51
|
Nolz JC, Richer MJ. Control of memory CD8 + T cell longevity and effector functions by IL-15. Mol Immunol 2019; 117:180-188. [PMID: 31816491 DOI: 10.1016/j.molimm.2019.11.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/12/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022]
Abstract
IL-15 is a member of the common gamma chain family of cytokines and plays important roles in regulating several aspects of innate and adaptive immunity. Besides its established role in controlling homeostatic proliferation and survival of memory CD8+ T cells and natural killer cells, recent findings demonstrate that inflammatory IL-15 can also stimulate a variety of effector functions, such as enhanced cytotoxicity, entry into the cell cycle, and trafficking into non-lymphoid tissues. Here, we discuss how IL-15 is critical in regulating many functions of memory CD8+ T cells and how these processes act collectively to ensure optimal protective cellular immunity against re-infections.
Collapse
Affiliation(s)
- Jeffrey C Nolz
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, United States; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, United States; Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States.
| | - Martin J Richer
- Department of Microbiology & Immunology, McGill University, 712 McIntyre Medical Building, 3655 promenade Sir William Osler, Montreal, Quebec, Canada; Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
52
|
Quéméner A, Morisseau S, Sousa RP, Trillet K, Maillasson M, Leray I, Jacques Y, Dion J, Barbieux I, Frutoso M, Laurent AD, Le Questel JY, Mortier E. IL-15Rα membrane anchorage in either cis or trans is required for stabilization of IL-15 and optimal signaling. J Cell Sci 2019; 133:jcs.236802. [PMID: 31653781 DOI: 10.1242/jcs.236802] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023] Open
Abstract
Interleukin (IL)-15 plays an important role in the communication between immune cells. It delivers its signal through different modes involving three receptor chains: IL-15Rα, IL-2Rβ and IL-2Rγc. The combination of the different chains result in the formation of IL-15Rα/IL-2Rβ/γc trimeric or IL-2Rβ/γc dimeric receptors. In this study, we have investigated the role of the IL-15Rα chain in stabilizing the cytokine in the IL-2Rβ/γc dimeric receptor. By analyzing the key amino acid residues of IL-15 facing IL-2Rβ, we provide evidence of differential interfaces in the presence or in the absence of membrane-anchored IL-15Rα. Moreover, we found that the anchorage of IL-15Rα to the cell surface regardless its mode of presentation - i.e. cis or trans - is crucial for complete signaling. These observations show how the cells can finely modulate the intensity of cytokine signaling through the quality and the level of expression of the receptor chains.
Collapse
Affiliation(s)
- Agnès Quéméner
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Sébastien Morisseau
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France.,CHU, Nantes Hospital, Nantes, France
| | - Rui P Sousa
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France.,CEISAM UMR CNRS 6230, UFR Sciences et Techniques, University of Nantes, Nantes, France
| | - Kilian Trillet
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Mike Maillasson
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France.,Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, FED 4203, Inserm UMS 016, CNRS UMS 3556, IMPACT Platform, Nantes, France
| | - Isabelle Leray
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Yannick Jacques
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Johann Dion
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, FED 4203, Inserm UMS 016, CNRS UMS 3556, IMPACT Platform, Nantes, France
| | - Isabelle Barbieux
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Marie Frutoso
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France.,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France
| | - Adèle D Laurent
- CEISAM UMR CNRS 6230, UFR Sciences et Techniques, University of Nantes, Nantes, France
| | - Jean-Yves Le Questel
- CEISAM UMR CNRS 6230, UFR Sciences et Techniques, University of Nantes, Nantes, France
| | - Erwan Mortier
- CRCINA, CNRS, Inserm, University of Angers, University of Nantes, Nantes, France .,LabEx IGO, Immunotherapy, Graft, Oncology, Nantes, France.,Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, FED 4203, Inserm UMS 016, CNRS UMS 3556, IMPACT Platform, Nantes, France
| |
Collapse
|
53
|
IL-15/IL-15Rα/CD80-expressing AML cell vaccines eradicate minimal residual disease in leukemic mice. Blood Adv 2019; 2:3177-3192. [PMID: 30482760 DOI: 10.1182/bloodadvances.2018019026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/14/2018] [Indexed: 12/17/2022] Open
Abstract
Engineered autologous acute myeloid leukemia (AML) cells present multiple leukemia-associated and patient-specific antigens and as such hold promise as immunotherapeutic vaccines. However, prior vaccines have not reliably induced effective antileukemic immunity, in part because AML blasts have immune inhibitory effects and lack expression of the critical costimulatory molecule CD80. To enhance induction of leukemia-specific cytolytic activity, 32Dp210 murine AML cells were engineered to express either CD80 alone, or the immunostimulatory cytokine interleukin-15 (IL-15) with its receptor α (IL-15Rα), or heterodimeric IL-15/IL-15Rα together with CD80 and tested as irradiated cell vaccines. IL-15 is a γc-chain cytokine, with unique properties suited to stimulating antitumor immunity, including stimulation of both natural killer and CD8+ memory T cells. Coexpression of IL-15 and IL-15Rα markedly increases IL-15 stability and secretion. Non-tumor-bearing mice vaccinated with irradiated 32Dp210-IL-15/IL-15Rα/CD80 and challenged with 32Dp210 leukemia had greater survival than did mice treated with 32Dp210-CD80 or 32Dp210-IL-15/IL-15Rα vaccines, whereas no unvaccinated mice inoculated with leukemia survived. In mice with established leukemia, treatment with 32Dp210-IL-15/IL-15Rα/CD80 vaccination stimulated unprecedented antileukemic immunity enabling 80% survival, an effect that was abrogated by anti-CD8 antibody-mediated depletion in vivo. Because, clinically, AML vaccines are administered as postremission therapy, we established a novel model in which mice with high leukemic burdens were treated with cytotoxic therapy to induce remission (<5% marrow blasts). Postremission vaccination with 32Dp210-IL-15/IL-15Rα/CD80 achieved 50% overall survival in these mice, whereas all unvaccinated mice achieving remission subsequently relapsed. These studies demonstrate that combined expression of IL-15/IL-15Rα and CD80 by syngeneic AML vaccines stimulates effective and long-lasting antileukemic immunity.
Collapse
|
54
|
Frutoso M, Mortier E. NK Cell Hyporesponsiveness: More Is Not Always Better. Int J Mol Sci 2019; 20:ijms20184514. [PMID: 31547251 PMCID: PMC6770168 DOI: 10.3390/ijms20184514] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 08/30/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
Natural Killer (NK) cells are a type of cytotoxic lymphocytes that play an important role in the innate immune system. They are of particular interest for their role in elimination of intracellular pathogens, viral infection and tumor cells. As such, numerous strategies are being investigated in order to potentiate their functions. One of these techniques aims at promoting the function of their activating receptors. However, different observations have revealed that providing activation signals could actually be counterproductive and lead to NK cells’ hyporesponsiveness. This phenomenon can occur during the NK cell education process, under pathological conditions, but also after treatment with different agents, including cytokines, that are promising tools to boost NK cell function. In this review, we aim to highlight the different circumstances where NK cells become hyporesponsive and the methods that could be used to restore their functionality.
Collapse
Affiliation(s)
- Marie Frutoso
- CRCINA, CNRS, Inserm, University of Nantes, F-44200 Nantes, France.
- LabEX IGO, Immuno-Onco-Greffe, Nantes, France.
| | - Erwan Mortier
- CRCINA, CNRS, Inserm, University of Nantes, F-44200 Nantes, France.
- LabEX IGO, Immuno-Onco-Greffe, Nantes, France.
| |
Collapse
|
55
|
Microenvironmental immune cell signatures dictate clinical outcomes for PTCL-NOS. Blood Adv 2019; 2:2242-2252. [PMID: 30194138 DOI: 10.1182/bloodadvances.2018018754] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/23/2018] [Indexed: 12/23/2022] Open
Abstract
Peripheral T-cell lymphoma (PTCL), not otherwise specified (PTCL-NOS) is among the most common disease subtypes of PTCL, one that exhibits heterogeneous clinicopathological features. Although multiple disease-stratification models, including the cell-of-origin or gene-expression profiling methods, have been proposed for this condition, their clinical significance remains unclear. To establish a clinically meaningful stratification model, we analyzed gene-expression signatures of tumors and tumor-infiltrating immune cells using the nCounter system, which enables accurate quantification of low abundance and/or highly fragmented transcripts. To do so, we assessed transcripts of 120 genes related to cancer or immune cells using tumor samples from 68 newly diagnosed PTCL-NOS patients and validated findings by immunofluorescence in tumor sections. We show that gene-expression signatures representing tumor-infiltrating immune cells, but not those of cancerous T cells, dictate patient clinical outcomes. Cases exhibiting both B-cell and dendritic cell (DC) signatures (BD subgroup) showed favorable clinical outcomes, whereas those exhibiting neither B-cell nor DC signatures (non-BD subgroup) showed extremely poor prognosis. Notably, half of the non-BD cases exhibited a macrophage signature, and macrophage infiltration was evident in those cases, as revealed by immunofluorescence. Importantly, tumor-infiltrating macrophages expressed the immune-checkpoint molecules programmed death ligand 1/2 and indoleamine 2, 3-dioxygenase 1 at high levels, suggesting that checkpoint inhibitors could serve as therapeutic options for patients in this subgroup. Our study identifies clinically distinct subgroups of PTCL-NOS and suggests a novel therapeutic strategy for 1 subgroup associated with a poor prognosis. Our data also suggest functional interactions between cancerous T cells and tumor-infiltrating immune cells potentially relevant to PTCL-NOS pathogenesis.
Collapse
|
56
|
Chen X, Guo W, Chang Y, Chen J, Kang P, Yi X, Cui T, Guo S, Xiao Q, Jian Z, Li K, Gao T, Li S, Liu L, Li C. Oxidative stress-induced IL-15 trans-presentation in keratinocytes contributes to CD8 + T cells activation via JAK-STAT pathway in vitiligo. Free Radic Biol Med 2019; 139:80-91. [PMID: 31078730 DOI: 10.1016/j.freeradbiomed.2019.05.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 01/06/2023]
Abstract
Oxidative stress and effector memory CD8+ T cells have been greatly implicated in vitiligo pathogenesis. However, the crosstalk between these two crucial pathogenic factors has been merely investigated. IL-15 has been regarded as an important cytokine exerting its facilitative effect on memory CD8+ T cells function in various autoimmune diseases. In the present study, we initially discovered that the IL-15 expression was significantly increased in vitiligo epidermis and highly associated with epidermal H2O2 content. In addition, epidermal IL-15 expression was mainly derived from keratinocytes. Then, we showed that oxidative stress promoted IL-15 and IL-15Rα expression as well as IL-15 trans-presentation by activating NF-κB signaling in keratinocytes. What's more, the trans-presented IL-15, rather than the secreted one, was accounted for the potentiation of CD8+ TEMs activation. We further investigated the mechanism underlying trans-presented IL-15 in potentiating CD8+ TEMs activation and found that the blockage of IL-15-JAK-STAT signaling could be a potent therapeutic approach. Taken together, our results demonstrate that oxidative stress-induced IL-15 trans-presentation in keratinocytes contributes to the activation of CD8+ TEMs, providing a novel mechanism by which oxidative stress initiates autoimmunity in vitiligo.
