201
|
T Cells Engineered With Chimeric Antigen Receptors Targeting NKG2D Ligands Display Lethal Toxicity in Mice. Mol Ther 2015; 23:1600-10. [PMID: 26122933 DOI: 10.1038/mt.2015.119] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/18/2015] [Indexed: 12/13/2022] Open
Abstract
Ligands for the NKG2D receptor are overexpressed on tumors, making them interesting immunotherapy targets. To assess the tumoricidal properties of T cells directed to attack NKG2D ligands, we engineered murine T cells with two distinct NKG2D-based chimeric antigen receptors (CARs): (i) a fusion between the NKG2D receptor and the CD3ζ chain and (ii) a conventional second-generation CAR, where the extracellular domain of NKG2D was fused to CD28 and CD3ζ. To enhance the CAR surface expression, we also engineered T cells to coexpress DAP10. In vitro functionality and surface expression levels of all three CARs was greater in BALB/c T cells than C57BL/6 T cells, indicating strain-specific differences. Upon adoptive transfer of NKG2D-CAR-T cells into syngeneic animals, we observed significant clinical toxicity resulting in morbidity and mortality. The severity of these toxicities varied between the CAR configurations and paralleled their in vitro NKG2D surface expression. BALB/c mice were more sensitive to these toxicities than C57BL/6 mice, consistent with the higher in vitro functionality of BALB/c T cells. Treatment with cyclophosphamide prior to adoptive transfer exacerbated the toxicity. We conclude that while NKG2D ligands may be useful targets for immunotherapy, the pursuit of NKG2D-based CAR-T cell therapies should be undertaken with caution.
Collapse
|
202
|
Kellner C, Günther A, Humpe A, Repp R, Klausz K, Derer S, Valerius T, Ritgen M, Brüggemann M, van de Winkel JG, Parren PW, Kneba M, Gramatzki M, Peipp M. Enhancing natural killer cell-mediated lysis of lymphoma cells by combining therapeutic antibodies with CD20-specific immunoligands engaging NKG2D or NKp30. Oncoimmunology 2015; 5:e1058459. [PMID: 26942070 DOI: 10.1080/2162402x.2015.1058459] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 02/06/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) mediated through the IgG Fc receptor FcγRIIIa represents a major effector function of many therapeutic antibodies. In an attempt to further enhance natural killer (NK) cell-mediated ADCC, we combined therapeutic antibodies against CD20 and CD38 with recombinant immunoligands against the stimulatory NK cell receptors NKG2D or NKp30. These immunoligands, respectively designated as ULBP2:7D8 and B7-H6:7D8, contained the CD20 scFv 7D8 as a targeting moiety and a cognate ligand for either NKG2D or NKp30 (i.e. ULBP2 and B7-H6, respectively). Both the immunoligands synergistically augmented ADCC in combination with the CD20 antibody rituximab and the CD38 antibody daratumumab. Combinations with ULBP2:7D8 resulted in higher cytotoxicity compared to combinations with B7-H6:7D8, suggesting that coligation of FcγRIIIa with NKG2D triggered NK cells more efficiently than with NKp30. Addition of B7-H6:7D8 to ULBP2:7D8 and rituximab in a triple combination did not further increase the extent of tumor cell lysis. Importantly, immunoligand-mediated enhancement of ADCC was also observed for tumor cells and autologous NK cells from patients with hematologic malignancies, in which, again, ULBP2:7D8 was particularly active. In summary, co-targeting of NKG2D was more effective in promoting rituximab or daratumumab-mediated ADCC by NK cells than co-ligation of NKp30. The observed increase in the ADCC activity of these therapeutic antibodies suggests promise for a 'dual-dual-targeting' approach in which tumor cell surface antigens are targeted in concert with two distinct activating NK cell receptors (i.e. FcγRIIIa and NKG2D or B7-H6).
Collapse
Affiliation(s)
- Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Andreas Günther
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Andreas Humpe
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Roland Repp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Matthias Ritgen
- 2nd Department of Medicine; Christian-Albrechts-University Kiel ; Kiel, Germany
| | - Monika Brüggemann
- 2nd Department of Medicine; Christian-Albrechts-University Kiel ; Kiel, Germany
| | - Jan Gj van de Winkel
- Genmab; Utrecht, the Netherlands; Department of Immunology; University Medical Center Utrecht; Utrecht, the Netherlands
| | - Paul Whi Parren
- Genmab; Utrecht, the Netherlands; Department of Cancer and Inflammation Research; Institute of Molecular Medicine; University of Southern Denmark; Odense, Denmark; Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden, the Netherlands
| | - Michael Kneba
- 2nd Department of Medicine; Christian-Albrechts-University Kiel ; Kiel, Germany
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine; Christian-Albrechts-University Kiel; Kiel, Germany
| |
Collapse
|
203
|
Yan C, Jie L, Yongqi W, Weiming X, Juqun X, Yanbing D, Li Q, Xingyuan P, Mingchun J, Weijuan G. Delivery of human NKG2D-IL-15 fusion gene by chitosan nanoparticles to enhance antitumor immunity. Biochem Biophys Res Commun 2015; 463:336-43. [PMID: 26022121 DOI: 10.1016/j.bbrc.2015.05.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 01/22/2023]
Abstract
Nanoparticles are becoming promising carriers for gene delivery because of their high capacity in gene loading and low cell cytotoxicity. In this study, a chitosan-based nanoparticle encapsulated within a recombinant pcDNA3.1-dsNKG2D-IL-15 plasmid was generated. The fused dsNKG2D-IL-15 gene fragment consisted of double extracellular domains of NKG2D with IL-15 gene at downstream. The average diameter of the gene nanoparticles ranged from 200 nm to 400 nm, with mean zeta potential value of 53.8 ± 6.56 mV. The nanoparticles which were loaded with the dsNKG2D-IL-15 gene were uptaken by tumor cells with low cytotoxicity. Tumor cells pre-transfected by gene nanopartilces stimulated NK and T cells in vitro. Intramuscular injection of gene nanoparticles suppressed tumor growth and prolonged survival of tumor-bearing mice through activation of NK and CD8(+) T cells. Thus, chitosan-based nanoparticle delivery of dsNKG2D-IL-15 gene vaccine can be potentially used for tumor therapy.
Collapse
Affiliation(s)
- Chen Yan
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Leng Jie
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Wang Yongqi
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Xiao Weiming
- Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, 225009, PR China
| | - Xi Juqun
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225009, PR China
| | - Ding Yanbing
- Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, 225009, PR China
| | - Qian Li
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Pan Xingyuan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, PR China
| | - Ji Mingchun
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Gong Weijuan
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, PR China; Department of Gastroenterology, The Second Clinical Medical College, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China.
| |
Collapse
|
204
|
Hagberg N, Theorell J, Hjorton K, Spee P, Eloranta ML, Bryceson YT, Rönnblom L. Functional anti-CD94/NKG2A and anti-CD94/NKG2C autoantibodies in patients with systemic lupus erythematosus. Arthritis Rheumatol 2015; 67:1000-11. [PMID: 25510434 DOI: 10.1002/art.38999] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 12/11/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Recently we serendipitously identified a patient with systemic lupus erythematosus (SLE) who was positive for autoantibodies to CD94/natural killer receptor group 2A (NKG2A). The present study was undertaken to investigate the occurrence and function of autoantibodies targeting lectin-like NK cell receptors in SLE. METHODS Sera from 203 SLE patients and 90 healthy individuals were analyzed, by flow cytometry, for Ig binding to Ba/F3 cells transfected with CD94/NKG2A, CD94/NKG2C, or NKG2D. Autoantibodies identified were characterized with regard to interference with HLA-E binding, effect on NK cell activation in response to HLA-E-transfected K562 cells, and capacity to facilitate antibody-dependent cell-mediated cytotoxicity (ADCC). Levels of autoantibodies were determined in longitudinally sampled sera, and correlations with disease activity (SLE Disease Activity Index 2000) and severity (Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage Index) were investigated. RESULTS Anti-CD94/NKG2A autoantibodies were identified in 7 SLE patients. The autoantibodies from 6 patients inhibited binding of HLA-E to CD94/NKG2A, whereas those from the seventh patient augmented this binding. Autoantibodies from 2 patients also reacted with the activating receptor CD94/NKG2C, with inhibition of the binding of HLA-E to CD94/NKG2C observed in 1 case and enhancement of this binding in the other. None of the sera contained anti-NKG2D autoantibodies. The levels of anti-CD94/NKG2A and anti-CD94/NKG2C autoantibodies correlated with disease activity and with a more severe SLE phenotype. Mechanistically, anti-CD94/NKG2A and anti-CD94/NKG2C autoantibodies both interfered with HLA-E-mediated regulation of NK cell activation and facilitated the elimination of target cells expressing CD94/NKG2A or CD94/NKG2C through ADCC. CONCLUSION Anti-CD94/NKG2A and anti-CD94/NKG2C autoantibodies occur in a subset of patients with clinically active SLE. Given their capacity to deplete certain NK cell subsets and interfere with particular NK cell function, such autoantibodies may promote the pathogenesis of SLE.
