151
|
Schwartz DM, Bonelli M, Gadina M, O'Shea JJ. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat Rev Rheumatol 2016; 12:25-36. [PMID: 26633291 PMCID: PMC4688091 DOI: 10.1038/nrrheum.2015.167] [Citation(s) in RCA: 459] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines are major drivers of autoimmunity, and biologic agents targeting cytokines have revolutionized the treatment of immune-mediated diseases. Despite the effectiveness of these drugs, they do not induce complete remission in all patients, prompting the development of alternative strategies - including targeting of intracellular signal transduction pathways downstream of cytokines. Many cytokines that bind type I and type II cytokine receptors are critical regulators of immune-mediated diseases and employ the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway to exert their effect. Pharmacological inhibition of JAKs blocks the actions of type I/II cytokines, and within the past 3 years therapeutic JAK inhibitors, or Jakinibs, have become available to rheumatologists. Jakinibs have proven effective for the treatment of rheumatoid arthritis and other inflammatory diseases. Adverse effects of these agents are largely related to their mode of action and include infections and hyperlipidemia. Jakinibs are currently being investigated for a number of new indications, and second-generation selective Jakinibs are being developed and tested. Targeting STATs could be a future avenue for the treatment of rheumatologic diseases, although substantial challenges remain. Nonetheless, the ability to therapeutically target intracellular signalling pathways has already created a new paradigm for the treatment of rheumatologic disease.
Collapse
Affiliation(s)
- Daniella M Schwartz
- Molecular Immunology &Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis &Musculoskeletal and Skin Diseases, NIH, Building 10, 9000 Rockville Pike, Bethesda, Maryland 20892-1616, USA
| | - Michael Bonelli
- Medical University of Vienna, Department of Rheumatology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Massimo Gadina
- Molecular Immunology &Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis &Musculoskeletal and Skin Diseases, NIH, Building 10, 9000 Rockville Pike, Bethesda, Maryland 20892-1616, USA
| | - John J O'Shea
- Molecular Immunology &Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis &Musculoskeletal and Skin Diseases, NIH, Building 10, 9000 Rockville Pike, Bethesda, Maryland 20892-1616, USA
| |
Collapse
|
152
|
Regulation and Immune Function of IL-27. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:191-211. [DOI: 10.1007/978-94-024-0921-5_9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
153
|
Sénécal V, Deblois G, Beauseigle D, Schneider R, Brandenburg J, Newcombe J, Moore CS, Prat A, Antel J, Arbour N. Production of IL-27 in multiple sclerosis lesions by astrocytes and myeloid cells: Modulation of local immune responses. Glia 2015; 64:553-69. [PMID: 26649511 DOI: 10.1002/glia.22948] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/20/2015] [Accepted: 11/17/2015] [Indexed: 01/22/2023]
Abstract
The mechanisms whereby human glial cells modulate local immune responses are not fully understood. Interleukin-27 (IL-27), a pleiotropic cytokine, has been shown to dampen the severity of experimental autoimmune encephalomyelitis, but it is still unresolved whether IL-27 plays a role in the human disease multiple sclerosis (MS). IL-27 contribution to local modulation of immune responses in the brain of MS patients was investigated. The expression of IL-27 subunits (EBI3 and p28) and its cognate receptor IL-27R (the gp130 and TCCR chains) was elevated within post-mortem MS brain lesions compared with normal control brains. Moreover, astrocytes (GFAP(+) cells) as well as microglia and macrophages (Iba1(+) cells) were important sources of IL-27. Brain-infiltrating CD4 and CD8 T lymphocytes expressed the IL-27R specific chain (TCCR) implying that these cells could respond to local IL-27 sources. In primary cultures of human astrocytes inflammatory cytokines increased IL-27 production, whereas myeloid cell inflammatory M1 polarization and inflammatory cytokines enhanced IL-27 expression in microglia and macrophages. Astrocytes in postmortem tissues and in vitro expressed IL-27R. Moreover, IL-27 triggered the phosphorylation of the transcription regulator STAT1, but not STAT3 in human astrocytes; indeed IL-27 up-regulated MHC class I expression on astrocytes in a STAT1-dependent manner. These findings demonstrated that IL-27 and its receptor were elevated in MS lesions and that local IL-27 can modulate immune properties of astrocytes and infiltrating immune cells. Thus, therapeutic strategies targeting IL-27 may influence not only peripheral but also local inflammatory responses within the brain of MS patients.
Collapse
Affiliation(s)
- Vincent Sénécal
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| | - Gabrielle Deblois
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| | - Diane Beauseigle
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| | - Raphael Schneider
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| | - Jonas Brandenburg
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| | - Jia Newcombe
- NeuroResource, UCL Institute of Neurology, University College London, London, WC1N 1PJ, England
| | - Craig S Moore
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Alexandre Prat
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Nathalie Arbour
- Department of Neurosciences, Université De Montréal and CRCHUM Montreal, Quebec, Canada, H2X 0A9
| |
Collapse
|
154
|
Meka RR, Venkatesha SH, Dudics S, Acharya B, Moudgil KD. IL-27-induced modulation of autoimmunity and its therapeutic potential. Autoimmun Rev 2015; 14:1131-1141. [PMID: 26253381 PMCID: PMC4628569 DOI: 10.1016/j.autrev.2015.08.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/01/2015] [Indexed: 11/26/2022]
Abstract
Interleukin-27 (IL-27) is a new member of the IL-12 family. It is produced by activated antigen-presenting cells and plays an important role in the regulation of CD4+ T cell differentiation and immune response. IL-27 activates multiple signaling cascades, including the JAK-STAT and p38 MAPK pathways. Several studies have revealed that IL-27 promotes the differentiation of Th1 and Tr1, but inhibits Th2, Th17, and Treg cells. However, a few studies have shown an opposite effect on certain T cell subsets, such as Treg. IL-27 displays both pro- and anti- inflammatory activities in different autoimmune diseases. Here, we have discussed the role of IL-27 in rheumatoid arthritis, multiple sclerosis, colitis, lupus, psoriasis, type 1 diabetes, and uveitis. Most of this information is derived from experimental models of these autoimmune diseases. The mechanistic basis of the dual role of IL-27 in inflammation and autoimmunity is still not fully defined. In general, the pro-/anti-inflammatory activity of IL-27 is influenced by the underlying immune effector pathways, the phase of the disease, the presence or absence of counter-regulatory cytokines/T cell subsets, and the tissue/cell type under study. Despite a spectrum of outcomes in various autoimmune diseases, mostly anti-inflammatory and immunomodulatory effects of IL-27 have been observed in this category of diseases. Accordingly, IL-27 represents a novel, promising target/agent for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Rakeshchandra R. Meka
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Shivaprasad H. Venkatesha
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Steven Dudics
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Bodhraj Acharya
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Kamal D. Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
155
|
Li MS, Liu Z, Liu JQ, Zhu X, Liu Z, Bai XF. The Yin and Yang aspects of IL-27 in induction of cancer-specific T-cell responses and immunotherapy. Immunotherapy 2015; 7:191-200. [PMID: 25713993 DOI: 10.2217/imt.14.95] [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: 12/18/2022] Open
Abstract
Accumulating evidences from animal studies have indicated that both endogenous and exogenous IL-27, an IL-12 family of cytokine, can increase antitumor T-cell activities and inhibit tumor growth. IL-27 can modulate Treg responses, and program effector T cells into a unique T-effector stem cell (TSEC) phenotype, which enhances T-cell survival in the tumor microenvironment. However, animal studies also suggest that IL-27 induces molecular pathways such as IL-10, PD-L1 and CD39, which may downregulate tumor-specific T-cell responses. In this review paper, we will discuss the Yin and Yang aspects of IL-27 in the induction of tumor-specific T-cell responses, and the potential impacts of these functions of IL-27 in the design of cancer immunotherapy.
Collapse
Affiliation(s)
- Ming-Song Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
156
|
Lupar E, Brack M, Garnier L, Laffont S, Rauch KS, Schachtrup K, Arnold SJ, Guéry JC, Izcue A. Eomesodermin Expression in CD4+ T Cells Restricts Peripheral Foxp3 Induction. THE JOURNAL OF IMMUNOLOGY 2015; 195:4742-52. [PMID: 26453746 DOI: 10.4049/jimmunol.1501159] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
CD4(+) T cells polarize into effector Th subsets characterized by signature transcription factors and cytokines. Although T-bet drives Th1 responses and represses the alternative Th2, Th17, and Foxp3(+) regulatory T cell fates, the role of the T-bet-related transcription factor eomesodermin (Eomes) in CD4(+) T cells is less well understood. In this study, we analyze the expression and effects of Eomes in mouse CD4(+) T lymphocytes. We find that Eomes is readily expressed in activated CD4(+) Th1 T cells in vivo. Eomes(+) CD4(+) T cells accumulated in old mice, under lymphopenic conditions in a T cell transfer model of colitis, and upon oral Ag administration. However, despite its expression, genetic deletion of Eomes in CD4(+) T cells did not impact on IFN-γ production nor increase Th2 or Th17 responses. In contrast, Eomes deficiency favored the accumulation of Foxp3(+) cells in old mice, after in vivo differentiation of Eomes-deficient naive CD4(+) T cells, and in response to oral Ag in a cell-intrinsic way. Enforced Eomes expression during in vitro regulatory T cell induction also reduced Foxp3 transcription. Likewise, bystander Eomes-deficient CD4(+) T cells were more efficient at protecting from experimental autoimmune encephalitis compared with wild-type CD4(+) T cells. This enhanced capacity of Eomes-deficient CD4(+) T cells to inhibit EAE in trans was associated with an enhanced frequency of Foxp3(+) cells. Our data identify a novel role for Eomes in CD4(+) T cells and indicate that Eomes expression may act by limiting Foxp3 induction, which may contribute to the association of EOMES to susceptibility to multiple sclerosis.
