51
|
Jeffery HC, Braitch MK, Bagnall C, Hodson J, Jeffery LE, Wawman RE, Wong LL, Birtwistle J, Bartlett H, Lohse AW, Hirschfield GM, Dyson J, Jones D, Hubscher SG, Klenerman P, Adams DH, Oo YH. Changes in natural killer cells and exhausted memory regulatory T Cells with corticosteroid therapy in acute autoimmune hepatitis. Hepatol Commun 2018; 2:421-436. [PMID: 29619420 PMCID: PMC5880196 DOI: 10.1002/hep4.1163] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 12/24/2022] Open
Abstract
Autoimmune hepatitis (AIH) is an immune-mediated liver disease currently treated by immunosuppressive medications with significant side effects. Thus, novel mechanistic treatments are greatly needed. We performed prospective deep immunophenotyping of blood immune cells in patients with acute AIH before and after corticosteroid therapy. Blood samples from 26 patients with acute AIH (United Kingdom-AIH Consortium) were phenotyped by flow cytometry at baseline and 4 months after starting corticosteroids. Pretreatment liver tissues were stained for forkhead box P3-positive (FOXP3POS) regulatory T cells (Tregs), clusters of differentiation (CD)56POS natural killer (NK) cells, and chemokine (C-X-C motif) ligand 10. Chemokine secretion by cultured primary hepatocyte and biliary epithelial cells was measured by enzyme-linked immunosorbent assay. Functional coculture assays with stimulated NK cells and Tregs were performed. CD161 ligand, lectin-like transcript-1 expression by intrahepatic immune cells was demonstrated with flow cytometry. Frequencies of NKbright cells declined with therapy (P < 0.001) and correlated with levels of alanine aminotransferase (P = 0.023). The Treg:NKbright ratio was lower pretreatment, and Tregs had an activated memory phenotype with high levels of CD39, cytotoxic T lymphocyte antigen 4, and FOXP3 but also high programmed death ligand 1, indicating exhaustion. Coculture experiments suggested the Tregs could not efficiently suppress interferon-γ secretion by NK cells. Both Tregs and NK cells had high expression of liver infiltration and T helper 17 plasticity-associated marker CD161 (P = 0.04). Pretreatment and CD161pos NK cells expressed high levels of perforin and granzyme B, consistent with an activated effector phenotype (P < 0.05). Lectin-like transcript 1, a ligand for CD161, is expressed on intrahepatic B cells, monocytes, and neutrophils. Conclusion: Activated effector NK cells, which correlate with biochemical measurements of hepatitis, and exhausted memory Tregs are increased in the blood of patients with treatment-naive AIH and decline with corticosteroid therapy. Inadequate regulation of NK cells by exhausted FOXP3pos Tregs may play a role in AIH pathogenesis and contribute to liver injury. (Hepatology Communications 2018;2:421-436).
Collapse
Affiliation(s)
- Hannah C. Jeffery
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | - Manjit K. Braitch
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | - Chris Bagnall
- Human Biomaterials Resource CentreUniversity of BirminghamUnited Kingdom
| | - James Hodson
- Institute of Translational MedicineUniversity Hospitals Birmingham National Health Services Foundation Trust, University of BirminghamBirminghamUnited Kingdom
| | - Louisa E. Jeffery
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | - Rebecca E. Wawman
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- School of Life Sciences, Faculty of Health and Life SciencesCoventry UniversityCoventryUnited Kingdom
| | - Lin Lee Wong
- Newcastle Biomedical Research Centre and Newcastle UniversityNewcastleUnited Kingdom
| | - Jane Birtwistle
- Clinical Immunology DepartmentUniversity of BirminghamBirminghamUnited Kingdom
| | - Helen Bartlett
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
| | | | - Gideon M. Hirschfield
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Liver Transplantation and Hepatobiliary Unit, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| | - Jessica Dyson
- Newcastle Biomedical Research Centre and Newcastle UniversityNewcastleUnited Kingdom
| | - David Jones
- Newcastle Biomedical Research Centre and Newcastle UniversityNewcastleUnited Kingdom
| | - Stefan G. Hubscher
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Department of Histopathology, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| | - Paul Klenerman
- Peter Medawar Building of Pathogen ResearchUniversity of OxfordOxfordUnited Kingdom
| | - David H. Adams
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Liver Transplantation and Hepatobiliary Unit, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| | - Ye H. Oo
- Centre for Liver Research, Institute of Immunology and Immunotherapy and National Institute of Health Research Inflammation Biomedical Research Centre BirminghamUniversity of BirminghamBirminghamUnited Kingdom
- Liver Transplantation and Hepatobiliary Unit, Queen Elizabeth HospitalUniversity Hospitals Birmingham National Health Service Foundation TrustBirminghamUnited Kingdom
| |
Collapse
|
52
|
Gajovic N, Jurisevic M, Pantic J, Radosavljevic G, Arsenijevic N, Lukic ML, Jovanovic I. Attenuation of NK cells facilitates mammary tumor growth in streptozotocin-induced diabetes in mice. Endocr Relat Cancer 2018; 25:493-507. [PMID: 29459428 DOI: 10.1530/erc-17-0529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 02/19/2018] [Indexed: 11/08/2022]
Abstract
Diabetic patients have higher incidence and mortality of cancer. Recent study revealed that hyperglycemia-induced oxidative stress is involved in the acceleration of tumor metastasis. We used model of high-dose streptozotocin-induced diabetes to investigate its effect on tumor growth and modulation of antitumor immune response of 4T1 murine breast cancer in BALB/c mice. Diabetes accelerated tumor appearance, growth and weight, which was associated with decreased NK cells cytotoxicity against 4T1 tumor cells in vitro Diabetes reduced frequencies of systemic NKG2D+, perforin+, granzyme+, IFN-γ+ and IL-17+ NK cells, while increased level of PD-1 expression and production of IL-10 in NK cells. Diabetes decreased percentage of NKG2D+NK cells and increased percentage of PD-1+ NK cells also in primary tumor. Diabetes increased accumulation of IL-10+ Tregs and TGF-β+ myeloid-derived suppressor cells (MDSCs) in spleen and tumor. Diabetic sera in vitro significantly increased the percentage of KLRG-1+ and PD-1+ NK cells, decreased the percentage of IFN-γ+NK cells, expression of NKp46 and production of perforin, granzyme, CD107a and IL-17 per NK cell in comparison to glucose-added mouse sera and control sera. Significantly increased percentages of inducible nitric oxide synthase (iNOS) and indoleamine 2,3-dioxygenase (IDO) producing MDSCs and dendritic cells (DC) were found in the spleens of diabetic mice prior to tumor induction. 1-methyl-DL-tryptophan, specific IDO inhibitor, almost completely restored phenotype of NK cells cultivated in diabetic sera. These findings indicate that diabetes promotes breast cancer growth at least in part through increased accumulation of immunosuppressive cells and IDO-mediated attenuation of NK cells.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Female
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Nitric Oxide Synthase Type II/metabolism
- Spleen/metabolism
Collapse
|
53
|
T reg cells limit IFN-γ production to control macrophage accrual and phenotype during skeletal muscle regeneration. Proc Natl Acad Sci U S A 2018; 115:E2585-E2593. [PMID: 29476012 DOI: 10.1073/pnas.1800618115] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Skeletal muscle regeneration is a highly orchestrated process that depends on multiple immune-system cell types, notably macrophages (MFs) and Foxp3+CD4+ regulatory T (Treg) cells. This study addressed how Treg cells rein in MFs during regeneration of murine muscle after acute injury with cardiotoxin. We first delineated and characterized two subsets of MFs according to their expression of major histocompatibility complex class II (MHCII) molecules, i.e., their ability to present antigens. Then, we assessed the impact of Treg cells on these MF subsets by punctually depleting Foxp3+ cells during the regenerative process. Treg cells controlled both the accumulation and phenotype of the two types of MFs. Their absence after injury promoted IFN-γ production, primarily by NK and effector T cells, which ultimately resulted in MF dysregulation and increased inflammation and fibrosis, pointing to compromised muscle repair. Thus, we uncovered an IFN-γ-centered regulatory layer by which Treg cells keep MFs in check and dampen inflammation during regeneration of skeletal muscle.
Collapse
|
54
|
Ishikawa T, Okayama T, Sakamoto N, Ideno M, Oka K, Enoki T, Mineno J, Yoshida N, Katada K, Kamada K, Uchiyama K, Handa O, Takagi T, Konishi H, Kokura S, Uno K, Naito Y, Itoh Y. Phase I clinical trial of adoptive transfer of expanded natural killer cells in combination with IgG1 antibody in patients with gastric or colorectal cancer. Int J Cancer 2018; 142:2599-2609. [PMID: 29388200 DOI: 10.1002/ijc.31285] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/06/2018] [Accepted: 01/18/2018] [Indexed: 12/16/2022]
Abstract
Natural killer (NK) cells exhibit strong cytotoxic activity against tumor cells without prior sensitization, and have the potential to exert antibody-dependent cellular cytotoxicity (ADCC). In this clinical trial, we examined the safety and efficacy of the use of NK cells, generated using a novel expansion system, in combination with IgG1 antibodies for the treatment of advanced gastric or colorectal cancers. Treatment consisted of trastuzumab- or cetuximab-based chemotherapy, plus adoptive NK cell therapy. For administration of expanded NK cells, dose escalation with a sequential 3 + 3 design was performed in three steps, at doses of 0.5 × 109 , 1.0 × 109 , and 2.0 × 109 cells/injection (N = 9). After 3 days of IgG1 antibody administration, patients were infused with expanded NK cells three times at triweekly intervals. NK cell populations expanded with our system were confirmed as being enriched in NK cells (median 92.9%) with high expression of NKG2D (97.6%) and CD16 (69.6%). The combination therapy was very well tolerated with no severe adverse events. Among six evaluable patients, four presented stable disease (SD) and two presented progressive disease. Of the four SD patients, three showed an overall decrease in tumor size after combination therapy. Immune monitoring suggested that combination therapy enhanced whole blood IFN-γ production and reduced peripheral regulatory T cells (Tregs). In conclusion, this phase I trial provides evidence of good tolerability, induction of Th1 immune responses, and preliminary anti-tumor activity for this combination therapy, in patients with advanced gastric and colorectal cancer that have received previous therapy.