Collapse
|
205
|
Giannattasio A, Weil S, Kloess S, Ansari N, Stelzer EHK, Cerwenka A, Steinle A, Koehl U, Koch J. Cytotoxicity and infiltration of human NK cells in in vivo-like tumor spheroids. BMC Cancer 2015; 15:351. [PMID: 25933805 PMCID: PMC4422268 DOI: 10.1186/s12885-015-1321-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/17/2015] [Indexed: 02/12/2023] Open
Abstract
Background The complex cellular networks within tumors, the cytokine milieu, and tumor immune escape mechanisms affecting infiltration and anti-tumor activity of immune cells are of great interest to understand tumor formation and to decipher novel access points for cancer therapy. However, cellular in vitro assays, which rely on monolayer cultures of mammalian cell lines, neglect the three-dimensional architecture of a tumor, thus limiting their validity for the in vivo situation. Methods Three-dimensional in vivo-like tumor spheroid were established from human cervical carcinoma cell lines as proof of concept to investigate infiltration and cytotoxicity of NK cells in a 96-well plate format, which is applicable for high-throughput screening. Tumor spheroids were monitored for NK cell infiltration and cytotoxicity by flow cytometry. Infiltrated NK cells, could be recovered by magnetic cell separation. Results The tumor spheroids were stable over several days with minor alterations in phenotypic appearance. The tumor spheroids expressed high levels of cellular ligands for the natural killer (NK) group 2D receptor (NKG2D), mediating spheroid destruction by primary human NK cells. Interestingly, destruction of a three-dimensional tumor spheroid took much longer when compared to the parental monolayer cultures. Moreover, destruction of tumor spheroids was accompanied by infiltration of a fraction of NK cells, which could be recovered at high purity. Conclusion Tumor spheroids represent a versatile in vivo-like model system to study cytotoxicity and infiltration of immune cells in high-throughput screening. This system might proof useful for the investigation of the modulatory potential of soluble factors and cells of the tumor microenvironment on immune cell activity as well as profiling of patient-/donor-derived immune cells to personalize cellular immunotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1321-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ariane Giannattasio
- NK Cell Biology, Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.
| | - Sandra Weil
- NK Cell Biology, Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.
| | - Stephan Kloess
- Institute for Cellular therapeutics, IFB-Tx, Hannover Medical School, Hannover, Germany.
| | - Nariman Ansari
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences, Goethe Universität, Frankfurt, Germany.
| | - Ernst H K Stelzer
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences, Goethe Universität, Frankfurt, Germany.
| | - Adelheid Cerwenka
- Innate Immunity, German Cancer Research Center, Heidelberg, Germany.
| | - Alexander Steinle
- Institute for Molecular Medicine, Johann Wolfgang Goethe-University, Frankfurt, Germany.
| | - Ulrike Koehl
- Institute for Cellular therapeutics, IFB-Tx, Hannover Medical School, Hannover, Germany.
| | - Joachim Koch
- NK Cell Biology, Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.
| |
Collapse
|
206
|
Affiliation(s)
- Alexander Steinle
- Institute for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
207
|
Gras Navarro A, Björklund AT, Chekenya M. Therapeutic potential and challenges of natural killer cells in treatment of solid tumors. Front Immunol 2015; 6:202. [PMID: 25972872 PMCID: PMC4413815 DOI: 10.3389/fimmu.2015.00202] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/14/2015] [Indexed: 12/22/2022] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells that hold tremendous potential for effective immunotherapy for a broad range of cancers. Due to the mode of NK cell killing, requiring one-to-one target engagement and site-directed release of cytolytic granules, the therapeutic potential of NK cells has been most extensively explored in hematological malignancies. However, their ability to precisely kill antibody coated cells, cancer stem cells, and genotoxically altered cells, while maintaining tolerance to healthy cells makes them appealing therapeutic effectors for all cancer forms, including metastases. Due to their release of pro-inflammatory cytokines, NK cells may potently reverse the anti-inflammatory tumor microenvironment (TME) and augment adaptive immune responses by promoting differentiation, activation, and/or recruitment of accessory immune cells to sites of malignancy. Nevertheless, integrated and coordinated mechanisms of subversion of NK cell activity against the tumor and its microenvironment exist. Although our understanding of the receptor ligand interactions that regulate NK cell functionality has evolved remarkably, the diversity of ligands and receptors is complex, as is their mechanistic foundations in regulating NK cell function. In this article, we review the literature and highlight how the TME manipulates the NK cell phenotypes, genotypes, and tropism to evade tumor recognition and elimination. We discuss counter strategies that may be adopted to augment the efficacy of NK cell anti-tumor surveillance, the clinical trials that have been undertaken so far in solid malignancies, critically weighing the challenges and opportunities with this approach.
Collapse
Affiliation(s)
| | - Andreas T Björklund
- Karolinska University Hospital, Hematology Center and Karolinska Institute , Stockholm , Sweden
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen , Bergen , Norway
| |
Collapse
|
208
|
Wu AA, Drake V, Huang HS, Chiu S, Zheng L. Reprogramming the tumor microenvironment: tumor-induced immunosuppressive factors paralyze T cells. Oncoimmunology 2015; 4:e1016700. [PMID: 26140242 DOI: 10.1080/2162402x.2015.1016700] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/02/2015] [Accepted: 02/03/2015] [Indexed: 02/08/2023] Open
Abstract
It has become evident that tumor-induced immuno-suppressive factors in the tumor microenvironment play a major role in suppressing normal functions of effector T cells. These factors serve as hurdles that limit the therapeutic potential of cancer immunotherapies. This review focuses on illustrating the molecular mechanisms of immunosuppression in the tumor microenvironment, including evasion of T-cell recognition, interference with T-cell trafficking, metabolism, and functions, induction of resistance to T-cell killing, and apoptosis of T cells. A better understanding of these mechanisms may help in the development of strategies to enhance the effectiveness of cancer immunotherapies.
Collapse
Key Words
- 1MT, 1-methyltryptophan
- COX2, cyclooxygenase-2
- GM-CSF, granulocyte macrophage colony-stimulating factor
- GPI, glycosylphosphatidylinositol
- Gal1, galectin-1
- HDACi, histone deacetylase inhibitor
- HLA, human leukocyte antigen
- IDO, indoleamine-2,3- dioxygenase
- IL-10, interleukin-10
- IMC, immature myeloid cell
- MDSC, myeloid-derived suppressor cells
- MHC, major histocompatibility
- MICA, MHC class I related molecule A
- MICB, MHC class I related molecule B
- NO, nitric oxide
- PARP, poly ADP-ribose polymerase
- PD-1, program death receptor-1
- PD-L1, programmed death ligand 1
- PGE2, prostaglandin E2
- RCAS1, receptor-binding cancer antigen expressed on Siso cells 1
- RCC, renal cell carcinoma
- SOCS, suppressor of cytokine signaling
- STAT3, signal transducer and activator of transcription 3
- SVV, survivin
- T cells
- TCR, T-cell receptor
- TGF-β, transforming growth factor β
- TRAIL, TNF-related apoptosis-inducing ligand
- VCAM-1, vascular cell adhesion molecule-1
- XIAP, X-linked inhibitor of apoptosis protein
- iNOS, inducible nitric-oxide synthase
- immunosuppression
- immunosuppressive factors
- immunotherapy
- tumor microenvironment
Collapse
Affiliation(s)
- Annie A Wu
- Department of Oncology; The Johns Hopkins University School of Medicine ; Baltimore, MD USA
| | - Virginia Drake
- School of Medicine; University of Maryland ; Baltimore, MD USA
| | | | - ShihChi Chiu
- College of Medicine; National Taiwan University ; Taipei, Taiwan
| | - Lei Zheng
- Department of Oncology; The Johns Hopkins University School of Medicine ; Baltimore, MD USA
| |
Collapse
|
209
|
NKG2D expression by CD8+ T cells contributes to GVHD and GVT effects in a murine model of allogeneic HSCT. Blood 2015; 125:3655-63. [PMID: 25788701 DOI: 10.1182/blood-2015-02-629006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/05/2015] [Indexed: 12/27/2022] Open
Abstract
In allogeneic hematopoietic stem cell transplantation (HSCT), controlling graft-versus-host disease (GVHD) while maintaining graft-versus-tumor (GVT) responses is of critical importance. Using a mouse model of allogeneic HSCT, we hereby demonstrate that NKG2D expression by CD8(+) T cells plays a major role in mediating GVHD and GVT effects by promoting the survival and cytotoxic function of CD8(+) T cells. The expression of NKG2D ligands was not induced persistently on normal tissues of allogeneic HSCT-recipient mice treated with anti-NKG2D antibody, suggesting that transient NKG2D blockade might be sufficient to attenuate GVHD and allow CD8(+) T cells to regain their GVT function. Indeed, short-term treatment with anti-NKG2D antibody restored GVT effects while maintaining an attenuated GVHD state. NKG2D expression was also detected on CD8(+) T cells from allogeneic HSCT patients and trended to be higher in those with active GVHD. Together, these data support a novel role for NKG2D expression by CD8(+) T cells during allogeneic HSCT, which could be potentially therapeutically exploited to separate GVHD from GVT effects.