Collapse
Affiliation(s)
- Xuguang Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuqian Chang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiaxi Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pan Kang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tingting Cui
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qian Xiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kai Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuli Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Ling Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
57
|
Stocks BT, Wilson CS, Marshall AF, Hoopes EM, Moore DJ. Regulation of Diabetogenic Immunity by IL-15-Activated Regulatory CD8 T Cells in Type 1 Diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:158-166. [PMID: 31127035 PMCID: PMC6581590 DOI: 10.4049/jimmunol.1800976] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 05/01/2019] [Indexed: 01/10/2023]
Abstract
Unchecked collaboration between islet-reactive T and B lymphocytes drives type 1 diabetes (T1D). In the healthy setting, CD8 T regulatory cells (Tregs) terminate ongoing T-B interactions. We determined that specific CD8 Tregs from NOD mice lack suppressive function, representing a previously unreported regulatory cell deficit in this T1D-prone strain. NOD mice possess 11-fold fewer Ly-49+ CD8 Tregs than nonautoimmune mice, a deficiency that worsens as NOD mice age toward diabetes and leaves them unable to regulate CD4 T follicular helper cells. As IL-15 is required for Ly-49+ CD8 Treg development, we determined that NOD macrophages inadequately trans-present IL-15. Despite reduced IL-15 trans-presentation, NOD Ly-49+ CD8 Tregs can effectively transduce IL-15-mediated survival signals when they are provided. Following stimulation with an IL-15/IL-15Ra superagonist complex, Ly-49+ CD8 Tregs expanded robustly and became activated to suppress the Ag-specific Ab response. IL-15/IL-15Ra superagonist complex-activated CD8+CD122+ T cells also delayed diabetes transfer, indicating the presence of an underactivated CD8 T cell subset with regulatory capacity against late stage T1D. We identify a new cellular contribution to anti-islet autoimmunity and demonstrate the correction of this regulatory cell deficit. Infusion of IL-15-activated CD8 Tregs may serve as an innovative cellular therapy for the treatment of T1D.
Collapse
Affiliation(s)
- Blair T Stocks
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN 37232; and
| | - Christopher S Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232
| | - Andrew F Marshall
- Ian Burr Division of Endocrinology and Diabetes, Department of Pediatrics, Vanderbilt University, Nashville, TN 37232
| | - Emilee M Hoopes
- Ian Burr Division of Endocrinology and Diabetes, Department of Pediatrics, Vanderbilt University, Nashville, TN 37232
| | - Daniel J Moore
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232;
- Ian Burr Division of Endocrinology and Diabetes, Department of Pediatrics, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
58
|
Vidard L, Dureuil C, Baudhuin J, Vescovi L, Durand L, Sierra V, Parmantier E. CD137 (4-1BB) Engagement Fine-Tunes Synergistic IL-15- and IL-21-Driven NK Cell Proliferation. THE JOURNAL OF IMMUNOLOGY 2019; 203:676-685. [PMID: 31201235 DOI: 10.4049/jimmunol.1801137] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 05/27/2019] [Indexed: 12/22/2022]
Abstract
To understand and dissect the mechanisms driving human NK cell proliferation, we exploited the methodology used in cell therapy to numerically expand NK cells in the presence of K562-derived artificial APC (aAPCs) and cytokines. For four consecutive weeks, high expression of CD137L by a K562-derived aAPC cell line could sustain NK cell expansion by 3 × 105-fold, whereas low expression of CD137L by the parental K562 cell line only supported the expansion by 2 × 103-fold. The level of expression of CD137L, however, did not modulate the sensitivity of K562 cells to the intrinsic cytotoxicity of NK cells. Similarly, the low NK cell proliferation in the presence of the parental K562 cell line and cytokines was increased by adding agonistic anti-CD137 Abs to levels similar to CD137L-expressing K562-derived aAPCs. Finally, synergy between IL-15 and IL-21 was observed only upon CD137 engagement and the presence of aAPCs. Therefore, we conclude that NK cell proliferation requires cell-to-cell contact, activation of the CD137 axis, and presence of IL-15 (or its membranous form) and IL-21. By analogy with the three-signal model required to activate T cells, we speculate that the cell-to-cell contact represents "signal 1," CD137 represents "signal 2," and cytokines represent "signal 3." The precise nature of signal 1 remains to be defined.
Collapse
Affiliation(s)
- Laurent Vidard
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| | - Christine Dureuil
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| | - Jérémy Baudhuin
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| | - Lionel Vescovi
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| | - Laurence Durand
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| | - Véronique Sierra
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| | - Eric Parmantier
- Department of Immuno-Oncology, Sanofi, 94403 Vitry-sur-Seine, France
| |
Collapse
|
59
|
Van Acker HH, Versteven M, Lichtenegger FS, Roex G, Campillo-Davo D, Lion E, Subklewe M, Van Tendeloo VF, Berneman ZN, Anguille S. Dendritic Cell-Based Immunotherapy of Acute Myeloid Leukemia. J Clin Med 2019; 8:E579. [PMID: 31035598 PMCID: PMC6572115 DOI: 10.3390/jcm8050579] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/16/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a type of blood cancer characterized by the uncontrolled clonal proliferation of myeloid hematopoietic progenitor cells in the bone marrow. The outcome of AML is poor, with five-year overall survival rates of less than 10% for the predominant group of patients older than 65 years. One of the main reasons for this poor outcome is that the majority of AML patients will relapse, even after they have attained complete remission by chemotherapy. Chemotherapy, supplemented with allogeneic hematopoietic stem cell transplantation in patients at high risk of relapse, is still the cornerstone of current AML treatment. Both therapies are, however, associated with significant morbidity and mortality. These observations illustrate the need for more effective and less toxic treatment options, especially in elderly AML and have fostered the development of novel immune-based strategies to treat AML. One of these strategies involves the use of a special type of immune cells, the dendritic cells (DCs). As central orchestrators of the immune system, DCs are key to the induction of anti-leukemia immunity. In this review, we provide an update of the clinical experience that has been obtained so far with this form of immunotherapy in patients with AML.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Maarten Versteven
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Felix S Lichtenegger
- Department of Medicine III, LMU Munich, University Hospital, 80799 Munich, Germany.
| | - Gils Roex
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Marion Subklewe
- Department of Medicine III, LMU Munich, University Hospital, 80799 Munich, Germany.
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Antwerp, Belgium.
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, 2610 Wilrijk, Antwerp, Belgium.
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Antwerp, Belgium.
| |
Collapse
|
60
|
Abstract
Natural killer (NK) cells have evolved to complement T and B cells in host defense against pathogens and cancer. They recognize infected cells and tumors using a sophisticated array of activating, costimulatory, and inhibitory receptors that are expressed on NK cell subsets to create extensive functional diversity. NK cells can be targeted to kill with exquisite antigen specificity by antibody-dependent cellular cytotoxicity. NK and T cells share many of the costimulatory and inhibitory receptors that are currently under evaluation in the clinic for cancer immunotherapy. As with T cells, genetic engineering is being employed to modify NK cells to specifically target them to tumors and to enhance their effector functions. As the selective pressures exerted by immunotherapies to augment CD8+T cell responses may result in loss of MHC class I, NK cells may provide an important fail-safe to eliminate these tumors by their capacity to eliminate tumors that are “missing self.”
Collapse
Affiliation(s)
- Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California 94143, USA
- The Parker Institute for Cancer Immunotherapy, San Francisco, California 94143, USA
| |
Collapse
|
61
|
Nayyar G, Chu Y, Cairo MS. Overcoming Resistance to Natural Killer Cell Based Immunotherapies for Solid Tumors. Front Oncol 2019; 9:51. [PMID: 30805309 PMCID: PMC6378304 DOI: 10.3389/fonc.2019.00051] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/18/2019] [Indexed: 12/22/2022] Open
Abstract
Despite advances in the diagnostic and therapeutic modalities, the prognosis of several solid tumor malignancies remains poor. Different factors associated with solid tumors including a varied genetic signature, complex molecular signaling pathways, defective cross talk between the tumor cells and immune cells, hypoxic and immunosuppressive effects of tumor microenvironment result in a treatment resistant and metastatic phenotype. Over the past several years, immunotherapy has emerged as an attractive therapeutic option against multiple malignancies. The unique ability of natural killer (NK) cells to target cancer cells without antigen specificity makes them an ideal candidate for use against solid tumors. However, the outcomes of adoptive NK cell infusions into patients with solid tumors have been disappointing. Extensive studies have been done to investigate different strategies to improve the NK cell function, trafficking and tumor targeting. Use of cytokines and cytokine analogs has been well described and utilized to enhance the proliferation, stimulation and persistence of NK cells. Other techniques like blocking the human leukocyte antigen-killer cell receptors (KIR) interactions with anti-KIR monoclonal antibodies, preventing CD16 receptor shedding, increasing the expression of activating NK cell receptors like NKG2D, and use of immunocytokines and immune checkpoint inhibitors can enhance NK cell mediated cytotoxicity. Using genetically modified NK cells with chimeric antigen receptors and bispecific and trispecific NK cell engagers, NK cells can be effectively redirected to the tumor cells improving their cytotoxic potential. In this review, we have described these strategies and highlighted the need to further optimize these strategies to improve the clinical outcome of NK cell based immunotherapy against solid tumors.
Collapse
Affiliation(s)
- Gaurav Nayyar
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, United States.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, United States.,Department of Medicine, New York Medical College, Valhalla, NY, United States.,Department of Pathology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
62
|
Verboogen DRJ, Revelo NH, ter Beest M, van den Bogaart G. Interleukin-6 secretion is limited by self-signaling in endosomes. J Mol Cell Biol 2019; 11:144-157. [PMID: 30016456 PMCID: PMC6392102 DOI: 10.1093/jmcb/mjy038] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 05/04/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
Cells producing cytokines often express the receptor for the same cytokine, which makes them prone to autocrine signaling. How cytokine release and signaling are regulated in the same cell is not understood. In this study, we demonstrate that signaling by exogenous and self-synthesized inflammatory cytokine interleukin-6 (IL-6) within endosomal compartments acts as a cellular brake that limits the synthesis of IL-6. Our data show that IL-6 is internalized by dendritic cells and signals from endosomal compartments containing the IL-6 receptor. Newly synthesized IL-6 also traffics via these endosomal compartments and signals in transit to the plasma membrane. This allows activation of STAT3 which in turn limits toll-like receptor 4 stimulant lipopolysaccharide (LPS) triggered transcription of IL-6. Long-term exposure to LPS removes this brake via inhibition of STAT3 by increased expression of suppressor of cytokine signaling 3 and results in fully fledged IL-6 production. This transient regulation could prevent excessive IL-6 production during early infections.
Collapse
Affiliation(s)
- Daniëlle R J Verboogen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Natalia H Revelo
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, Groningen, The Netherlands
| |
Collapse
|
63
|
IL-15 is a component of the inflammatory milieu in the tumor microenvironment promoting antitumor responses. Proc Natl Acad Sci U S A 2018; 116:599-608. [PMID: 30587590 DOI: 10.1073/pnas.1814642116] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Previous studies have provided evidence that IL-15 expression within human tumors is crucial for optimal antitumor responses; however, the regulation of IL-15 within the tumor microenvironment (TME) is unclear. We report herein, in analyses of mice implanted with various tumor cell lines, soluble IL-15/IL-15Rα complexes (sIL-15 complexes) are abundant in the interstitial fluid of tumors with expression preceding the infiltration of tumor-infiltrating lymphocytes. Moreover, IL-15 as well as type I IFN, which regulates IL-15, was required for establishing normal numbers of CD8 T cells and natural killer cells in tumors. Depending on tumor type, both the tumor and the stroma are sources of sIL-15 complexes. In analyses of IL-15 reporter mice, most myeloid cells in the TME express IL-15 with CD11b+Ly6Chi cells being the most abundant, indicating there is a large source of IL-15 protein in tumors that lies sequestered within the tumor stroma. Despite the abundance of IL-15-expressing cells, the relative levels of sIL-15 complexes are low in advanced tumors but can be up-regulated by local stimulator of IFN genes (STING) activation. Furthermore, while treatment of tumors with STING agonists leads to tumor regression, optimal STING-mediated immunity and regression of distant secondary tumors required IL-15 expression. Overall, our study reveals the dynamic regulation of IL-15 in the TME and its importance in antitumor immunity. These findings provide insight into an unappreciated attribute of the tumor landscape that contributes to antitumor immunity, which can be manipulated therapeutically to enhance antitumor responses.
