S, Conde-Garrosa R, Sancho D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur J Immunol 2014; 45:119-29. [PMID: 25312824 PMCID: PMC4316187 DOI: 10.1002/eji.201444651] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 08/24/2014] [Accepted: 10/08/2014] [Indexed: 02/02/2023]
Abstract
The role of different DC subsets in priming and maintenance of immunity against Leishmania major (L. major) infection is debated. The transcription factor basic leucine zipper transcription factor, ATF-like 3 (Batf3) is essential for the development of mouse CD103(+) DCs and some functions of CD8α(+) DCs. We found that CD103(+) DCs were significantly reduced in the dermis of Batf3-deficient C57BL/6 mice. Batf3(-/-) mice developed exacerbated and unresolved cutaneous pathology following a low dose of intradermal L. major infection in the ear pinnae. Parasite load was increased 1000-fold locally and expanded systemically. Batf3 deficiency did not affect L. major antigen presentation to T cells, which was directly exerted by CD8α(-) conventional DCs (cDCs) in the skin draining LN. However, CD4(+) T-cell differentiation in the LN and skin was skewed to nonprotective Treg- and Th2-cell subtypes. CD103(+) DCs are major IL-12 producers during L. major infection. Local Th1 immunity was severely hindered, correlating with impaired IL-12 production and reduction in CD103(+) DC numbers. Adoptive transfer of WT but not IL-12p40(-/-) Batf3-dependent DCs significantly improved anti-L. major response in infected Batf3(-/-) mice. Our results suggest that IL-12 production by Batf3-dependent CD103(+) DCs is crucial for maintenance of local Th1 immunity against L. major infection.
Collapse
Affiliation(s)
- María Martínez-López
- Department of Vascular Biology and Inflammation, CNIC-Fundación Centro Nacional de Investigaciones Cardiovasculares "Carlos III", Madrid, Spain
| | | | | | | |
Collapse
|
31
|
Lymphoid tissue and plasmacytoid dendritic cells and macrophages do not share a common macrophage-dendritic cell-restricted progenitor. Immunity 2014; 41:104-15. [PMID: 25035955 DOI: 10.1016/j.immuni.2014.05.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 05/19/2014] [Indexed: 12/17/2022]
Abstract
The relationship between dendritic cells (DCs) and macrophages is often debated. Here we ask whether steady-state, lymphoid-tissue-resident conventional DCs (cDCs), plasmacytoid DCs (pDCs), and macrophages share a common macrophage-DC-restricted precursor (MDP). Using new clonal culture assays combined with adoptive transfer, we found that MDP fractions isolated by previous strategies are dominated by precursors of macrophages and monocytes, include some multipotent precursors of other hematopoietic lineages, but contain few precursors of resident cDCs and pDCs and no detectable common precursors restricted to these DC types and macrophages. Overall we find no evidence for a common restricted MDP leading to both macrophages and FL-dependent, resident cDCs and pDCs.
Collapse
|
32
|
Abstract
Despite significant effort, the development of effective vaccines inducing strong and durable T-cell responses against intracellular pathogens and cancer cells has remained a challenge. The initiation of effector CD8+ T-cell responses requires the presentation of peptides derived from internalized antigen on class I major histocompatibility complex molecules by dendritic cells (DCs) in a process called cross-presentation. A current strategy to enhance the effectiveness of vaccination is to deliver antigens directly to DCs. This is done via selective targeting of antigen using monoclonal antibodies directed against endocytic receptors on the surface of the DCs. In this review, we will discuss considerations relevant to the design of such vaccines: the existence of DC subsets with specialized functions, the impact of the antigen intracellular trafficking on cross-presentation, and the influence of maturation signals received by DCs on the outcome of the immune response.