Collapse
|
210
|
Zhang J, Basher F, Wu JD. NKG2D Ligands in Tumor Immunity: Two Sides of a Coin. Front Immunol 2015; 6:97. [PMID: 25788898 PMCID: PMC4349182 DOI: 10.3389/fimmu.2015.00097] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/18/2015] [Indexed: 11/13/2022] Open
Abstract
The activating/co-stimulatory receptor NKG2D (natural-killer group 2, member D) is expressed on the surface of all human NK, NKT, CD8(+) T, and subsets of γδ(+) T cells. The significance of NKG2D function in tumor immunity has been well demonstrated in experimental animal models. However, the role of human NKG2D ligands in regulating tumor immunity and cancer prognosis had been controversial in the literature. In this review, we summarize the latest advancement, discuss the controversies, and present evidence that membrane-bound and soluble NKG2D ligands oppositely regulate tumor immunity. We also discuss new perspectives of targeting NKG2D ligands for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Fahmin Basher
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Jennifer D Wu
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA ; Cancer Immunology Program, Hollings Cancer Center , Charleston, SC , USA
| |
Collapse
|
211
|
Xiao G, Wang X, Sheng J, Lu S, Yu X, Wu JD. Soluble NKG2D ligand promotes MDSC expansion and skews macrophage to the alternatively activated phenotype. J Hematol Oncol 2015; 8:13. [PMID: 25887583 PMCID: PMC4342005 DOI: 10.1186/s13045-015-0110-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/14/2015] [Indexed: 12/19/2022] Open
Abstract
Expression of surface NKG2D ligand MIC on tumor cells is deemed to stimulate NK and co-stimulate CD8 T cell anti-tumor immunity. Human cancer cells however frequently adopt a proteinase-mediated shedding strategy to generate soluble MIC (sMIC) to circumvent host immunity. High levels of sMIC have been shown to correlate with advanced disease stages in cancer patients. The underlying mechanism is currently understood as systemic downregulation of NKG2D expression on CD8 T and NK cells and perturbing NK cell periphery maintenance. Herein we report a novel mechanism by which sMIC poses immune suppressive effect on host immunity and tumor microenvironment. We demonstrate that sMIC facilitates expansion of myeloid-derived suppressor cells (MDSCs) and skews macrophages to the more immune suppressive alternative phenotype through activation of STAT3. These findings further endorse that sMIC is an important therapeutic target for cancer immunotherapy.
Collapse
Affiliation(s)
- Gang Xiao
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA. .,Present address: The Third Hospital of South Medical University, Guangzhou, China.
| | - Xuanjun Wang
- Key Lab for Puer Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China.
| | - Jun Sheng
- Key Lab for Puer Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Shengjun Lu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA. .,Present Address: Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuezhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA. .,Cancer Immunology Program, Hollings Cancer Center, Charleston, SC, USA.
| | - Jennifer D Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA. .,Cancer Immunology Program, Hollings Cancer Center, Charleston, SC, USA.
| |
Collapse
|
212
|
Gütgemann SA, Sandusky MM, Wingert S, Claus M, Watzl C. Recruitment of activating NK-cell receptors 2B4 and NKG2D to membrane microdomains in mammalian cells is dependent on their transmembrane regions. Eur J Immunol 2015; 45:1258-69. [DOI: 10.1002/eji.201444741] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 11/21/2014] [Accepted: 12/22/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Stephan A. Gütgemann
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Mina M. Sandusky
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Sabine Wingert
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Maren Claus
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| | - Carsten Watzl
- IfADo-Leibniz Research Centre for Working Environment and Human Factors; Dortmund Germany
| |
Collapse
|
213
|
Korrer MJ, Routes JM. Adenovirus serotype 5 E1A expressing tumor cells elicit a tumor-specific CD8+ T cell response independent of NKG2D. RESULTS IN IMMUNOLOGY 2015; 5:1-5. [PMID: 25685658 PMCID: PMC4309920 DOI: 10.1016/j.rinim.2015.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 11/22/2022]
Abstract
The expression of the Adenovirus serotype 2 or serotype 5 (Ad2/5) E1A gene in tumor cells upregulates ligands that are recognized by the NKG2D activating receptor, which is expressed on NK cells and T cells, and reduces their tumorigenicity, a process dependent on NK cells and T cells. In some model systems, the forced overexpression of NKG2D ligands on tumor cells induced antigen-specific CD8+ T cells that mediated anti-tumor immunity. We wanted to determine if the interaction of NKG2D ligands on tumor cells that express E1A with NKG2D on immune cells contributed to the ability of E1A to induce a CD8+ T cell anti-tumor response or reduce tumorigenicity. To address these questions, we used the MCA-205 tumor cell line or MCA-205 cells that expressed Ad5 E1A (MCA-205-E1A cells), a fusion protein of E1A and ovalbumin (MCA-205-E1A-OVA) or OVA (MCA-205-OVA). We found that the expression of E1A or E1A–OVA, but not OVA, upregulated the expression of the NKG2D ligand RAE-1 on the surface of MCA-205 cells. Additionally, MCA-205-E1A cells and MCA-205-E1A-OVA cells were more sensitive to NK cell lysis than MCA-205 or MCA-205-OVA cells in WT B6 mice, but not NKG2D deficient B6 mice. Next, we adoptively transferred WT or NKG2D deficient OT-1 T cells (CD8 T cells that recognize OVA residues 257–264) into WT B6 mice or B6 mice that were deficient in NKG2D respectively and measured the expansion of OT-1 cells following immunization with MCA-205-E1A-OVA or MCA-205-OVA cells. We found that the expansion of OT-1 cells following immunization of either OVA-expressing MCA-205 cell lines was not affected by the presence or absence of NKG2D in B6 mice. Finally, we found that the capacity of E1A to reduce the tumorigenicity of MCA-205 cells was not impaired in B6-NKG2D deficient mice as compared to WT B6 mice. Our results suggest that the ability of E1A to reduce the tumorigenicity of MCA-205 cells, or induce an antigen-specific CD8+ T cell response, is independent of the interaction of NKG2D ligands with the NKG2D receptor.
Collapse
Key Words
- Ad, adenovirus
- Adenovirus E1A
- B6, C57BL/6
- CD8+ T cells
- E1A, early region 1 A
- MCA, methylcholanthrene
- NK cells
- NK, natural killer
- NKG2D
- NKG2D ligands
- NKG2D, natural killer group 2 D
- OVA, ovalbumin
- RAE-1, retinoic acid early inducible
- TPD50, tumor producing dose 50
- Tumor immunology
- WT, wildtype
Collapse
Affiliation(s)
- Michael J. Korrer
- Department of Pediatrics, Division of Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Correspondence to: Medical College of Wisconsin, 9000 W. Wisconsin Avenue, Milwaukee, WI 53226-4874, USA.
| | - John M. Routes
- Department of Pediatrics, Division of Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Correspondence to: Medical College of Wisconsin, 9000 W. Wisconsin Avenue, Milwaukee, WI 53226-4874, USA.
| |
Collapse
|
214
|
Abadie V, Jabri B. Immunopathology of Celiac Disease. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00080-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
215
|
Fujita H, Hatanaka Y, Sutoh Y, Suzuki Y, Oba K, Hatanaka KC, Mitsuhashi T, Otsuka N, Fugo K, Kasahara M, Matsuno Y. Immunohistochemical validation and expression profiling of NKG2D ligands in a wide spectrum of human epithelial neoplasms. J Histochem Cytochem 2014; 63:217-27. [PMID: 25473094 DOI: 10.1369/0022155414563800] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The MHC class I-chain-related proteins (MICs) and the UL16-binding proteins (ULBPs) are inducible stress response molecules that work as activators of a specific receptor, NKG2D, which is expressed on effector cells, such as NK cells and subsets of T cells. In this study, we sought to explore the biological significance of NKG2D ligands in human neoplasms by comprehensively examining the immunohistochemical expression profile of NKG2D ligands in a variety of human epithelial neoplasms. Following careful validation of the immunohistochemical specificity and availability of anti-human ULBP antibodies for formalin-fixed paraffin-embedded (FFPE) materials, the expression of NKG2D ligands was analyzed in FFPE tissue microarrays comprising 22 types of epithelial neoplastic tissue with their non-neoplastic counterpart from various organs. Hierarchical cluster analysis demonstrated a positive relationship among ULBP2/6, ULBP3, ULBP1, and ULBP5, whose expression patterns were similar across all of the neoplastic tissues examined. In contrast, MICA/B, as well as ULBP4, did not appear to be related to any other ligand. These expression profiles of NKG2D ligands in human neoplasms based on well-validated specific antibodies, followed by hierarchical cluster analysis, should help to clarify some functional aspects of these molecules in cancer biology, and also provide a path to the development of novel tumor-type-specific treatment strategies.
Collapse
Affiliation(s)
- Hiromi Fujita
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM),Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Yoichi Sutoh
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM),Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Yuta Suzuki
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Koji Oba
- Translational Research and Clinical Trial Center, Hokkaido University Hospital, Sapporo, Japan (KO)
| | - Kanako C Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Tomoko Mitsuhashi
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Noriyuki Otsuka
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Kazunori Fugo
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Masanori Kasahara
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| |
Collapse
|
216
|
LIU CHANG, QI YING, MA YANPING, HE RONG, SUN ZHENGRONG, HUANG YUJING, JI YAOHUA, RUAN QIANG. Interaction between the human cytomegalovirus-encoded UL142 and cellular Snapin proteins. Mol Med Rep 2014; 11:1069-72. [DOI: 10.3892/mmr.2014.2829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 09/24/2014] [Indexed: 11/06/2022] Open
|
217
|
Huyan T, Li Q, Ye LJ, Yang H, Xue XP, Zhang MJ, Huang QS, Yin DC, Shang P. Inhibition of human natural killer cell functional activity by human aspartyl β-hydroxylase. Int Immunopharmacol 2014; 23:452-9. [PMID: 25281391 DOI: 10.1016/j.intimp.2014.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 09/13/2014] [Accepted: 09/15/2014] [Indexed: 11/30/2022]
Abstract
Natural killer (NK) cells are a key component of the innate immune system and play pivotal roles as inflammatory regulators and in tumor surveillance. Human aspartyl β-hydroxylase (HAAH) is a plasma membrane and endoplasmic reticulum protein with hydroxylation activity, which is over-expressed in many malignant neoplasms and can be detected from the sera of tumor patients. HAAH is involved in regulating tumor cell infiltration and metastasis. Escaping from immune surveillance may help tumor cell infiltration and metastasis. However, the effects of HAAH on tumor immune surveillance have not yet been investigated carefully. The present study investigated the potential use of HAAH as an immune regulator of human NK cells. We assessed the effects of recombinant HAAH (r-HAAH) on primary human NK cell morphology, viability, cytotoxicity, apoptosis, receptors expression and cytokine/cytolytic proteins production. Our results demonstrated that r-HAAH negatively affects NK cell activity in a time and dose-dependent manner. It noticeably reduces the viability of the NK cells by increasing apoptosis and necrosis via caspase signaling pathways. Moreover, r-HAAH reduces the NK cell cytotoxicity by inhibiting surface expression of NKG2D, NKp44 and IFN-γ secretion. These findings suggest that one of the ways by which HAAH actively promotes tumor formation and proliferation is by inhibiting NK cell-surveillance activity.