Collapse
|
64
|
Gauthier SD, Moutuou MM, Daudelin F, Leboeuf D, Guimond M. IL-7 Is the Limiting Homeostatic Factor that Constrains Homeostatic Proliferation of CD8 + T Cells after Allogeneic Stem Cell Transplantation and Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 25:648-655. [PMID: 30576835 DOI: 10.1016/j.bbmt.2018.12.066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/11/2018] [Indexed: 11/29/2022]
Abstract
Immune reconstitution after allogeneic hematopoietic stem cell transplantation relies primarily on homeostatic proliferation (HP) of mature T lymphocytes, but this process is typically impaired during graft-versus-host disease (GVHD). We previously showed that low IL-7 levels combined with lack of dendritic cell (DC) regeneration constrain CD4+ T cell HP during GVHD. However, it is not clear whether these alterations to the peripheral CD4+ T cell niche also contribute to impair CD8+ T cell regeneration during GVHD. We found that IL-7 therapy was sufficient for restoring CD8+ T cell HP in GVHD hosts while forcing DC regeneration with Flt3-L had only a modest effect on CD8+ T cell HP in IL-7 treated mice. Using bone marrow chimeras, we showed that HP of naïve CD8+ T cells is primarily regulated by MHC class I on radio-resistant stromal cells, yet optimal recovery of CD8+ T cell counts still requires expression of MHC class I on both radio-resistant and radio-sensitive hematopoietic cells. Thus, IL-7 level is the primary limiting factor that constrains naïve CD8+ T cell HP during GVHD, and accessibility of MHC class I on stromal cells explains how IL-7 therapy, as a single agent, can induce robust CD8 + T cell HP in the absence of DCs.
Collapse
Affiliation(s)
- Simon-David Gauthier
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Moutuaata M Moutuou
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Francis Daudelin
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Dominique Leboeuf
- Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada; Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Martin Guimond
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada; Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.
| |
Collapse
|
65
|
Gómez-Fernández P, Urtasun A, Paton AW, Paton JC, Borrego F, Dersh D, Argon Y, Alloza I, Vandenbroeck K. Long Interleukin-22 Binding Protein Isoform-1 Is an Intracellular Activator of the Unfolded Protein Response. Front Immunol 2018; 9:2934. [PMID: 30619294 PMCID: PMC6302113 DOI: 10.3389/fimmu.2018.02934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/29/2018] [Indexed: 12/26/2022] Open
Abstract
The human IL22RA2 gene co-produces three protein isoforms in dendritic cells [IL-22 binding protein isoform-1 (IL-22BPi1), IL-22BPi2, and IL-22BPi3]. Two of these, IL-22BPi2 and IL-22BPi3, are capable of neutralizing the biological activity of IL-22. The function of IL-22BPi1, which differs from IL-22BPi2 through an in-frame 32-amino acid insertion provided by an alternatively spliced exon, remains unknown. Using transfected human cell lines, we demonstrate that IL-22BPi1 is secreted detectably, but at much lower levels than IL-22BPi2, and unlike IL-22BPi2 and IL-22BPi3, is largely retained in the endoplasmic reticulum (ER). As opposed to IL-22BPi2 and IL-22BPi3, IL-22BPi1 is incapable of neutralizing or binding to IL-22 measured in bioassay or assembly-induced IL-22 co-folding assay. We performed interactome analysis to disclose the mechanism underlying the poor secretion of IL-22BPi1 and identified GRP78, GRP94, GRP170, and calnexin as main interactors. Structure-function analysis revealed that, like IL-22BPi2, IL-22BPi1 binds to the substrate-binding domain of GRP78 as well as to the middle domain of GRP94. Ectopic expression of wild-type GRP78 enhanced, and ATPase-defective GRP94 mutant decreased, secretion of both IL-22BPi1 and IL-22BPi2, while neither of both affected IL-22BPi3 secretion. Thus, IL-22BPi1 and IL-22BPi2 are bona fide clients of the ER chaperones GRP78 and GRP94. However, only IL-22BPi1 activates an unfolded protein response (UPR) resulting in increased protein levels of GRP78 and GRP94. Cloning of the IL22RA2 alternatively spliced exon into an unrelated cytokine, IL-2, bestowed similar characteristics on the resulting protein. We also found that CD14++/CD16+ intermediate monocytes produced a higher level of IL22RA2 mRNA than classical and non-classical monocytes, but this difference disappeared in immature dendritic cells (moDC) derived thereof. Upon silencing of IL22RA2 expression in moDC, GRP78 levels were significantly reduced, suggesting that native IL22RA2 expression naturally contributes to upregulating GRP78 levels in these cells. The IL22RA2 alternatively spliced exon was reported to be recruited through a single mutation in the proto-splice site of a Long Terminal Repeat retrotransposon sequence in the ape lineage. Our work suggests that positive selection of IL-22BPi1 was not driven by IL-22 antagonism as in the case of IL-22BPi2 and IL-22BPi3, but by capacity for induction of an UPR response.
Collapse
Affiliation(s)
- Paloma Gómez-Fernández
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Andoni Urtasun
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Adrienne W. Paton
- Research for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - James C. Paton
- Research for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - Francisco Borrego
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Basque Center for Transfusion and Human Tissues, Galdakao, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Devin Dersh
- Division of Cell Pathology, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yair Argon
- Division of Cell Pathology, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Iraide Alloza
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Koen Vandenbroeck
- Neurogenomiks Group, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
66
|
Abstract
Cytokines that control the immune response were shown to have efficacy in preclinical murine cancer models. Interferon (IFN)-α is approved for treatment of hairy cell leukemia, and interleukin (IL)-2 for the treatment of advanced melanoma and metastatic renal cancer. In addition, IL-12, IL-15, IL-21, and granulocyte macrophage colony-stimulating factor (GM-CSF) have been evaluated in clinical trials. However, the cytokines as monotherapy have not fulfilled their early promise because cytokines administered parenterally do not achieve sufficient concentrations in the tumor, are often associated with severe toxicities, and induce humoral or cellular checkpoints. To circumvent these impediments, cytokines are being investigated clinically in combination therapy with checkpoint inhibitors, anticancer monoclonal antibodies to increase the antibody-dependent cellular cytotoxicity (ADCC) of these antibodies, antibody cytokine fusion proteins, and anti-CD40 to facilitate tumor-specific immune responses.
Collapse
Affiliation(s)
- Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Clinical Center, Bethesda, Maryland 20892-1374
| |
Collapse
|
67
|
Molecular characterization and expression analysis of interleukin 15 (IL15) and interleukin-15 receptor subunit alpha (IL15Rα) in dojo loach (Misgurnus anguillicaudatus): Their salient roles during bacterial, parasitic and fungal infection. Mol Immunol 2018; 103:293-305. [DOI: 10.1016/j.molimm.2018.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/07/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022]
|
68
|
Finch DK, Stolberg VR, Ferguson J, Alikaj H, Kady MR, Richmond BW, Polosukhin VV, Blackwell TS, McCloskey L, Curtis JL, Freeman CM. Lung Dendritic Cells Drive Natural Killer Cytotoxicity in Chronic Obstructive Pulmonary Disease via IL-15Rα. Am J Respir Crit Care Med 2018; 198:1140-1150. [PMID: 29676596 PMCID: PMC6221577 DOI: 10.1164/rccm.201712-2513oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/19/2018] [Indexed: 02/02/2023] Open
Abstract
RATIONALE Lung natural killer cells (NKs) kill a greater percentage of autologous lung parenchymal cells in chronic obstructive pulmonary disease (COPD) than in nonobstructed smokers. To become cytotoxic, NKs require priming, typically by dendritic cells (DCs), but whether priming occurs in the lungs in COPD is unknown. METHODS We used lung tissue and in some cases peripheral blood from patients undergoing clinically indicated resections to determine in vitro killing of CD326+ lung epithelial cells by isolated lung CD56+ NKs. We also measured the cytotoxicity of unprimed blood NKs after preincubation with lung DCs. To investigate mechanisms of DC-mediated priming, we used murine models of COPD induced by cigarette smoke (CS) exposure or by polymeric immunoglobulin receptor (pIgR) deficiency, and blocked IL-15Rα (IL-15 receptor α subunit) trans-presentation by genetic and antibody approaches. RESULTS Human lung NKs killed isolated autologous lung epithelial cells; cytotoxicity was increased (P = 0.0001) in COPD, relative to smokers without obstruction. Similarly, increased lung NK cytotoxicity compared with control subjects was observed in CS-exposed mice and pIgR-/- mice. Blood NKs both from smokers without obstruction and subjects with COPD showed minimal epithelial cell killing, but in COPD, preincubation with lung DCs increased cytotoxicity. NKs were primed by CS-exposed murine DCs in vitro and in vivo. Inhibiting IL-15Rα trans-presentation eliminated NK priming both by murine CS-exposed DCs and by lung DCs from subjects with COPD. CONCLUSIONS Heightened NK cytotoxicity against lung epithelial cells in COPD results primarily from lung DC-mediated priming via IL-15 trans-presentation on IL-15Rα. Future studies are required to test whether increased NK cytotoxicity contributes to COPD pathogenesis.
Collapse
Affiliation(s)
- Donna K. Finch
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Valerie R. Stolberg
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - John Ferguson
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Henrih Alikaj
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Mohamed R. Kady
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Bradley W. Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell and Developmental Biology and
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Lisa McCloskey
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Jeffrey L. Curtis
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan; and
- Pulmonary and Critical Care Medicine Section, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Christine M. Freeman
- Research Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan; and
| |
Collapse
|
69
|
Beilin C, Choudhuri K, Bouma G, Malinova D, Llodra J, Stokes DL, Shimaoka M, Springer TA, Dustin ML, Thrasher AJ, Burns SO. Dendritic cell-expressed common gamma-chain recruits IL-15 for trans-presentation at the murine immunological synapse. Wellcome Open Res 2018; 3:84. [PMID: 30483599 PMCID: PMC6234741 DOI: 10.12688/wellcomeopenres.14493.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Mutations of the common cytokine receptor gamma chain (γc) cause Severe Combined Immunodeficiency characterized by absent T and NK cell development. Although stem cell therapy restores these lineages, residual immune defects are observed that may result from selective persistence of γc-deficiency in myeloid lineages. However, little is known about the contribution of myeloid-expressed γc to protective immune responses. Here we examine the importance of γc for myeloid dendritic cell (DC) function. Methods: We utilize a combination of in vitro DC/T-cell co-culture assays and a novel lipid bilayer system mimicking the T cell surface to delineate the role of DC-expressed γc during DC/T-cell interaction. Results: We observed that γc in DC was recruited to the contact interface following MHCII ligation, and promoted IL-15Rα colocalization with engaged MHCII. Unexpectedly, trans-presentation of IL-15 was required for optimal CD4+T cell activation by DC and depended on DC γc expression. Neither recruitment of IL-15Rα nor IL-15 trans-signaling at the DC immune synapse (IS), required γc signaling in DC, suggesting that γc facilitates IL-15 transpresentation through induced intermolecular cis associations or cytoskeletal reorganization following MHCII ligation. Conclusions: These findings show that DC-expressed γc is required for effective antigen-induced CD4+ T cell activation. We reveal a novel mechanism for recruitment of DC IL-15/IL-15Rα complexes to the IS, leading to CD4+ T cell costimulation through localized IL-15 transpresentation that is coordinated with antigen-recognition.