Collapse
Affiliation(s)
- Ekaterina Lupar
- Max-Planck-Institute of Immunobiology and Epigenetics, D-79108 Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany
| | - Maria Brack
- Max-Planck-Institute of Immunobiology and Epigenetics, D-79108 Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany
| | - Laure Garnier
- INSERM, U1043, Toulouse, F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse, F-31300, France; Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, F-31300, France
| | - Sophie Laffont
- INSERM, U1043, Toulouse, F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse, F-31300, France; Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, F-31300, France
| | - Katharina S Rauch
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany
| | - Kristina Schachtrup
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany
| | - Sebastian J Arnold
- University Medical Centre, Renal Department, Centre for Clinical Research, D-79106 Freiburg, Germany; BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, D-79104 Freiburg, Germany; and Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, D-79102 Freiburg, Germany
| | - Jean-Charles Guéry
- INSERM, U1043, Toulouse, F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse, F-31300, France; Université de Toulouse, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan, Toulouse, F-31300, France
| | - Ana Izcue
- Max-Planck-Institute of Immunobiology and Epigenetics, D-79108 Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, D-79106 Freiburg, Germany;
| |
Collapse
|
157
|
Jeon YT, Na H, Ryu H, Chung Y. Modulation of Dendritic Cell Activation and Subsequent Th1 Cell Polarization by Lidocaine. PLoS One 2015; 10:e0139845. [PMID: 26445366 PMCID: PMC4596553 DOI: 10.1371/journal.pone.0139845] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/17/2015] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells play an essential role in bridging innate and adaptive immunity by recognizing cellular stress including pathogen- and damage-associated molecular patterns and by shaping the types of antigen-specific T cell immunity. Although lidocaine is widely used in clinical settings that trigger cellular stress, it remains unclear whether such treatment impacts the activation of innate immune cells and subsequent differentiation of T cells. Here we showed that lidocaine inhibited the production of IL–6, TNFα and IL–12 from dendritic cells in response to toll-like receptor ligands including lipopolysaccharide, poly(I:C) and R837 in a dose-dependent manner. Notably, the differentiation of Th1 cells was significantly suppressed by the addition of lidocaine while the same treatment had little effect on the differentiation of Th17, Th2 and regulatory T cells in vitro. Moreover, lidocaine suppressed the ovalbumin-specific Th1 cell responses in vivo induced by the adoptive transfer of ovalbumin-pulsed dendritic cells. These results demonstrate that lidocaine inhibits the activation of dendritic cells in response to toll-like receptor signals and subsequently suppresses the differentiation of Th1 cell responses.
Collapse
Affiliation(s)
- Young-Tae Jeon
- Department of Anesthesiology and Pain Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyeongjin Na
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Heeju Ryu
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
158
|
Chong WP, van Panhuys N, Chen J, Silver PB, Jittayasothorn Y, Mattapallil MJ, Germain RN, Caspi RR. NK-DC crosstalk controls the autopathogenic Th17 response through an innate IFN-γ-IL-27 axis. J Exp Med 2015; 212:1739-52. [PMID: 26347474 PMCID: PMC4577839 DOI: 10.1084/jem.20141678] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 08/11/2015] [Indexed: 12/13/2022] Open
Abstract
IFN-γ is a pathogenic cytokine involved in inflammation. Paradoxically, its deficiency exacerbates experimental autoimmune encephalomyelitis, uveitis, and arthritis. Here, we demonstrate using IFN-γ(-/-) mice repleted with IFN-γ +/+: NK cells that innate production of IFN-γ from NK cells is necessary and sufficient to trigger an endogenous regulatory circuit that limits autoimmunity. After immunization, DCs recruited IFN-γ-producing NK cells to the draining lymph node and interacted with them in a CXCR3-dependent fashion. The interaction caused DCs to produce IL-27, which in turn enhanced IFN-γ production by NK cells, forming a self-amplifying positive feedback loop. IL-10, produced by the interacting cells themselves, was able to limit this process. The NK-DC-dependent IL-27 inhibited development of the adaptive pathogenic IL-17 response and induced IL-10-producing Tr1-like cells, which ameliorated disease in an IL-10-dependent manner. Our data reveal that an early NK-DC interaction controls the adaptive Th17 response and limits tissue-specific autoimmunity through an innate IFN-γ-IL-27 axis.
Collapse
Affiliation(s)
- Wai Po Chong
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Nicholas van Panhuys
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmical Center, Sun Yat-sen University, Guangzhou 510060, China Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Phyllis B Silver
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Yingyos Jittayasothorn
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mary J Mattapallil
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ronald N Germain
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
159
|
Wang WB, Yen ML, Liu KJ, Hsu PJ, Lin MH, Chen PM, Sudhir PR, Chen CH, Chen CH, Sytwu HK, Yen BL. Interleukin-25 Mediates Transcriptional Control of PD-L1 via STAT3 in Multipotent Human Mesenchymal Stromal Cells (hMSCs) to Suppress Th17 Responses. Stem Cell Reports 2015; 5:392-404. [PMID: 26321145 PMCID: PMC4618596 DOI: 10.1016/j.stemcr.2015.07.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/29/2015] [Accepted: 07/30/2015] [Indexed: 02/08/2023] Open
Abstract
Multipotent human mesenchymal stromal cells (hMSCs) harbor immunomodulatory properties that are therapeutically relevant. One of the most clinically important populations of leukocytes is the interleukin-17A (IL-17A)-secreting T (Th17) lymphocytes. However, mechanisms of hMSC and Th17 cell interactions are incompletely resolved. We found that, along with Th1 responses, hMSCs strongly suppressed Th17 responses and this required both IL-25—also known as IL-17E—as well as programmed death ligand-1 (PD-L1), a potent cell surface ligand for tolerance induction. Knockdown of IL-25 expression in hMSCs abrogated Th17 suppression in vitro and in vivo. However, IL-25 alone was insufficient to significantly suppress Th17 responses, which also required surface PD-L1 expression. Critically, IL-25 upregulated PD-L1 surface expression through the signaling pathways of JNK and STAT3, with STAT3 found to constitutively occupy the proximal region of the PD-L1 promoter. Our findings demonstrate the complexities of hMSC-mediated Th17 suppression, and highlight the IL-25/STAT3/PD-L1 axis as a candidate therapeutic target. hMSC-secreted IL-25 suppress Th17 responses in vitro and in vivo IL-25 alone is insufficient to significantly suppress Th17 responses IL-25 upregulates PD-L1 expression in hMSCs to suppress Th17 cells IL-25-mediated PD-L1 expression can be driven by STAT3
Collapse
Affiliation(s)
- Wei-Bei Wang
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 35053, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics/Gynecology, National Taiwan University Hospital and School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, NHRI, Tainan 70403, Taiwan; Taipei Medical University, Taipei 10031, Taiwan.
| | - Pei-Ju Hsu
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 35053, Taiwan
| | - Ming-Hong Lin
- Graduate Institute of Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Pei-Min Chen
- Department of Obstetrics/Gynecology, National Taiwan University Hospital and School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | | | - Chein-Hung Chen
- Genomic Research Center, Academia Sinica, Taipei 11529, Taiwan
| | | | - Huei-Kang Sytwu
- Graduate Institute of Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 35053, Taiwan; Department of Obstetrics/Gynecology, Cathay General Hospital Shiji, Taipei 21174, Taiwan.
| |
Collapse
|
160
|
Yoshimoto T, Chiba Y, Furusawa JI, Xu M, Tsunoda R, Higuchi K, Mizoguchi I. Potential clinical application of interleukin-27 as an antitumor agent. Cancer Sci 2015; 106:1103-10. [PMID: 26132605 PMCID: PMC4582978 DOI: 10.1111/cas.12731] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/17/2015] [Accepted: 06/25/2015] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapies such as sipuleucel-T and ipilimumab are promising new treatments that harness the power of the immune system to fight cancer and achieve long-lasting remission. Interleukin (IL)-27, a member of the IL-12 heterodimeric cytokine family, has pleiotropic functions in the regulation of immune responses with both pro-inflammatory and anti-inflammatory properties. Evidence obtained using a variety of preclinical mouse models indicates that IL-27 possesses potent antitumor activity against various types of tumors through multiple mechanisms without apparent adverse effects. These mechanisms include those mediated not only by CD8+ T cells, natural killer cells and macrophages, but also by antibody-dependent cell-mediated cytotoxicity, antiangiogenesis, direct antiproliferative effects, inhibition of expression of cyclooxygenase-2 and prostaglandin E2, and suppression of epithelial–mesenchymal transition, depending on the characteristics of individual tumors. However, the endogenous role of IL-27 subunits and one of its receptor subunits, WSX-1, in the susceptibility to tumor development after transplantation of tumor cell lines or endogenously arising tumors seems to be more complicated. IL-27 functions as a double-edged sword: IL-27 increases IL-10 production and the expression of programmed death ligand 1 and T-cell immunoglobulin and mucin domain-3, and promotes the generation of regulatory T cells, and IL-27 receptor α singling enhances transformation; IL-27 may augment protumor effects as well. Here, we review both facets of IL-27, antitumor effects and protumor effects, and discuss the potential clinical application of IL-27 as an antitumor agent.
Collapse
Affiliation(s)
- Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yukino Chiba
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Jun-Ichi Furusawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Ren Tsunoda
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Kaname Higuchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
161
|
Yuan S, Chen QP. Th17 cells and intestinal mucosal immunity. Shijie Huaren Xiaohua Zazhi 2015; 23:3094-3100. [DOI: 10.11569/wcjd.v23.i19.3094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
T helper cell 17 (Th17) cells are identified as a new subset of T helper cells. Their differentiation is associated with a variety of cytokines and transcription factors, and they can secrete a variety of cytokines, such as interleukin (IL)-17 and IL-22, both of which can promote inflammation in the intestinal mucosa barrier and have a protective effect on organs. Probiotics
have been confirmed to have anti-inflammatory effects in the intestinal tract, the role of which may be associated with inhibiting Th17 cell activity. However, the stable number of Th17 cells requires the presence of intestinal symbiotic microbita. This paper will review the differentiation of Th17 cells and their role in intestinal mucosal immunity.
Collapse
|
162
|
Aparicio-Siegmund S, Garbers C. The biology of interleukin-27 reveals unique pro- and anti-inflammatory functions in immunity. Cytokine Growth Factor Rev 2015. [PMID: 26195434 DOI: 10.1016/j.cytogfr.2015.07.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin (IL)-27 is a multifaceted heterodimeric cytokine with pronounced pro- and anti-inflammatory as well as immunoregulatory functions. It consists of the two subunits p28/IL-30 and Epstein Bar virus-induced protein 3 (EBI3). EBI3 functions as a soluble α-receptor, and IL-27 can therefore directly activate its target cells through a heterodimer of glycoprotein 130 (gp130) and WSX-1. Being a heterodimeric cytokine that signals through gp130, IL-27 is either grouped into the IL-6 or the IL-12 family of cytokines. Originally identified as an IL-12-like cytokine that induces proliferation of CD4+ T cells and production of IFN-γ more than ten years ago, subsequent research revealed a much broader role of IL-27 in inflammation, cancer development and regulation and differentiation of immune cells. In this review, we summarize the current biochemical and molecular knowledge about the signal transduction of IL-27. Based on this, we highlight functional overlaps and plasticity with other cytokines and cytokine receptors of the IL-6/IL-12 superfamily, and describe the important role of IL-27 with regard to the differentiation of T cells, infections and cancer development. We further discuss IL-27 as a therapeutic target and how specific blockade of this cytokine could be achieved.