Collapse
Affiliation(s)
- Ting Huyan
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Qi Li
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Lin-Jie Ye
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Hui Yang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Xiao-Ping Xue
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Ming-Jie Zhang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China; Laboratory of Molecular Virology, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 1401 Rockville Pike, Rockville, MD, USA
| | - Qing-Sheng Huang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China.
| | - Peng Shang
- Key Laboratory for Space Bioscience and Space Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 YouyiXilu, Xi'an 710072, Shaanxi, PR China
| |
Collapse
|
218
|
Lundholm M, Schröder M, Nagaeva O, Baranov V, Widmark A, Mincheva-Nilsson L, Wikström P. Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8+ T cells: mechanism of immune evasion. PLoS One 2014; 9:e108925. [PMID: 25268476 PMCID: PMC4182531 DOI: 10.1371/journal.pone.0108925] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/31/2014] [Indexed: 11/19/2022] Open
Abstract
Tumor-derived exosomes, which are nanometer-sized extracellular vesicles of endosomal origin, have emerged as promoters of tumor immune evasion but their role in prostate cancer (PC) progression is poorly understood. In this study, we investigated the ability of prostate tumor-derived exosomes to downregulate NKG2D expression on natural killer (NK) and CD8+ T cells. NKG2D is an activating cytotoxicity receptor whose aberrant loss in cancer plays an important role in immune suppression. Using flow cytometry, we found that exosomes produced by human PC cells express ligands for NKG2D on their surface. The NKG2D ligand-expressing prostate tumor-derived exosomes selectively induced downregulation of NKG2D on NK and CD8+ T cells in a dose-dependent manner, leading to impaired cytotoxic function in vitro. Consistent with these findings, patients with castration-resistant PC (CRPC) showed a significant decrease in surface NKG2D expression on circulating NK and CD8+ T cells compared to healthy individuals. Tumor-derived exosomes are likely involved in this NKG2D downregulation, since incubation of healthy lymphocytes with exosomes isolated from serum or plasma of CRPC patients triggered downregulation of NKG2D expression in effector lymphocytes. These data suggest prostate tumor-derived exosomes as down-regulators of the NKG2D-mediated cytotoxic response in PC patients, thus promoting immune suppression and tumor escape.
Collapse
Affiliation(s)
- Marie Lundholm
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| | - Mona Schröder
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Olga Nagaeva
- Department of Clinical Microbiology, Clinical Immunology, Umeå University, Umeå, Sweden
| | - Vladimir Baranov
- Department of Clinical Microbiology, Clinical Immunology, Umeå University, Umeå, Sweden
| | - Anders Widmark
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | | | - Pernilla Wikström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
219
|
Goldberger T, Mandelboim O. The use of microRNA by human viruses: lessons from NK cells and HCMV infection. Semin Immunopathol 2014; 36:659-74. [PMID: 25234555 DOI: 10.1007/s00281-014-0447-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 08/28/2014] [Indexed: 12/21/2022]
Abstract
Depending on ethnicity and on social conditions, between 40 and 90 % of the population is infected with human cytomegalovirus (HCMV). In immunocompetent patients, the virus may cause an acute disease and then revert to a state of latency, which enables its coexistence with the human host. However, in cases of immunosuppression or in neonatal infections, HCMV can cause serious long-lasting illnesses. HCMV has developed multiple mechanisms in order to escape its elimination by the immune system, specifically by two killer cell types of the adaptive and the innate immune systems; cytotoxic T lymphocytes (CTL) and natural killer (NK) cells, respectively. Another fascinating aspect of HCMV is that like other highly developed herpesviruses, it expresses its own unique set of microRNAs. Here, we initially describe how the activity of NK cells is regulated under normal conditions and during infection. Then, we discuss what is currently known about HCMV microRNA-mediated interactions, with special emphasis on immune modulation and NK cell evasion. We further illustrate the significant modulation of cellular microRNAs during HCMV infection. Although, the full target spectrum of HCMV microRNAs is far from being completely elucidated, it can already be concluded that HCMV uses its "multitasking" microRNAs to globally affect its own life cycle, as well as important cellular and immune-related pathways.
Collapse
Affiliation(s)
- Tal Goldberger
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC, Jerusalem, 91120, Israel
| | | |
Collapse
|
220
|
Moretta L, Montaldo E, Vacca P, Del Zotto G, Moretta F, Merli P, Locatelli F, Mingari MC. Human natural killer cells: origin, receptors, function, and clinical applications. Int Arch Allergy Immunol 2014; 164:253-64. [PMID: 25323661 DOI: 10.1159/000365632] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells are important effectors playing a relevant role in innate immunity, primarily in tumor surveillance and in defenses against viruses. Human NK cells recognize HLA class I molecules through surface receptors (KIR and NKG2A) that inhibit NK cell function and kill target cells that have lost (or underexpress) HLA class I molecules as it occurs in tumors or virus-infected cells. NK cell activation is mediated by an array of activating receptors and co-receptors that recognize ligands expressed primarily on tumors or virus-infected cells. In vivo anti-tumor NK cell activity may be suppressed by tumor or tumor-associated cells. Alloreactive NK cells (i.e. those that are not inhibited by the HLA class I alleles of the patient) derived from HSC of haploidentical donors play a major role in the cure of high-risk leukemia, by killing leukemia blasts and patient's DC, thus preventing tumor relapses and graft-versus-host disease. The expression of the HLA-C2-specific activating KIR2DS1 may also contribute to NK alloreactivity in patients expressing C2 alleles. A clear correlation has been proven between the size of the alloreactive NK cell population and the clinical outcome. Recently, haplo-HSCT has been further improved with the direct infusion, together with HSC, of donor-derived, mature alloreactive NK cells and TCRγδ(+) T cells - both contributing to a prompt anti-leukemia effect together with an efficient defense against pathogens during the 6- to 8-week interval required for the generation of alloreactive NK cells from HSC.
Collapse
Affiliation(s)
- Lorenzo Moretta
- Istituto Giannina Gaslini, Università di Genova, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
221
|
Sakaguchi M, Murata H, Aoyama Y, Hibino T, Putranto EW, Ruma IMW, Inoue Y, Sakaguchi Y, Yamamoto KI, Kinoshita R, Futami J, Kataoka K, Iwatsuki K, Huh NH. DNAX-activating protein 10 (DAP10) membrane adaptor associates with receptor for advanced glycation end products (RAGE) and modulates the RAGE-triggered signaling pathway in human keratinocytes. J Biol Chem 2014; 289:23389-402. [PMID: 25002577 DOI: 10.1074/jbc.m114.573071] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is involved in the pathogenesis of many inflammatory, degenerative, and hyperproliferative diseases, including cancer. Previously, we revealed mechanisms of downstream signaling from ligand-activated RAGE, which recruits TIRAP/MyD88. Here, we showed that DNAX-activating protein 10 (DAP10), a transmembrane adaptor protein, also binds to RAGE. By artificial oligomerization of RAGE alone or RAGE-DAP10, we found that RAGE-DAP10 heterodimer formation resulted in a marked enhancement of Akt activation, whereas homomultimeric interaction of RAGE led to activation of caspase 8. Normal human epidermal keratinocytes exposed to S100A8/A9, a ligand for RAGE, at a nanomolar concentration mimicked the pro-survival response of RAGE-DAP10 interaction, although at a micromolar concentration, the cells mimicked the pro-apoptotic response of RAGE-RAGE. In transformed epithelial cell lines, A431 and HaCaT, in which endogenous DAP10 was overexpressed, and S100A8/A9, even at a micromolar concentration, led to cell growth and survival due to RAGE-DAP10 interaction. Functional blocking of DAP10 in the cell lines abrogated the Akt phosphorylation from S100A8/A9-activated RAGE, eventually leading to an increase in apoptosis. Finally, S100A8/A9, RAGE, and DAP10 were overexpressed in the psoriatic epidermis. Our findings indicate that the functional interaction between RAGE and DAP10 coordinately regulates S100A8/A9-mediated survival and/or apoptotic response of keratinocytes.