Collapse
Affiliation(s)
- Chiara Beilin
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Kaushik Choudhuri
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - Gerben Bouma
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Dessislava Malinova
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Jaime Llodra
- Program in Structural Biology, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - David L. Stokes
- Program in Structural Biology, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - Motumu Shimaoka
- Immune Disease Institute, Children's Hospital Boston, Boston, MA, 02115, USA
| | - Timothy A. Springer
- Program in Structural Biology, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - Michael L. Dustin
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, OX3 7FY, UK
| | - Adrian J. Thrasher
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Siobhan O. Burns
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
- University College London Institute of Immunity and Transplantation, Department of Immunology, Royal Free London NHS Foundation Trust, London, NW3 2PF, UK
| |
Collapse
|
70
|
Gupta R, Yan XJ, Barrientos J, Kolitz JE, Allen SL, Rai K, Chiorazzi N, Mongini PKA. Mechanistic Insights into CpG DNA and IL-15 Synergy in Promoting B Cell Chronic Lymphocytic Leukemia Clonal Expansion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1570-1585. [PMID: 30068596 PMCID: PMC6103916 DOI: 10.4049/jimmunol.1800591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/26/2018] [Indexed: 12/30/2022]
Abstract
Malignant cell growth within patients with B cell chronic lymphocytic leukemia (B-CLL) is largely restricted to lymphoid tissues, particularly lymph nodes. The recent in vitro finding that TLR-9 ligand (oligodeoxynucleotide [ODN]) and IL-15 exhibit strong synergy in promoting B-CLL growth may be particularly relevant to growth in these sites. This study shows IL-15-producing cells are prevalent within B-CLL-infiltrated lymph nodes and, using purified B-CLL cells from blood, investigates the mechanism for ODN and IL-15 synergy in driving B-CLL growth. ODN boosts baseline levels of phospho-RelA(S529) in B-CLL and promotes NF-κB-driven increases in IL15RA and IL2RB mRNA, followed by elevated IL-15Rα and IL-2/IL-15Rβ (CD122) protein. IL-15→CD122 signaling during a critical interval, 20 to 36-48 h following initial ODN exposure, is required for optimal induction of the cycling process. Furthermore, experiments with neutralizing anti-IL-15 and anti-CD122 mAbs indicate that clonal expansion requires continued IL-15/CD122 signaling during cycling. The latter is consistent with evidence of heightened IL2RB mRNA in the fraction of recently proliferated B-CLL cells within patient peripheral blood. Compromised ODN+IL-15 growth with limited cell density is consistent with a role for upregulated IL-15Rα in facilitating homotypic trans IL-15 signaling, although there may be other explanations. Together, the findings show that ODN and IL-15 elicit temporally distinct signals that function in a coordinated manner to drive B-CLL clonal expansion.
Collapse
Affiliation(s)
- Rashmi Gupta
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Xiao J Yan
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Jacqueline Barrientos
- Department of Medicine, North Shore University Hospital-Long Island Jewish Medical Center, Northwell Health, Manhasset, NY 11303
| | - Jonathan E Kolitz
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
- Department of Medicine, North Shore University Hospital-Long Island Jewish Medical Center, Northwell Health, Manhasset, NY 11303
- Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549; and
| | - Steven L Allen
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
- Department of Medicine, North Shore University Hospital-Long Island Jewish Medical Center, Northwell Health, Manhasset, NY 11303
- Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549; and
| | - Kanti Rai
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
- Department of Medicine, North Shore University Hospital-Long Island Jewish Medical Center, Northwell Health, Manhasset, NY 11303
- Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549; and
| | - Nicholas Chiorazzi
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
- Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549; and
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549
| | - Patricia K A Mongini
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030;
| |
Collapse
|
71
|
Beilin C, Choudhuri K, Bouma G, Malinova D, Llodra J, Stokes DL, Shimaoka M, Springer TA, Dustin ML, Thrasher AJ, Burns SO. Dendritic cell-expressed common gamma-chain recruits IL-15 for trans-presentation at the murine immunological synapse. Wellcome Open Res 2018; 3:84. [PMID: 30483599 PMCID: PMC6234741 DOI: 10.12688/wellcomeopenres.14493.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2018] [Indexed: 10/05/2023] Open
Abstract
Background: Mutations of the common cytokine receptor gamma chain (γc) cause Severe Combined Immunodeficiency characterized by absent T and NK cell development. Although stem cell therapy restores these lineages, residual immune defects are observed that may result from selective persistence of γc-deficiency in myeloid lineages. However, little is known about the contribution of myeloid-expressed γc to protective immune responses. Here we examine the importance of γc for myeloid dendritic cell (DC) function. Methods: We utilize a combination of in vitro DC/T-cell co-culture assays and a novel lipid bilayer system mimicking the T cell surface to delineate the role of DC-expressed γc during DC/T-cell interaction. Results: We observed that γc in DC was recruited to the contact interface following MHCII ligation, and promoted IL-15Rα colocalization with engaged MHCII. Unexpectedly, trans-presentation of IL-15 was required for optimal CD4+T cell activation by DC and depended on DC γc expression. Neither recruitment of IL-15Rα nor IL-15 trans-signaling at the DC immune synapse (IS), required γc signaling in DC, suggesting that γc facilitates IL-15 transpresentation through induced intermolecular cis associations or cytoskeletal reorganization following MHCII ligation. Conclusions: These findings show that DC-expressed γc is required for effective antigen-induced CD4+ T cell activation. We reveal a novel mechanism for recruitment of DC IL-15/IL-15Rα complexes to the IS, leading to CD4+ T cell costimulation through localized IL-15 transpresentation that is coordinated with antigen-recognition.
Collapse
Affiliation(s)
- Chiara Beilin
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Kaushik Choudhuri
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - Gerben Bouma
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Dessislava Malinova
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Jaime Llodra
- Program in Structural Biology, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - David L. Stokes
- Program in Structural Biology, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - Motumu Shimaoka
- Immune Disease Institute, Children's Hospital Boston, Boston, MA, 02115, USA
| | - Timothy A. Springer
- Program in Structural Biology, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
| | - Michael L. Dustin
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York University, New York, NY, 10016, USA
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, OX3 7FY, UK
| | - Adrian J. Thrasher
- Molecular Immunology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Siobhan O. Burns
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
- University College London Institute of Immunity and Transplantation, Department of Immunology, Royal Free London NHS Foundation Trust, London, NW3 2PF, UK
| |
Collapse
|
72
|
Frutoso M, Morisseau S, Tamzalit F, Quéméner A, Meghnem D, Leray I, Jacques Y, Mortier E. Emergence of NK Cell Hyporesponsiveness after Two IL-15 Stimulation Cycles. THE JOURNAL OF IMMUNOLOGY 2018; 201:493-506. [PMID: 29848756 DOI: 10.4049/jimmunol.1800086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022]
Abstract
IL-15 is a cytokine playing a crucial role in the function of immune cells, including NK and CD8 T cells. In this study, we demonstrated that in vivo, in mice, IL-15-prestimulated NK cells were no longer able to respond to a second cycle of IL-15 stimulation. This was illustrated by defects in cell maturation, proliferation, and activation, seemingly linked to the environment surrounding NK cells but not related to the presence of CD4 regulatory T cells, TGF-β, or IL-10. Moreover, NK cells from immunodeficient mice could respond to two cycles of IL-15 stimulation, whereas an adoptive transfer of CD44+CD8+ cells impaired their responsiveness to the second cycle. Conversely, in immunocompetent mice, NK cell responsiveness to a second IL-15 stimulation was restored by the depletion of CD8+ cells. These biological findings refine our understanding of the complex mode of action of NK cells in vivo, and they should be taken into consideration for IL-15-based therapy.
Collapse
Affiliation(s)
- Marie Frutoso
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| | - Sébastien Morisseau
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and.,Centre Hospitalier Universitaire, 44000 Nantes, France
| | - Fella Tamzalit
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| | - Agnès Quéméner
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| | - Dihia Meghnem
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| | - Isabelle Leray
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| | - Yannick Jacques
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| | - Erwan Mortier
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, INSERM, Université de Nantes, 44007 Nantes, France; and
| |
Collapse
|
73
|
Beffinger M, Tallón de Lara P, Tugues S, Vermeer M, Montagnolo Y, Ohs I, Cecconi V, Lucchiari G, Gagliardi A, Misljencevic N, Sutton J, Spörri R, Becher B, Gupta A, van den Broek M. CSF1R-dependent myeloid cells are required for NK‑mediated control of metastasis. JCI Insight 2018; 3:97792. [PMID: 29769439 DOI: 10.1172/jci.insight.97792] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/13/2018] [Indexed: 12/20/2022] Open
Abstract
Myeloid leukocytes are essentially involved in both tumor progression and control. We show that neo-adjuvant treatment of mice with an inhibitor of CSF1 receptor (CSF1R), a drug that is used to deplete tumor-associated macrophages, unexpectedly promoted metastasis. CSF1R blockade indirectly diminished the number of NK cells due to a paucity of myeloid cells that provide the survival factor IL-15 to NK cells. Reduction of the number of NK cells resulted in increased seeding of metastatic tumor cells to the lungs but did not impact on progression of established metastases. Supplementation of mice treated with CSF1R-inhibitor with IL-15 restored numbers of NK cells and diminished metastasis. Our data suggest that CSF1R blockade should be combined with administration of IL-15 to reduce the risk of metastasis.
Collapse
Affiliation(s)
- Michal Beffinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Sònia Tugues
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marijne Vermeer
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Yannick Montagnolo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Isabel Ohs
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Giulia Lucchiari
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Aron Gagliardi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Nikola Misljencevic
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - James Sutton
- Novartis Institutes for Biomedical Research, Emeryville, California, USA
| | - Roman Spörri
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Anurag Gupta
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
74
|
Bergamaschi C, Watson DC, Valentin A, Bear J, Peer CJ, Figg WD, Felber BK, Pavlakis GN. Optimized administration of hetIL-15 expands lymphocytes and minimizes toxicity in rhesus macaques. Cytokine 2018; 108:213-224. [PMID: 29402721 PMCID: PMC6657354 DOI: 10.1016/j.cyto.2018.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
The common γ-chain cytokine interleukin-15 (IL-15) plays a significant role in regulating innate and adaptive lymphocyte homeostasis and can stimulate anti-tumor activity of leukocytes. We have previously shown that the circulating IL-15 in the plasma is the heterodimeric form (hetIL-15), produced upon co-expression of IL-15 and IL-15 Receptor alpha (IL-15Rα) polypeptides in the same cell, heterodimerization of the two chains and secretion. We investigated the pharmacokinetic and pharmacodynamic profile and toxicity of purified human hetIL-15 cytokine upon injection in rhesus macaques. We compared the effects of repeated hetIL-15 administration during a two-week dosing cycle, using different subcutaneous dosing schemata, i.e. fixed doses of 0.5, 5 and 50 μg/kg or a doubling step-dose scheme ranging from 2 to 64 μg/kg. Following a fixed-dose regimen, dose-dependent peak plasma IL-15 levels decreased significantly between the first and last injection. The trough plasma IL-15 levels measured at 48 h after injections were significantly higher after the first dose, compared to subsequent doses. In contrast, following the step-dose regimen, the systemic exposure increased by more than 1 log between the first injection given at 2 μg/kg and the last injection given at 64 μg/kg, and the trough levels were comparable after each injection. Blood lymphocyte cell count, proliferation, and plasma IL-18 levels peaked at day 8 when hetIL-15 was provided at fixed doses, and at the end of the cycle following a step-dose regimen, suggesting that sustained expansion of target cells requires increasing doses of cytokine. Macaques treated with a 50 μg/kg dose showed moderate and transient toxicity, including fever, signs of capillary leak syndrome and renal dysfunction. In contrast, these effects were mild or absent using the step-dose regimen. The results provide a new method of optimal administration of this homeostatic cytokine and may have applications for the delivery of other cytokines.