Collapse
Affiliation(s)
| | - Christoph Garbers
- Institute of Biochemistry, Kiel University, Olshausenstrasse 40, Kiel 24098, Germany.
| |
Collapse
|
163
|
Karachaliou N, Cao MG, Teixidó C, Viteri S, Morales-Espinosa D, Santarpia M, Rosell R. Understanding the function and dysfunction of the immune system in lung cancer: the role of immune checkpoints. Cancer Biol Med 2015; 12:79-86. [PMID: 26175923 PMCID: PMC4493378 DOI: 10.7497/j.issn.2095-3941.2015.0029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/10/2015] [Indexed: 12/17/2022] Open
Abstract
Survival rates for metastatic lung cancer, including non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), are poor with 5-year survivals of less than 5%. The immune system has an intricate and complex relationship with tumorigenesis; a groundswell of research on the immune system is leading to greater understanding of how cancer progresses and presenting new ways to halt disease progress. Due to the extraordinary power of the immune system-with its capacity for memory, exquisite specificity and central and universal role in human biology-immunotherapy has the potential to achieve complete, long-lasting remissions and cures, with few side effects for any cancer patient, regardless of cancer type. As a result, a range of cancer therapies are under development that work by turning our own immune cells against tumors. However deeper understanding of the complexity of immunomodulation by tumors is key to the development of effective immunotherapies, especially in lung cancer.
Collapse
Affiliation(s)
- Niki Karachaliou
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Maria Gonzalez Cao
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Cristina Teixidó
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Santiago Viteri
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Daniela Morales-Espinosa
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Mariacarmela Santarpia
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Rafael Rosell
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| |
Collapse
|
164
|
Hirahara K, Onodera A, Villarino AV, Bonelli M, Sciumè G, Laurence A, Sun HW, Brooks SR, Vahedi G, Shih HY, Gutierrez-Cruz G, Iwata S, Suzuki R, Mikami Y, Okamoto Y, Nakayama T, Holland SM, Hunter CA, Kanno Y, O'Shea JJ. Asymmetric Action of STAT Transcription Factors Drives Transcriptional Outputs and Cytokine Specificity. Immunity 2015; 42:877-89. [PMID: 25992861 PMCID: PMC11037422 DOI: 10.1016/j.immuni.2015.04.014] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/14/2015] [Accepted: 04/27/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-6 (IL-6) and IL-27 signal through a shared receptor subunit and employ the same downstream STAT transcription proteins, but yet are ascribed unique and overlapping functions. To evaluate the specificity and redundancy for these cytokines, we quantified their global transcriptomic changes and determined the relative contributions of STAT1 and STAT3 using genetic models and chromatin immunoprecipitation-sequencing (ChIP-seq) approaches. We found an extensive overlap of the transcriptomes induced by IL-6 and IL-27 and few examples in which the cytokines acted in opposition. Using STAT-deficient cells and T cells from patients with gain-of-function STAT1 mutations, we demonstrated that STAT3 is responsible for the overall transcriptional output driven by both cytokines, whereas STAT1 is the principal driver of specificity. STAT1 cannot compensate in the absence of STAT3 and, in fact, much of STAT1 binding to chromatin is STAT3 dependent. Thus, STAT1 shapes the specific cytokine signature superimposed upon STAT3's action.
Collapse
Affiliation(s)
- Kiyoshi Hirahara
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA; Department of Advanced Allergology of the Airway, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Japan
| | - Alejandro V Villarino
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Bonelli
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Giuseppe Sciumè
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arian Laurence
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Golnaz Vahedi
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Han-Yu Shih
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gustavo Gutierrez-Cruz
- Laboratory of Muscle Stem Cells and Gene Regulation, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shigeru Iwata
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryo Suzuki
- Laboratory of Molecular Immunogenetics, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yohei Mikami
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Chiba, Japan
| | - Steven M Holland
- The Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuka Kanno
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA.
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
165
|
Biswas PS, Aggarwal R, Levesque MC, Maers K, Ramani K. Type I interferon and T helper 17 cells co-exist and co-regulate disease pathogenesis in lupus patients. Int J Rheum Dis 2015; 18:646-53. [DOI: 10.1111/1756-185x.12636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Partha S. Biswas
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Rohit Aggarwal
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Marc C. Levesque
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Kelly Maers
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Kritika Ramani
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| |
Collapse
|
166
|
Decreased IL-27 Negatively Correlated with Th17 Cells in Non-Small-Cell Lung Cancer Patients. Mediators Inflamm 2015; 2015:802939. [PMID: 25969628 PMCID: PMC4417605 DOI: 10.1155/2015/802939] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/22/2015] [Indexed: 11/29/2022] Open
Abstract
The presence of Th17 cells and IL-27 is observed in a variety of inflammatory associated cancers. However, there are some data on the role of Th17 cells and IL-27 in the regulation of immune reactions in non-small-cell lung cancer (NSCLC). The aim of this study is to assess the variation of Th17 cells and IL-27 in the peripheral blood (PB) of patients with NSCLC. The proportion of Th17 cells in peripheral blood mononuclear cells (PBMCs) was evaluated by flow cytometry. The serum concentrations of IL-27 and IL-17 were measured by enzyme-linked immunosorbent assay (ELISA). The mRNA expression of RORγt and IL-27 in the peripheral blood was examined by real-time quantitative polymerase chain reaction (QPCR). Expression of IL-27 was lower in NSCLC patients compared with normal controls. The frequency of Th17 cells was increased in NSCLC patients, accompanied by the upregulation of IL-17 and RORγt. IL-27 negatively correlated with the number of Th17 cells and the RORγt mRNA. Our results indicate that IL-27 might inhibit Th17 differentiation in NSCLC patients and better understanding of the regulatory effects of IL-27 on Th17 cells may shed light on potential new targets in cancer prevention and therapy.
Collapse
|
167
|
Duan Y, Jia Y, Wang T, Wang Y, Han X, Liu L. Potent therapeutic target of inflammation, virus and tumor: focus on interleukin-27. Int Immunopharmacol 2015; 26:139-46. [PMID: 25812768 DOI: 10.1016/j.intimp.2015.03.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 03/13/2015] [Accepted: 03/13/2015] [Indexed: 10/23/2022]
Abstract
Interleukin (IL)-27 is an important pleiotropic immunological regulator for having dual effects on the immune responses. Several distinct findings have been highlighted that in certain conditions, neutralizing IL-27 or its subunit IL-27p28 might be a useful strategy to limit inflammation. Recently more insights have revealed that IL-27 could also exert potent inhibitory functions in some other immune circumstances including virus infection and tumor immunity. In this review, we describe IL-27 receptor subunits and the mechanisms of individual IL-27, and summarize the advances of their preclinical application trials. In addition, the potential role of IL-27 as a clinical therapeutic target will also be discussed.
Collapse
Affiliation(s)
- Yuqing Duan
- Department of Biotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, China
| | - Yunlong Jia
- Department of Biotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, China
| | - Tingting Wang
- Department of Biotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, China
| | - Yu Wang
- Department of Biotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, China
| | - Xiaonan Han
- Department of Biotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, China
| | - Lihua Liu
- Department of Biotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, China.
| |
Collapse
|
168
|
Buddhisa S, Rinchai D, Ato M, Bancroft GJ, Lertmemongkolchai G. Programmed death ligand 1 on Burkholderia pseudomallei-infected human polymorphonuclear neutrophils impairs T cell functions. THE JOURNAL OF IMMUNOLOGY 2015; 194:4413-21. [PMID: 25801435 DOI: 10.4049/jimmunol.1402417] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/18/2015] [Indexed: 01/21/2023]
Abstract
Polymorphonuclear neutrophils (PMNs) are terminally differentiated cells that are involved in innate immune responses and form an early line of defense against pathogens. More recently, it has been shown that PMNs have immunosuppressive abilities on other immune cells. However, the effect of PMNs on T cell responses during bacterial infection remains to be determined. In this report, we examined the interaction of PMNs and T cells in response to infection with Burkholderia pseudomallei, the causative agent of human melioidosis. We observed that CD4(+) T cell proliferation and IFN-γ production in response to polyclonal activators is significantly inhibited by uninfected PMNs, and to a greater extent B. pseudomallei-infected PMNs. Programmed death ligand 1 (PD-L1), a known regulator of T cell activation, is increased in mRNA expression in the blood of patients and upon infection of PMNs in vitro. The increased expression of PD-L1 was correlated with the degree of T cell inhibition in individuals with type 2 diabetes, a major risk factor of melioidosis. In vitro, addition of anti-PD-L1 Abs blocked this inhibitory activity and restored proliferation of CD4(+) T cells and IFN-γ production, suggesting that PD-L1 on B. pseudomallei-infected PMNs is a regulatory molecule for the functions of T cells and may be involved in pathogenesis versus control of melioidosis.
Collapse
Affiliation(s)
- Surachat Buddhisa
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Darawan Rinchai
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; and
| | - Gregory J Bancroft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Ganjana Lertmemongkolchai
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand;
| |
Collapse
|
169
|
Affiliation(s)
- Hiroki Yoshida
- Department of Biomolecular Sciences, Division of Molecular and Cellular Immunoscience, Saga University Faculty of Medicine, Saga 849-8501, Japan;
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-4539;
| |
Collapse
|
170
|
Song MY, Hong CP, Park SJ, Kim JH, Yang BG, Park Y, Kim SW, Kim KS, Lee JY, Lee SW, Jang MH, Sung YC. Protective effects of Fc-fused PD-L1 on two different animal models of colitis. Gut 2015; 64:260-71. [PMID: 24902766 DOI: 10.1136/gutjnl-2014-307311] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Programmed death-ligand 1 (PD-L1) has been shown to negatively regulate immune responses via its interaction with PD-1 receptor. In this study, we investigated the effects of PD-L1-Fc treatment on intestinal inflammation using two murine models of inflammatory colitis induced by dextran sulfate sodium (DSS) and T-cell transfer. DESIGN The anti-colitis effect of adenovirus expressing Fc-conjugated PD-L1 (Ad/PD-L1-Fc) and recombinant PD-L1-Fc protein was evaluated in DSS-treated wild-type and Rag-1 knockout (KO) mice. We examined differentiation of T-helper cells, frequency of innate immune cells, and cytokine production by dendritic cells (DCs) in the colon from DSS-treated mice after PD-L1-Fc administration. In Rag-1 KO mice reconstituted with CD4 CD45RB(high) T cells, we assessed the treatment effect of PD-L1-Fc protein on the development of colitis. RESULTS Administration of Ad/PD-L1-Fc significantly ameliorated DSS-induced colitis, which was accompanied by diminished frequency of interleukin (IL)-17A-producing CD4 T cells and increased interferon-γ-producing CD4 T cells in the colon of DSS-fed mice. The anti-colitic effect of PD-L1-Fc treatment was also observed in DSS-treated Rag-1 KO mice, indicating lymphoid cell independency. PD-L1-Fc modulated cytokine production by colonic DCs and the effect was dependent on PD-1 expression. Furthermore, PD-L1-Fc protein could significantly reduce the severity of colitis in CD4 CD45RB(high) T-cell-transferred Rag-1 KO mice. CONCLUSIONS Based on the protective effect of PD-L1-Fc against DSS-induced and T-cell-induced colitis, our results suggest that PD-1-mediated inhibitory signals have a crucial role in limiting the development of colonic inflammation. This implicates that PD-L1-Fc may provide a novel therapeutic approach to treat inflammatory bowel disease.