Collapse
Affiliation(s)
- Takeshi Ishikawa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Okayama
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoyuki Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Kaname Oka
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | - Naohisa Yoshida
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Katada
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Kamada
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Handa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohisa Takagi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideyuki Konishi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Kokura
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuko Uno
- Division of Basic Research, Louis Pasteur Center for Medical Research, Kyoto, Japan
| | - Yuji Naito
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
55
|
Duggleby R, Danby RD, Madrigal JA, Saudemont A. Clinical Grade Regulatory CD4 + T Cells (Tregs): Moving Toward Cellular-Based Immunomodulatory Therapies. Front Immunol 2018; 9:252. [PMID: 29487602 PMCID: PMC5816789 DOI: 10.3389/fimmu.2018.00252] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/29/2018] [Indexed: 12/26/2022] Open
Abstract
Regulatory T cells (Tregs) are CD4+ T cells that are key players of immune tolerance. They are powerful suppressor cells, able to impact the function of numerous immune cells, including key effectors of inflammation such as effector T cells. For this reason, Tregs are an ideal candidate for the development of cell therapy approaches to modulate immune responses. Treg therapy has shown promising results so far, providing key knowledge on the conditions in which these cells can provide protection and demonstrating that they could be an alternative to current pharmacological immunosuppressive therapies. However, a more comprehensive understanding of their characteristics, isolation, activation, and expansion is needed to be able design cost effective therapies. Here, we review the practicalities of making Tregs a viable cell therapy, in particular, discussing the challenges faced in isolating and manufacturing Tregs and defining what are the most appropriate applications for this new therapy.
Collapse
Affiliation(s)
- Richard Duggleby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Robert David Danby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom.,Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| |
Collapse
|
56
|
Yuan X, Dong Y, Tsurushita N, Tso JY, Fu W. CD122 blockade restores immunological tolerance in autoimmune type 1 diabetes via multiple mechanisms. JCI Insight 2018; 3:96600. [PMID: 29367461 DOI: 10.1172/jci.insight.96600] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/12/2017] [Indexed: 12/16/2022] Open
Abstract
Signaling through IL-2/IL-15Rβ (CD122) is essential for the differentiation and function of T cells and NK cells. A mAb against CD122 has been implicated to suppress autoimmune type 1 diabetes (T1D) development in animal models. However, the mechanisms remain poorly understood. We find that in vivo administration of an anti-CD122 mAb (CD122 blockade) restores immune tolerance in nonobese diabetic (NOD) mice via multiple mechanisms. First, CD122 blockade selectively ablates pathogenic NK cells and memory phenotype CD8+ T cells from pancreatic islets. In contrast, islet CD4+Foxp3+ Tregs are only mildly affected. Second, CD122 blockade suppresses IFN-γ production in islet immune cells. Third, CD122 blockade inhibits the conversion of islet Th17 cells into diabetogenic Th1 cells. Furthermore, a combination of anti-CD122 mAb and Treg-trophic cytokines (IL-2 or IL-33) enhances the abundance and function of islet Tregs. In summary, these data provide crucial mechanistic insights into CD122 blockade-mediated immunoregulation and support therapeutic benefits of this combinational treatment in T1D.
Collapse
Affiliation(s)
- Xiaomei Yuan
- Pediatric Diabetes Research Center, Department of Pediatrics, UCSD, La Jolla, California, USA
| | - Yi Dong
- Pediatric Diabetes Research Center, Department of Pediatrics, UCSD, La Jolla, California, USA
| | | | - J Yun Tso
- JN Biosciences, Mountain View, California, USA
| | - Wenxian Fu
- Pediatric Diabetes Research Center, Department of Pediatrics, UCSD, La Jolla, California, USA.,Institute for Diabetes and Metabolic Health and.,Moores Cancer Center, UCSD, La Jolla, California, USA
| |
Collapse
|
57
|
Abstract
A group of impressive immunotherapies for cancer treatment, including immune checkpoint-blocking antibodies, gene therapy and immune cell adoptive cellular immunotherapy, have been established, providing new weapons to fight cancer. Natural killer (NK) cells are a component of the first line of defense against tumors and virus infections. Studies have shown dysfunctional NK cells in patients with cancer. Thus, restoring NK cell antitumor functionality could be a promising therapeutic strategy. NK cells that are activated and expanded ex vivo can supplement malfunctional NK cells in tumor patients. Therapeutic antibodies, chimeric antigen receptor (CAR), or bispecific proteins can all retarget NK cells precisely to tumor cells. Therapeutic antibody blockade of the immune checkpoints of NK cells has been suggested to overcome the immunosuppressive signals delivered to NK cells. Oncolytic virotherapy provokes antitumor activity of NK cells by triggering antiviral immune responses. Herein, we review the current immunotherapeutic approaches employed to restore NK cell antitumor functionality for the treatment of cancer.
Collapse
Affiliation(s)
- Yangxi Li
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Rui Sun
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
58
|
Lu J, Zhang C, Li L, Xue W, Zhang C, Zhang X. Unique Features of Pancreatic-Resident Regulatory T Cells in Autoimmune Type 1 Diabetes. Front Immunol 2017; 8:1235. [PMID: 29033948 PMCID: PMC5626883 DOI: 10.3389/fimmu.2017.01235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/19/2017] [Indexed: 12/18/2022] Open
Abstract
Recent progress in regulatory T cells (Tregs) biology emphasizes the importance of understanding tissue-resident Tregs in response to tissue-specific environment. Now, emerging evidence suggests that pancreatic-resident forkhead box P3+ Tregs have distinguishable effects on the suppression of over-exuberant immune responses in autoimmune type 1 diabetes (T1D). Thus, there is growing interest in elucidating the role of pancreatic-resident Tregs that function and evolve in the local environment. In this review, we discuss the phenotype and function of Tregs residing in pancreatic tissues and pancreatic lymph nodes, with emphasis on the unique subpopulations of Tregs that control the disease progression in the context of T1D. Specifically, we discuss known and possible modulators that influence the survival, migration, and maintenance of pancreatic Tregs.
Collapse
Affiliation(s)
- Jingli Lu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaoqi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lifeng Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhua Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chengliang Zhang
- Department of Pharmacy, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
59
|
Mohan JF, Kohler RH, Hill JA, Weissleder R, Mathis D, Benoist C. Imaging the emergence and natural progression of spontaneous autoimmune diabetes. Proc Natl Acad Sci U S A 2017; 114:E7776-E7785. [PMID: 28839093 PMCID: PMC5604023 DOI: 10.1073/pnas.1707381114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes in the nonobese diabetic mouse stems from an infiltration of the pancreatic islets by a mixed population of immunocytes, which results in the impairment and eventual destruction of insulin-producing β-cells. Little is known about the dynamics of lymphocyte movement in the pancreas during disease progression. Using advanced intravital imaging approaches and newly created reporter mice (Flt3-BFP2, Mertk-GFP-DTR, Cd4-tdTomato, Cd8a-tdTomato), we show that the autoimmune process initiates first with a T cell infiltration into the islets, where they have restricted mobility but reside and are activated in apposition to CX3CR1+ macrophages. The main expansion then occurs in the connective tissue outside the islet, which remains more or less intact. CD4+ and CD8+ T cells, Tregs, and dendritic cells (DCs) are highly mobile, going along microvascular tracks, while static macrophages (MF) form a more rigid structure, often encasing the islet cell mass. Transient cell-cell interactions are formed between T cells and both MFs and DCs, but also surprisingly between MFs and DCs themselves, possibly denoting antigen transfer. In later stages, extensive islet destruction coincides with preferential antigen presentation to, and activation of, CD8+ T cells. Throughout the process, Tregs patrol the active compartments, consistent with the notion that they control the activation of many cell types.
Collapse
Affiliation(s)
- James F Mohan
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Jonathan A Hill
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114;
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115;
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
60
|
Wu Y, Tian Z, Wei H. Developmental and Functional Control of Natural Killer Cells by Cytokines. Front Immunol 2017; 8:930. [PMID: 28824650 PMCID: PMC5543290 DOI: 10.3389/fimmu.2017.00930] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cells are effective in combating infections and tumors and as such are tempting for adoptive transfer therapy. However, they are not homogeneous but can be divided into three main subsets, including cytotoxic, tolerant, and regulatory NK cells, with disparate phenotypes and functions in diverse tissues. The development and functions of such NK cells are controlled by various cytokines, such as fms-like tyrosine kinase 3 ligand (FL), kit ligand (KL), interleukin (IL)-3, IL-10, IL-12, IL-18, transforming growth factor-β, and common-γ chain family cytokines, which operate at different stages by regulating distinct signaling pathways. Nevertheless, the specific roles of each cytokine that regulates NK cell development or that shapes different NK cell functions remain unclear. In this review, we attempt to describe the characteristics of each cytokine and the existing protocols to expand NK cells using different combinations of cytokines and feeder cells. A comprehensive understanding of the role of cytokines in NK cell development and function will aid the generation of better efficacy for adoptive NK cell treatment.
Collapse
Affiliation(s)
- Yang Wu
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|
61
|
CD11a/ICAM-1 blockade combined with IL-2 targeting therapy causes a paradoxical acceleration of type 1 diabetes. Immunol Cell Biol 2017; 95:803-813. [PMID: 28611472 DOI: 10.1038/icb.2017.49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 06/06/2017] [Accepted: 06/07/2017] [Indexed: 12/12/2022]
Abstract
Enhancement of regulatory T-cell (Treg) function is the goal of many immunotherapies aimed at treating type 1 diabetes (T1D). The use of interleukin (IL)-2 is hindered by its effects on other populations such as effector T cells and NK cells. Combination therapies aimed at suppressing effector T cells while using IL-2 to expand Tregs could be beneficial and have been trialed in T1D patients. We have investigated a combination therapy using IL-2 and αCD11a blocking antibody to simultaneously expand Tregs and suppress the activation and migration of autoreactive T cells. When non-obese diabetic mice were treated with low-dose IL-2/anti-IL-2 complexes (IL-2c) and αCD11a, significant Treg expansion occurred in both the spleen and pancreas. Activation and IFNγ production by islet-specific T cells was robustly suppressed in the periphery following IL-2c/αCD11a treatment. Surprisingly, combination therapy accelerated diabetes onset compared with control treatments. Analysis of IL-2 responsive populations found that combination therapy increased the activation of CD8+ T cells and natural killer (NK) cells specifically within the pancreas despite concomitant Treg expansion. Blocking effector T-cell migration with the inhibitor FTY720 together with IL-2c treatment also resulted in intra-pancreatic expansion of effector cell populations. Thus, inhibiting effector T-cell migration into the islets unleashes islet-resident pathogenic effectors in the presence of low doses of exogenous IL-2.