Collapse
Affiliation(s)
- Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Dionysios C Watson
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Cody J Peer
- Clinical Pharmacology Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - William D Figg
- Clinical Pharmacology Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA.
| |
Collapse
|
75
|
Bálint Š, Lopes FB, Davis DM. A nanoscale reorganization of the IL-15 receptor is triggered by NKG2D in a ligand-dependent manner. Sci Signal 2018; 11:11/525/eaal3606. [PMID: 29636390 DOI: 10.1126/scisignal.aal3606] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Natural killer group 2D (NKG2D), an activating receptor on natural killer (NK) cells and a subset of T cells, recognizes stress-inducible proteins, including MICA and ULBP2, which are present on infected or transformed cells. Whether each NKG2D ligand (NKG2DL) has a distinct biological role is not clear. Using superresolution microscopy, we found that NKG2D is constitutively arranged in nanoclusters at the surface of human primary NK cells. Nanoclusters of NKG2D became smaller upon ligation with MICA but became larger upon activation by ULBP2. In addition, ULBP2 induced the reorganization of nanoclusters of the cytokine receptor subunit for both interleukin-2 (IL-2) and IL-15 (IL-2/IL-15Rβ), such that these cytokine receptor subunits coalesced with nanoclusters of NKG2D. Functionally, the response of NK cells activated by ULBP2 was augmented by an interaction between ULBP2-bound NKG2D and IL-15R ligated by IL-15 (trans-presented by IL-15Rα-coated surfaces). These data suggest that NKG2DLs are not equivalent in their capacity to activate NKG2D and establish a previously unknown paradigm in how ligand-induced changes to the nanoscale organization of the cell surface can affect immune responses.
Collapse
Affiliation(s)
- Štefan Bálint
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Filipa B Lopes
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK.
| |
Collapse
|
76
|
Keating N, Nicholson SE. SOCS-mediated immunomodulation of natural killer cells. Cytokine 2018; 118:64-70. [PMID: 29609875 DOI: 10.1016/j.cyto.2018.03.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 10/17/2022]
Abstract
Natural killer (NK) cells are innate immune cells with an intrinsic ability to detect and kill infected and cancerous cells. The success of therapies targeting immune checkpoints on CD8 cells has intensified interest in harnessing the cytolytic effector functions of NK cells for new cancer treatments. NK cell development, survival and effector activity is dependent on exposure to the cytokine interleukin (IL)-15. The suppressor of cytokine (SOCS) proteins (CIS; SOCS1-7) are important negative regulators of cytokine signaling, and both CIS and SOCS2 are reported to have roles in regulating NK cell responses. Their immunomodulatory effects on NK cells suggest that these SOCS proteins are promising targets that can potentially form the basis of novel cancer therapies. Here we discuss the role of NK cells in tumor immunity as well as review the role of the SOCS proteins in regulating IL-15 signaling and NK cell function.
Collapse
Affiliation(s)
- Narelle Keating
- Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, Australia
| | - Sandra E Nicholson
- Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, University of Melbourne, Melbourne 3010, Australia.
| |
Collapse
|
77
|
Huang H, Luo Y, Baradei H, Liu S, Haenssen KK, Sanglikar S, Kumar S, Cini J. A novel strategy to produce high level and high purity of bioactive IL15 fusion proteins from mammalian cells. Protein Expr Purif 2018; 148:30-39. [PMID: 29596991 DOI: 10.1016/j.pep.2018.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 01/28/2023]
Abstract
IL15, a member of the common γ chain receptor (γc) cytokine family, is gaining attention in recent years as one of the most promising anti-tumor agents. IL15 regulates T cell activation and proliferation, promotes the survival of CD8+ CD44hi memory T cells and is also essential for NK cell expansion and development. Despite the attraction of developing IL15 as an anti-cancer agent, production of recombinant IL15 has proven to be difficult due to the stringent control of IL15 expression at the transcriptional, translational and the post-translational levels. Furthermore, the bioactivity of IL15 fused to an extra functional domain that is isolated from mammalian cells is generally inferior to recombinant IL15 produced by E. coli. In this study, we report that Lysine 86 in IL15 is responsible for the instability in mammalian cells when its C-terminus is fused to the albumin binding scFv (IL15-A10m3). We demonstrate that K86A or K86R mutants increased the expression of the fusion protein from HEK293 cells. When the wild type IL15 is used for the fusion, no recombinant IL15 fusion was detected in the culture media. Additionally, we determined that the residue 112 in IL15 is critical for the bioactivity of IL15-A10m3. Examination of single and double mutants provides a better understanding of how IL15 engages with its receptor complex to achieve full signaling capacity. The results of our experiments were successfully applied to scale up production to levels up to 50 mg/L and >10 mg/L of >95% pure monomeric recombinant fusion proteins after a 2-step purification from culture media. More importantly, the recombinant fusion protein produced is fully active in stimulating T cell proliferation, when compared to the recombinant wild type IL15.
Collapse
Affiliation(s)
- Haomin Huang
- Sonnet Biotherapeutics, 1 Duncan Drive, Cranbury, NJ 08512, USA.
| | - Yuying Luo
- Sonnet Biotherapeutics, 1 Duncan Drive, Cranbury, NJ 08512, USA
| | - Hanna Baradei
- Sonnet Biotherapeutics, 1 Duncan Drive, Cranbury, NJ 08512, USA
| | - Shan Liu
- Sonnet Biotherapeutics, 1 Duncan Drive, Cranbury, NJ 08512, USA
| | | | | | - Senthil Kumar
- Sonnet Biotherapeutics, 1 Duncan Drive, Cranbury, NJ 08512, USA
| | - John Cini
- Sonnet Biotherapeutics, 1 Duncan Drive, Cranbury, NJ 08512, USA
| |
Collapse
|
78
|
Al-Attar A, Presnell SR, Clasey JL, Long DE, Walton RG, Sexton M, Starr ME, Kern PA, Peterson CA, Lutz CT. Human Body Composition and Immunity: Visceral Adipose Tissue Produces IL-15 and Muscle Strength Inversely Correlates with NK Cell Function in Elderly Humans. Front Immunol 2018; 9:440. [PMID: 29559978 PMCID: PMC5845694 DOI: 10.3389/fimmu.2018.00440] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/19/2018] [Indexed: 01/11/2023] Open
Abstract
Natural killer (NK) lymphocyte-mediated cytotoxicity and cytokine secretion control infections and cancers, but these crucial activities decline with age. NK cell development, homeostasis, and function require IL-15 and its chaperone, IL-15 receptor alpha (IL-15Rα). Macrophages and dendritic cells (DC) are major sources of these proteins. We had previously postulated that additional IL-15 and IL-15Rα is made by skeletal muscle and adipose tissue. These sources may be important in aging, when IL-15-producing immune cells decline. NK cells circulate through adipose tissue, where they may be exposed to local IL-15. The objectives of this work were to determine (1) if human muscle, subcutaneous adipose tissue (SAT), and visceral adipose tissue (VAT) are sources of IL-15 and IL-15 Rα, and (2) whether any of these tissues correlate with NK cell activity in elderly humans. We first investigated IL-15 and IL-15Rα RNA expression in paired muscle and SAT biopsies from healthy human subjects. Both tissues expressed these transcripts, but IL-15Rα RNA levels were higher in SAT than in skeletal muscle. We also investigated tissue obtained from surgeries and found that SAT and VAT expressed equivalent amounts of IL-15 and IL-15Rα RNA, respectively. Furthermore, stromal vascular fraction cells expressed more IL-15 RNA than did adipocytes. To test if these findings related to circulating IL-15 protein and NK cell function, we tested 50 healthy adults aged > 70 years old. Plasma IL-15 levels significantly correlated with abdominal VAT mass in the entire cohort and in non-obese subjects. However, plasma IL-15 levels did not correlate with skeletal muscle cross-sectional area and correlated inversely with muscle strength. Plasma IL-15 did correlate with NK cell cytotoxic granule exocytosis and with CCL4 (MIP-1β) production in response to NKp46-crosslinking. Additionally, NK cell responses to K562 leukemia cells correlated inversely with muscle strength. With aging, immune function declines while infections, cancers, and deaths increase. We propose that VAT-derived IL-15 and IL-15Rα is a compensatory NK cell support mechanism in elderly humans.
Collapse
Affiliation(s)
- Ahmad Al-Attar
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Steven R. Presnell
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Jody L. Clasey
- Department of Kinesiology and Health Promotion, College of Education, University of Kentucky, Lexington, KY, United States
| | - Douglas E. Long
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - R. Grace Walton
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - Morgan Sexton
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Marlene E. Starr
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Philip A. Kern
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Charlotte A. Peterson
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - Charles T. Lutz
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
79
|
Wagstaffe HR, Nielsen CM, Riley EM, Goodier MR. IL-15 Promotes Polyfunctional NK Cell Responses to Influenza by Boosting IL-12 Production. THE JOURNAL OF IMMUNOLOGY 2018; 200:2738-2747. [PMID: 29491009 PMCID: PMC5890538 DOI: 10.4049/jimmunol.1701614] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/31/2018] [Indexed: 11/19/2022]
Abstract
IL-15 is a key regulator of NK cell maintenance and proliferation and synergizes with other myeloid cell–derived cytokines to enhance NK cell effector function. At low concentrations, trans-presentation of IL-15 by dendritic cells can activate NK cells, whereas at higher concentrations it can act directly on NK cells, independently of accessory cells. In this study, we investigate the potential for IL-15 to boost responses to influenza virus by promoting accessory cell function. We find that coculture of human PBMCs with inactivated whole influenza virus (A/Victoria/361/2011) in the presence of very low concentrations of IL-15 results in increased production of myeloid cell–derived cytokines, including IL-12, IFN-α2, GM-CSF, and IL-1β, and an increased frequency of polyfunctional NK cells (defined by the expression of two or more of CD107a, IFN-γ, and CD25). Neutralization experiments demonstrate that IL-15–mediated enhancement of NK cell responses is primarily dependent on IL-12 and partially dependent on IFN-αβR1 signaling. Critically, IL-15 boosted the production of IL-12 in influenza-stimulated blood myeloid dendritic cells. IL-15 costimulation also restored the ability of less-differentiated NK cells from human CMV-seropositive individuals to respond to influenza virus. These data suggest that very low concentrations of IL-15 play an important role in boosting accessory cell function to support NK cell effector functions.
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Carolyn M Nielsen
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Eleanor M Riley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom
| | - Martin R Goodier
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom;
| |
Collapse
|
80
|
Watson DC, Moysi E, Valentin A, Bergamaschi C, Devasundaram S, Fortis SP, Bear J, Chertova E, Bess J, Sowder R, Venzon DJ, Deleage C, Estes JD, Lifson JD, Petrovas C, Felber BK, Pavlakis GN. Treatment with native heterodimeric IL-15 increases cytotoxic lymphocytes and reduces SHIV RNA in lymph nodes. PLoS Pathog 2018; 14:e1006902. [PMID: 29474450 PMCID: PMC5825155 DOI: 10.1371/journal.ppat.1006902] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
B cell follicles in secondary lymphoid tissues represent an immune privileged sanctuary for AIDS viruses, in part because cytotoxic CD8+ T cells are mostly excluded from entering the follicles that harbor infected T follicular helper (TFH) cells. We studied the effects of native heterodimeric IL-15 (hetIL-15) treatment on uninfected rhesus macaques and on macaques that had spontaneously controlled SHIV infection to low levels of chronic viremia. hetIL-15 increased effector CD8+ T lymphocytes with high granzyme B content in blood, mucosal sites and lymph nodes, including virus-specific MHC-peptide tetramer+ CD8+ cells in LN. Following hetIL-15 treatment, multiplexed quantitative image analysis (histo-cytometry) of LN revealed increased numbers of granzyme B+ T cells in B cell follicles and SHIV RNA was decreased in plasma and in LN. Based on these properties, hetIL-15 shows promise as a potential component in combination immunotherapy regimens to target AIDS virus sanctuaries and reduce long-term viral reservoirs in HIV-1 infected individuals. TRIAL REGISTRATION ClinicalTrials.gov NCT02452268.