Collapse
Affiliation(s)
- Lillian Cohn
- Laboratory of Molecular Immunology, Rockefeller University , New York, NY , USA
| | | |
Collapse
|
33
|
The closely related CD103+ dendritic cells (DCs) and lymphoid-resident CD8+ DCs differ in their inflammatory functions. PLoS One 2014; 9:e91126. [PMID: 24637385 PMCID: PMC3956455 DOI: 10.1371/journal.pone.0091126] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/07/2014] [Indexed: 01/01/2023] Open
Abstract
Migratory CD103+ and lymphoid-resident CD8+ dendritic cells (DCs) share many attributes, such as dependence on the same transcription factors, cross-presenting ability and expression of certain surface molecules, such that it has been proposed they belong to a common sub-lineage. The functional diversity of the two DC types is nevertheless incompletely understood. Here we reveal that upon skin infection with herpes simplex virus, migratory CD103+ DCs from draining lymph nodes were more potent at inducing Th17 cytokine production by CD4+ T cells than CD8+ DCs. This superior capacity to drive Th17 responses was also evident in CD103+ DCs from uninfected mice. Their differential potency to induce Th17 differentiation was reflected by higher production of IL-1β and IL-6 by CD103+ DCs compared with CD8+ DCs upon stimulation. The two types of DCs from isolated lymph nodes also differ in expression of certain pattern recognition receptors. Furthermore, elevated levels of GM-CSF, typical of those found in inflammation, substantially increased the pool size of CD103+ DCs in lymph nodes and skin. We argue that varied levels of GM-CSF may explain the contrasting reports regarding the positive role of GM-CSF in regulating development of CD103+ DCs. Together, we find that these two developmentally closely-related DC subsets display functional differences and that GM-CSF has differential effect on the two types of DCs.
Collapse
|
34
|
Xu Y, Lindemann P, Vega-Ramos J, Zhang JG, Villadangos JA. Developmental regulation of synthesis and dimerization of the amyloidogenic protease inhibitor cystatin C in the hematopoietic system. J Biol Chem 2014; 289:9730-40. [PMID: 24570004 DOI: 10.1074/jbc.m113.538041] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cysteine protease inhibitor cystatin C is thought to be secreted by most cells and eliminated in the kidneys, so its concentration in plasma is diagnostic of kidney function. Low extracellular cystatin C is linked to pathologic protease activity in cancer, arthritis, atherosclerosis, aortic aneurism, and emphysema. Cystatin C forms non-inhibitory dimers and aggregates by a mechanism known as domain swapping, a property that reportedly protects against Alzheimer disease but can also cause amyloid angiopathy. Despite these clinical associations, little is known about the regulation of cystatin C production, dimerization, and secretion. We show that hematopoietic cells are major contributors to extracellular cystatin C levels in healthy mice. Among these cells, macrophages and dendritic cells (DC) are the predominant producers of cystatin C. Both cell types synthesize monomeric and dimeric cystatin C in vivo, but only secrete monomer. Dimerization occurs co-translationally in the endoplasmic reticulum and is regulated by the levels of reactive oxygen species (ROS) derived from mitochondria. Drugs or stimuli that reduce the intracellular concentration of ROS inhibit cystatin C dimerization. The extracellular concentration of inhibitory cystatin C is thus partly dependent on the abundance of macrophages and DC, and the ROS levels. These results have implications for the diagnostic use of serum cystatin C as a marker of kidney function during inflammatory processes that induce changes in DC or macrophage abundance. They also suggest an important role for macrophages, DC, and ROS in diseases associated with the protease inhibitory activity or amyloidogenic properties of cystatin C.
Collapse
|
35
|
Globisch T, Steiner N, Fülle L, Lukacs-Kornek V, Degrandi D, Dresing P, Alferink J, Lang R, Pfeffer K, Beyer M, Weighardt H, Kurts C, Ulas T, Schultze JL, Förster I. Cytokine-dependent regulation of dendritic cell differentiation in the splenic microenvironment. Eur J Immunol 2014; 44:500-10. [PMID: 24136200 DOI: 10.1002/eji.201343820] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/02/2013] [Accepted: 09/18/2013] [Indexed: 12/21/2022]
Abstract
The DC-derived chemokine CCL17, a ligand of CCR4, has been shown to promote various inflammatory diseases such as atopic dermatitis, atherosclerosis, and inflammatory bowel disease. Under steady-state conditions, and even after systemic stimulation with LPS, CCL17 is not expressed in resident splenic DCs as opposed to CD8α⁻CD11b⁺ LN DCs, which produce large amounts of CCL17 in particular after maturation. Upon systemic NKT cell activation through α-galactosylceramide stimulation however, CCL17 can be upregulated in both CD8α⁻ and CD8α⁺ splenic DC subsets and enhances cross-presentation of exogenous antigens. Based on genome-wide expression profiling, we now show that splenic CD11b⁺ DCs are susceptible to IFN-γ-mediated suppression of CCL17, whereas LN CD11b⁺CCL17⁺ DCs downregulate the IFN-γR and are much less responsive to IFN-γ. Under inflammatory conditions, particularly in the absence of IFN-γ signaling in IFN-γRKO mice, CCL17 expression is strongly induced in a major proportion of splenic DCs by the action of GM-CSF in concert with IL-4. Our findings demonstrate that the local cytokine milieu and differential cytokine responsiveness of DC subsets regulate lymphoid organ specific immune responses at the level of chemokine expression.