Collapse
|
62
|
Trojan K, Zhu L, Aly M, Weimer R, Bulut N, Morath C, Opelz G, Daniel V. Association of peripheral NK cell counts with Helios + IFN-γ - T regs in patients with good long-term renal allograft function. Clin Exp Immunol 2017; 188:467-479. [PMID: 28194759 DOI: 10.1111/cei.12945] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 12/16/2022] Open
Abstract
Little is known about a possible interaction of natural killer (NK) cells with regulatory T cells (Treg ) in long-term stable kidney transplant recipients. Absolute counts of lymphocyte and Treg subsets were studied in whole blood samples of 136 long-term stable renal transplant recipients and 52 healthy controls using eight-colour fluorescence flow cytometry. Patients were 1946 ± 2201 days (153-10 268 days) post-transplant and showed a serum creatinine of 1·7 ± 0·7 mg/dl. Renal transplant recipients investigated > 1·5 years post-transplant showed higher total NK cell counts than recipients studied < 1·5 years after transplantation (P = 0·006). High NK cells were associated with high glomerular filtration rate (P = 0·002) and low serum creatinine (P = 0·005). Interestingly, high NK cells were associated with high CD4+ CD25+ CD127- forkhead box protein 3 (FoxP3+ ) Treg that co-express the phenotype Helios+ interferon (IFN)-γ- and appear to have stable FoxP3 expression and originate from the thymus. Furthermore, high total NK cells were associated with Treg that co-express the phenotypes interleukin (IL)-10- transforming growth factor (TGF)-β+ (P = 0·013), CD183+ CD62L- (P = 0·003), CD183+ CD62+ (P = 0·001), CD183- CD62L+ (P = 0·002), CD252- CD152+ (P < 0·001), CD28+ human leucocyte antigen D-related (HLA-DR- ) (P = 0·002), CD28+ HLA-DR+ (P < 0·001), CD95+ CD178- (P < 0·001) and CD279- CD152+ (P < 0·001), suggesting that these activated Treg home in peripheral tissues and suppress effector cells via TGF-β and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). The higher numbers of NK and Treg cell counts in patients with long-term good allograft function and the statistical association of these two lymphocyte subsets with each other suggest a direct or indirect (via DC) interaction of these cell subpopulations that contributes to good long-term allograft acceptance. Moreover, we speculate that regulatory NK cells are formed late post-transplant that are able to inhibit graft-reactive effector cells.
Collapse
Affiliation(s)
- K Trojan
- Transplantation-Immunology, Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - L Zhu
- Transplantation-Immunology, Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Hematology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - M Aly
- Transplantation-Immunology, Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany.,Nephrology Unit, Internal Medicine Department, Assiut University, Egypt
| | - R Weimer
- Department of Internal Medicine, University of Giessen, Giessen, Germany
| | - N Bulut
- Department of Internal Medicine, University of Giessen, Giessen, Germany
| | - C Morath
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - G Opelz
- Transplantation-Immunology, Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - V Daniel
- Transplantation-Immunology, Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
63
|
Gasteiger G, Ataide M, Kastenmüller W. Lymph node - an organ for T-cell activation and pathogen defense. Immunol Rev 2016; 271:200-20. [PMID: 27088916 DOI: 10.1111/imr.12399] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The immune system is a multicentered organ that is characterized by intimate interactions between its cellular components to efficiently ward off invading pathogens. A key constituent of this organ system is the distinct migratory activity of its cellular elements. The lymph node represents a pivotal meeting point of immune cells where adaptive immunity is induced and regulated. Additionally, besides barrier tissues, the lymph node is a critical organ where invading pathogens need to be eliminated in order to prevent systemic distribution of virulent microbes. Here, we explain how the lymph node is structurally and functionally organized to fulfill these two critical functions - pathogen defense and orchestration of adaptive immunity. We will discuss spatio-temporal aspects of cellular immune responses focusing on CD8 T cells and review how and where these cells are activated in the context of viral infections, as well as how viral antigen expression kinetics and different antigen presentation pathways are involved. Finally, we will describe how such responses are regulated and 'helped', and discuss how this relates to intranodal positioning and cellular migration of the various cellular components that are involved in these processes.
Collapse
Affiliation(s)
- Georg Gasteiger
- Institute of Medical Microbiology and Hygiene & FZI Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Marco Ataide
- Institute of Experimental Immunology, University of Bonn, Bonn, Germany
| | | |
Collapse
|
64
|
Roussey JA, Olszewski MA, Osterholzer JJ. Immunoregulation in Fungal Diseases. Microorganisms 2016; 4:microorganisms4040047. [PMID: 27973396 PMCID: PMC5192530 DOI: 10.3390/microorganisms4040047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 02/07/2023] Open
Abstract
This review addresses specific regulatory mechanisms involved in the host immune response to fungal organisms. We focus on key cells and regulatory pathways involved in these responses, including a brief overview of their broader function preceding a discussion of their specific relevance to fungal disease. Important cell types discussed include dendritic cells and regulatory T cells, with a focus on specific studies relating to their effects on immune responses to fungi. We highlight the interleukin-10, programmed cell death 1, and cytotoxic T lymphocyte-associated protein 4 signaling pathways and emphasize interrelationships between these pathways and the regulatory functions of dendritic cells and regulatory T cells. Throughout our discussion, we identify selected studies best illustrating the role of these cells and pathways in response to specific fungal pathogens to provide a contextual understanding of the tightly-controlled network of regulatory mechanisms critical to determining the outcome of exposure to fungal pathogens. Lastly, we discuss two unique phenomena relating to immunoregulation, protective tolerance and immune reactivation inflammatory syndrome. These two clinically-relevant conditions provide perspective as to the range of immunoregulatory mechanisms active in response to fungi.
Collapse
Affiliation(s)
- Jonathan A Roussey
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA.
- Pulmonary Section, Medical Service, VA Ann Arbor Health System, Ann Arbor, MI 48105, USA.
| | - Michal A Olszewski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA.
- Pulmonary Section, Medical Service, VA Ann Arbor Health System, Ann Arbor, MI 48105, USA.
- Graduate Program in Immunology, University of Michigan Health System, Ann Arbor, MI 48109, USA.
| | - John J Osterholzer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA.
- Pulmonary Section, Medical Service, VA Ann Arbor Health System, Ann Arbor, MI 48105, USA.
- Graduate Program in Immunology, University of Michigan Health System, Ann Arbor, MI 48109, USA.
| |
Collapse
|
65
|
Singular role for T-BET+CXCR3+ regulatory T cells in protection from autoimmune diabetes. Proc Natl Acad Sci U S A 2016; 113:14103-14108. [PMID: 27872297 PMCID: PMC5150376 DOI: 10.1073/pnas.1616710113] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Foxp3+ regulatory T (Treg) cells are crucial for restraining inflammation in a variety of autoimmune diseases, including type 1 diabetes (T1D). However, the transcriptional and functional phenotypes of Treg cells within the pancreatic lesion remain poorly understood. Here we characterized pancreas-infiltrating Treg cells in the NOD mouse model of T1D and uncovered a substantial enrichment of the Treg subpopulation expressing the chemokine receptor CXCR3. Accumulation of CXCR3+ Treg cells within pancreatic islets was dependent on the transcription factor T-BET, and genetic ablation of T-BET increased the onset and penetrance of disease, abrogating the sex bias normally seen in the NOD model. Both male and female mice lacking T-BET+ Treg cells showed a more aggressive insulitic infiltrate, reflected most prominently by elevated production of type 1 cytokines. Our results suggest the possibility of fine therapeutic targeting of Treg cells, in a tissue- and cell-subset-specific fashion, as a more focused immunotherapy for T1D.
Collapse
|
66
|
Abstract
Treatments for autoimmune diseases including type 1 diabetes (T1D) are aimed at resetting the immune system, especially its adaptive arm. The innate immune system is often ignored in the design of novel immune-based therapies. There is increasing evidence for multiple natural killer (NK) subpopulations, but their role is poorly understood in autoimmunity and likely is contributing to the controversial role reported for NKs. In this review, we will summarize NK subsets and their roles in tolerance, autoimmune diabetes, and immunotherapy.
Collapse
Affiliation(s)
- Chris Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
67
|
Mfarrej BG, Battaglia M. The “Unusual Suspects” in Allograft Rejection: Will T Regulatory Cell Therapy Arrest Them? CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0108-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
68
|
Chemoattractant Signals and Adhesion Molecules Promoting Human Regulatory T Cell Recruitment to Porcine Endothelium. Transplantation 2016; 100:753-62. [PMID: 26720299 DOI: 10.1097/tp.0000000000001034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Human CD4+CD25+Foxp3+ T regulatory cells (huTreg) suppress CD4+ T cell-mediated antipig xenogeneic responses in vitro and might therefore be used to induce xenograft tolerance. The present study investigated the role of the adhesion molecules, their porcine ligands, and the chemoattractant factors that may promote the recruitment of huTreg to porcine aortic endothelial cells (PAEC) and their capacity to regulate antiporcine natural killer (NK) cell responses. METHODS Interactions between ex vivo expanded huTreg and PAEC were studied by static chemotaxis assays and flow-based adhesion and transmigration assays. In addition, the suppressive function of huTreg on human antiporcine NK cell responses was analyzed. RESULTS The TNFα-activated PAEC released factors that induce huTreg chemotaxis, partially inhibited by antihuman CXCR3 blocking antibodies. Coating of PAEC with human CCL17 significantly increased the transmigration of CCR4+ huTreg under physiological shear stress. Under static conditions, transendothelial Treg migration was inhibited by blocking integrin sub-units (CD18, CD49d) on huTreg, or their respective porcine ligands intercellular adhesion molecule 2 (CD102) and vascular cell adhesion molecule 1 (CD106). Finally, huTreg partially suppressed xenogeneic human NK cell adhesion, NK cytotoxicity and degranulation (CD107 expression) against PAEC; however, this inhibition was modest, and there was no significant change in the production of IFNγ. CONCLUSIONS Recruitment of huTreg to porcine endothelium depends on particular chemokine receptors (CXCR3, CCR4) and integrins (CD18 and CD49d) and was increased by CCL17 coating. These results will help to develop new strategies to enhance the recruitment of host huTreg to xenogeneic grafts to regulate cell-mediated xenograft rejection including NK cell responses.