Collapse
Affiliation(s)
- Dionysios C. Watson
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Eirini Moysi
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section; Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Santhi Devasundaram
- Human Retrovirus Pathogenesis Section; Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Sotirios P. Fortis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section; Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Elena Chertova
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Julian Bess
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ray Sowder
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - David J. Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, Maryland, United States of America
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Constantinos Petrovas
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section; Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| |
Collapse
|
81
|
Nicholson SE, Keating N, Belz GT. Natural killer cells and anti-tumor immunity. Mol Immunol 2017; 110:40-47. [PMID: 29233542 DOI: 10.1016/j.molimm.2017.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/20/2017] [Accepted: 12/01/2017] [Indexed: 01/10/2023]
Abstract
Immune checkpoint inhibitors harness the power of the immune system to fight cancer. The clinical success achieved with antibodies against the inhibitory T cell receptors PD-1 and CTLA4 has focused attention on the possibility of manipulating other immune cells, in particular those involved in innate immunity. Here we review the role of innate lymphoid cells (ILCs) and their contribution to tumor immunity. As the prototypical ILC, the natural killer (NK) cell has an intrinsic ability to detect and kill cancer cells. NK cells are dependent on the cytokine interleukin (IL)-15 for their development and effector activity. We discuss the role of the Suppressor of cytokine (SOCS) proteins in negatively regulating IL-15 and NK cell responses and the potential for targeting these small intracellular regulators as new immune checkpoints.
Collapse
Affiliation(s)
- Sandra E Nicholson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; and Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia.
| | - Narelle Keating
- Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; and Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; and Department of Medical Biology, University of Melbourne, Melbourne, 3010, Australia.
| |
Collapse
|
82
|
Inflammatory Ly6C high Monocytes Protect against Candidiasis through IL-15-Driven NK Cell/Neutrophil Activation. Immunity 2017. [PMID: 28636955 DOI: 10.1016/j.immuni.2017.05.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neutrophils play a crucial role in defense against systemic candidiasis, a disease associated with a high mortality rate in patients receiving immunosuppressive therapy, although the early immune mechanisms that boost the candidacidal activity of neutrophils remain to be defined in depth. Here, we used a murine model of systemic candidiasis to explore the role of inflammatory Ly6Chigh monocytes in NK cell-mediated neutrophil activation during the innate immune response against C. albicans. We found that efficient anti-Candida immunity required a collaborative response between the spleen and kidney, which relied on type I interferon-dependent IL-15 production by spleen inflammatory Ly6Chigh monocytes to drive efficient activation and GM-CSF release by spleen NK cells; this in turn was necessary to boost the Candida killing potential of kidney neutrophils. Our findings unveil a role for IL-15 as a critical mediator in defense against systemic candidiasis and hold promise for the design of IL-15-based antifungal immunotherapies.
Collapse
|
83
|
Abstract
Dendritic cells (DCs) play critical roles in activating innate immune cells and initiating adaptive immune responses. The functions of DCs were originally obscured by their overlap with other mononuclear phagocytes, but new mouse models have allowed for the selective ablation of subsets of DCs and have helped to identify their non-redundant roles in the immune system. These tools have elucidated the functions of DCs in host defense against pathogens, autoimmunity, and cancer. This review will describe the mouse models generated to interrogate the role of DCs and will discuss how their use has progressively clarified our understanding of the unique functions of DC subsets.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
84
|
Guo Y, Luan L, Patil NK, Sherwood ER. Immunobiology of the IL-15/IL-15Rα complex as an antitumor and antiviral agent. Cytokine Growth Factor Rev 2017; 38:10-21. [PMID: 28888485 DOI: 10.1016/j.cytogfr.2017.08.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-15 is essential for natural killer (NK), NKT and memory (m) CD8+ T cell development and function, and is currently under investigation as an immunotherapeutic agent for the treatment of cancer. Recently, the creation of IL-15 superagonist by complexing IL-15 and its high affinity receptor alpha (IL-15 Rα) in solution, inspired by the natural trans-presentation of IL-15, advances the potential of IL-15-based tumor immunotherapy. IL-15 superagonist shows promising advantages over monomeric IL-15 such as sustaining high circulating concentrations due to prolonged half-life and more potently stimulating NK and CD8+ T effector lymphocytes. So far, there are three different forms of recombinant IL-15 superagonist fusion protein based on configurational modifications. Gene therapy using engineered cells co-expressing IL-15/IL-15 Rα complex for cancer treatment is also emerging. All forms have demonstrated efficacy in causing tumor regression in animal studies, which provides strong rationale for advancing IL-15 superagonist through clinical trials. To date, there are fourteen phase I/II IL-15 superagonist trials in cancer patients and one phase I trial in HIV patients. Information generated by ongoing trials regarding the toxicity and efficacy of IL-15 superagonist is awaited. Finally, we elaborate on immunotoxicity caused by IL-15 superagonist in preclinical studies and discuss important safety considerations.
Collapse
Affiliation(s)
- Yin Guo
- Departments of Pathology, Microbiology and Immunology. Vanderbilt University, Nashville, TN, USA; Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Liming Luan
- Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naeem K Patil
- Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA
| | - Edward R Sherwood
- Departments of Pathology, Microbiology and Immunology. Vanderbilt University, Nashville, TN, USA; Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
85
|
Saunderson SC, McLellan AD. Role of Lymphocyte Subsets in the Immune Response to Primary B Cell-Derived Exosomes. THE JOURNAL OF IMMUNOLOGY 2017; 199:2225-2235. [PMID: 28842467 DOI: 10.4049/jimmunol.1601537] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 07/26/2017] [Indexed: 12/15/2022]
Abstract
Exosomes are lipid nanovesicles released after fusion of the endosomal limiting membrane with the plasma membrane. In this study, we investigated the requirement for CD4 T cells, B cells, and NK cells to provide help for CD8 T cell-mediated response to B cell-derived exosomes. CTL responses to Ag-loaded exosomes were dependent on host MHC class I, with a critical role for splenic langerin+ CD8α+ dendritic cells (DCs) in exosomal Ag cross-presentation. In addition, there was an absolute dependence on the presence of CD4 T cells, CD8 T cells, and NK cells, where the loss of any one of these subsets led to a complete loss of CTL response. Interestingly, NK cell depletion experiments demonstrated a critical cutoff point for depletion efficacy, with low-level residual NK cells providing sufficient help to allow optimal CD8 T cell proliferative responses to exosomal protein. Despite the potential role for B cells in the response to B cell-derived exosomal proteins, B cell depletion did not alter the exosome-induced CTL response. Similarly, a possible role for the BCR or circulating Ab in mediating CTL responses to B cell-derived exosomes was ruled out using DHLMP2A mice, which lack secreted and membrane-bound Ab, yet harbor marginal zone and follicular B cells. In contrast, CTL responses to DC-derived exosomes were significantly inhibited within Ab-deficient DHLMP2A mice compared with wild-type mice. However, this response was not restored upon serum transfer, implicating a role for the BCR, but not circulating Ab, in DC-derived exosome responses.
Collapse
Affiliation(s)
- Sarah C Saunderson
- Department of Microbiology and Immunology, University of Otago, Dunedin 9010, Otago, New Zealand
| | - Alexander D McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin 9010, Otago, New Zealand
| |
Collapse
|
86
|
Stabile H, Nisti P, Peruzzi G, Fionda C, Pagliara D, Brescia PL, Merli P, Locatelli F, Santoni A, Gismondi A. Reconstitution of multifunctional CD56 lowCD16 low natural killer cell subset in children with acute leukemia given α/β T cell-depleted HLA-haploidentical haematopoietic stem cell transplantation. Oncoimmunology 2017; 6:e1342024. [PMID: 28932646 DOI: 10.1080/2162402x.2017.1342024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/06/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022] Open
Abstract
We recently described the CD56lowCD16low subset of Natural Killer (NK) cells that both mediate cytotoxic activity and produce IFNγ, being more abundant in bone marrow (BM) than in peripheral blood (PB) of pediatric normal subjects. Given the multifunctional properties of this subset, we examined its development and functional recovery in a cohort of children undergoing α/β T-cell depleted HLA-haploidentical haematopoietic stem cell transplantation (HSCT). The results obtained indicate that CD56lowCD16low NK cells are present in both PB and BM already at one month post-HSCT, with an increased frequency in BM of graft recipients as compared with normal subjects. During the first 6 months after HSCT, no difference in CD56lowCD16low NK cells distribution between PB and BM was observed. In comparison to normal subjects, CD56lowCD16low NK cells from transplanted patients show lower expression levels of CD25 and CD127 and higher levels of CD122, and accordingly, produce higher amounts of IFNγ after stimulation with IL-12 plus IL-15. The recovery of NK-cell cytotoxicity after HSCT was strictly restricted to CD56lowCD16low NK cells, and their ability to degranulate against K562 target cells or autologous leukemic blasts was completely restored only one year after HSCT. Based on the phenotypic and functional ability of reconstituted CD56lowCD16low NK cells, we suggest that they play an important role in host defense against leukemia relapse and infections after HSCT, and represent an ideal candidate for approaches of adoptive immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Daria Pagliara
- Dept. Pediatric Hematology/Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome
| | - Pomonia Letizia Brescia
- Dept. Pediatric Hematology/Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome
| | - Pietro Merli
- Dept. Pediatric Hematology/Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome
| | - Franco Locatelli
- Dept. Pediatric Hematology/Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome.,University of Pavia, Italy
| | | | - Angela Gismondi
- Dept. Molecular Medicine.,Eleonora Lorillard Spencer Cenci Foundation
| |
Collapse
|
87
|
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: 192] [Impact Index Per Article: 27.4] [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
|
88
|
Van den Bergh JMJ, Smits ELJM, Versteven M, De Reu H, Berneman ZN, Van Tendeloo VFI, Lion E. Characterization of Interleukin-15-Transpresenting Dendritic Cells for Clinical Use. J Immunol Res 2017; 2017:1975902. [PMID: 28785596 PMCID: PMC5530419 DOI: 10.1155/2017/1975902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/04/2017] [Indexed: 11/17/2022] Open
Abstract
Personalized dendritic cell- (DC-) based vaccination has proven to be safe and effective as second-line therapy against various cancer types. In terms of overall survival, there is still room for improvement of DC-based therapies, including the development of more immunostimulatory DC vaccines. In this context, we redesigned our currently clinically used DC vaccine generation protocol to enable transpresentation of interleukin- (IL-) 15 to IL-15Rβγ-expressing cells aiming at boosting the antitumor immune response. In this study, we demonstrate that upon electroporation with both IL-15 and IL-15Rα-encoding messenger RNA, mature DC become highly positive for surface IL-15, without influencing the expression of prototypic mature DC markers and with preservation of their cytokine-producing capacity and their migratory profile. Functionally, we show that IL-15-transpresenting DC are equal if not better inducers of T-cell proliferation and are superior in tumor antigen-specific T-cell activation compared with DC without IL-15 conditioning. In view of the clinical use of DC vaccines, we evidence with a time- and cost-effective manner that clinical grade DC can be safely engineered to transpresent IL-15, hereby gaining the ability to transfer the immune-stimulating IL-15 signal towards antitumor immune effector cells.