Collapse
Affiliation(s)
| | | | - Yumi Aoyama
- Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kita-ku, Okayama 700-8558
| | - Toshihiko Hibino
- the Shiseido Research Center, Advanced Science Research, 2-2-1 Hayabuchi, Tsuzuki-ku, Yokohama 224-8558
| | | | | | - Yusuke Inoue
- the Faculty of Science and Technology, Division of Molecular Science, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515
| | - Yoshihiko Sakaguchi
- the Interdisciplinary Research Organization, University of Miyazaki, Kiyotakecho, Miyazaki 889-1692
| | | | - Rie Kinoshita
- the Department of Biotechnology, Division of Chemistry and Biochemistry, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, and
| | - Junichiro Futami
- the Department of Biotechnology, Division of Chemistry and Biochemistry, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, and
| | - Ken Kataoka
- the Department of Life Science, Faculty of Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama 700-0005, Japan
| | - Keiji Iwatsuki
- Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kita-ku, Okayama 700-8558
| | - Nam-Ho Huh
- From the Departments of Cell Biology and
| |
Collapse
|
222
|
Natural killer cell function and dysfunction in hepatitis C virus infection. BIOMED RESEARCH INTERNATIONAL 2014; 2014:903764. [PMID: 25057504 PMCID: PMC4095668 DOI: 10.1155/2014/903764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 04/14/2014] [Indexed: 01/06/2023]
Abstract
Viruses must continually adapt against dynamic innate and adaptive responses of the host immune system to establish chronic infection. Only a small minority (~20%) of those exposed to hepatitis C virus (HCV) spontaneously clear infection, leaving approximately 200 million people worldwide chronically infected with HCV. A number of recent research studies suggest that establishment and maintenance of chronic HCV infection involve natural killer (NK) cell dysfunction. This relationship is illustrated in vitro by disruption of typical NK cell responses including both cell-mediated cytotoxicity and cytokine production. Expression of a number of activating NK cell receptors in vivo is also affected in chronic HCV infection. Thus, direct in vivo and in vitro evidence of compromised NK function in chronic HCV infection in conjunction with significant epidemiological associations between the outcome of HCV infection and certain combinations of NK cell regulatory receptor and class I human histocompatibility linked antigen (HLA) genotypes indicate that NK cells are important in the immune response against HCV infection. In this review, we highlight evidence suggesting that selective impairment of NK cell activity is related to establishment of chronic HCV infection.
Collapse
|
223
|
Molfetta R, Quatrini L, Capuano C, Gasparrini F, Zitti B, Zingoni A, Galandrini R, Santoni A, Paolini R. c-Cbl regulates MICA- but not ULBP2-induced NKG2D down-modulation in human NK cells. Eur J Immunol 2014; 44:2761-70. [PMID: 24846123 DOI: 10.1002/eji.201444512] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/14/2014] [Accepted: 05/12/2014] [Indexed: 12/24/2022]
Abstract
The NKG2D activating receptor on human NK cells mediates "altered self" recognition, as its ligands (NKG2DLs) are upregulated on target cells in a variety of stress conditions. Evidence collected in the past years shows that, even though expression of NKG2DLs acts as a danger signal that renders tumor cells susceptible to cytotoxicity, chronic exposure to soluble or membrane-bound NKG2DLs can lead to down-modulation of receptor expression and impairment of NKG2D-mediated cell functions. Here, we evaluated whether different cell-bound NKG2DLs, namely MICA and ULBP2, are equivalently able to induce NKG2D down-modulation on human NK cells. We found that although both ligands reduce NKG2D surface expression, MICA promotes a stronger receptor down-modulation than ULBP2, leading to a severe impairment of NKG2D-dependent NK-cell cytotoxicity. We also provide evidence that the ubiquitin pathway and c-Cbl direct MICA-induced but not ULBP2-induced NKG2D internalization and degradation, thus identifying a molecular mechanism to explain the differential effects of MICA and ULBP2 on NKG2D expression. A better understanding of the molecular mechanisms employed by the different NKG2DLs to control NKG2D surface expression could be useful for the development of anti-tumor strategies to restore a normal level of NKG2D receptors on human NK cells.
Collapse
Affiliation(s)
- Rosa Molfetta
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Serrano-Pertierra E, Cernuda-Morollón E, Brdička T, Hoøejši V, López-Larrea C. L-plastin is involved in NKG2D recruitment into lipid rafts and NKG2D-mediated NK cell migration. J Leukoc Biol 2014; 96:437-45. [PMID: 24803550 DOI: 10.1189/jlb.2a1013-564r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Membrane rafts are microdomains of the plasma membrane that have multiple biological functions. The involvement of these structures in the biology of T cells, namely in signal transduction by the TCR, has been widely studied. However, the role of membrane rafts in immunoreceptor signaling in NK cells is less well known. We studied the distribution of the activating NKG2D receptor in lipid rafts by isolating DRMs in a sucrose density gradient or by raft fractionation by β-OG-selective solubility in the NKL cell line. We found that the NKG2D-DAP10 complex and pVav are recruited into rafts upon receptor stimulation. Qualitative proteomic analysis of these fractions showed that the actin cytoskeleton is involved in this process. In particular, we found that the actin-bundling protein L-plastin plays an important role in the clustering of NKG2D into lipid rafts. Moreover, coengagement of the inhibitory receptor NKG2A partially disrupted NKG2D recruitment into rafts. Furthermore, we demonstrated that L-plastin participates in NKG2D-mediated inhibition of NK cell chemotaxis.
Collapse
Affiliation(s)
| | - Eva Cernuda-Morollón
- Neurology Departments, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Tomáš Brdička
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; and
| | - Václav Hoøejši
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic; and
| | | |
Collapse
|
225
|
Luo J, Tian W, Pan F, Liu X, Li L. Allelic and haplotypic diversity of 5′promoter region of the MICA gene. Hum Immunol 2014; 75:383-8. [DOI: 10.1016/j.humimm.2013.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/08/2013] [Accepted: 12/17/2013] [Indexed: 10/25/2022]
|
226
|
Bezbradica JS, Rosenstein RK, DeMarco RA, Brodsky I, Medzhitov R. A role for the ITAM signaling module in specifying cytokine-receptor functions. Nat Immunol 2014; 15:333-42. [PMID: 24608040 PMCID: PMC4137873 DOI: 10.1038/ni.2845] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 02/11/2014] [Indexed: 12/12/2022]
Abstract
Diverse cellular responses to external cues are controlled by a small number of signal-transduction pathways, but how the specificity of functional outcomes is achieved remains unclear. Here we describe a mechanism for signal integration based on the functional coupling of two distinct signaling pathways widely used in leukocytes: the ITAM pathway and the Jak-STAT pathway. Through the use of the receptor for interferon-γ (IFN-γR) and the ITAM adaptor Fcγ as an example, we found that IFN-γ modified responses of the phagocytic antibody receptor FcγRI (CD64) to specify cell-autonomous antimicrobial functions. Unexpectedly, we also found that in peritoneal macrophages, IFN-γR itself required tonic signaling from Fcγ through the kinase PI(3)K for the induction of a subset of IFN-γ-specific antimicrobial functions. Our findings may be generalizable to other ITAM and Jak-STAT signaling pathways and may help explain signal integration by those pathways.
Collapse
Affiliation(s)
- Jelena S Bezbradica
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rachel K Rosenstein
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Igor Brodsky
- 1] Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA. [2] Present address: Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ruslan Medzhitov
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
227
|
Pan F, Li L, Luo J, Liu X, Tian W. The 5′ promoter region of MHC class I chain-related gene B. ACTA ACUST UNITED AC 2014; 83:337-43. [DOI: 10.1111/tan.12348] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/05/2014] [Accepted: 03/04/2014] [Indexed: 01/07/2023]
Affiliation(s)
- F. Pan
- Immunogenetics Research Group, Department of Immunology, College of Basic Medical Sciences; Central South University; Changsha China
| | - L. Li
- Immunogenetics Research Group, Department of Immunology, College of Basic Medical Sciences; Central South University; Changsha China
| | - J. Luo
- Immunogenetics Research Group, Department of Immunology, College of Basic Medical Sciences; Central South University; Changsha China
| | - X. Liu
- Immunogenetics Research Group, Department of Immunology, College of Basic Medical Sciences; Central South University; Changsha China
| | - W. Tian
- Immunogenetics Research Group, Department of Immunology, College of Basic Medical Sciences; Central South University; Changsha China
| |
Collapse
|
228
|
Huergo-Zapico L, Acebes-Huerta A, López-Soto A, Villa-Álvarez M, Gonzalez-Rodriguez AP, Gonzalez S. Molecular Bases for the Regulation of NKG2D Ligands in Cancer. Front Immunol 2014; 5:106. [PMID: 24711808 PMCID: PMC3968767 DOI: 10.3389/fimmu.2014.00106] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/03/2014] [Indexed: 01/10/2023] Open
Abstract
NKG2D is an activating receptor expressed by NK and T cells primarily involved in the elimination of transformed and infected cells. NKG2D ligands are self-proteins restrictedly expressed in healthy tissues, but induced in response to signaling pathways commonly associated with transformation. Proliferative, tumor suppressor, and stress signaling pathways linked to the tumorigenic process induce the expression of NKG2D ligands, initiating an immune response against the incipient tumor. Nevertheless, the activity of NKG2D ligands is counter-regulated in vivo by the immunoediting of cancer cells, resulting in the expression of multiple mechanisms of immune evasion in advanced tumors. The redundancy of NKG2D ligands, besides increasing the complexity of their regulation, may impair the generation of these immune evasion mechanisms. In this review, we attempt to integrate the mechanisms and pathways involved in the regulation of NKG2D ligand expression in cancer.