Collapse
|
69
|
Ni J, Hölsken O, Miller M, Hammer Q, Luetke-Eversloh M, Romagnani C, Cerwenka A. Adoptively transferred natural killer cells maintain long-term antitumor activity by epigenetic imprinting and CD4 + T cell help. Oncoimmunology 2016; 5:e1219009. [PMID: 27757318 DOI: 10.1080/2162402x.2016.1219009] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cell infusions can induce remissions in subsets of patients with different types of cancer. The optimal strategies for NK cell activation prior to infusion are still under debate. There is recent evidence that NK cells can acquire long-term functional competence by preactivation with the cytokines IL-12/15/18. The mechanisms supporting the maintenance of long-term NK cell antitumor activity are incompletely under-stood. Here, we show that NK cells preactivated in vitro with IL-12/15/18, but not with IL-15 alone, maintained high antitumor activity even 1 mo after transfer into lymphopenic RAG-2-/-γc-/- mice. The NK cell intrinsic ability for IFNγ production coincided with demethylation of the conserved non-coding sequence (CNS) 1 in the Ifng locus, previously shown to enhance transcription of Ifng. In a xenograft melanoma mouse model, human IL-12/15/18-preactivated NK cells rejected tumors more efficiently. In RAG-2-/-γc-/- mice, co-transfer of CD4+ T cells further improved the long-term competence of NK cells for IFNγ production that was dependent on IL-2. CD4+ T cell activation during homeostatic proliferation required macrophages and further promoted the long-term NK cell antitumor activity. Thus, NK cells can "remember" a previous exposure to cytokines by epigenetic imprinting resulting in a remarkable stability of the IFNγ-producing phenotype after adoptive transfer. In addition, our results support combination of cytokine-preactivated NK cells with CD4+ T cell activation upon lymphopenic conditioning to achieve long-term NK cell effector function for cancer immunotherapy.
Collapse
Affiliation(s)
- Jing Ni
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| | - Oliver Hölsken
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| | - Matthias Miller
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| | - Quirin Hammer
- Innate Immunity, Deutsches Rheuma-Forschungszentrum - A Leibniz Institute , Berlin, Germany
| | - Merlin Luetke-Eversloh
- Innate Immunity, Deutsches Rheuma-Forschungszentrum - A Leibniz Institute , Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum - A Leibniz Institute , Berlin, Germany
| | - Adelheid Cerwenka
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| |
Collapse
|
70
|
Khor B. Regulatory T Cells: Central Concepts from Ontogeny to Therapy. Transfus Med Rev 2016; 31:36-44. [PMID: 27523957 DOI: 10.1016/j.tmrv.2016.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/06/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023]
Abstract
The balanced differentiation of naive CD4+ T cells into either pro- or anti-inflammatory fates is a central regulator of immune homeostasis, dysregulation of which can lead to inflammatory disease or cancer. Accordingly, the development of diagnostics and therapeutics to measure and modulate this balance is of great interest. In this review, we focus on the predominant anti-inflammatory subset, regulatory T cells, discussing key concepts including development, function, antigen specificity, and lineage stability. In particular, we highlight how these notions are shaping the evolution of therapeutics, especially in the context of the transfusion medicine specialist, and identify several key areas that urgently need to be addressed.
Collapse
Affiliation(s)
- Bernard Khor
- Department of Pathology, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
71
|
Kadri N, Wagner AK, Ganesan S, Kärre K, Wickström S, Johansson MH, Höglund P. Dynamic Regulation of NK Cell Responsiveness. Curr Top Microbiol Immunol 2016; 395:95-114. [PMID: 26658943 DOI: 10.1007/82_2015_485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Natural killer (NK) cells deliver cytotoxic granules and immunomodulatory cytokines in response to transformed and virally infected cells. NK cell functions are under the control of a large number of germline-encoded receptors that recognize various ligands on target cells, but NK cells also respond to cytokines in the surrounding environment. The interaction between NK cell receptors and their ligands delivers either inhibitory or activating signals. The cytokine milieu further shapes NK cell responses, either directly or by influencing the way inhibitory or activating signals are perceived by NK cells. In this review, we discuss how NK cell function is controlled by inhibitory receptors and MHC-I molecules, how activating receptors contribute to NK cell education, and finally, how cytokines secreted by the surrounding cells affect NK cell responsiveness. Inputs at these three levels involve different cell types and are seamlessly integrated to form a functional NK cell population.
Collapse
Affiliation(s)
- Nadir Kadri
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Arnika Kathleen Wagner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Sridharan Ganesan
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Klas Kärre
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Stina Wickström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Maria H Johansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Petter Höglund
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 141 86, Stockholm, Sweden.
| |
Collapse
|
72
|
Pallmer K, Oxenius A. Recognition and Regulation of T Cells by NK Cells. Front Immunol 2016; 7:251. [PMID: 27446081 PMCID: PMC4919350 DOI: 10.3389/fimmu.2016.00251] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/13/2016] [Indexed: 12/22/2022] Open
Abstract
Regulation of T cell responses by innate lymphoid cells (ILCs) is increasingly documented and studied. Direct or indirect crosstalk between ILCs and T cells early during and after T cell activation can affect their differentiation, polarization, and survival. Natural killer (NK) cells that belong to the ILC1 group were initially described for their function in recognizing and eliminating "altered self" and as source of early inflammatory cytokines, most notably type II interferon. Using signals conveyed by various germ-line encoded activating and inhibitory receptors, NK cells are geared to sense sudden cellular changes that can be caused by infection events, malignant transformation, or cellular stress responses. T cells, when activated by TCR engagement (signal 1), costimulation (signal 2), and cytokines (signal 3), commit to a number of cellular alterations, including entry into rapid cell cycling, metabolic changes, and acquisition of effector functions. These abrupt changes may alert NK cells, and T cells might thereby expose themselves as NK cell targets. Here, we review how activated T cells can be recognized and regulated by NK cells and what consequences such regulation bears for T cell immunity in the context of vaccination, infection, or autoimmunity. Conversely, we will discuss mechanisms by which activated T cells protect themselves against NK cell attack and outline the significance of this safeguard mechanism.
Collapse
Affiliation(s)
| | - Annette Oxenius
- Institute of Microbiology, ETH Zürich , Zürich , Switzerland
| |
Collapse
|
73
|
Abstract
Dysregulation of the immune system contributes to the breakdown of immune regulation, leading to autoimmune diseases, such as type 1 diabetes (T1D). Current therapies for T1D include daily insulin, due to pancreatic β-cell destruction to maintain blood glucose levels, suppressive immunotherapy to decrease the symptoms associated with autoimmunity, and islet transplantation. Genetic risks for T1D have been linked to IL-2 and IL-2R signaling pathways that lead to the breakdown of self-tolerance mechanisms, primarily through altered regulatory T cell (Treg) function and homeostasis. In attempt to correct such deficits, therapeutic administration of IL-2 at low doses has gained attention due to the capacity to boost Tregs without the unwanted stimulation of effector T cells. Preclinical and clinical studies utilizing low-dose IL-2 have shown promising results to expand Tregs due to their high selective sensitivity to respond to IL-2. These results suggest that low-dose IL-2 therapy represents a new class of immunotherapy for T1D by promoting immune regulation rather than broadly suppressing unwanted and beneficial immune responses.
Collapse
Affiliation(s)
- Connor J Dwyer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA
| | - Natasha C Ward
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA
| | - Alberto Pugliese
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA.
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, 33101, USA.
| |
Collapse
|
74
|
Barsoumian HB, Yolcu ES, Shirwan H. 4-1BB Signaling in Conventional T Cells Drives IL-2 Production That Overcomes CD4+CD25+FoxP3+ T Regulatory Cell Suppression. PLoS One 2016; 11:e0153088. [PMID: 27049955 PMCID: PMC4822835 DOI: 10.1371/journal.pone.0153088] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/23/2016] [Indexed: 12/22/2022] Open
Abstract
Costimulation with the recombinant SA-4-1BBL agonist of 4-1BB receptor on conventional CD4+ T cells (Tconvs) overcomes the suppression mediated by naturally occurring CD4+CD25+FoxP3+ T regulatory cells (Tregs). The mechanistic basis of this observation has remained largely unknown. Herein we show that Tconvs, but not Tregs, are the direct target of SA-4-1BBL-mediated evasion of Treg suppression. IL-2 produced by Tconvs in response to 4-1BB signaling is both necessary and sufficient for overcoming Treg suppression. Supernatant from Tconvs stimulated with SA-4-1BBL contains high levels of IL-2 and overcomes Treg suppression in ex vivo Tconv:Treg cocultures. Removal of IL-2 from such supernatant restores Treg suppression and repletion of Tconv:Treg cocultures with exogenous recombinant IL-2 overcomes suppression. This study establishes 4-1BB signaling as a key circuit that regulates physical and functional equilibrium between Tregs and Tconvs with important implications for immunotherapy for indications where a fine balance between Tregs and Teffs plays a decisive role.