Collapse
Affiliation(s)
- J. M. J. Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - E. L. J. M. Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
- Center for Oncological Research Antwerp, Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - M. Versteven
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - H. De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Z. N. Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - V. F. I. Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - E. Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
89
|
Lemire P, Galbas T, Thibodeau J, Segura M. Natural Killer Cell Functions during the Innate Immune Response to Pathogenic Streptococci. Front Microbiol 2017; 8:1196. [PMID: 28706510 PMCID: PMC5489694 DOI: 10.3389/fmicb.2017.01196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/12/2017] [Indexed: 01/21/2023] Open
Abstract
Dendritic cells (DCs) and NK cells play a crucial role in the first phase of host defense against infections. Group B Streptococcus (GBS) and Streptococcus suis are encapsulated streptococci causing severe systemic inflammation, leading to septicemia and meningitis. Yet, the involvement of NK cells in the innate immune response to encapsulated bacterial infection is poorly characterized. Here, it was observed that these two streptococcal species rapidly induce the release of IFN-γ and that NK cells are the major cell type responsible for this production during the acute phase of the infection. Albeit S. suis capacity to activate NK cells was lower than that of GBS, these cells partially contribute to S. suis systemic infection; mainly through amplification of the inflammatory loop. In contrast, such a role was not observed during GBS systemic infection. IFN-γ release by NK cells required the presence of DCs, which in turn had a synergistic effect on DC cytokine production. These responses were mainly mediated by direct DC-NK cell contact and partially dependent on soluble factors. Though IL-12 and LFA-1 were shown to be critical in S. suis-mediated activation of the DC-NK cell crosstalk, different or redundant molecular pathways modulate DC-NK interactions during GBS infection. The bacterial capsular polysaccharides also differently modulated NK cell activation. Together, these results demonstrated a role of NK cells in the innate immune response against encapsulated streptococcal infections; yet the molecular pathways governing NK activation seem to differ upon the pathogen and should not be generalized when studying bacterial infections.
Collapse
Affiliation(s)
- Paul Lemire
- Laboratory of Immunology of the Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of MontrealSt-Hyacinthe, QC, Canada
| | - Tristan Galbas
- Laboratory of Molecular Immunology, Faculty of Medicine, University of MontrealMontreal, QC, Canada
| | - Jacques Thibodeau
- Laboratory of Molecular Immunology, Faculty of Medicine, University of MontrealMontreal, QC, Canada
| | - Mariela Segura
- Laboratory of Immunology of the Swine and Poultry Infectious Diseases Research Centre, Faculty of Veterinary Medicine, University of MontrealSt-Hyacinthe, QC, Canada
| |
Collapse
|
90
|
Meghnem D, Morisseau S, Frutoso M, Trillet K, Maillasson M, Barbieux I, Khaddage S, Leray I, Hildinger M, Quéméner A, Jacques Y, Mortier E. Cutting Edge: Differential Fine-Tuning of IL-2- and IL-15-Dependent Functions by Targeting Their Common IL-2/15Rβ/γc Receptor. THE JOURNAL OF IMMUNOLOGY 2017; 198:4563-4568. [PMID: 28507024 DOI: 10.4049/jimmunol.1700046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/21/2017] [Indexed: 11/19/2022]
Abstract
Interleukin 2 and IL-15 are two closely related cytokines, displaying important functions in the immune system. They share the heterodimeric CD122/CD132 receptor to deliver their signals within target cells. Their specificity of action is conferred by their α receptor chains, IL-2Rα and IL-15Rα. By combining an increased affinity for CD122 and an impaired recruitment of CD132, we have generated an original molecule named IL-2Rβ/γ (CD122/CD132) inhibitor (BiG), targeting the CD122/CD132 receptor. BiG efficiently inhibited IL-15- and IL-2-dependent functions of primary cells, including CD8 T and NK cells, in vitro and in vivo. We also report a differential dynamic of action of these cytokines by highlighting a major role played by the IL-2Rα receptor. Interestingly, due to the presence of IL-2Rα, BiG had no impact on IL-2-dependent regulatory T cell proliferation. Thus, by acting as a fine switch in the immune system, BiG emphasizes the differential roles of these two cytokines.
Collapse
Affiliation(s)
- Dihia Meghnem
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Sébastien Morisseau
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France.,Centre Hospitalier Universitaire, Nantes 44000, France; and
| | - Marie Frutoso
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Kilian Trillet
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Mike Maillasson
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Isabelle Barbieux
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Sarah Khaddage
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Isabelle Leray
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | | | - Agnès Quéméner
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Yannick Jacques
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France
| | - Erwan Mortier
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, CNRS, Inserm, Université de Nantes, Nantes 44007, France;
| |
Collapse
|
91
|
Mathias CB, Schramm CM, Guernsey LA, Wu CA, Polukort SH, Rovatti J, Ser-Dolansky J, Secor E, Schneider SS, Thrall RS, Aguila HL. IL-15-deficient mice develop enhanced allergic responses to airway allergen exposure. Clin Exp Allergy 2017; 47:639-655. [PMID: 28093832 PMCID: PMC5407912 DOI: 10.1111/cea.12886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/15/2016] [Accepted: 12/18/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Interleukin-15 is a pleiotropic cytokine that is critical for the development and survival of multiple haematopoietic lineages. Mice lacking IL-15 have selective defects in populations of several pro-allergic immune cells including natural killer (NK) cells, NKT cells, and memory CD8+ T cells. We therefore hypothesized that IL-15-/- mice will have reduced inflammatory responses during the development of allergic airway disease (AAD). OBJECTIVE To determine whether IL-15-/- mice have attenuated allergic responses in a mouse model of AAD. METHODS C57BL/6 wild-type (WT) and IL-15-/- mice were sensitized and challenged with ovalbumin (OVA), and the development of AAD was ascertained by examining changes in airway inflammatory responses, Th2 responses, and lung histopathology. RESULTS Here, we report that IL-15-/- mice developed enhanced allergic responses in an OVA-induced model of AAD. In the absence of IL-15, OVA-challenged mice exhibited enhanced bronchial eosinophilic inflammation, elevated IL-13 production, and severe lung histopathology in comparison with WT mice. In addition, increased numbers of CD4+ T and B cells in the spleens and bronchoalveolar lavage (BAL) were also observed. Examination of OVA-challenged IL-15Rα-/- animals revealed a similar phenotype resulting in enhanced airway eosinophilia compared to WT mice. Adoptive transfer of splenic CD8+ T cells from OVA-sensitized WT mice suppressed the enhancement of eosinophilia in IL-15-/- animals to levels observed in WT mice, but had no further effects. CONCLUSION AND CLINICAL RELEVANCE These data demonstrate that mice with an endogenous IL-15 deficiency are susceptible to the development of severe, enhanced Th2-mediated AAD, which can be regulated by CD8+ T cells. Furthermore, the development of disease as well as allergen-specific Th2 responses occurs despite deficiencies in several IL-15-dependent cell types including NK, NKT, and γδ T cells, suggesting that these cells or their subsets are dispensable for the induction of AAD in IL-15-deficient mice.
Collapse
Affiliation(s)
- Clinton B. Mathias
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy, Western New England University, Springfield, MA 01119
| | - Craig M. Schramm
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Linda A. Guernsey
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Carol A. Wu
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Stephanie H. Polukort
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy, Western New England University, Springfield, MA 01119
| | - Jeffrey Rovatti
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy, Western New England University, Springfield, MA 01119
| | - Jennifer Ser-Dolansky
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA 01199
| | - Eric Secor
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA 01199
| | - Roger S. Thrall
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Hector L. Aguila
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
92
|
Suppressor of Cytokine Signaling 2 Negatively Regulates NK Cell Differentiation by Inhibiting JAK2 Activity. Sci Rep 2017; 7:46153. [PMID: 28383049 PMCID: PMC5382670 DOI: 10.1038/srep46153] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/09/2017] [Indexed: 01/19/2023] Open
Abstract
Suppressor of cytokine signaling (SOCS) proteins are negative regulators of cytokine responses. Although recent reports have shown regulatory roles for SOCS proteins in innate and adaptive immunity, their roles in natural killer (NK) cell development are largely unknown. Here, we show that SOCS2 is involved in NK cell development. SOCS2−/− mice showed a high frequency of NK cells in the bone marrow and spleen. Knockdown of SOCS2 was associated with enhanced differentiation of NK cells in vitro, and the transplantation of hematopoietic stem cells (HSCs) into congenic mice resulted in enhanced differentiation in SOCS2−/− HSCs. We found that SOCS2 could inhibit Janus kinase 2 (JAK2) activity and JAK2-STAT5 signaling pathways via direct interaction with JAK2. Furthermore, SOCS2−/− mice showed a reduction in lung metastases and an increase in survival following melanoma challenge. Overall, our findings suggest that SOCS2 negatively regulates the development of NK cells by inhibiting JAK2 activity via direct interaction.
Collapse
|
93
|
Multi-cellular natural killer (NK) cell clusters enhance NK cell activation through localizing IL-2 within the cluster. Sci Rep 2017; 7:40623. [PMID: 28074895 PMCID: PMC5225448 DOI: 10.1038/srep40623] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/08/2016] [Indexed: 12/16/2022] Open
Abstract
Multi-cellular cluster formation of natural killer (NK) cells occurs during in vivo priming and potentiates their activation to IL-2. However, the precise mechanism underlying this synergy within NK cell clusters remains unclear. We employed lymphocyte-laden microwell technologies to modulate contact-mediated multi-cellular interactions among activating NK cells and to quantitatively assess the molecular events occurring in multi-cellular clusters of NK cells. NK cells in social microwells, which allow cell-to-cell contact, exhibited significantly higher levels of IL-2 receptor (IL-2R) signaling compared with those in lonesome microwells, which prevent intercellular contact. Further, CD25, an IL-2R α chain, and lytic granules of NK cells in social microwells were polarized toward MTOC. Live cell imaging of lytic granules revealed their dynamic and prolonged polarization toward neighboring NK cells without degranulation. These results suggest that IL-2 bound on CD25 of one NK cells triggered IL-2 signaling of neighboring NK cells. These results were further corroborated by findings that CD25-KO NK cells exhibited lower proliferation than WT NK cells, and when mixed with WT NK cells, underwent significantly higher level of proliferation. These data highlights the existence of IL-2 trans-presentation between NK cells in the local microenvironment where the availability of IL-2 is limited.
Collapse
|
94
|
Viant C, Guia S, Hennessy RJ, Rautela J, Pham K, Bernat C, Goh W, Jiao Y, Delconte R, Roger M, Simon V, Souza-Fonseca-Guimaraes F, Grabow S, Belz GT, Kile BT, Strasser A, Gray D, Hodgkin PD, Beutler B, Vivier E, Ugolini S, Huntington ND. Cell cycle progression dictates the requirement for BCL2 in natural killer cell survival. J Exp Med 2017; 214:491-510. [PMID: 28057804 PMCID: PMC5294858 DOI: 10.1084/jem.20160869] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/27/2016] [Accepted: 12/12/2016] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells with antitumor functions. Using an N-ethyl-N-nitrosourea (ENU)-induced mutagenesis screen in mice, we identified a strain with an NK cell deficiency caused by a hypomorphic mutation in the Bcl2 (B cell lymphoma 2) gene. Analysis of these mice and the conditional deletion of Bcl2 in NK cells revealed a nonredundant intrinsic requirement for BCL2 in NK cell survival. In these mice, NK cells in cycle were protected against apoptosis, and NK cell counts were restored in inflammatory conditions, suggesting a redundant role for BCL2 in proliferating NK cells. Consistent with this, cycling NK cells expressed higher MCL1 (myeloid cell leukemia 1) levels in both control and BCL2-null mice. Finally, we showed that deletion of BIM restored survival in BCL2-deficient but not MCL1-deficient NK cells. Overall, these data demonstrate an essential role for the binding of BCL2 to BIM in the survival of noncycling NK cells. They also favor a model in which MCL1 is the dominant survival protein in proliferating NK cells.