Collapse
Affiliation(s)
- Mi-Young Song
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Chun-Pyo Hong
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Republic of Korea
| | - Seong Jeong Park
- Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jung-Hwan Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Republic of Korea
| | - Bo-Gie Yang
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Republic of Korea
| | - Yunji Park
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sae Won Kim
- Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Kwang Soon Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Republic of Korea
| | - Ji Yeung Lee
- Research Institute, Genexine Co., Seongnam, Republic of Korea
| | - Seung-Woo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Myoung Ho Jang
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Republic of Korea
| | - Young-Chul Sung
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea Research Institute, Genexine Co., Seongnam, Republic of Korea
| |
Collapse
|
171
|
Guilleminault L, Carmier D, Heuzé-Vourc'h N, Diot P, Pichon E. [Immunotherapy in non-small cell lung cancer: inhibition of PD1/PDL1 pathway]. REVUE DE PNEUMOLOGIE CLINIQUE 2015; 71:44-56. [PMID: 25687821 DOI: 10.1016/j.pneumo.2014.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 10/13/2014] [Accepted: 11/01/2014] [Indexed: 06/04/2023]
Abstract
Despite recent advances in targeted therapy of non-small cell lung cancer (NSCLC), many patients do not benefit from these therapies. Inhibition of PD1/PDL1 is an interesting therapeutic target which restores the immune system against tumor cells. PD1 is located on lymphocytes and PDL1 on the antigen presenting cells. PD1 and PDL1 are co-inhibition molecules and their interaction results in immune tolerance against tumor cells. Anti-PD1 and anti-PDL1 antibodies have been developed to restore immune system in solid cancer including NSCLC. In phase I, studies assessing nivolumab, an anti-PD1 antibody, objective responses were observed in 13 to 18% of NSCLC patients failing previous treatment. The data obtained with anti-PDL1 antibodies is similar with objective responses ranging from 6 to 22%. The encouraging results of phase I/II studies must be confirmed in ongoing phase III studies. Anti-PD1 and anti-PDL1 antibodies exposed to new adverse events including auto-immune diseases whose support is not codified. Questions about treatment duration and criteria evaluation are not resolved. These treatments pave the way for immunomodulation in NSCLC treatment.
Collapse
Affiliation(s)
- L Guilleminault
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France; Centre d'étude des pathologies respiratoires, UMR 1100/EA6305, 37032 Tours, France; EA6305, université François-Rabelais de Tours, 37032 Tours, France.
| | - D Carmier
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France
| | - N Heuzé-Vourc'h
- Centre d'étude des pathologies respiratoires, UMR 1100/EA6305, 37032 Tours, France; EA6305, université François-Rabelais de Tours, 37032 Tours, France
| | - P Diot
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France; Centre d'étude des pathologies respiratoires, UMR 1100/EA6305, 37032 Tours, France; EA6305, université François-Rabelais de Tours, 37032 Tours, France
| | - E Pichon
- Service de pneumologie et d'explorations fonctionnelles, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex, France
| |
Collapse
|
172
|
Iwasaki Y, Fujio K, Okamura T, Yamamoto K. Interleukin-27 in T cell immunity. Int J Mol Sci 2015; 16:2851-63. [PMID: 25633106 PMCID: PMC4346869 DOI: 10.3390/ijms16022851] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 01/19/2015] [Accepted: 01/22/2015] [Indexed: 12/03/2022] Open
Abstract
Interleukin (IL)-27, a member of IL-12/IL-23 heterodimeric family of cytokines, has pleiotropic properties that can enhance or limit immune responses. IL-27 acts on various cell types, including T cells, B cells, macrophages, dendritic cells, natural killer (NK) cells and non-hematopoietic cells. Intensive studies have been conducted especially on T cells, revealing that various subsets of T cells respond uniquely to IL-27. IL-27 induces expansion of Th1 cells by activating signal transducer and activator of transcription (STAT) 1-mediated T-bet signaling pathway. On the other hand, IL-27 suppresses immune responses through inhibition of the development of T helper (Th) 17 cells and induction of IL-10 production in a STAT1- and STAT3-dependent manner. IL-27 is a potentially promising cytokine for therapeutic approaches on various human diseases. Here, we provide an overview of the biology of IL-27 related to T cell subsets, its structure, and production mechanism.
Collapse
Affiliation(s)
- Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
173
|
Zhu C, Sakuishi K, Xiao S, Sun Z, Zaghouani S, Gu G, Wang C, Tan DJ, Wu C, Rangachari M, Pertel T, Jin HT, Ahmed R, Anderson AC, Kuchroo VK. An IL-27/NFIL3 signalling axis drives Tim-3 and IL-10 expression and T-cell dysfunction. Nat Commun 2015; 6:6072. [PMID: 25614966 PMCID: PMC4311884 DOI: 10.1038/ncomms7072] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/09/2014] [Indexed: 12/18/2022] Open
Abstract
The inhibitory receptor T-cell immunoglobulin and mucin domain-3 (Tim-3) has emerged as a critical regulator of the T-cell dysfunction that develops in chronic viral infections and cancers. However, little is known regarding the signalling pathways that drive Tim-3 expression. Here, we demonstrate that interleukin (IL)-27 induces nuclear factor, interleukin 3 regulated (NFIL3), which promotes permissive chromatin remodelling of the Tim-3 locus and induces Tim-3 expression together with the immunosuppressive cytokine IL-10. We further show that the IL-27/NFIL3 signalling axis is crucial for the induction of Tim-3 in vivo. IL-27-conditioned T helper 1 cells exhibit reduced effector function and are poor mediators of intestinal inflammation. This inhibitory effect is NFIL3 dependent. In contrast, tumour-infiltrating lymphocytes from IL-27R(-/-) mice exhibit reduced NFIL3, less Tim-3 expression and failure to develop dysfunctional phenotype, resulting in better tumour growth control. Thus, our data identify an IL-27/NFIL3 signalling axis as a key regulator of effector T-cell responses via induction of Tim-3, IL-10 and T-cell dysfunction.
Collapse
Affiliation(s)
- Chen Zhu
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Kaori Sakuishi
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Sheng Xiao
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Zhiyi Sun
- New England Biolabs Inc., 240 County Road, Ipswich, MA 01938, USA
| | - Sarah Zaghouani
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Guangxiang Gu
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Chao Wang
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Dewar J. Tan
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Chuan Wu
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Manu Rangachari
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Thomas Pertel
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Hyun-Tak Jin
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Ana C. Anderson
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| |
Collapse
|
174
|
Ji M, Liu Y, Li Q, Li XD, Zhao WQ, Zhang H, Zhang X, Jiang JT, Wu CP. PD-1/PD-L1 pathway in non-small-cell lung cancer and its relation with EGFR mutation. J Transl Med 2015; 13:5. [PMID: 25592115 PMCID: PMC4302082 DOI: 10.1186/s12967-014-0373-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/26/2014] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy has become a crucial modality for non-small-cell lung cancer treatment. Recently, two immune checkpoints, PD-1 and PD-L1, have emerged as important targets for immunotherapy. Their antitumor efficacy has been confirmed by in vitro and in vivo studies. But the correlation between PD-1/PD-L1 expression and EGFR expression was controversial and needs more evidences to support the combination of PD-1/PD-L1 inhibitors and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Mei Ji
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Yan Liu
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Xiao-Dong Li
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Wei-Qing Zhao
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Hanze Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, USA.
| | - Xiaofei Zhang
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| | - Jing-Ting Jiang
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Chang-Ping Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
175
|
Huang YH, Zhu C, Kondo Y, Anderson AC, Gandhi A, Russell A, Dougan SK, Petersen BS, Melum E, Pertel T, Clayton KL, Raab M, Chen Q, Beauchemin N, Yazaki PJ, Pyzik M, Ostrowski MA, Glickman JN, Rudd CE, Ploegh HL, Franke A, Petsko GA, Kuchroo VK, Blumberg RS. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 2015; 517:386-90. [PMID: 25363763 PMCID: PMC4297519 DOI: 10.1038/nature13848] [Citation(s) in RCA: 535] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 09/08/2014] [Indexed: 02/05/2023]
Abstract
T-cell immunoglobulin domain and mucin domain-3 (TIM-3, also known as HAVCR2) is an activation-induced inhibitory molecule involved in tolerance and shown to induce T-cell exhaustion in chronic viral infection and cancers. Under some conditions, TIM-3 expression has also been shown to be stimulatory. Considering that TIM-3, like cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1), is being targeted for cancer immunotherapy, it is important to identify the circumstances under which TIM-3 can inhibit and activate T-cell responses. Here we show that TIM-3 is co-expressed and forms a heterodimer with carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1), another well-known molecule expressed on activated T cells and involved in T-cell inhibition. Biochemical, biophysical and X-ray crystallography studies show that the membrane-distal immunoglobulin-variable (IgV)-like amino-terminal domain of each is crucial to these interactions. The presence of CEACAM1 endows TIM-3 with inhibitory function. CEACAM1 facilitates the maturation and cell surface expression of TIM-3 by forming a heterodimeric interaction in cis through the highly related membrane-distal N-terminal domains of each molecule. CEACAM1 and TIM-3 also bind in trans through their N-terminal domains. Both cis and trans interactions between CEACAM1 and TIM-3 determine the tolerance-inducing function of TIM-3. In a mouse adoptive transfer colitis model, CEACAM1-deficient T cells are hyper-inflammatory with reduced cell surface expression of TIM-3 and regulatory cytokines, and this is restored by T-cell-specific CEACAM1 expression. During chronic viral infection and in a tumour environment, CEACAM1 and TIM-3 mark exhausted T cells. Co-blockade of CEACAM1 and TIM-3 leads to enhancement of anti-tumour immune responses with improved elimination of tumours in mouse colorectal cancer models. Thus, CEACAM1 serves as a heterophilic ligand for TIM-3 that is required for its ability to mediate T-cell inhibition, and this interaction has a crucial role in regulating autoimmunity and anti-tumour immunity.