Collapse
Affiliation(s)
- Leticia Huergo-Zapico
- Department of Functional Biology, University Institute of Oncology (IUOPA), University of Oviedo , Oviedo , Spain
| | - Andrea Acebes-Huerta
- Department of Functional Biology, University Institute of Oncology (IUOPA), University of Oviedo , Oviedo , Spain
| | - Alejandro López-Soto
- Department of Functional Biology, University Institute of Oncology (IUOPA), University of Oviedo , Oviedo , Spain
| | - Mónica Villa-Álvarez
- Department of Functional Biology, University Institute of Oncology (IUOPA), University of Oviedo , Oviedo , Spain
| | | | - Segundo Gonzalez
- Department of Functional Biology, University Institute of Oncology (IUOPA), University of Oviedo , Oviedo , Spain
| |
Collapse
|
229
|
López-Soto A, Huergo-Zapico L, Acebes-Huerta A, Villa-Alvarez M, Gonzalez S. NKG2D signaling in cancer immunosurveillance. Int J Cancer 2014; 136:1741-50. [DOI: 10.1002/ijc.28775] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/23/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Alejandro López-Soto
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| | | | - Andrea Acebes-Huerta
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| | - Mónica Villa-Alvarez
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| | - Segundo Gonzalez
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| |
Collapse
|
230
|
Hu J, Zhu S, Xia X, Zhang L, Kleinerman ES, Li S. CD8+T cell-specific induction of NKG2D receptor by doxorubicin plus interleukin-12 and its contribution to CD8+T cell accumulation in tumors. Mol Cancer 2014; 13:34. [PMID: 24565056 PMCID: PMC3938086 DOI: 10.1186/1476-4598-13-34] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 02/03/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Increased infiltration of CD8+T cells into tumors has a positive impact on survival. Our previous study showed that doxorubicin (Dox) plus interleukin-12 (IL-12) boosted the accumulation of CD8+T cells in tumors and had a greater antitumor effect than did either agent alone. The purpose of this study was to determine the impact of NKG2D expression on CD8+T cell infiltration and antitumor efficacy. METHODS Tumor-bearing mice were administered Dox, IL-12 plasmid DNA, or both via intraperitoneal injection or intramuscular electroporation. The induction of NKG2D on CD8+T cells and other lymphocytes was analyzed via flow cytometry, and NKG2D-positive CD8+T cell-specific localization in tumors was determined by using immunofluorescence staining in various types of immune cell-depleted mice. RESULTS The combination of Dox plus IL-12 specifically increased expression of NKG2D in CD8+T cells but not in other types of immune cells, including NK cells, which naturally express NKG2D. This induced NKG2D expression in CD8+T cells was associated with increased accumulation of CD8+T cells in murine tumors. Administration of NKG2D-blocking antibody or CD8+T cell-depletion antibody abrogated the NKG2D+CD8+T cell detection in tumors, whereas administration of NK cell-depletion antibody had no effect. Increased NKG2D expression in CD8+T cells was associated with increased antitumor efficacy in vivo. CONCLUSION We conclude that Dox plus IL-12 induces NKG2D in CD8+T cells in vivo and boosts NKG2D+CD8+T-dependent antitumor immune surveillance. This discovery reveals a novel mechanism for how chemoimmunotherapy synergistically promotes T cell-mediated antitumor immune surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | - Shulin Li
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
231
|
Kawano M, Nakayama M, Aoshima Y, Nakamura K, Ono M, Nishiya T, Nakamura S, Takeda Y, Dobashi A, Takahashi A, Endo M, Ito A, Ueda K, Sato N, Higuchi S, Kondo T, Hashimoto S, Watanabe M, Watanabe M, Takahashi T, Sasaki K, Nakamura M, Sasazuki T, Narushima T, Suzuki R, Ogasawara K. NKG2D⁺ IFN-γ⁺ CD8⁺ T cells are responsible for palladium allergy. PLoS One 2014; 9:e86810. [PMID: 24533050 PMCID: PMC3922723 DOI: 10.1371/journal.pone.0086810] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 12/19/2013] [Indexed: 11/18/2022] Open
Abstract
Nickel, cobalt, and chromium are well known to be causal agents of allergic contact dermatitis. Palladium (Pd) can also cause allergic disease and exposure results from wide use of this metal in dental restorations and jewelry. Metal allergy is categorized as a delayed-type hypersensitivity, and metal-responsive T cell clones have been isolated from allergic patients. However, compared to nickel, little is known about the pathology of allergic disease mediated by Pd, and pathogenic T cells are poorly understood. To identify the pathogenic T cells that are responsible for onset of Pd allergy, we enriched metal-responsive lymphocytes by sequential adoptive transfer of involved lymph node cells. Here we show that sequential adoptive transfer gradually increased the incidence and the intensity of Pd allergy, and CD8+ T cells are responsible for the disease as CD8+ T cell-depleted mice and β2-microglobulin-deficient mice did not develop Pd allergy. In addition, we found that draining lymph node cells skewed toward CD8+ T cells in response to Pd challenge in 8th adoptive transferred recipient mice. The CD8+ T cells expressed NKG2D, a costimulatory molecule involved in the production of IFN-γ. NKG2D ligand was also induced in Pd-injected tissues. Furthermore, both NKG2D ligand-transgenic mice, where NKG2D is downmodulated, and IFN-γ-deficient mice showed impaired Pd allergy. Taken together, these results indicate that IFN-γ-producing NKG2D+ CD8+ T cells are responsible for Pd allergy and suggest that NKG2D is a potential therapeutic target for treatment of metal allergy.
Collapse
Affiliation(s)
- Mitsuko Kawano
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Masafumi Nakayama
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Yusuke Aoshima
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, Aramakiaza, Aoba-ku, Sendai, Miyagi, Japan
| | - Kyohei Nakamura
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Mizuho Ono
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Tadashi Nishiya
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Syou Nakamura
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Yuri Takeda
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Graduate School of Dentistry, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Akira Dobashi
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Akiko Takahashi
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Misato Endo
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Akiyo Ito
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Kyosuke Ueda
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, Aramakiaza, Aoba-ku, Sendai, Miyagi, Japan
| | - Naoki Sato
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Shigehito Higuchi
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Takeru Kondo
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Suguru Hashimoto
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Masamichi Watanabe
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Makoto Watanabe
- Graduate School of Dentistry, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Tetsu Takahashi
- Graduate School of Dentistry, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Keiichi Sasaki
- Graduate School of Dentistry, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Masanori Nakamura
- Graduate School of Dentistry, Showa University, Shinagawa-ku, Tokyo, Japan
| | - Takehiko Sasazuki
- Institute for Advanced Study, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Takayuki Narushima
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, Aramakiaza, Aoba-ku, Sendai, Miyagi, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Minami-ku, Sagamihara, Kanagawa, Japan
| | - Kouetsu Ogasawara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- * E-mail:
| |
Collapse
|
232
|
Divergence and diversity of ULBP2 genes in rhesus and cynomolgus macaques. Immunogenetics 2014; 66:161-70. [PMID: 24469065 DOI: 10.1007/s00251-014-0760-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 01/13/2014] [Indexed: 10/25/2022]
Abstract
Non-human primates such as rhesus macaque and cynomolgus macaque are important animals for medical research fields and they are classified as Old World monkey, in which genome structure is characterized by gene duplications. In the present study, we investigated polymorphisms in two genes for ULBP2 molecules that are ligands for NKG2D. A total of 15 and 11 ULBP2.1 alleles and 11 and 10 ULBP2.2 alleles were identified in rhesus macaques and cynomolgus macaques, respectively. Nucleotide sequences of exons for extra cellular domain were highly polymorphic and more than 70 % were non-synonymous variations in both ULBP2.1 and ULBP2.2. In addition, phylogenetic analyses revealed that the ULBP2.2 was diverged from a branch of ULBP2.1 along with ULBP2s of higher primates. Moreover, when 3D structural models were constructed for the rhesus ULBP2 molecules, residues at presumed contact sites with NKG2D were polymorphic in ULBP2.1 and ULBP2.2 in the rhesus macaque and cynomolgus macaque, respectively. These observations suggest that amino acid replacements at the interaction sites with NKG2D might shape a specific nature of ULBP2 molecules in the Old World monkeys.
Collapse
|
233
|
Chen DY, Yao L, Chen YM, Lin CC, Huang KC, Chen ST, Lan JL, Hsieh SLE. A potential role of myeloid DAP12-associating lectin (MDL)-1 in the regulation of inflammation in rheumatoid arthritis patients. PLoS One 2014; 9:e86105. [PMID: 24465901 PMCID: PMC3897620 DOI: 10.1371/journal.pone.0086105] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 12/06/2013] [Indexed: 12/14/2022] Open
Abstract
The pathogenic roles of myeloid DAP12-associating lectin-1(MDL-1) and DAP12 in human rheumatoid arthritis (RA) remain unknown. Frequencies of MDL-1-expressing monocytes in 22 active RA patients, 16 inactive RA patients, 12 osteoarthritis (OA) patients and 10 healthy controls (HC) were determined by flow-cytometry analysis. The mRNA expression levels of MDL-1 and DAP12 on PBMCs were evaluated by quantitative PCR, and their protein expression levels in the synovium were examined by immunohistochemistry. Significantly higher median percentages of circulating MDL-1-expressing monocytes were observed in active RA patients (53.6%) compared to inactive RA patients (34.1%), OA patients (27.9%), and HC (21.2%). Levels of MDL-1 and DAP12 gene expression in PBMCs and their protein expression in the synovium were significantly higher in active RA patients than in inactive RA or OA patients. MDL-1 levels were positively correlated with parameters of disease activity, articular damage, and levels of proinflammatory cytokines. MDL-1 activator (Dengue virus type 2 antigen) stimulation on PBMCs resulted in significantly enhanced levels of proinflammatory cytokines in RA patients compared to those in OA patients or HC, indicating that MDL-1 activation is functional. Frequencies of MDL-1-expressing monocytes and levels of MDL-1 and DAP12 gene expression significantly decreased after effective therapy. Concordant overexpression of MDL-1 and DAP12 were correlated with increased production of proinflammatory cytokines in RA patients, suggesting their roles in regulating articular inflammation.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Arthritis, Rheumatoid/blood
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- C-Reactive Protein/metabolism
- Cytokines/blood
- Female
- Flow Cytometry
- Gene Expression Regulation
- Humans
- Immunohistochemistry
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation Mediators/blood
- Joints/pathology
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Leukocytes, Mononuclear/metabolism
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Middle Aged
- Monocytes/metabolism
- Monocytes/pathology
- Osteoarthritis/genetics
- Osteoarthritis/metabolism
- Osteoarthritis/pathology
- Polymerase Chain Reaction
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
Collapse
Affiliation(s)
- Der-Yuan Chen
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C.