Collapse
Affiliation(s)
- Hampartsoum B. Barsoumian
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, 40202, United States of America
| | - Esma S. Yolcu
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, 40202, United States of America
- * E-mail: (HS); (EY)
| | - Haval Shirwan
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, 40202, United States of America
- * E-mail: (HS); (EY)
| |
Collapse
|
75
|
Pietra G, Vitale C, Pende D, Bertaina A, Moretta F, Falco M, Vacca P, Montaldo E, Cantoni C, Mingari MC, Moretta A, Locatelli F, Moretta L. Human natural killer cells: news in the therapy of solid tumors and high-risk leukemias. Cancer Immunol Immunother 2016; 65:465-76. [PMID: 26289090 PMCID: PMC11028670 DOI: 10.1007/s00262-015-1744-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/18/2015] [Indexed: 12/28/2022]
Abstract
It is well established that natural killer (NK) cells play an important role in the immunity against cancer, while the involvement of other recently identified, NK-related innate lymphoid cells is still poorly defined. In the haploidentical hematopoietic stem cell transplantation for the therapy of high-risk leukemias, NK cells have been shown to exert a key role in killing leukemic blasts residual after conditioning. While the clinical results in the cure of leukemias are excellent, the exploitation of NK cells in the therapy of solid tumors is still limited and unsatisfactory. In solid tumors, NK cell function may be inhibited via different mechanisms, occurring primarily at the tumor site. The cellular interactions in the tumor microenvironment involve tumor cells, stromal cells and resident or recruited leukocytes and may favor tumor evasion from the host's defenses. In this context, a number of cytokines, growth factors and enzymes synthesized by tumor cells, stromal cells, suppressive/regulatory myeloid and lymphoid cells may substantially impair the function of different tumor-reactive effector cells, including NK cells. The identification and characterization of such mechanisms may offer clues for the development of new immunotherapeutic strategies to restore effective anti-tumor responses. In order to harness NK cell-based immunotherapies, several approaches have been proposed, including reinforcement of NK cell cytotoxicity by means of specific cytokines, antibodies or drugs. These new tools may improve NK cell function and/or increase tumor susceptibility to NK-mediated killing. Hence, the integration of NK-based immunotherapies with conventional anti-tumor therapies may increase chances of successful cancer treatment.
Collapse
Affiliation(s)
- Gabriella Pietra
- IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Chiara Vitale
- IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | | | | | - Francesca Moretta
- Department of Medicine, University of Verona, Verona, Italy
- Ospedale Sacro Cuore, Negrar, Verona, Italy
| | - Michela Falco
- Istituto Giannina Gaslini, Via G. Gaslini n. 5, 16147, Genoa, Italy
| | - Paola Vacca
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Elisa Montaldo
- Istituto Giannina Gaslini, Via G. Gaslini n. 5, 16147, Genoa, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Istituto Giannina Gaslini, Via G. Gaslini n. 5, 16147, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | - Maria Cristina Mingari
- IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | - Alessandro Moretta
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | | | | |
Collapse
|
76
|
Abstract
Innate lymphoid cells (ILCs) are innate immune cells that provide an early source of cytokines to initiate and tailor the immune response to the type of the encountered pathogen or insult. The group 1 ILCs are comprised of conventional natural killer (cNK) cells and subsets of "unconventional NK cells," termed ILC1s. Although cNK cells and ILC1s share many features, such as certain phenotypic markers and the ability to produce IFN-γ upon activation, it is now becoming apparent that these two subsets develop from different progenitors and show unique tissue distribution and functional characteristics. Recent studies have aimed at elucidating the individual contributions of cNK cells and ILC1s during protective host responses as well as during chronic inflammation. This review provides an overview of the current knowledge of the developmental origins as well as of the phenotypic and functional characteristics of ILC1s.
Collapse
Affiliation(s)
- Anja Fuchs
- Department of Surgery, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
77
|
Pedroza-Pacheco I, Shah D, Domogala A, Luevano M, Blundell M, Jackson N, Thrasher A, Madrigal A, Saudemont A. Regulatory T cells inhibit CD34+ cell differentiation into NK cells by blocking their proliferation. Sci Rep 2016; 6:22097. [PMID: 26915707 PMCID: PMC4768165 DOI: 10.1038/srep22097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/08/2016] [Indexed: 02/05/2023] Open
Abstract
Graft versus Host Disease (GvHD) remains one of the main complications after hematopoietic stem cell transplantation (HSCT). Due to their ability to suppress effector cells, regulatory T cells (Tregs) have been proposed as a cellular therapy to prevent GvHD, however they also inhibit the functions of natural killer (NK) cells, key effectors of the Graft versus Leukemia effect. In this study, we have explored whether a Tregs therapy will also impact on NK cell differentiation. Using an in vitro model of hematopoietic stem cell (HSC) differentiation into NK cells, we found that activated Tregs led to a 90% reduction in NK cell numbers when added at the time of commitment to the NK cell lineage. This effect was contact dependent and was reversible upon Tregs depletion. The few NK cells that developed in these cultures were mature and exhibited normal functions. Furthermore, adoptive transfer of activated Tregs in rag(-/-) γc(-/-) mice abrogated HSC differentiation into NK cells thus confirming our in vitro findings. Collectively, these results demonstrate for the first time that activated Tregs can inhibit NK cell differentiation from HSC under specific conditions.
Collapse
Affiliation(s)
- Isabela Pedroza-Pacheco
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Divya Shah
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Anna Domogala
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Martha Luevano
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Michael Blundell
- Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Nicola Jackson
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Adrian Thrasher
- Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Alejandro Madrigal
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Aurore Saudemont
- Anthony Nolan Research Institute and University College London, Royal Free Campus, Pond Street, London NW3 2QG, UK
| |
Collapse
|
78
|
Pelletier AN, Guilbault L, Guimont-Desrochers F, Hillhouse EE, Lesage S. NK Cell Proportion and Number Are Influenced by Genetic Loci on Chromosomes 8, 9, and 17. THE JOURNAL OF IMMUNOLOGY 2016; 196:2627-36. [DOI: 10.4049/jimmunol.1502284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/06/2016] [Indexed: 11/19/2022]
|
79
|
Reduced interleukin-2 responsiveness impairs the ability of Treg cells to compete for IL-2 in nonobese diabetic mice. Immunol Cell Biol 2016; 94:509-19. [PMID: 26763864 DOI: 10.1038/icb.2016.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/06/2016] [Accepted: 01/10/2015] [Indexed: 12/11/2022]
Abstract
Enhancement of regulatory T cell (Treg cell) frequency and function is the goal of many therapeutic strategies aimed at treating type 1 diabetes (T1D). The interleukin-2 (IL-2) pathway, which has been strongly implicated in T1D susceptibility in both humans and mice, is a master regulator of Treg cell homeostasis and function. We investigated how IL-2 pathway defects impact Treg cells in T1D-susceptible nonobese diabetic (NOD) mice in comparison with protected C57BL/6 and NOD congenic mice. NOD Treg cells were reduced in frequency specifically in the lymph nodes and expressed lower levels of CD25 and CD39/CD73 immunosuppressive molecules. In the spleen and blood, Treg cell frequency was preserved through expansion of CD25(low), effector phenotype Treg cells. Reduced CD25 expression led to decreased IL-2 signaling in NOD Treg cells. In vivo, treatment with IL-2-anti-IL-2 antibody complexes led to effective upregulation of suppressive molecules on NOD Treg cells in the spleen and blood, but had reduced efficacy on lymph node Treg cells. In contrast, NOD CD8(+) and CD4(+) effector T cells were not impaired in their response to IL-2 therapy. We conclude that NOD Treg cells have an impaired responsiveness to IL-2 that reduces their ability to compete for a limited supply of IL-2.
Collapse
|
80
|
Wiesner DL, Smith KD, Kotov DI, Nielsen JN, Bohjanen PR, Nielsen K. Regulatory T Cell Induction and Retention in the Lungs Drives Suppression of Detrimental Type 2 Th Cells During Pulmonary Cryptococcal Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:365-74. [PMID: 26590316 PMCID: PMC4685009 DOI: 10.4049/jimmunol.1501871] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022]
Abstract
Lethal disease caused by the fungus Cryptococcus neoformans is a consequence of the combined failure to control pulmonary fungal replication and immunopathology caused by induced type 2 Th2 cell responses in animal models. In order to gain insights into immune regulatory networks, we examined the role of regulatory T (Treg) cells in suppression of Th2 cells using a mouse model of experimental cryptococcosis. Upon pulmonary infection with Cryptococcus, Treg cells accumulated in the lung parenchyma independently of priming in the draining lymph node. Using peptide-MHC class II molecules to identify Cryptococcus-specific Treg cells combined with genetic fate-mapping, we noted that a majority of the Treg cells found in the lungs were induced during the infection. Additionally, we found that Treg cells used the transcription factor, IFN regulatory factor 4, to dampen harmful Th2 cell responses, as well as mediate chemokine retention of Treg cells in the lungs. Taken together, induction and IFN regulatory factor 4-dependent localization of Treg cells in the lungs allow Treg cells to suppress the deleterious effects of Th2 cells during cryptococcal infection.
Collapse
Affiliation(s)
- Darin L Wiesner
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Kyle D Smith
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Dmitri I Kotov
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455; Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Judith N Nielsen
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599; and
| | - Paul R Bohjanen
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455; Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Center for Infectious Diseases and Translational Research, University of Minnesota, Minneapolis, MN 55455
| | - Kirsten Nielsen
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455; Center for Infectious Diseases and Translational Research, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
81
|
Pérol L, Piaggio E. New Molecular and Cellular Mechanisms of Tolerance: Tolerogenic Actions of IL-2. Methods Mol Biol 2016; 1371:11-28. [PMID: 26530792 DOI: 10.1007/978-1-4939-3139-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Interleukin-2 (IL-2) is an old molecule with brand new functions. Indeed, IL-2 has been first described as a T-cell growth factor but recent data pointed out that its main function in vivo is the maintenance of immune tolerance. Mechanistically, IL-2 is essential for the development and function of CD4(+) Foxp3(+) regulatory T cells (Treg cells) that are essential players in the control of immune responded to self, tumors, microbes and grafts. Treg cells are exquisitely sensitive to IL-2 due to their constitutive expression of the high affinity IL-2 receptor (IL-2R) and the new paradigm suggests that low-doses of IL-2 could selectively boost Treg cells in vivo. Consequently, a growing body of clinical research is aiming at using IL-2 at low doses as a tolerogenic drug to boost endogenous Treg cells in patients suffering from autoimmune or inflammatory conditions. In this manuscript, we briefly review IL-2/IL-2R biology and the role of IL-2 in the development, maintenance, and function of Treg cells; and also its effects on other immune cell populations such as CD4(+) T helper cells and CD8(+) memory T cells. Then, focusing on type 1 diabetes, we review the preclinical studies and clinical trials supporting the use of low-doses IL-2 as a tolerogenic immunotherapy. Finally, we discuss the limitations and future directions for IL-2 based immunotherapy.