Collapse
MESH Headings
- Animals
- Antigens, CD/chemistry
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Autoimmunity/immunology
- Cell Adhesion Molecules/chemistry
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Cell Line
- Colorectal Neoplasms/immunology
- Disease Models, Animal
- Female
- Hepatitis A Virus Cellular Receptor 2
- Humans
- Immune Tolerance/immunology
- Inflammation/immunology
- Inflammation/pathology
- Ligands
- Male
- Membrane Proteins/chemistry
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Models, Molecular
- Mucous Membrane/immunology
- Mucous Membrane/pathology
- Protein Conformation
- Protein Multimerization
- Receptors, Virus/chemistry
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Yu-Hwa Huang
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Chen Zhu
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Harvard Institutes of Medicine, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Yasuyuki Kondo
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Harvard Institutes of Medicine, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Amit Gandhi
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Andrew Russell
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, USA
| | - Stephanie K Dougan
- Whitehead Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Britt-Sabina Petersen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Espen Melum
- 1] Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA [2] Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Oslo 0424, Norway
| | - Thomas Pertel
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Harvard Institutes of Medicine, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Kiera L Clayton
- Department of Immunology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Monika Raab
- Cell Signalling Section, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Qiang Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nicole Beauchemin
- Goodman Cancer Research Centre, McGill University, Montreal H3G 1Y6, Canada
| | - Paul J Yazaki
- Beckman Institute, City of Hope, Duarte, California 91010, USA
| | - Michal Pyzik
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Mario A Ostrowski
- 1] Department of Immunology, University of Toronto, Toronto, Ontario M5S1A8, Canada [2] Keenan Research Centre of St. Michael's Hospital, Toronto, Ontario M5S1A8, Canada
| | | | - Christopher E Rudd
- Cell Signalling Section, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Hidde L Ploegh
- Whitehead Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Gregory A Petsko
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Harvard Institutes of Medicine, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, USA
| |
Collapse
|
176
|
Abdalla AE, Li Q, Xie L, Xie J. Biology of IL-27 and its role in the host immunity against Mycobacterium tuberculosis. Int J Biol Sci 2015; 11:168-75. [PMID: 25561899 PMCID: PMC4279092 DOI: 10.7150/ijbs.10464] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/16/2014] [Indexed: 11/09/2022] Open
Abstract
IL-27, a heterodimeric cytokine of IL-12 family, regulates both innate and adaptive immunity largely via Jak-Stat signaling. IL-27 can induce IFN-γ and inflammatory mediators from T lymphocytes and innate immune cells. IL-27 has unique anti-inflammatory properties via both Tr1 cells dependent and independent mechanisms. Here the role and biology of IL-27 in innate and adaptive immunity are summarized, with special interest with immunity against Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Abualgasim Elgaili Abdalla
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
- 2. Department of Clinical Microbiology, College of Medical Laboratory Sciences, Omdurman Islamic University, Omdurman, Khartoum, Sudan
| | - Qiming Li
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Longxiang Xie
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Jianping Xie
- 1. Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| |
Collapse
|
177
|
Clarke EV, Weist BM, Walsh CM, Tenner AJ. Complement protein C1q bound to apoptotic cells suppresses human macrophage and dendritic cell-mediated Th17 and Th1 T cell subset proliferation. J Leukoc Biol 2015; 97:147-60. [PMID: 25381385 PMCID: PMC4377823 DOI: 10.1189/jlb.3a0614-278r] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/17/2022] Open
Abstract
A complete genetic deficiency of the complement protein C1q results in SLE with nearly 100% penetrance in humans, but the molecular mechanisms responsible for this association have not yet been fully determined. C1q opsonizes ACs for enhanced ingestion by phagocytes, such as Mϕ and iDCs, avoiding the extracellular release of inflammatory DAMPs upon loss of the membrane integrity of the dying cell. We previously showed that human monocyte-derived Mϕ and DCs ingesting autologous, C1q-bound LALs (C1q-polarized Mϕ and C1q-polarized DCs), enhance the production of anti-inflammatory cytokines, and reduce proinflammatory cytokines relative to Mϕ or DC ingesting LAL alone. Here, we show that C1q-polarized Mϕ have elevated PD-L1 and PD-L2 and suppressed surface CD40, and C1q-polarized DCs have higher surface PD-L2 and less CD86 relative to Mϕ or DC ingesting LAL alone, respectively. In an MLR, C1q-polarized Mϕ reduced allogeneic and autologous Th17 and Th1 subset proliferation and demonstrated a trend toward increased Treg proliferation relative to Mϕ ingesting LAL alone. Moreover, relative to DC ingesting AC in the absence of C1q, C1q-polarized DCs decreased autologous Th17 and Th1 proliferation. These data demonstrate that a functional consequence of C1q-polarized Mϕ and DC is the regulation of Teff activation, thereby "sculpting" the adaptive immune system to avoid autoimmunity, while clearing dying cells. It is noteworthy that these studies identify novel target pathways for therapeutic intervention in SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Elizabeth V Clarke
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| | - Brian M Weist
- Department of Molecular & Cell Biology, University of California-Berkeley, Berkeley, California, USA
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| |
Collapse
|
178
|
Forrester MA, Robertson L, Bayoumi N, Keavney BD, Barker RN, Vickers MA. Human interleukin-27: wide individual variation in plasma levels and complex inter-relationships with interleukin-17A. Clin Exp Immunol 2014; 178:373-83. [PMID: 24975574 DOI: 10.1111/cei.12408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2014] [Indexed: 12/14/2022] Open
Abstract
Although it is widely believed that interleukin (IL)-27 is anti-inflammatory, its role in controlling human immune responses is not fully established. In particular, its interactions with T helper type 17 (Th)17 cytokines are unclear. Our aims were to establish the relationships between IL-27 and proinflammatory cytokines, including IL-17A, in human sera and cultures of peripheral blood mononuclear cells. Plasma IL-27 levels in 879 healthy humans from 163 families varied widely, but with relatively low heritability (19%). Despite IL-27 including a subunit encoded by Epstein-Barr virus-induced gene 3 (EBI3), there was no correlation of levels with serological evidence of infection with the virus. Although IL-27 has been reported to inhibit IL-17A production, we demonstrated a strong positive correlation in sera, but lower correlations of IL-27 with other proinflammatory cytokines. We verified that IL-27 inhibited IL-17A production by human peripheral blood T cells in vitro, but not that it stimulated IL-10 secretion. Importantly, addition of IL-17A decreased IL-27 production by stimulated T cells but had the opposite effect on resting T cells. Together, these data suggest a model whereby IL-27 and IL-17A exerts complex reciprocal effects to boost inflammatory responses, but restrain resting cells to prevent inappropriate activation.
Collapse
Affiliation(s)
- M A Forrester
- Immunity, Infection and Inflammation, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | | | | | | |
Collapse
|
179
|
Chen Y, Chen S, Liu LY, Zou ZL, Cai YJ, Wang JG, Chen B, Xu LM, Lin Z, Wang XD, Chen YP. Mesenchymal stem cells ameliorate experimental autoimmune hepatitis by activation of the programmed death 1 pathway. Immunol Lett 2014; 162:222-8. [PMID: 25445618 DOI: 10.1016/j.imlet.2014.10.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 01/01/2023]
Abstract
Previous studies have shown beneficial effects of mesenchymal stem cells (MSCs) transplantation in many autoimmune diseases. However, few studies have focused on the effects of MSCs on autoimmune hepatitis. In our study, we investigated the therapeutic effects of BMSCs (bone mesenchymal stem cells) transplantation in mouse experimental autoimmune hepatitis (EAH) and explored the potential mechanism. BMSCs were injected intravenously into EAH mice. Then, serum levels of ALT and AST, and pathologic alteration of liver tissue were measured to evaluate the liver function and inflammation degree. The expressions of programmed death ligand 1, IL-17 and IL-23 were detected by enzyme-linked immunosorbent assay (ELISA), reverse transcription-polymerase chain reaction (RT-PCR), and western blotting. Upon serum biochemical levels and pathological examination, the BMSCs-treated mice especially with multiple dosing administration showed significantly reduction of liver damage. Moreover, the expression of IL-17 was down-regulated by BMSCs intervention as compared to the model group, whereas the PD-L1 and IL-23 were up-regulated following the administration of MSCs. In conclusion, the results of this study suggest that BMSCs transplantation, especially on multiple dosing, may exert immunosuppression effect to ameliorate EAH through the inhibition of IL-17 and up-regulation of PD-L1.