Collapse
Affiliation(s)
- Theresa Globisch
- Department of Molecular Immunology, IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany; Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mineharu Y, Castro MG, Lowenstein PR, Sakai N, Miyamoto S. Dendritic cell-based immunotherapy for glioma: multiple regimens and implications in clinical trials. Neurol Med Chir (Tokyo) 2013; 53:741-54. [PMID: 24140772 PMCID: PMC3926207 DOI: 10.2176/nmc.ra2013-0234] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High grade glioma is a highly invasive brain tumor and recurrence is almost inevitable, even after radical resection of the tumor mass. Cytotoxic immune responses and immunological memory induced by immunotherapy might prevent tumor recurrence. Dendritic cells (DCs) are professional antigen-presenting cells of the innate immune system with the potential to generate robust antigen-specific T cell immune responses. DC-based immunotherapeutic strategies have been intensively studied in both preclinical and clinical settings. Although advances have been made in the experimental use of DCs, there are still considerable challenges that need to be addressed for clinical translation. In this review, we describe the variability of regimens currently available for DC-based immunotherapy and then review strategies to optimize DC therapeutic efficacy against glioma.
Collapse
Affiliation(s)
- Yohei Mineharu
- Division of Neuroendovascular Therapy, Institute of Biomedical Research and Innovation
| | | | | | | | | |
Collapse
|
37
|
Loss of DAP12 and FcRγ drives exaggerated IL-12 production and CD8(+) T cell response by CCR2(+) Mo-DCs. PLoS One 2013; 8:e76145. [PMID: 24155889 PMCID: PMC3796521 DOI: 10.1371/journal.pone.0076145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/19/2013] [Indexed: 11/24/2022] Open
Abstract
Dap12 and FcRγ, the two transmembrane ITAM-containing signaling adaptors expressed in dendritic cells (DC), are implicated in the regulation of DC function. Several activating and adhesion receptors including integrins require these chains for their function in triggering downstream signaling and effector pathways, however the exact role(s) for Dap12 and FcRγ remains elusive as their loss can lead to both attenuating and enhancing effects. Here, we report that mice congenitally lacking both Dap12 and FcRγ chains (DF) show a massively enhanced effector CD8+ T cell response to protein antigen immunization or West Nile Virus (WNV) infection. Thus, immunization of DF mice with MHCI-restricted OVA peptide leads to accumulation of IL-12-producing monocyte-derived dendritic cells (Mo-DC) in draining lymph nodes, followed by vastly enhanced generation of antigen-specific IFNγ-producing CD8+ T cells. Moreover, DF mice show increased viral clearance in the WNV infection model. Depletion of CCR2+ monocytes/macrophages in vivo by administration anti-CCR2 antibodies or clodronate liposomes completely prevents the exaggerated CD8+ T cell response in DF mice. Mechanistically, we show that the loss of Dap12 and FcRγ-mediated signals in Mo-DC leads to a disruption of GM-CSF receptor-induced STAT5 activation resulting in upregulation of expression of IRF8, a transcription factor. Consequently, Dap12- and FcRγ-deficiency exacerbates GM-CSF-driven monocyte differentiation and production of inflammatory Mo-DC. Our data suggest a novel cross-talk between DC-ITAM and GM-CSF signaling pathways, which controls Mo-DC differentiation, IL-12 production, and CD8+ T cell responses.