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan, R.O.C.
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
- Infection and Immunity Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C.
| | - Ling Yao
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C.
| | - Yi-Ming Chen
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C.
| | - Chi-Chen Lin
- Institute of Biomedical Science, National Chung-Hsing University, Taichung, Taiwan, R.O.C.
| | - Kui-Chou Huang
- Department of Orthopedics and Traumatology, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C.
| | - Szu-Ting Chen
- Infection and Immunity Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C.
| | - Joung-Liang Lan
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C.
- Division of Immunology and Rheumatology, China Medical University Hospital, Taichung, Taiwan, R.O.C.
- College of Chinese Medicine, China Medical University, Taichung, Taiwan, R.O.C.
- * E-mail: (JLL); (SLEH)
| | - Shie-Liang Edmond Hsieh
- Infection and Immunity Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei City, Taiwan
- * E-mail: (JLL); (SLEH)
| |
Collapse
|
234
|
Le Bert N, Gasser S. Advances in NKG2D ligand recognition and responses by NK cells. Immunol Cell Biol 2014; 92:230-6. [PMID: 24445601 DOI: 10.1038/icb.2013.111] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 02/06/2023]
Abstract
The natural killer (NK) group 2 member D (NKG2D) is an activating immune receptor expressed on NK cells, cytotoxic T cells and a subset of other T cells. It has an important role in the recognition and lysis of a variety of infected and tumor cells. Despite significant gains in our understanding of NKG2D, the relevance of NKG2D and its ligands in human diseases has only recently started to emerge. Here, we present an overview of the recent advances in NKG2D biology, discuss the expression of NKG2D ligands in cancer patients and evaluate the diagnostic and prognostic potential of NKG2D ligands.
Collapse
Affiliation(s)
- Nina Le Bert
- Immunology Programme, Department of Microbiology, National University of Singapore, Singapore
| | - Stephan Gasser
- Immunology Programme, Department of Microbiology, National University of Singapore, Singapore
| |
Collapse
|
235
|
|
236
|
Marcus A, Gowen BG, Thompson TW, Iannello A, Ardolino M, Deng W, Wang L, Shifrin N, Raulet DH. Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol 2014; 122:91-128. [PMID: 24507156 PMCID: PMC4228931 DOI: 10.1016/b978-0-12-800267-4.00003-1] [Citation(s) in RCA: 273] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, roles of the immune system in immune surveillance of cancer have been explored using a variety of approaches. The roles of the adaptive immune system have been a major emphasis, but increasing evidence supports a role for innate immune effector cells such as natural killer (NK) cells in tumor surveillance. Here, we discuss some of the evidence for roles in tumor surveillance of innate immune cells. In particular, we focus on NK cells and other immune cells that express germline-encoded receptors, often labeled NK receptors. The impact of these receptors and the cells that express them on tumor suppression is summarized. We discuss in detail some of the pathways and events in tumor cells that induce or upregulate cell-surface expression of the ligands for these receptors, and the logic of how those pathways serve to identify malignant, or potentially malignant cells. How tumors often evade tumor suppression mediated by innate killer cells is another major subject of the review. We end with a discussion on some of the implications of the various findings with respect to possible therapeutic approaches.
Collapse
Affiliation(s)
- Assaf Marcus
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Benjamin G Gowen
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Thornton W Thompson
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Alexandre Iannello
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Michele Ardolino
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Weiwen Deng
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Lin Wang
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Nataliya Shifrin
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - David H Raulet
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA.
| |
Collapse
|
237
|
The role of T cell immunoglobulin mucin domains 1 and 4 in a herpes simplex virus-induced Behçet's disease mouse model. Mediators Inflamm 2013; 2013:903948. [PMID: 24453431 PMCID: PMC3888750 DOI: 10.1155/2013/903948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/24/2013] [Accepted: 11/18/2013] [Indexed: 01/04/2023] Open
Abstract
The T cell immunoglobulin mucin (TIM) proteins regulate T cell activation and tolerance. TIM-1 plays an important role in the regulation of immune responses and the development of autoimmune diseases. TIM-4 is a natural ligand of TIM-1, and the interaction of TIM-1 and TIM-4 is involved in the regulation of T helper (Th) cell responses and modulation of the Th1/Th2 cytokine balance. Behçet's disease (BD) is a chronic, multisystemic inflammatory disorder with arthritic, intestinal, mucocutaneous, ocular, vascular, and central nervous system involvement. Tim-1 expression was lower in a herpes simplex virus-induced BD mouse model compared to that in asymptomatic BD normal (BDN) mice. Tim-4 expression was higher in BD mice than that in BDN mice. In this study, we investigated the Tim expression in a BD mouse model with BD-like symptoms. Tim-1 and Tim-4 expression was regulated by an expression vector or siRNA injected into the BD mouse model. The Tim-1 vector injected into BD mice resulted in changes in BD-like symptoms and decreased the severity score. Treatment with Tim-4 siRNA also improved BD-like symptoms and decreased the severity score accompanied by upregulation of regulatory T cells. We showed that regulating Tim-1 or Tim-4 affected BD-like symptoms in mice.
Collapse
|
238
|
Rothe A, Jachimowicz RD, Borchmann S, Madlener M, Keßler J, Reiners KS, Sauer M, Hansen HP, Ullrich RT, Chatterjee S, Borchmann P, Yazaki P, Koslowsky TC, Engert A, Heukamp LC, Hallek M, von Strandmann EP. The bispecific immunoligand ULBP2-aCEA redirects natural killer cells to tumor cells and reveals potent anti-tumor activity against colon carcinoma. Int J Cancer 2013; 134:2829-40. [PMID: 24242212 DOI: 10.1002/ijc.28609] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 10/25/2013] [Indexed: 02/03/2023]
Abstract
NKG2D, an activating receptor expressed on NK cells and T cells, is critically involved in tumor immunosurveillance. In this study, we explored the potential therapeutic utility of the NKG2D ligand ULBP2 for the treatment of colon carcinoma. To this end we designed a fusion protein consisting of human ULBP2 and an antibody-derived single chain targeting the tumor carcinoembryonic antigen (CEA). The bispecific recombinant fusion protein re-directed NK cells towards malignant cells by binding to both, tumor cells and NK cells, and triggered NK cell-mediated target cell killing in vitro. Moreover, tumor growth was significantly delayed in a syngeneic colon carcinoma mouse model in response to immunoligand treatment. The anti-tumor activity could be attributed to the stimulation of immune cells with an elevated expression of the activation marker CD69 on NK, T and NKT cells and the infiltration of CD45+ immune cells into the solid tumor. In summary, it was demonstrated that immunoligands provide specific tumor targeting by NK cells and exert anti-tumor activity in vitro and in vivo. This technology represents a novel immunotherapeutic strategy for solid tumors with the potential to be further developed for clinical applications.
Collapse
Affiliation(s)
- Achim Rothe
- Department I of Internal Medicine, Innate Immunity Group, University Hospital Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
The narrow-spectrum HDAC inhibitor entinostat enhances NKG2D expression without NK cell toxicity, leading to enhanced recognition of cancer cells. Pharm Res 2013; 32:779-92. [PMID: 24203492 DOI: 10.1007/s11095-013-1231-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 10/14/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE Natural killer (NK) cell cytotoxicity correlates with the ligation of activating receptors (e.g., NKG2D) by their ligands (e.g., MHC class I-related chains [MIC] A and B) on target cells. Histone deacetylase inhibitors (HDACi) at high concentrations inhibit tumor growth and can increase NKG2D ligand expression on tumor targets, but are widely regarded as toxic to NK cells. METHODS We investigated the mechanism of entinostat, a benzamide-derivative narrow-spectrum HDACi, in augmenting the cytotoxicity of NK cells against human colon carcinoma and sarcoma by assessing gene and protein expression, histone acetylation, and cytotoxicity in in vitro and murine models. RESULTS We observed that entinostat dose- and time-dependent increase in MIC expression in tumor targets and NKG2D in primary human NK cells, both correlating with increased acetylated histone 3 (AcH3) binding to associated promoters. Entinostat pretreatment of colon carcinoma and sarcoma cells, NK cells, or both led to enhanced overall cytotoxicity in vitro, which was reversed by NKG2D blockade, and inhibited growth of tumor xenografts. Lastly, we showed decreased expression of MICA and ULBP2 transcription in primary human osteosarcoma. CONCLUSIONS Entinostat enhances NK cell killing of cancer cells through upregulation of both NKG2D and its ligands, suggesting an attractive approach for augmenting NK cell immunotherapy of solid tumors such as colon carcinoma and sarcomas.
Collapse
|
240
|
Serrano-Pertierra E, Cernuda-Morollón E, López-Larrea C. NKG2D- and CD28-mediated costimulation regulate CD8+
T cell chemotaxis through different mechanisms: the role of Cdc42/N-WASp. J Leukoc Biol 2013; 95:487-95. [DOI: 10.1189/jlb.0613316] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
241
|
Ullrich E, Koch J, Cerwenka A, Steinle A. New prospects on the NKG2D/NKG2DL system for oncology. Oncoimmunology 2013; 2:e26097. [PMID: 24353908 PMCID: PMC3862635 DOI: 10.4161/onci.26097] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 08/06/2013] [Accepted: 08/09/2013] [Indexed: 11/19/2022] Open
Abstract
The activating immunoreceptor NKG2D endows cytotoxic lymphocytes with the capacity to recognize and eliminate infected or malignant cells. The recognition of such harmful cells is enabled by binding of NKG2D to various MHC class I-related glycoproteins, which are upregulated in the course of viral infection or malignant transformation. The past years have witnessed substantial progress in our understanding of the mechanisms underlying the regulation of NKG2D ligands (NKG2DLs) by malignant cells, of tumor-associated countermeasures promoting escape from NKG2D-dependent immunosurveillance, and of therapeutic measures that may bolster the NKG2D/NKG2DL system against malignancies. Here, we summarize the current knowledge on the NKG2D/NKG2DL system and outline opportunities to exploit the tumoricidal function of NKG2D for anticancer immunotherapy.