Collapse
Affiliation(s)
- Yi Chen
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Si Chen
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Li-Yuan Liu
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuo-Lin Zou
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi-Jing Cai
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jin-Guo Wang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bi Chen
- Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lan-Man Xu
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuo Lin
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao-Dong Wang
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yong-Ping Chen
- Hepatology Institute of Wenzhou Medical University, Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
180
|
Ellestad KK, Thangavelu G, Ewen CL, Boon L, Anderson CC. PD-1 is not required for natural or peripherally induced regulatory T cells: Severe autoimmunity despite normal production of regulatory T cells. Eur J Immunol 2014; 44:3560-72. [DOI: 10.1002/eji.201444688] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/06/2014] [Accepted: 09/16/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Kristofor K. Ellestad
- Department of Medical Microbiology and Immunology; University of Alberta; Edmonton AB Canada
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
| | - Govindarajan Thangavelu
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
- Department of Surgery; University of Alberta; Edmonton AB Canada
| | - Catherine L. Ewen
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
| | | | - Colin C. Anderson
- Department of Medical Microbiology and Immunology; University of Alberta; Edmonton AB Canada
- Alberta Diabetes and Transplant Institutes; University of Alberta; Edmonton AB Canada
- Department of Surgery; University of Alberta; Edmonton AB Canada
| |
Collapse
|
181
|
Pawaria S, Ramani K, Maers K, Liu Y, Kane LP, Levesque MC, Biswas PS. Complement component C5a permits the coexistence of pathogenic Th17 cells and type I IFN in lupus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3288-3295. [PMID: 25149466 DOI: 10.4049/jimmunol.1401322] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is a type I IFN (IFN-I)-driven autoimmune disorder with exaggerated B and Th cell responses. Th17 cells, a recently identified Th cell subset, have been strongly implicated in the pathogenesis of SLE. Because IFN-I suppresses the generation and expansion of Th17 cells in an IL-27-dependent manner, it is unclear how pathogenic Th17 cells are generated in SLE in the presence of an environment characterized by high IFN-I levels. In this study, we showed that activation of c5aR on murine macrophages blocked IFN-I-mediated IL-27 production, thus permitting the development of Th17 cells. C5aR activation on IFN-I-responsive macrophages inhibits IRF-1-mediated transactivation of IL-27 gene expression via the PI3K/Akt pathway. Consistently, C5aR-deficient mice exhibited increased IL-27 expression and fewer Th17 cells and consequently developed reduced lupus nephritis in comparison with wild-type mice. In support of these findings in mice, we found that C5a inhibited IFN-I-induced IL-27 production from macrophages of lupus subjects. Moreover, the level of serum C5a correlated with Th17 frequency in peripheral blood. Collectively, these data indicate an essential role for C5a in the generation of pathogenic Th17 responses in SLE. Thus, therapeutic strategies to block C5aR activation may be beneficial for controlling pathogenic Th17-mediated inflammation in SLE.
Collapse
Affiliation(s)
- Sudesh Pawaria
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kritika Ramani
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kelly Maers
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Youhua Liu
- Division of Pathology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261; and
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Marc C Levesque
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261;
| |
Collapse
|
182
|
Casaluce F, Sgambato A, Sacco PC, Palazzolo G, Maione P, Rossi A, Ciardiello F, Gridelli C. Emerging drugs targeting PD-1 and PD-L1: reality or hope? Expert Opin Emerg Drugs 2014; 19:557-69. [DOI: 10.1517/14728214.2014.964682] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
183
|
Abstract
Optimal T cell response is dependent not only on T cell receptor activation, but also on additional signaling from coreceptors. The main coreceptors include B7 and tumor necrosis factor family members. They exert costimulatory or coinhibitory effects, and their balance determines the fate of T cell response. In normal conditions, costimulators facilitate the development of protective immune response, whereas coinhibitors dampen inflammation to avoid organ/tissue damage from excessive immune reaction. In the tumor microenvironment, the balance is garbled: inhibitory pathways predominate, and T cell response is impaired. The importance of cosignaling in the tumor immune response has been experimentally and clinically demonstrated. New therapeutic strategies targeting T cell cosignaling, especially coinhibitory molecules, are under active experimental and clinical investigation. This review summarizes the functions of main T cell cosignaling axes and discusses their clinical application.
Collapse
|
184
|
Fujiwara H, Maeda Y, Kobayashi K, Nishimori H, Matsuoka KI, Fujii N, Kondo E, Tanaka T, Chen L, Azuma M, Yagita H, Tanimoto M. Programmed death-1 pathway in host tissues ameliorates Th17/Th1-mediated experimental chronic graft-versus-host disease. THE JOURNAL OF IMMUNOLOGY 2014; 193:2565-73. [PMID: 25080485 DOI: 10.4049/jimmunol.1400954] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chronic graft-versus-host disease (GVHD) is a major cause of late death and morbidity after allogeneic hematopoietic cell transplantation, but its pathogenesis remains unclear. We investigated the role of the programmed death-1 (PD-1) pathway in chronic GVHD using a well-defined mouse model of B10.D2 (H-2(d)) donor to BALB/c (H-2(d)) recipients. PD-1 expression on allogeneic donor T cells was upregulated continuously in chronic GVHD development, whereas PD-L1 expression in host tissues was transiently upregulated and declined to basal levels in the late posttransplant period. Blockade of the PD-1 pathway by anti-PD-1, anti-PD-L1, or anti-PD-L2 mAbs exacerbated clinical and pathologic chronic GVHD. Chimeric mice revealed that PD-L1 expression in host tissues suppressed expansion of IL-17(+)IFN-γ(+) T cells, and that PD-L1 expression on hematopoietic cells plays a role in the development of regulatory T cells only during the early transplantation period but does not affect the severity of chronic GVHD. Administration of the synthetic retinoid Am80 overcame the IL-17(+)IFN-γ(+) T cell expansion caused by PD-L1 deficiency, resulting in reduced chronic GVHD damage in PD-L1(-/-) recipients. Stimulation of the PD-1 pathway also alleviated chronic GVHD. These results suggest that the PD-1 pathway contributes to the suppression of Th17/Th1-mediated chronic GVHD and may represent a new target for the prevention or treatment of chronic GVHD.
Collapse
Affiliation(s)
- Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558;
| | - Koichiro Kobayashi
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Eisei Kondo
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Takehiro Tanaka
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| | - Lieping Chen
- Department of Immunobiology and Yale Comprehensive Cancer Center, Yale University, New Haven, CT 06519
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan 113-8549; and
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan 113-8421
| | - Mitsune Tanimoto
- Department of Hematology and Oncology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan 700-8558
| |
Collapse
|
185
|
TH17 cells in human recurrent pregnancy loss and pre-eclampsia. Cell Mol Immunol 2014; 11:564-70. [PMID: 25027967 DOI: 10.1038/cmi.2014.54] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/06/2014] [Accepted: 06/06/2014] [Indexed: 12/21/2022] Open
Abstract
T helper 17 (TH17) cells have been identified as a new lineage of helper T cells and have been shown to be important in host defense against extracellular infectious agents, autoimmune disease and chronic inflammatory diseases. Recently, TH17 cells have also been shown to participate in successful pregnancy, as well as in the pathogenesis of diseases of pregnancy, such as recurrent spontaneous abortion (RSA) and pre-eclampsia (PE). Here, we review our current knowledge of TH17 cells in human RSA and PE. We also discuss how the local uterine microenvironment affects the differentiation of TH17 cells and the mechanisms that regulate TH17 cells during pregnancy. Research into TH17 cells will not only advance our understanding of TH17-related pregnancy complications, but will also facilitate the design of novel therapies for reproductive diseases.
Collapse
|
186
|
Takeda A, Hasegawa E, Fukuhara T, Hirakawa S, Yamada H, Yang Y, Yoshimura T, Hisatomi T, Oshima Y, Yoshida H, Sonoda KH, Ishibashi T. EBI3 is pivotal for the initiation of experimental autoimmune uveitis. Exp Eye Res 2014; 125:107-13. [PMID: 24929202 DOI: 10.1016/j.exer.2014.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 05/22/2014] [Accepted: 06/03/2014] [Indexed: 11/16/2022]
Abstract
Murine experimental autoimmune uveitis (EAU) is a model for human autoimmune uveitis, whose pathogenesis is caused by both Th1 and Th17 cell responses. Epstein-Barr virus-induced gene 3 (EBI3) is a component of the heterodimeric cytokines: interleukin (IL)-27 and IL-35. Although IL-27 was shown to initiate Th1 cell development, it is also recognized as a negative regulator of fully activated CD4+ T cells, including Th17 cells. Recently, IL-35 also has also been reported to play immunosuppressive roles in autoimmunity. To investigate the roles of EBI3 in EAU, EBI3(-/-) mice were immunized with human interphotoreceptor retinoid binding protein peptide 1-20 (IRBP) to induce EAU. We observed that the clinical score in EBI3(-/-) mice was diminished compared with that in EBI3(+/+) mice up to day 22 after immunization, whereas the score in EBI3(-/-) mice reached the same levels as that of EBI3(+/+) mice after day 28. Histological analysis revealed a significant reduction of cellular infiltration into the retina in EBI3(-/-) mice on day 16. Although Th1 cell responses and IRBP-specific IL-10 production were reduced in EBI3(-/-) mice, the development of Th17 cell responses was unaffected on day 9. On day 21, Th1 cell responses and IRBP-specific IL-10 production was restored to the same levels as that in EBI3(+/+) mice, and Th17 cell responses significantly increased in EBI3(-/-) mice. Furthermore, Foxp3 expression in CD4+ T cells was comparable between EBI3(+/+) and EBI3(-/-) mice on days 9 and 21. Therefore, these results indicate that EBI3 may be important in EAU initiation by Th1 cell responses and may suppress EAU by inhibition of both Th1 and Th17 cell responses in the late/maintenance phase.