Collapse
Affiliation(s)
- Louis Pérol
- INSERM U932, 26 rue d'Ulm, 75005, Paris, France.
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005, Paris, France.
| | - Eliane Piaggio
- INSERM U932, 26 rue d'Ulm, 75005, Paris, France
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005, Paris, France
| |
Collapse
|
82
|
Spence A, Klementowicz JE, Bluestone JA, Tang Q. Targeting Treg signaling for the treatment of autoimmune diseases. Curr Opin Immunol 2015; 37:11-20. [PMID: 26432763 PMCID: PMC4679451 DOI: 10.1016/j.coi.2015.09.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/30/2015] [Accepted: 09/10/2015] [Indexed: 12/16/2022]
Abstract
Regulatory T (Treg) cells are crucial players in the prevention of autoimmunity. Treg lineage commitment and functional stability are influenced by selected extracellular signals from the local environment, shaped by distinctive intracellular signaling network, and secured by their unique epigenetic profile. Recent advances in our understanding of the complex processes of Treg lineage differentiation, maintenance, and function has paved the way for developing strategies to manipulate these important cells for therapeutic benefit in many diseases. In this review, we will summarize recent advances in our understanding of Treg biology as well as Treg-targeted therapies in the context of autoimmune disease.
Collapse
Affiliation(s)
- Allyson Spence
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joanna E Klementowicz
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey A Bluestone
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
83
|
Zhang M, Tang Q. Manipulating IL-2 and IL-2R in autoimmune diseases and transplantation. Immunotherapy 2015; 7:1231-4. [PMID: 26601714 DOI: 10.2217/imt.15.94] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mei Zhang
- Department of Endocrinology & Metabolism, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, 210029, Jiangsu Province, China
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, 513 Parnassus Ave, HSE520, Box 0780, San Francisco CA 94143, USA
| |
Collapse
|
84
|
Arima N, Nakamura F, Yabe T, Tanaka J, Fuji S, Ohashi K, Fukuda T, Miyamura K, Iwato K, Eto T, Mori T, Kobayashi N, Hoshino T, Kato C, Kanamori H, Nakamae H, Atsuta Y, Morishima Y, Kanda Y. Influence of Differently Licensed KIR2DL1-Positive Natural Killer Cells in Transplant Recipients with Acute Leukemia: A Japanese National Registry Study. Biol Blood Marrow Transplant 2015; 22:423-31. [PMID: 26456260 DOI: 10.1016/j.bbmt.2015.09.029] [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: 07/26/2015] [Accepted: 09/28/2015] [Indexed: 10/22/2022]
Abstract
Licensing by self MHC class I ligands is required for proper natural killer (NK) cell response. NK cells with inhibitory killer cell immunoglobulin-like receptors for nonself MHC exhibit transient alloreactivity after hematopoietic stem cell transplantation (HSCT). We analyzed 3866 recipients in the Japan national registry who underwent their first allogeneic HSCT for acute myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL) from HLA-A, -B, and -DRB1 allele-genomatched unrelated donors. By classifying them into 5 independent groups based on HLA-C group matching and assumed donor NK cell status, we found that for HLA-C-matched HSCT for AML in HLA-C1/C1 recipients, in whom transient alloreactivity against HLA-C2-negative leukemic cells was expected, the relapse rate was significantly lower than it was in HLA-C-matched HSCT for AML in HLA-C1/C2 recipients (hazard ratio [HR], .72; P = .011). This difference was not observed in HLA-C-matched HSCT for ALL. Compared with HLA-C-matched HSCT, significantly higher mortality was observed in HLA-C1/C1 AML patients who received transplants from HLA-C-mismatched HLA-C1/C1 donors (HR, 1.37; P = .001) and in HLA-C1/C1 ALL patients who received transplants from HLA-C2-positive donors (HR, 2.13; P = .005). In conclusion, donor selection based on leukemic subtype and donor HLA-C group matching improves transplantation outcome after HLA-C-mismatched HSCT.
Collapse
Affiliation(s)
- Nobuyoshi Arima
- Department of Hematology, Medical Research Institute Kitano Hospital, Osaka, Japan.
| | - Fumiaki Nakamura
- Department of Public Health/Health Policy, The University of Tokyo, Tokyo, Japan
| | - Toshio Yabe
- Hematology Division, Japanese Red Cross Tokyo Metropolitan Blood Center, Tokyo, Japan
| | - Junji Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Shigeo Fuji
- Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuteru Ohashi
- Hematology Division, Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Takahiro Fukuda
- Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Koichi Miyamura
- Department of Hematology, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Koji Iwato
- Department of Hematology, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Takehiko Mori
- Department of Hematology, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Kobayashi
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Takumi Hoshino
- Leukemia Research Center, Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Chiaki Kato
- Department of Hematology, Meitetsu hospital, Nagoya, Japan
| | - Heiwa Kanamori
- Department of Hematology, Kanagawa Cancer Center, Yokohama, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka City University Hospital, Osaka, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation/Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuo Morishima
- Department of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
85
|
Ovcinnikovs V, Walker LSK. Regulatory T Cells in Autoimmune Diabetes: Mechanisms of Action and Translational Potential. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:245-77. [PMID: 26615100 DOI: 10.1016/bs.pmbts.2015.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Since the discovery of specialized T cells with regulatory function, harnessing the power of these cells to ameliorate autoimmunity has been a major goal. Here we collate the evidence that regulatory T cells (Treg) can inhibit Type 1 diabetes in animal models and humans. We discuss the anatomical sites and molecular mechanisms of Treg suppressive function in the Type 1 diabetes setting, citing evidence that Treg can function in both the pancreatic lymph nodes and within the pancreatic lesion. Involvement of the CTLA-4 pathway, as well as TGF-β and IL-2 deprivation will be considered. Finally, we summarize current efforts to manipulate Treg therapeutically in individuals with Type 1 diabetes. The translation of this research area from bench to bedside is still in its infancy, but the remarkable therapeutic potential of successfully manipulating Treg populations is clear to see.
Collapse
Affiliation(s)
- Vitalijs Ovcinnikovs
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, London, United Kingdom.
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, London, United Kingdom
| |
Collapse
|
86
|
Cantoni C, Grauwet K, Pietra G, Parodi M, Mingari MC, Maria AD, Favoreel H, Vitale M. Role of NK cells in immunotherapy and virotherapy of solid tumors. Immunotherapy 2015; 7:861-82. [PMID: 26314197 DOI: 10.2217/imt.15.53] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Although natural killer (NK) cells are endowed with powerful cytolytic activity against cancer cells, their role in different therapies against solid tumors has not yet been fully elucidated. Their interactions with various elements of the tumor microenvironment as well as their possible effects in contributing to and/or limiting oncolytic virotherapy render this potential immunotherapeutic tool still difficult to exploit at the bedside. Here, we will review the current literature with the aim of providing new hints to manage this powerful cell type in future innovative therapies, such as the use of NK cells in combination with new cytokines, specific mAbs (inducing ADCC), Tyr-Kinase inhibitors, immunomodulatory drugs and/or the design of oncolytic viruses aimed at optimizing the effect of NK cells in virotherapy.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,Istituto Giannina Gaslini, Genova, Italy
| | - Korneel Grauwet
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Monica Parodi
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Andrea De Maria
- Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Herman Favoreel
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | | |
Collapse
|
87
|
von Burg N, Turchinovich G, Finke D. Maintenance of Immune Homeostasis through ILC/T Cell Interactions. Front Immunol 2015; 6:416. [PMID: 26322047 PMCID: PMC4534831 DOI: 10.3389/fimmu.2015.00416] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/29/2015] [Indexed: 12/19/2022] Open
Abstract
Innate lymphoid cells (ILCs) have emerged as a new family of immune cells with crucial functions in innate and adaptive immunity. ILC subsets mirror the cytokine and transcriptional profile of CD4+ T helper (TH) cell subsets. Hence, group 1 (ILC1), group 2 (ILC2), and group 3 (ILC3) ILCs can be distinguished by the production of TH1, TH2, and TH17-type cytokines, respectively. Cytokine release by ILCs not only shapes early innate immunity but can also orchestrate TH immune responses to microbial or allergen exposure. Recent studies have identified an unexpected effector function of ILCs as antigen presenting cells. Both ILC2s and ILC3s are able to process and present foreign antigens (Ags) via major histocompatibility complex class II, and to induce cognate CD4+ T cell responses. In addition, Ag-stimulated T cells promote ILC activation and effector functions indicating a reciprocal interaction between the adaptive and innate immune system. A fundamental puzzle in ILC function is how ILC/T cell interactions promote host protection and prevent autoimmune diseases. Furthermore, the way in which microenvironmental and inflammatory signals determine the outcome of ILC/T cell immune responses in various tissues is not yet understood. This review focuses on recent advances in understanding the mechanisms that coordinate the collaboration between ILCs and T cells under homeostatic and inflammatory conditions. We also discuss the potential roles of T cells and other immune cells to regulate ILC functions and to maintain homeostasis in mucosal tissues.