Collapse
|
38
|
Li BZ, Ye QL, Xu WD, Li JH, Ye DQ, Xu Y. GM-CSF alters dendritic cells in autoimmune diseases. Autoimmunity 2013; 46:409-18. [PMID: 23786272 DOI: 10.3109/08916934.2013.803533] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Autoimmune diseases arise from an inappropriate immune response against self components, including macromolecules, cells, tissues, organs etc. They are often triggered or accompanied by inflammation, during which the levels of granulocyte macrophage colony-stimulating factor (GM-CSF) are elevated. GM-CSF is an inflammatory cytokine that has profound impact on the differentiation of immune system cells of myeloid lineage, especially dendritic cells (DCs) that play critical roles in immune initiation and tolerance, and is involved in the pathogenesis of autoimmune diseases. Although GM-CSF was discovered decades ago, recent studies with some new findings have shed an interesting light on the old hematopoietic growth factor. In the inflammatory autoimmune diseases, GM-CSF redirects the normal developmental pathway of DCs, conditions their antigen presentation capacities and endows them with unique cytokine signatures to affect autoimmune responses. Here we review the latest advances in the field, with the aim of demonstrating the effects of GM-CSF on DCs and their influences on autoimmune diseases. The summarized knowledge will help to design DC-based strategies for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University , Anhui , PR China
| | | | | | | | | | | |
Collapse
|
39
|
Cohn L, Chatterjee B, Esselborn F, Smed-Sörensen A, Nakamura N, Chalouni C, Lee BC, Vandlen R, Keler T, Lauer P, Brockstedt D, Mellman I, Delamarre L. Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation. ACTA ACUST UNITED AC 2013; 210:1049-63. [PMID: 23569326 PMCID: PMC3646496 DOI: 10.1084/jem.20121251] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human BDCA3(+) dendritic cells (DCs), the proposed equivalent to mouse CD8α(+) DCs, are widely thought to cross present antigens on MHC class I (MHCI) molecules more efficiently than other DC populations. If true, it is unclear whether this reflects specialization for cross presentation or a generally enhanced ability to present antigens on MHCI. We compared presentation by BDCA3(+) DCs with BDCA1(+) DCs using a quantitative approach whereby antigens were targeted to distinct intracellular compartments by receptor-mediated internalization. As expected, BDCA3(+) DCs were superior at cross presentation of antigens delivered to late endosomes and lysosomes by uptake of anti-DEC205 antibody conjugated to antigen. This difference may reflect a greater efficiency of antigen escape from BDCA3(+) DC lysosomes. In contrast, if antigens were delivered to early endosomes through CD40 or CD11c, BDCA1(+) DCs were as efficient at cross presentation as BDCA3(+) DCs. Because BDCA3(+) DCs and BDCA1(+) DCs were also equivalent at presenting peptides and endogenously synthesized antigens, BDCA3(+) DCs are not likely to possess mechanisms for cross presentation that are specific to this subset. Thus, multiple DC populations may be comparably effective at presenting exogenous antigens to CD8(+) T cells as long as the antigen is delivered to early endocytic compartments.
Collapse
|
40
|
Moore AJ, Anderson MK. Dendritic cell development: a choose-your-own-adventure story. Adv Hematol 2013; 2013:949513. [PMID: 23476654 PMCID: PMC3588201 DOI: 10.1155/2013/949513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/27/2012] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are essential components of the immune system and contribute to immune responses by activating or tolerizing T cells. DCs comprise a heterogeneous mixture of subsets that are located throughout the body and possess distinct and specialized functions. Although numerous defined precursors from the bone marrow and spleen have been identified, emerging data in the field suggests many alternative routes of DC differentiation from precursors with multilineage potential. Here, we discuss how the combinatorial expression of transcription factors can promote one DC lineage over another as well as the integration of cytokine signaling in this process.
Collapse
Affiliation(s)
- Amanda J. Moore
- Division of Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, Canada M4N 3M5
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Michele K. Anderson
- Division of Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, Canada M4N 3M5
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
41
|
Xu WD, Pan HF, Ye DQ, Xu Y. Targeting IRF4 in autoimmune diseases. Autoimmun Rev 2012; 11:918-24. [DOI: 10.1016/j.autrev.2012.08.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 08/14/2012] [Indexed: 12/28/2022]
|