Collapse
Affiliation(s)
- Atsunobu Takeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Eiichi Hasegawa
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takako Fukuhara
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Sayaka Hirakawa
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hisakata Yamada
- Division of Host Defense, Research Center for Prevention of Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yang Yang
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeru Yoshimura
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Toshio Hisatomi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuji Oshima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunosciences, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima Saga, Saga 849-8501, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tatsuro Ishibashi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
187
|
Visperas A, Do J, Bulek K, Li X, Min B. IL-27, targeting antigen-presenting cells, promotes Th17 differentiation and colitis in mice. Mucosal Immunol 2014; 7:625-33. [PMID: 24129161 PMCID: PMC3989480 DOI: 10.1038/mi.2013.82] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/11/2013] [Indexed: 02/04/2023]
Abstract
T helper type 17 (Th17) cells have been implicated in autoimmunity and inflammatory bowel disease (IBD). Antigen-presenting cell (APC) -derived cytokines such as interleukin (IL)-1β and IL-6 are key mediators supporting Th17 differentiation, yet how these factors are induced in vivo remains unclear. Here, we show that IL-27 acting on APCs enhances IL-6 and IL-1β production and Th17 differentiation. IL-27Rα-/- T-cell receptor (TCR)β-/- recipients fail to develop gut inflammation following naive CD4 T-cell transfer, whereas IL-27Rα+/+ TCRβ-/- recipients develop severe colitis. Investigation of T-cell responses exhibits that IL-27Rα-/- TCRβ-/- mice do not support Th17 differentiation with significantly decreased levels of IL-6 and IL-1β by APCs. Our study has identified a novel proinflammatory role for IL-27 in vivo that promotes Th17 differentiation by inducing Th17-supporting cytokines in APCs.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigens/immunology
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Differentiation
- Colitis/genetics
- Colitis/immunology
- Colitis/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Disease Models, Animal
- Interleukin-27/genetics
- Interleukin-27/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Interleukin/genetics
- Receptors, Interleukin/metabolism
- Th17 Cells/cytology
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Anabelle Visperas
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
- Department of Molecular Medicine, Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| | - Jeongsu Do
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Katarzyna Bulek
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Xiaoxia Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
- Department of Molecular Medicine, Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|
188
|
Bandaru A, Devalraju KP, Paidipally P, Dhiman R, Venkatasubramanian S, Barnes PF, Vankayalapati R, Valluri V. Phosphorylated STAT3 and PD-1 regulate IL-17 production and IL-23 receptor expression in Mycobacterium tuberculosis infection. Eur J Immunol 2014; 44:2013-24. [PMID: 24643836 DOI: 10.1002/eji.201343680] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 02/13/2014] [Accepted: 03/14/2014] [Indexed: 11/07/2022]
Abstract
We studied the factors that regulate IL-23 receptor expression and IL-17 production in human tuberculosis infection. Mycobacterium tuberculosis (M. tb)-stimulated CD4(+) T cells from tuberculosis patients secreted less IL-17 than did CD4(+) T cells from healthy tuberculin reactors (PPD(+) ). M. tb-cultured monocytes from tuberculosis patients and PPD(+) donors expressed equal amounts of IL-23p19 mRNA and protein, suggesting that reduced IL-23 production is not responsible for decreased IL-17 production by tuberculosis patients. Freshly isolated and M. tb-stimulated CD4(+) T cells from tuberculosis patients had reduced IL-23 receptor and phosphorylated STAT3 (pSTAT3) expression, compared with cells from PPD(+) donors. STAT3 siRNA reduced IL-23 receptor expression and IL-17 production by CD4(+) T cells from PPD(+) donors. Tuberculosis patients had increased numbers of PD-1(+) T cells compared with healthy PPD(+) individuals. Anti-PD-1 antibody enhanced pSTAT3 and IL-23R expression and IL-17 production by M. tb-cultured CD4(+) T cells of tuberculosis patients. Anti-tuberculosis therapy decreased PD-1 expression, increased IL-17 and IFN-γ production and pSTAT3 and IL-23R expression. These findings demonstrate that increased PD-1 expression and decreased pSTAT3 expression reduce IL-23 receptor expression and IL-17 production by CD4(+) T cells of tuberculosis patients.
Collapse
Affiliation(s)
- Anuradha Bandaru
- Blue Peter Research Center, LEPRA Society, Cherlapally, Hyderabad, India
| | | | | | | | | | | | | | | |
Collapse
|
189
|
Cochain C, Chaudhari SM, Koch M, Wiendl H, Eckstein HH, Zernecke A. Programmed cell death-1 deficiency exacerbates T cell activation and atherogenesis despite expansion of regulatory T cells in atherosclerosis-prone mice. PLoS One 2014; 9:e93280. [PMID: 24691202 PMCID: PMC3972211 DOI: 10.1371/journal.pone.0093280] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/03/2014] [Indexed: 12/14/2022] Open
Abstract
T cell activation represents a double-edged sword in atherogenesis, as it promotes both pro-inflammatory T cell activation and atheroprotective Foxp3+ regulatory T cell (Treg) responses. Here, we investigated the role of the co-inhibitory receptor programmed cell death-1 (PD-1) in T cell activation and CD4+ T cell polarization towards pro-atherogenic or atheroprotective responses in mice. Mice deficient for both low density lipoprotein receptor and PD-1 (Ldlr−/−Pd1−/−) displayed striking increases in systemic CD4+ and CD8+ T cell activation after 9 weeks of high fat diet feeding, associated with an expansion of both pro-atherogenic IFNγ-secreting T helper 1 cells and atheroprotective Foxp3+ Tregs. Importantly, PD-1 deficiency did not affect Treg suppressive function in vitro. Notably, PD-1 deficiency exacerbated atherosclerotic lesion growth and entailed a massive infiltration of T cells in atherosclerotic lesions. In addition, aggravated hypercholesterolemia was observed in Ldlr−/−Pd1−/− mice. In conclusion, we here demonstrate that although disruption of PD-1 signaling enhances both pro- and anti-atherogenic T cell responses in Ldlr−/− mice, pro-inflammatory T cell activation prevails and enhances dyslipidemia, vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Clément Cochain
- Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany
- Department of Vascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sweena M. Chaudhari
- Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany
- Department of Vascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Miriam Koch
- Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Hans-Henning Eckstein
- Department of Vascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Alma Zernecke
- Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany
- Department of Vascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
190
|
Attenuation of intestinal inflammation in interleukin-10-deficient mice infected with Citrobacter rodentium. Infect Immun 2014; 82:1949-58. [PMID: 24566625 DOI: 10.1128/iai.00066-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Interleukin-10 (IL-10) curtails immune responses to microbial infection and autoantigens and contributes to intestinal immune homeostasis, yet administration of IL-10 has not been effective at attenuating chronic intestinal inflammatory conditions, suggesting that its immune functions may be context dependent. To gain a broader understanding of the importance of IL-10 in controlling mucosal immune responses to infectious challenges, we employed the murine attaching and effacing pathogen Citrobacter rodentium, which colonizes primarily the surfaces of the cecum and colon and causes transient mucosal inflammation driven by Th17 and Th1 T helper cells. Infection induced macrophage and dendritic cell production of IL-10, which diminished antibacterial host defenses, because IL-10-deficient mice cleared infection faster than wild-type controls. In parallel, the mice had less acute infection-associated colitis and resolved it more rapidly than controls. Importantly, transient C. rodentium infection protected IL-10-deficient mice against the later development of spontaneous colitis that normally occurs with aging in these mice. Genome-wide expression studies revealed that IL-10 deficiency was associated with downregulation of proinflammatory pathways but increased expression of the anti-inflammatory cytokine IL-27 in response to infection. IL-27 was found to suppress in vitro Th17 and, to a lesser degree, Th1 differentiation independent of IL-10. Furthermore, neutralization of IL-27 resulted in more severe colitis in infected IL-10-deficient mice. Together, these findings indicate that IL-10 is dispensable for resolving C. rodentium-associated colitis and further suggest that IL-27 may be a critical factor for controlling intestinal inflammation and Th17 and Th1 development by IL-10-independent mechanisms.
Collapse
|
191
|
Morandi F, Di Carlo E, Ferrone S, Petretto A, Pistoia V, Airoldi I. IL-27 in human secondary lymphoid organs attracts myeloid dendritic cells and impairs HLA class I-restricted antigen presentation. THE JOURNAL OF IMMUNOLOGY 2014; 192:2634-42. [PMID: 24554774 DOI: 10.4049/jimmunol.1302656] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Different cytokines play crucial roles in inflammation and in polarizing immune responses, including IL-27 that exerts pro- and anti-inflammatory functions. Although the activity of IL-27 is well characterized in murine immune cells, only limited information is available regarding the natural cellular sources of IL-27 in humans and its effects on human immune cells. Dendritic cells (DCs) are the most potent professional APCs that in the immature state are positioned throughout peripheral tissues by acting as sentinels, sensing the presence of Ags. Activated DCs migrate into the lymph nodes and direct Ag-specific T cell responses, thus acting as key players in both adaptive and innate immunity. In this study we asked whether IL-27 is produced by human secondary lymphoid organs and what is its functional role on human DCs. To our knowledge, we provide the first evidence that 1) in lymph nodes, macrophages are the major source for IL-27; 2) immature and mature human DCs express functional IL-27R; 3) IL-27 exerts immunosuppressive activity by crippling the Ag processing machinery in immature DCs under steady-state conditions and after pulsing with a viral Ag; and 4) IL-27 is chemotactic for human DCs. Our findings highlight novel mechanisms underlying the immunosuppressive activity of IL-27, suggesting that this cytokine may function as a homeostatic cytokine in secondary lymphoid organs by limiting duration and/or intensity of ongoing adaptive immune responses. The results presented in this study pave the way to future studies aimed at investigating whether dysregulation of IL-27 expression and function may be involved in pathogenesis of autoimmune disease and cancer.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratorio di Oncologia, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | | | | | | | | | | |
Collapse
|
192
|
Abstract
Psoriasis is a T helper (Th)17/Th1-mediated autoimmune disease affecting the skin and joints. So far, distinct traditional oral compounds and modern biologics have been approved in most countries for the treatment of patients with moderate to severe psoriasis or psoriatic arthritis. Yet, the anti-psoriatic therapeutic spectrum is to be extended by a number of novel targeted therapies including biologics and modern oral compounds. The next set of anti-psoriatic biologics targets mainly Th17-associated cytokines such as IL-17 or IL-23. In contrast, modern oral anti-psoriatics, such as dimethyl fumarate (DMF), apremilast or Janus kinase (JAK) inhibitors interfere with intracellular proteins and affect signaling pathways. Here we summarize the current systemic therapies for psoriasis and their immunological mechanism. The recent advances in psoriasis therapy will help treat our patients efficiently and complete our understanding of disease pathogenesis.
Collapse
|
193
|
Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu YX. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 2014. [PMID: 24382348 DOI: 10.1172/jci67313ds1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
High-dose ionizing irradiation (IR) results in direct tumor cell death and augments tumor-specific immunity, which enhances tumor control both locally and distantly. Unfortunately, local relapses often occur following IR treatment, indicating that IR-induced responses are inadequate to maintain antitumor immunity. Therapeutic blockade of the T cell negative regulator programmed death-ligand 1 (PD-L1, also called B7-H1) can enhance T cell effector function when PD-L1 is expressed in chronically inflamed tissues and tumors. Here, we demonstrate that PD-L1 was upregulated in the tumor microenvironment after IR. Administration of anti-PD-L1 enhanced the efficacy of IR through a cytotoxic T cell-dependent mechanism. Concomitant with IR-mediated tumor regression, we observed that IR and anti-PD-L1 synergistically reduced the local accumulation of tumor-infiltrating myeloid-derived suppressor cells (MDSCs), which suppress T cells and alter the tumor immune microenvironment. Furthermore, activation of cytotoxic T cells with combination therapy mediated the reduction of MDSCs in tumors through the cytotoxic actions of TNF. Our data provide evidence for a close interaction between IR, T cells, and the PD-L1/PD-1 axis and establish a basis for the rational design of combination therapy with immune modulators and radiotherapy.