Collapse
Affiliation(s)
- Nicole von Burg
- Department of Biomedicine, University of Basel , Basel , Switzerland ; University of Basel Children's Hospital , Basel , Switzerland
| | - Gleb Turchinovich
- Department of Biomedicine, University of Basel , Basel , Switzerland ; University of Basel Children's Hospital , Basel , Switzerland
| | - Daniela Finke
- Department of Biomedicine, University of Basel , Basel , Switzerland ; University of Basel Children's Hospital , Basel , Switzerland
| |
Collapse
|
88
|
Brauner H, Hall HT, Flodström-Tullberg M, Kärre K, Höglund P, Johansson S. Depletion of IL-2 receptor β-positive cells protects from diabetes in non-obese diabetic mice. Immunol Cell Biol 2015; 94:177-84. [PMID: 26244831 DOI: 10.1038/icb.2015.78] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/25/2015] [Accepted: 07/22/2015] [Indexed: 01/12/2023]
Abstract
The destruction of β-cells in type 1 diabetes (T1D) progresses silently until only a minor fraction of the β-cells remain. A late acting therapy leading to the prevention of further β-cell killing would therefore be desirable. CD122, the β chain of the interleukin-2 receptor, is highly expressed on natural killer (NK) cells and on a subpopulation of CD8 T cells. In this study, we have treated non-obese diabetic (NOD) mice with a depleting antibody against CD122. The treatment protected from diabetes, even when initiated just before disease onset. The degree of leukocyte infiltration into islets was unaffected by the treatment, further supporting effectiveness late in the disease process. It effectively removed all NK cells from the spleen, pancreas and pancreatic lymph nodes and abolished NK cell activity. Interestingly, despite the lack of CD122 expression on CD8 T cells in the pancreas, the overall frequency of CD8 cells decreased in this organ, whereas it was unaffected in the spleen. T cells were also still capable to respond against a foreign antigen. Conclusively, targeting of CD122(+) cells could represent a novel treatment strategy against T1D.
Collapse
Affiliation(s)
- Hanna Brauner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Håkan T Hall
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Malin Flodström-Tullberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Klas Kärre
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Petter Höglund
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Sofia Johansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
89
|
Littwitz-Salomon E, Akhmetzyanova I, Vallet C, Francois S, Dittmer U, Gibbert K. Activated regulatory T cells suppress effector NK cell responses by an IL-2-mediated mechanism during an acute retroviral infection. Retrovirology 2015. [PMID: 26220086 PMCID: PMC4518534 DOI: 10.1186/s12977-015-0191-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background It is well established that effector T cell responses are crucial for the control of most virus infections, but they are often tightly controlled by regulatory T cells (Treg) to minimize immunopathology. NK cells also contribute to virus control but it is not known if their antiviral effect is influenced by virus-induced Tregs as well. We therefore analyzed whether antiretroviral NK cell functions are inhibited by Tregs during an acute Friend retrovirus infection of mice. Results Selective depletion of Tregs by using the transgenic DEREG mouse model resulted in improved NK cell proliferation, maturation and effector cell differentiation. Suppression of NK cell functions depended on IL-2 consumption by Tregs, which could be overcome by specific NK cell stimulation with an IL-2/anti-IL-2 mAb complex. Conclusions The current study demonstrates that virus-induced Tregs indeed inhibit antiviral NK cell responses and describes a targeted immunotherapy that can abrogate the suppression of NK cells by Tregs. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0191-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisabeth Littwitz-Salomon
- Institute of Virology of the University Hospital in Essen, University of Duisburg-Essen, Essen, Germany.
| | - Ilseyar Akhmetzyanova
- Institute of Virology of the University Hospital in Essen, University of Duisburg-Essen, Essen, Germany.
| | - Cecilia Vallet
- Institute of Virology of the University Hospital in Essen, University of Duisburg-Essen, Essen, Germany.
| | - Sandra Francois
- Institute of Virology of the University Hospital in Essen, University of Duisburg-Essen, Essen, Germany.
| | - Ulf Dittmer
- Institute of Virology of the University Hospital in Essen, University of Duisburg-Essen, Essen, Germany.
| | - Kathrin Gibbert
- Institute of Virology of the University Hospital in Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
90
|
Abstract
There is a clear need to develop strategies to induce tolerance without the need of chronic immunosuppression in transplant recipient and in patients with autoimmunity. Adoptive T regulatory cell (Treg) therapy offers the potential of long-lasting protection. However, based on results of clinical trials so far with ex vivo expanded autologous Tregs in type 1 diabetic (T1D) patients, it seems unlikely that single immunotherapy with Treg infusion without immunomodulation regimens that promote stable donor Treg engraftment and persistence would afford truly significant clinical benefit. Combination therapies could provide improved outcomes with consideration of the fundamental factors required for Treg generation, homeostasis, and function to promote long-term donor Treg persistence to provoke beneficial therapeutic outcomes.
Collapse
|
91
|
Rodríguez-Rodríguez N, Apostolidis SA, Penaloza-MacMaster P, Martín Villa JM, Barouch DH, Tsokos GC, Crispín JC. Programmed cell death 1 and Helios distinguish TCR-αβ+ double-negative (CD4-CD8-) T cells that derive from self-reactive CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:4207-14. [PMID: 25825451 DOI: 10.4049/jimmunol.1402775] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/01/2015] [Indexed: 01/20/2023]
Abstract
TCR-αβ(+) double-negative (DN; CD4(-)CD8(-)) T cells represent a poorly understood cellular subset suggested to contribute to the pathogenesis of the autoimmune disease systemic lupus erythematosus. DN T cells have been proposed to derive from CD8(+) cells. However, the conditions that govern the loss of CD8 expression after Ag encounter are unknown. In this study, we tracked the fate of CD8 T cells from transgenic TCR mice exposed to their cognate Ags as self or in the context of infection. We demonstrate that CD8 T cells lose CD8 expression and become DN only when cognate Ag is sensed as self. This process is restricted to tissues where the Ag is present. We also show that DN T cells derived from self-reactive CD8 cells express the inhibitory molecules PD-1 and Helios. These molecules identify a subset of DN T cells in normal mice. A similar population expands when CD8 T cells from repertoires enriched in self-reactive cells (Aire-deficient) are transferred into cognate hosts. Collectively, our data suggest that a subset of DN T cells, identified by the expression of PD-1 and Helios, represent self-reactive cells. Our results provide an explanation for the origin of DN T cells and introduce CD8 loss as a process associated with self-Ag encounter.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215; Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Sokratis A Apostolidis
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Pablo Penaloza-MacMaster
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
| | - José Manuel Martín Villa
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215;
| | - José C Crispín
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215;
| |
Collapse
|
92
|
Stat5 Exerts Distinct, Vital Functions in the Cytoplasm and Nucleus of Bcr-Abl+ K562 and Jak2(V617F)+ HEL Leukemia Cells. Cancers (Basel) 2015; 7:503-37. [PMID: 25809097 PMCID: PMC4381271 DOI: 10.3390/cancers7010503] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/09/2015] [Accepted: 03/12/2015] [Indexed: 01/09/2023] Open
Abstract
Signal transducers and activators of transcription (Stats) play central roles in the conversion of extracellular signals, e.g., cytokines, hormones and growth factors, into tissue and cell type specific gene expression patterns. In normal cells, their signaling potential is strictly limited in extent and duration. The persistent activation of Stat3 or Stat5 is found in many human tumor cells and contributes to their growth and survival. Stat5 activation plays a pivotal role in nearly all hematological malignancies and occurs downstream of oncogenic kinases, e.g., Bcr-Abl in chronic myeloid leukemias (CML) and Jak2(V617F) in other myeloproliferative diseases (MPD). We defined the mechanisms through which Stat5 affects growth and survival of K562 cells, representative of Bcr-Abl positive CML, and HEL cells, representative for Jak2(V617F) positive acute erythroid leukemia. In our experiments we suppressed the protein expression levels of Stat5a and Stat5b through shRNA mediated downregulation and demonstrated the dependence of cell survival on the presence of Stat5. Alternatively, we interfered with the functional capacities of the Stat5 protein through the interaction with a Stat5 specific peptide ligand. This ligand is a Stat5 specific peptide aptamer construct which comprises a 12mer peptide integrated into a modified thioredoxin scaffold, S5-DBD-PA. The peptide sequence specifically recognizes the DNA binding domain (DBD) of Stat5. Complex formation of S5-DBD-PA with Stat5 causes a strong reduction of P-Stat5 in the nuclear fraction of Bcr-Abl-transformed K562 cells and a suppression of Stat5 target genes. Distinct Stat5 mediated survival mechanisms were detected in K562 and Jak2(V617F)-transformed HEL cells. Stat5 is activated in the nuclear and cytosolic compartments of K562 cells and the S5-DBD-PA inhibitor most likely affects the viability of Bcr-Abl+ K562 cells through the inhibition of canonical Stat5 induced target gene transcription. In HEL cells, Stat5 is predominantly present in the cytoplasm and the survival of the Jak2(V617F)+ HEL cells is impeded through the inhibition of the cytoplasmic functions of Stat5.
Collapse
|
93
|
Mahne AE, Klementowicz JE, Chou A, Nguyen V, Tang Q. Therapeutic regulatory T cells subvert effector T cell function in inflamed islets to halt autoimmune diabetes. THE JOURNAL OF IMMUNOLOGY 2015; 194:3147-55. [PMID: 25732730 DOI: 10.4049/jimmunol.1402739] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Therapeutic regulatory T cells (Tregs) can reverse pre-established autoimmune pathology. In this study, using a mouse model of autoimmune diabetes, we aimed to determine the means by which therapeutic Tregs control islet inflammation. Islet Ag-specific Tregs infiltrated inflamed islets soon after infusion into prediabetic mice, which was quickly followed by a selective reduction of mRNA associated with effector T cells in the islets. This change was partially due to decreased CD8(+) T cell accumulation in the tissue. CD8(+) T cells that remained in the islets after Treg treatment were able to engage dendritic cells in a manner similar to that found in untreated mice, consistent with the retention of an activated phenotype by islet dendritic cells shortly after Treg treatment. Nonetheless, Treg treatment abrogated IFN-γ production by intraislet CD8(+) and CD4(+) T cells at the protein level with minimal effect on IFN-γ mRNA. Sustained expression of IFN-γ protein by effector T cells was dependent on common γ-chain cytokine activation of the mTOR pathway, which was suppressed in islet CD8(+) T cells in vivo after Treg treatment. These multifaceted mechanisms underlie the efficacy of therapeutic Treg subversion of effector T cell functions at the site of inflammation to restore normal tissue homeostasis.