Collapse
Affiliation(s)
- Charlotte Viant
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France
| | - Sophie Guia
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France
| | - Robert J Hennessy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Jai Rautela
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Kim Pham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Claire Bernat
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France
| | - Wilford Goh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Yuhao Jiao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia.,School of Medicine, Tsinghua University, Beijing 100084, China
| | - Rebecca Delconte
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Michael Roger
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France
| | - Vanina Simon
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France
| | - Fernando Souza-Fonseca-Guimaraes
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Stephanie Grabow
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Benjamin T Kile
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Daniel Gray
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Phillip D Hodgkin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric Vivier
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France.,Service Immunologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (APHM), 13288 Marseille, France
| | - Sophie Ugolini
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), 13288 Marseille, France
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia .,Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
95
|
Guo Y, Luan L, Patil NK, Wang J, Bohannon JK, Rabacal W, Fensterheim BA, Hernandez A, Sherwood ER. IL-15 Enables Septic Shock by Maintaining NK Cell Integrity and Function. THE JOURNAL OF IMMUNOLOGY 2016; 198:1320-1333. [PMID: 28031340 DOI: 10.4049/jimmunol.1601486] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/05/2016] [Indexed: 11/19/2022]
Abstract
Interleukin 15 is essential for the development and differentiation of NK and memory CD8+ (mCD8+) T cells. Our laboratory previously showed that NK and CD8+ T lymphocytes facilitate the pathobiology of septic shock. However, factors that regulate NK and CD8+ T lymphocyte functions during sepsis are not well characterized. We hypothesized that IL-15 promotes the pathogenesis of sepsis by maintaining NK and mCD8+ T cell integrity. To test our hypothesis, the pathogenesis of sepsis was assessed in IL-15-deficient (IL-15 knockout, KO) mice. IL-15 KO mice showed improved survival, attenuated hypothermia, and less proinflammatory cytokine production during septic shock caused by cecal ligation and puncture or endotoxin-induced shock. Treatment with IL-15 superagonist (IL-15 SA, IL-15/IL-15Rα complex) regenerated NK and mCD8+ T cells and re-established mortality of IL-15 KO mice during septic shock. Preventing NK cell regeneration attenuated the restoration of mortality caused by IL-15 SA. If given immediately prior to septic challenge, IL-15-neutralizing IgG M96 failed to protect against septic shock. However, M96 caused NK cell depletion if given 4 d prior to septic challenge and conferred protection. IL-15 SA treatment amplified endotoxin shock, which was prevented by NK cell or IFN-γ depletion. IL-15 SA treatment also exacerbated septic shock caused by cecal ligation and puncture when given after the onset of sepsis. In conclusion, endogenous IL-15 does not directly augment the pathogenesis of sepsis but enables the development of septic shock by maintaining NK cell numbers and integrity. Exogenous IL-15 exacerbates the severity of sepsis by activating NK cells and facilitating IFN-γ production.
Collapse
Affiliation(s)
- Yin Guo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37212; and
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jingbin Wang
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Whitney Rabacal
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37212; and
| | - Benjamin A Fensterheim
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37212; and
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Edward R Sherwood
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37212; and .,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
96
|
Ng SSM, Nagy BA, Jensen SM, Hu X, Alicea C, Fox BA, Felber BK, Bergamaschi C, Pavlakis GN. Heterodimeric IL15 Treatment Enhances Tumor Infiltration, Persistence, and Effector Functions of Adoptively Transferred Tumor-specific T Cells in the Absence of Lymphodepletion. Clin Cancer Res 2016; 23:2817-2830. [PMID: 27986749 DOI: 10.1158/1078-0432.ccr-16-1808] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022]
Abstract
Purpose: Adoptive cell transfer (ACT) is a promising immunotherapeutic approach for cancer. Host lymphodepletion is associated with favorable ACT therapy outcomes, but it may cause detrimental effects in humans. We tested the hypothesis that IL15 administration enhances ACT in the absence of lymphodepletion. We previously showed that bioactive IL15 in vivo comprises a stable complex of the IL15 chain with the IL15 receptor alpha chain (IL15Rα), termed heterodimeric IL15 (hetIL15).Experimental Design: We evaluated the effects of the combination regimen ACT + hetIL15 in the absence of lymphodepletion by transferring melanoma-specific Pmel-1 T cells into B16 melanoma-bearing mice.Results: hetIL15 treatment delayed tumor growth by promoting infiltration and persistence of both adoptively transferred Pmel-1 cells and endogenous CD8+ T cells into the tumor. In contrast, persistence of Pmel-1 cells was severely reduced following irradiation in comparison with mice treated with hetIL15. Importantly, we found that hetIL15 treatment led to the preferential enrichment of Pmel-1 cells in B16 tumor sites in an antigen-dependent manner. Upon hetIL15 administration, tumor-infiltrating Pmel-1 cells showed a "nonexhausted" effector phenotype, characterized by increased IFNγ secretion, proliferation, and cytotoxic potential and low level of PD-1. hetIL15 treatment also resulted in an improved ratio of Pmel-1 to Treg in the tumor.Conclusions: hetIL15 administration improves the outcome of ACT in lymphoreplete hosts, a finding with significant implications for improving cell-based cancer immunotherapy strategies. Clin Cancer Res; 23(11); 2817-30. ©2016 AACR.
Collapse
Affiliation(s)
- Sinnie Sin Man Ng
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Bethany A Nagy
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland.,Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Shawn M Jensen
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center, Portland, Oregon
| | - Xintao Hu
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Bernard A Fox
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center, Portland, Oregon
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland.
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland.
| |
Collapse
|
97
|
Lam VC, Lanier LL. NK cells in host responses to viral infections. Curr Opin Immunol 2016; 44:43-51. [PMID: 27984782 DOI: 10.1016/j.coi.2016.11.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/20/2016] [Indexed: 12/19/2022]
Abstract
Natural killer (NK) cells are cytotoxic innate lymphocytes that play an important role in viral clearance. NK cell responses to viral infections were originally believed to be non-specific and lacked immune memory recall responses. It is now appreciated that NK cell responses to viral infections can be specific and in some cases memory recall responses are established. Increasing evidence also illuminates the complexity of NK cell interactions with both innate and adaptive immune cells. Here, we summarize the evidence for NK cell-specific memory responses to viral infections and the intricate reciprocal interactions between NK cells and other immune cells that dictate their activation and effector functions.
Collapse
Affiliation(s)
- Viola C Lam
- Biomedical Sciences Graduate Program, San Francisco, CA 94143, United States; Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, United States
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, United States; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, United States.
| |
Collapse
|
98
|
Khameneh HJ, Ho AWS, Spreafico R, Derks H, Quek HQY, Mortellaro A. The Syk-NFAT-IL-2 Pathway in Dendritic Cells Is Required for Optimal Sterile Immunity Elicited by Alum Adjuvants. THE JOURNAL OF IMMUNOLOGY 2016; 198:196-204. [PMID: 27895176 DOI: 10.4049/jimmunol.1600420] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/28/2016] [Indexed: 12/29/2022]
Abstract
Despite a long history and extensive usage of insoluble aluminum salts (alum) as vaccine adjuvants, the molecular mechanisms underpinning Ag-specific immunity upon vaccination remain unclear. Dendritic cells (DCs) are crucial initiators of immune responses, but little is known about the molecular pathways used by DCs to sense alum and, in turn, activate T and B cells. In this article, we show that alum adjuvanticity requires IL-2 specifically released by DCs, even when T cell secretion of IL-2 is intact. We demonstrate that alum, as well as other sterile particulates, such as uric acid crystals, induces DCs to produce IL-2 following initiation of actin-mediated phagocytosis that leads to Src and Syk kinase activation, Ca2+ mobilization, and calcineurin-dependent activation of NFAT, the master transcription factor regulating IL-2 expression. Using chimeric mice, we show that DC-derived IL-2 is required for maximal Ag-specific proliferation of CD4+ T cells and optimal humoral responses following alum-adjuvanted immunization. These data identify DC-derived IL-2 as a key mediator of alum adjuvanticity in vivo and the Src-Syk pathway as a potential leverage point in the rational design of novel adjuvants.
Collapse
Affiliation(s)
- Hanif Javanmard Khameneh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Adrian W S Ho
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Roberto Spreafico
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Heidi Derks
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Hazel Q Y Quek
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Alessandra Mortellaro
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| |
Collapse
|
99
|
Rautela J, Huntington ND. IL-15 signaling in NK cell cancer immunotherapy. Curr Opin Immunol 2016; 44:1-6. [PMID: 27835762 DOI: 10.1016/j.coi.2016.10.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
Abstract
While cancer has been traditionally treated by chemotherapy, radiation, targeted therapies and surgery, a fifth pillar of cancer treatment, immunotherapy, has emerged over the past 10 years and revolutionized our war on cancer. The benchmark for drugs in this category has been set by the development of CD8 T cell checkpoint (CTLA-4 and PD-1/PD-L1) inhibitors. These therapies effectively expand and reactivate the pool of tumor-specific T cells leading to objective response rates of up to 50% in patients with certain cancers. However, the significant number of patients and cancer types that altogether fail or acquire resistance to these therapies highlights the need for novel immunotherapies that target alternate pathways and effector cells. Thus, there is renewed interest in harnessing the tumor-killing abilities of Natural Killer (NK) cells, though it has proven difficult to efficiently and specifically target these cells cancer patients. The commercial success of T cell checkpoint inhibitors has seen a swam of new biotech companies emerge with innovative or revised strategies that aim to harness the innate non-antigen dependent tumor lysis potential of NK cells. This review will focus on IL-15 biology in NK cells and proposes the development novel therapies aimed at this pathway in humans.
Collapse
Affiliation(s)
- Jai Rautela
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3010, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
100
|
Van den Bergh J, Willemen Y, Lion E, Van Acker H, De Reu H, Anguille S, Goossens H, Berneman Z, Van Tendeloo V, Smits E. Transpresentation of interleukin-15 by IL-15/IL-15Rα mRNA-engineered human dendritic cells boosts antitumoral natural killer cell activity. Oncotarget 2016; 6:44123-33. [PMID: 26675759 DOI: 10.18632/oncotarget.6536] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/28/2015] [Indexed: 01/20/2023] Open
Abstract
In cancer immunotherapy, the use of dendritic cell (DC)-based vaccination strategies can improve overall survival, but until now durable clinical responses remain scarce. To date, DC vaccines are designed primarily to induce effective T-cell responses, ignoring the antitumor activity potential of natural killer (NK) cells. Aiming to further improve current DC vaccination outcome, we engineered monocyte-derived DC to produce interleukin (IL)-15 and/or IL-15 receptor alpha (IL-15Rα) using mRNA electroporation. The addition of IL-15Rα to the protocol, enabling IL-15 transpresentation to neighboring NK cells, resulted in significantly better NK-cell activation compared to IL-15 alone. Next to upregulation of NK-cell membrane activation markers, IL-15 transpresentation resulted in increased NK-cell secretion of IFN-γ, granzyme B and perforin. Moreover, IL-15-transpresenting DC/NK cell cocultures from both healthy donors and acute myeloid leukemia (AML) patients in remission showed markedly enhanced cytotoxic activity against NK cell sensitive and resistant tumor cells. Blocking IL-15 transpresentation abrogated NK cell-mediated cytotoxicity against tumor cells, pointing to a pivotal role of IL-15 transpresentation by IL-15Rα to exert its NK cell-activating effects. In conclusion, we report an attractive approach to improve antitumoral NK-cell activity in DC-based vaccine strategies through the use of IL-15/IL-15Rα mRNA-engineered designer DC.
Collapse
Affiliation(s)
- Johan Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Heleen Van Acker
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Evelien Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Oncological Research Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|