Collapse
|
194
|
Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu YX. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 2014; 124:687-95. [PMID: 24382348 DOI: 10.1172/jci67313] [Citation(s) in RCA: 1635] [Impact Index Per Article: 148.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022] Open
Abstract
High-dose ionizing irradiation (IR) results in direct tumor cell death and augments tumor-specific immunity, which enhances tumor control both locally and distantly. Unfortunately, local relapses often occur following IR treatment, indicating that IR-induced responses are inadequate to maintain antitumor immunity. Therapeutic blockade of the T cell negative regulator programmed death-ligand 1 (PD-L1, also called B7-H1) can enhance T cell effector function when PD-L1 is expressed in chronically inflamed tissues and tumors. Here, we demonstrate that PD-L1 was upregulated in the tumor microenvironment after IR. Administration of anti-PD-L1 enhanced the efficacy of IR through a cytotoxic T cell-dependent mechanism. Concomitant with IR-mediated tumor regression, we observed that IR and anti-PD-L1 synergistically reduced the local accumulation of tumor-infiltrating myeloid-derived suppressor cells (MDSCs), which suppress T cells and alter the tumor immune microenvironment. Furthermore, activation of cytotoxic T cells with combination therapy mediated the reduction of MDSCs in tumors through the cytotoxic actions of TNF. Our data provide evidence for a close interaction between IR, T cells, and the PD-L1/PD-1 axis and establish a basis for the rational design of combination therapy with immune modulators and radiotherapy.
Collapse
|
195
|
Affiliation(s)
- Benjamin C. Creelan
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
196
|
Li M, Jiang J, Fu B, Chen J, Xue Q, Dong W, Gu Y, Tang L, Xue L, Fang Q, Wang M, Zhang X. PD-L1 is increased in the spinal cord and infiltrating lymphocytes in experimental allergic encephalomyelitis. Neural Regen Res 2013; 8:3296-305. [PMID: 25206651 PMCID: PMC4145942 DOI: 10.3969/j.issn.1673-5374.2013.35.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/06/2013] [Indexed: 11/18/2022] Open
Abstract
Experimental allergic encephalomyelitis is a mouse model of human multiple sclerosis with similar pathology and pathogenesis. Th1 cells play an important role in the pathogenesis of experimental allergic encephalomyelitis. This study determined the potential effect of programmed cell death 1 ligand 1 in the pathogenesis of experimental allergic encephalomyelitis induced by injecting myelin oligodendrocyte glycoprotein, complete Freund's adjuvant and Bordetella pertussis toxin into C57BL/6J mice. Experimental allergic encephalomyelitis mice developed disease and showed inflammatory changes in the central nervous system by hematoxylin-eosin staining of spinal cord pathological sections, demyelination by Luxol fast-blue staining and clinical manifestations. The expression of programmed cell death 1 ligand 1 in mice was detected by immunohistochemistry, flow cytometry and western blot analysis. The expression of programmed cell death 1 ligand 1 in the spinal cord and splenocytes of mice was significantly increased compared with normal mice. Our findings suggest the involvement of programmed cell death 1 ligand 1 in the pathogenesis of experimental allergic encephalomyelitis and suggest this should be studied in multiple sclerosis.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, Second People's Hospital of Lianyungang City, Lianyungang 222006, Jiangsu Province, China
| | - Jiandong Jiang
- Department of Neurology, Second People's Hospital of Lianyungang City, Lianyungang 222006, Jiangsu Province, China
| | - Bing Fu
- Department of Neurology, Second People's Hospital of Lianyungang City, Lianyungang 222006, Jiangsu Province, China
| | - Jiechun Chen
- Department of Neurology, Second People's Hospital of Lianyungang City, Lianyungang 222006, Jiangsu Province, China
| | - Qun Xue
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China ; Institute of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Wanli Dong
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China ; Institute of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yanzheng Gu
- Institute of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Lingtao Tang
- Department of Neurology, Third Hospital of Xingtai City, Xingtai 054000, Hebei Province, China
| | - Limin Xue
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qi Fang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Mingyuan Wang
- Red-Cross Blood Center of Suzhou City, Suzhou 215006, Jiangsu Province, China
| | - Xueguang Zhang
- Institute of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
197
|
Kochetkova I, Thornburg T, Callis G, Holderness K, Maddaloni M, Pascual DW. Oral Escherichia coli colonization factor antigen I fimbriae ameliorate arthritis via IL-35, not IL-27. THE JOURNAL OF IMMUNOLOGY 2013; 192:804-16. [PMID: 24337375 DOI: 10.4049/jimmunol.1302018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A Salmonella therapeutic expressing enterotoxigenic Escherichia coli colonization factor Ag I (CFA/I) fimbriae protects against collagen-induced arthritis (CIA) by eliciting two regulatory T cell (Treg) subsets: TGF-β-producing Foxp3(-)CD39(+)CD4(+) T cells and IL-10-producing Foxp3(+)CD39(+)CD4(+) T cells. However, it is unclear whether CFA/I fimbriae alone are protective and whether other regulatory cytokines are involved, especially in the context for the EBI3-sharing cytokines, Treg-derived IL-35 and APC-derived IL-27, both capable of suppressing Th17 cells and regulating autoimmune diseases. Subsequent evaluation revealed that a single oral dose of purified, soluble CFA/I fimbriae protected against CIA as effectively as did Salmonella-CFA/I and found that Foxp3(+)CD39(+)CD4(+) T cells were the source of secreted IL-35, whereas IL-27 production by CD11c(+) cells was inhibited. Inquiring into their relevance, CFA/I fimbriae-treated IL-27R-deficient (WSX-1(-/-)) mice were equally protected against CIA as were wild-type mice, suggesting a limited role for IL-27. In contrast, CFA/I fimbriae-mediated protection was abated in EBI3(-/-) mice, accompanied by the loss of TGF-β- and IL-10-producing Tregs. Adoptive transfer of C57BL/6 CD39(+)CD4(+) T cells to EBI3(-/-) mice with concurrent CFA/I plus IL-35 treatment effectively stimulated Tregs suppressing proinflammatory collagen II-specific Th cells. In contrast, recipients cotransferred with C57BL/6 and EBI3(-/-) CD39(+)CD4(+) T cells and treated with CFA/I plus IL-35 were not protected, implicating the importance of endogenous IL-35 for conferring CFA/I-mediated protection. Thus, CFA/I fimbriae stimulate IL-35 required for the coinduction of TGF-β and IL-10.
Collapse
Affiliation(s)
- Irina Kochetkova
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59717
| | | | | | | | | | | |
Collapse
|
198
|
Interleukin-17: a promoter in colorectal cancer progression. Clin Dev Immunol 2013; 2013:436307. [PMID: 24382972 PMCID: PMC3870650 DOI: 10.1155/2013/436307] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/23/2013] [Accepted: 10/30/2013] [Indexed: 12/13/2022]
Abstract
It is widely accepted that chronic inflammation plays an active role in cancer. Inflammatory immunocytes and related cytokines in the tumor microenvironment are supposed to be a “double-edged sword” in colorectal cancer (CRC) initiation and progression. Interleukin-17 (IL-17), a pleiotropic proinflammatory cytokine, can promote cancer-elicited inflammation and prevent cancer cells from immune surveillance. Despite controversy, IL-17 is generally considered to be a promoter in CRC progression. In this review, we devote to summarize the current progress regarding the role of IL-17 in tumor initiation and progression, as well as the prognostic value in CRC.
Collapse
|
199
|
Yang H, Lee SM, Gao B, Zhang J, Fang D. Histone deacetylase sirtuin 1 deacetylates IRF1 protein and programs dendritic cells to control Th17 protein differentiation during autoimmune inflammation. J Biol Chem 2013; 288:37256-66. [PMID: 24214980 DOI: 10.1074/jbc.m113.527531] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The type III histone deacetylase Sirt1 has recently emerged as a critical immune regulator by suppressing T cell immunity and macrophage activation during inflammation, but its role in dendritic cells (DCs) remains unknown. Here, we show that mice with genetic Sirt1 deletion specifically in DCs are resistant to MOG-induced experimental autoimmune encephalomyelitis. Loss of Sirt1 functions in DCs enhances their ability to produce IL-27 and interferon β (IFN-β). Co-cultivation of Sirt1-null DCs with CD4(+) T cells inhibited Th17 differentiation, which is reversed by anti-IL27 and anti-IFN-β neutralization antibodies. Sirt1 antagonizes acetylation of IRF1, a transcription factor that drives IL-27 production. Genetic deletion of IRF1 in Sirt1-null DCs abolishes IL-27 production and suppresses Th17 differentiation. Our results show that the histone deacetylase Sirt1 programs DCs to regulate Th17 differentiation during inflammation.
Collapse
Affiliation(s)
- Heeyoung Yang
- From the Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611 and
| | | | | | | | | |
Collapse
|
200
|
Astry B, Venkatesha SH, Moudgil KD. Temporal cytokine expression and the target organ attributes unravel novel aspects of autoimmune arthritis. Indian J Med Res 2013; 138:717-31. [PMID: 24434324 PMCID: PMC3928702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Susceptibility to autoimmunity is determined by multiple factors. Defining the contribution of the quantitative versus qualitative aspects of antigen-directed immune responses as well as the factors influencing target organ susceptibility is vital to advancing the understanding of the pathogenesis of autoimmunity. In a series of studies, we have addressed these issues using the adjuvant-induced arthritis (AA) model of human rheumatoid arthritis (RA). Lewis rats are susceptible to AA following immunization with heat-killed Mycobacterium tuberculosis H37Ra, whereas Wistar-Kyoto (WKY) rats of the same MHC (major histocompatibility complex) haplotype are resistant. Comparative studies on these and other susceptible/resistant rodent strains have offered interesting insights into differential cytokine responses in the face of comparable T cell proliferative response to the disease relevant antigens. Study of the cytokine kinetics have also permitted validation of the disease-protective versus disease-aggravating effects of specific cytokines by treatment of rats/mice with those cytokines at different phases of the disease. In regard to the target organ attributes, the migration of arthritogenic leukocytes into the joints; the expression of mediators of inflammation, angiogenesis, and tissue damage; the role of vascular permeability; and the characteristics of vascular endothelial cells have been examined. Further, various inhibitors of angiogenesis are effective in suppressing arthritis. Taken together, the differential cytokine responses and unique attributes of the target organ have revealed novel aspects of disease susceptibility and joint damage in AA. The translation of this basic research in animal models to RA patients would not only advance our understanding of the disease process, but also offer novel avenues for immunomodulation of this disease.
Collapse
Affiliation(s)
- Brian Astry
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shivaprasad H. Venkatesha
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kamal D. Moudgil
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA,Reprint requests: Dr Kamal D. Moudgil, Professor, Department of Microbiology & Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA e-mail:
| |
Collapse
|