Collapse
Affiliation(s)
- Ashley E Mahne
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Joanna E Klementowicz
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Annie Chou
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Vinh Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
94
|
Cichocki F, Verneris MR, Cooley S, Bachanova V, Brunstein CG, Blazar BR, Wagner J, Schlums H, Bryceson YT, Weisdorf DJ, Miller JS. The Past, Present, and Future of NK Cells in Hematopoietic Cell Transplantation and Adoptive Transfer. Curr Top Microbiol Immunol 2015; 395:225-43. [PMID: 26037048 DOI: 10.1007/82_2015_445] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hematopoietic cell transplantation (HCT) has been used as a part of cancer therapy for over half a decade. Beyond the necessity for donor-derived cells to reconstitute hematopoiesis after radiation and chemotherapy, immunologic reconstitution from allogeneic cells is important for the elimination of residual tumor cells. Natural killer (NK) cells are first among lymphocytes to reconstitute post-transplant and protect against cancer relapse. In this review, we provide a historical perspective on the role of NK cells in cancer control in the transplant setting and focus on current research aimed at improving NK cell responses for therapeutic benefit.
Collapse
Affiliation(s)
- Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Sarah Cooley
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - John Wagner
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Heinrich Schlums
- Centre for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yenan T Bryceson
- Centre for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Broeglmann Research Laboratory, Clinical Institute, University of Bergen, Bergen, Norway
| | - Daniel J Weisdorf
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA. .,MMC 806, Division of Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, Harvard Street at East River Road, Minneapolis, MN, 55455, USA.
| |
Collapse
|
95
|
Abstract
Natural killer (NK) cells are innate lymphocytes that survey the environment and protect the host from infected and cancerous cells. As their name implies, NK cells represent an early line of defense during pathogen invasion by directly killing infected cells and secreting inflammatory cytokines. Although the function of NK cells was first described more than four decades ago, the development of this cytotoxic lineage is not well understood. In recent years, we have begun to identify specific transcription factors that control each stage of development and maturation, from ontogeny of the NK cell progenitor to the effector functions of activated NK cells in peripheral organs. This chapter highlights the transcription factors that are unique to NK cells, or shared between NK cells and other hematopoietic cell lineages, but govern the biology of this cytolytic lymphocyte.
Collapse
Affiliation(s)
- Joseph C Sun
- Memorial Sloan Kettering Cancer Center, Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, 408 East 69th Street, ZRC-1402, New York, NY, 10065, USA.
| |
Collapse
|
96
|
Regulatory T cells promote a protective Th17-associated immune response to intestinal bacterial infection with C. rodentium. Mucosal Immunol 2014; 7:1290-301. [PMID: 24646939 DOI: 10.1038/mi.2014.17] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 02/17/2014] [Indexed: 02/04/2023]
Abstract
Intestinal infection with the mouse pathogen Citrobacter rodentium induces a strong local Th17 response in the colon. Although this inflammatory immune response helps to clear the pathogen, it also induces inflammation-associated pathology in the gut and thus, has to be tightly controlled. In this project, we therefore studied the impact of Foxp3(+) regulatory T cells (Treg) on the infectious and inflammatory processes elicited by the bacterial pathogen C. rodentium. Surprisingly, we found that depletion of Treg by diphtheria toxin in the Foxp3(DTR) (DEREG) mouse model resulted in impaired bacterial clearance in the colon, exacerbated body weight loss, and increased systemic dissemination of bacteria. Consistent with the enhanced susceptibility to infection, we found that the colonic Th17-associated T-cell response was impaired in Treg-depleted mice, suggesting that the presence of Treg is crucial for the establishment of a functional Th17 response after the infection in the gut. As a consequence of the impaired Th17 response, we also observed less inflammation-associated pathology in the colons of Treg-depleted mice. Interestingly, anti-interleukin (IL)-2 treatment of infected Treg-depleted mice restored Th17 induction, indicating that Treg support the induction of a protective Th17 response during intestinal bacterial infection by consumption of local IL-2.
Collapse
|
97
|
Hirohashi T, Chase CM, DellaPelle P, Sebastian D, Farkesh E, Colvin RB, Russell PS, Alessandrini A, Madsen JC. Depletion of T regulatory cells promotes natural killer cell-mediated cardiac allograft vasculopathy. Transplantation 2014; 98:828-34. [PMID: 25321164 PMCID: PMC4203423 DOI: 10.1097/tp.0000000000000329] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND A role for natural killer (NK) cells in cardiac allograft vasculopathy (CAV) was suggested by our earlier observation that CAV arises even in the absence of detectable antidonor T-cell or B-cell reactivity in parental to F1 mouse heart grafts. However, prevention of CAV in this setting required the depletion of both NK and CD4 T cells. METHODS To clarify the interrelationship between NK and CD4 cells, we analyzed early events and selective depletion of T regulatory cells (Tregs). Hearts from C57BL/6 (B6) donors were transplanted heterotopically into BALB/c x C57BL/6 (CB6F1) recipients and NK cells, CD4 T cells, and Tregs were depleted with anti-NK1.1 (PK136), anti-CD4 (GK1.5), or anti-CD25 (PC61), respectively. RESULTS In contrast to prior studies in which the prevention of CAV at 8 weeks required the codepletion of NK and CD4 T cells, NK cells depletion alone eliminated CAV at 3 weeks. Furthermore, depletion of CD25 cells accelerated the onset and maturation of CAV at both 2 and 3 weeks (P<0.02 and P<0.001, respectively). However, anti-NK1.1 treatment prevented lesions in CD25-depleted recipients. Finally, CD4 T cell depletion alone did not prevent or accelerate development of CAV but inhibited the effect of CD25 T cell depletion. CONCLUSION These data suggest that NK cells can play an important role in the early pathogenesis of CAV but that their ability to mediate early CAV can be modulated by Tregs.
Collapse
Affiliation(s)
- Tsutomu Hirohashi
- Transplantation Center and Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Catharine M. Chase
- Transplantation Center and Department of Surgery, Massachusetts General Hospital, Boston, MA
| | | | - Divya Sebastian
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Evan Farkesh
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Robert B. Colvin
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Paul S. Russell
- Transplantation Center and Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Alessandro Alessandrini
- Transplantation Center and Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Joren C. Madsen
- Transplantation Center and Department of Surgery, Massachusetts General Hospital, Boston, MA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
98
|
Donnelly RP, Loftus RM, Keating SE, Liou KT, Biron CA, Gardiner CM, Finlay DK. mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. THE JOURNAL OF IMMUNOLOGY 2014; 193:4477-84. [PMID: 25261477 DOI: 10.4049/jimmunol.1401558] [Citation(s) in RCA: 335] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mammalian target of rapamycin complex 1 (mTORC1) is a key regulator of cellular metabolism and also has fundamental roles in controlling immune responses. Emerging evidence suggests that these two functions of mTORC1 are integrally linked. However, little is known regarding mTORC1 function in controlling the metabolism and function of NK cells, lymphocytes that play key roles in antiviral and antitumor immunity. This study investigated the hypothesis that mTORC1-controlled metabolism underpins normal NK cell proinflammatory function. We demonstrate that mTORC1 is robustly stimulated in NK cells activated in vivo and in vitro. This mTORC1 activity is required for the production of the key NK cell effector molecules IFN-γ, which is important in delivering antimicrobial and immunoregulatory functions, and granzyme B, a critical component of NK cell cytotoxic granules. The data reveal that NK cells undergo dramatic metabolic reprogramming upon activation, upregulating rates of glucose uptake and glycolysis, and that mTORC1 activity is essential for attaining this elevated glycolytic state. Directly limiting the rate of glycolysis is sufficient to inhibit IFN-γ production and granzyme B expression. This study provides the highly novel insight that mTORC1-mediated metabolic reprogramming of NK cells is a prerequisite for the acquisition of normal effector functions.
Collapse
Affiliation(s)
- Raymond P Donnelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Róisín M Loftus
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sinéad E Keating
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kevin T Liou
- Division of Biology and Medicine; Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912; and
| | - Christine A Biron
- Division of Biology and Medicine; Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912; and
| | - Clair M Gardiner
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
99
|
Milanez-Almeida P, Klawonn F, Meyer-Hermann M, Huehn J. Differential control of immune cell homeostasis by Foxp3(+) regulatory T cells in murine peripheral lymph nodes and spleen. Eur J Microbiol Immunol (Bp) 2014; 4:147-55. [PMID: 25215190 DOI: 10.1556/eujmi-d-14-00022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 07/17/2014] [Indexed: 01/15/2023] Open
Abstract
Foxp3(+) regulatory T cells (Tregs) hamper efficient immune responses to tumors and chronic infections. Therefore, depletion of Foxp3(+) Tregs has been proposed as therapeutic option to boost immune responses and to improve vaccinations. Although Treg-mediated control of T cell homeostasis is well established, Foxp3(+) Treg interaction with other immune cell subsets is only incompletely understood. Thus, the present study aimed at examining dynamic effects of experimental Foxp3(+) Treg depletion on a broad range of immune cell subsets, including B cells, natural killer cells, and myeloid cells. Striking differences were observed when peripheral lymph nodes (LN) and spleen were compared. B cells, for example, showed a massive and long-lasting accumulation only in LN but not in spleen of transiently Treg-depleted mice. In contrast, monocyte-derived dendritic cells, which are potent inducers of T cell responses, also accumulated selectively, but only transiently in LN, suggesting that this cell population is under very strict control of Foxp3(+) Tregs. In summary, the observations described here provide insights into the dynamics of immune cells after selective depletion of Foxp3(+) Tregs. This will allow a better prediction of the impact of Treg ablation in translational studies that aim at boosting immune responses and vaccinations.
Collapse
|
100
|
Abstract
Innate lymphocytes - including natural killer cells and the recently discovered innate lymphoid cells - have crucial roles during infection, tissue injury and inflammation. Innate signals regulate the activation and homeostasis of innate lymphocytes. The contribution of the adaptive immune system to the coordination of innate lymphocyte responses is less well understood. In this Opinion article, we review our current understanding of the interactions between adaptive and innate lymphocytes, and propose a model in which T cells of the adaptive immune system function as antigen-specific sensors for the activation of innate lymphocytes to amplify and instruct local immune responses. We highlight the potential roles of regulatory and helper T cells in these processes, and discuss major questions in the emerging area of crosstalk between adaptive and innate lymphocytes.
Collapse
|