Collapse
Affiliation(s)
- Evelyn Ullrich
- Children's Hospital; Department of Pediatric Hematology and Oncology; Goethe-University Frankfurt am Main; Frankfurt am Main, Germany ; Center for Cell and Gene Therapy; Goethe University Frankfurt am Main; Frankfurt am Main, Germany
| | - Joachim Koch
- Center for Cell and Gene Therapy; Goethe University Frankfurt am Main; Frankfurt am Main, Germany ; Institute for Biomedical Research: Georg-Speyer-Haus; NK Cell Biology; Frankfurt am Main, Germany
| | - Adelheid Cerwenka
- German Cancer Research Center (DKFZ); Innate Immunity Group; Heidelberg, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine; Goethe-University Frankfurt am Main; Frankfurt am Main, Germany
| |
Collapse
|
242
|
Luo J, Tian W, Liu XX, Yu J, Li L, Pan F. The genetic architecture of 3′untranslated region of the MICA gene: Polymorphisms and haplotypes. Hum Immunol 2013; 74:1321-5. [DOI: 10.1016/j.humimm.2013.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/24/2013] [Accepted: 06/14/2013] [Indexed: 12/01/2022]
|
243
|
Hagberg N, Theorell J, Eloranta ML, Pascal V, Bryceson YT, Rönnblom L. Anti-NKG2A autoantibodies in a patient with systemic lupus erythematosus. Rheumatology (Oxford) 2013; 52:1818-23. [PMID: 23825044 DOI: 10.1093/rheumatology/ket220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
OBJECTIVES To characterize a novel anti-NKG2A autoantibody detected in a patient with SLE during a severe flare, and in a cross-sectional study investigate the occurrence of such autoantibodies in patients with SLE and primary SS (pSS). METHODS Serum or IgG from patients with SLE, pSS and healthy volunteers were assayed for blocking of anti-NKG2A or HLA-E binding to peripheral blood mononuclear cells or CD94/NKG2A- and CD94/NKG2C-transfected Ba/F3 cells. The anti-NKG2A autoantibodies were evaluated for effect on NK cell degranulation in response to HLA-E-transfected K562 cells. IFN-α was determined by an immunoassay and disease activity by the SLEDAI score. RESULTS Anti-NKG2A autoantibodies, which blocked binding of HLA-E tetramers to CD94/NKG2A-transfected cells and impaired NKG2A-mediated inhibition of NK cell activation, were observed in a patient with SLE. The presence of anti-NKG2A autoantibodies was associated with high SLE disease activity (SLEDAI score 14 and 16) and increased serum IFN-α. Of 94 SLE, 60 pSS and 30 healthy donor sera, only the index patient serum contained anti-NKG2A autoantibodies. CONCLUSION The presence of autoantibodies targeting NKG2A is a rare event, but when such autoantibodies occur they may promote excessive NK cell function. This can contribute to the pathogenesis by increasing the killing of cells and the release of autoantigens. Our findings highlight the possible importance of NK cells in the SLE disease process.
Collapse
Affiliation(s)
- Niklas Hagberg
- Department of Medical Sciences, Section of Rheumatology, Systemic Autoimmunity Group, Clinical Research Department 3, Entrance 85 3rd Floor, 751 85 Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
244
|
Chitadze G, Bhat J, Lettau M, Janssen O, Kabelitz D. Generation of soluble NKG2D ligands: proteolytic cleavage, exosome secretion and functional implications. Scand J Immunol 2013; 78:120-9. [PMID: 23679194 DOI: 10.1111/sji.12072] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/14/2013] [Indexed: 12/23/2022]
Abstract
The activating natural killer group 2 member D (NKG2D) receptor is expressed on NK cells, cytotoxic T cells and additional T cell subsets. Ligands for human NKG2D comprise two groups of MHC class I-related molecules, the MHC class I chain-related proteins A and B (MICA/B) and 6 UL16-binding proteins (ULBP1-6). While NKG2D ligands are absent from most normal cells, expression is induced upon stress and malignant transformation. In fact, most solid tumours and leukaemia/lymphomas constitutively express at least one NKG2D ligand and thereby are susceptible to NKG2D-dependent immunosurveillance. However, soluble NKG2D ligands are released from tumour cells and can down-modulate NKG2D activation as a means of tumour immune escape. In some tumour entities, levels of soluble NKG2D ligands in the serum correlate with tumour progression. NKG2D ligands can be proteolytically shed from the cell surface or liberated from the membrane by phospholipase C in the case of glycosylphosphatidylinositol (GPI)-anchored molecules. Moreover, NKG2D ligands can be secreted in exosomal microvesicles together with other tumour-derived molecules. Depending on the specific tumour/immune cell setting, these various forms of soluble and/or exosome-bound NKG2D ligands can exert multiple effects on NKG2D/NKG2D ligand interactions. In this review, we focus on the role of various proteases in the shedding of human NKG2D ligands from tumour cells and discuss the not completely unanimous reported functional implications of soluble and exosome-secreted NKG2D ligands for immunosurveillance.
Collapse
Affiliation(s)
- G Chitadze
- Institute of Immunology, University of Kiel, Kiel, Germany
| | | | | | | | | |
Collapse
|
245
|
Signaling by Fyn-ADAP via the Carma1-Bcl-10-MAP3K7 signalosome exclusively regulates inflammatory cytokine production in NK cells. Nat Immunol 2013; 14:1127-36. [PMID: 24036998 PMCID: PMC3855032 DOI: 10.1038/ni.2708] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/09/2013] [Indexed: 11/29/2022]
Abstract
Inflammation is a critical component of the immune response. However, acute or chronic inflammation can be highly destructive. Uncontrolled inflammation forms the basis for allergy, asthma, and multiple autoimmune disorders. Here, we identify a signaling pathway that is exclusively responsible for inflammatory cytokine production but not for cytotoxicity. Recognition of H60+ or CD137L+ tumor cells by murine NK cells led to efficient cytotoxicity and inflammatory cytokine production. Both of these effector functions required Lck, Fyn, PI(3)K-p85α, PI(3)K-p110δ, and PLC-γ2. However, the complex of Fyn and the adapter ADAP exclusively regulated inflammatory cytokine production but not cytotoxicity in NK cells. This unique function of ADAP required a Carma1-Bcl10-MAP3K7 signaling axis. Our results identify molecules that can be targeted to regulate inflammation without compromising NK cell cytotoxicity.
Collapse
|
246
|
Hosomi S, Chen Z, Baker K, Chen L, Huang YH, Olszak T, Zeissig S, Wang JH, Mandelboim O, Beauchemin N, Lanier LL, Blumberg RS. CEACAM1 on activated NK cells inhibits NKG2D-mediated cytolytic function and signaling. Eur J Immunol 2013; 43:2473-83. [PMID: 23696226 PMCID: PMC3775953 DOI: 10.1002/eji.201242676] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 04/09/2013] [Accepted: 05/17/2013] [Indexed: 12/29/2022]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is expressed on activated natural killer (NK) cells wherein it inhibits lysis of CEACAM1-bearing tumor cell lines. The mechanism for this is unknown. Here, we show that interleukin-2-induced expression of CEACAM1 on both mouse and primary human NK cells impairs the ability of NK gene complex group 2 member D (NKG2D) to stimulate cytolysis of CEACAM1-bearing cells. This process requires the expression of CEACAM1 on the NK cells and on the tumor cells, which is consistent with the involvement of trans-homophilic interactions between CEACAM1. Mechanistically, co-engagement of NKG2D and CEACAM1 results in a biochemical association between these two surface receptors and the recruitment of Src homology phosphatase 1 by CEACAM1 that leads to dephosphorylation of the guanine nucleotide exchange factor Vav1 and blockade of downstream signaling that is associated with the initiation of cytolysis. Thus, CEACAM1 on activated NK cells functions as an inhibitory receptor for NKG2D-mediated cytolysis, which has important implications for understanding the means by which CEACAM1 expression adversely affects tumor immunity.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhangguo Chen
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Immunology, National Jewish Health, University of Colorado Denver, Denver, CO, USA
| | - Kristi Baker
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lanfen Chen
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Yu-Hwa Huang
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Torsten Olszak
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastian Zeissig
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing H. Wang
- Department of Immunology, National Jewish Health, University of Colorado Denver, Denver, CO, USA
| | - Ofer Mandelboim
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Nicole Beauchemin
- Goodman Cancer Research Centre and Depts. of Biochemistry, Medicine and Oncology, McGill University, Montreal, Canada
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and the Cancer Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
247
|
NKG2D triggers cytotoxicity in murine epidermal γδ T cells via PI3K-dependent, Syk/ZAP70-independent signaling pathway. J Invest Dermatol 2013; 134:396-404. [PMID: 23962808 DOI: 10.1038/jid.2013.353] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 07/17/2013] [Accepted: 07/29/2013] [Indexed: 01/29/2023]
Abstract
Murine epidermal γδ T cells, known as dendritic epidermal T cells (DETCs), survey tissue stress through the invariant T-cell receptor (TCR) and non-clonotypic receptors such as NKG2D. NKG2D signaling via the DAP10-phosphatidylinositol 3-kinase (PI3K) pathway directly stimulates cytotoxicity in natural killer (NK) cells and costimulates CD8(+) T cells to augment TCR signals. In activated murine NK cells, NKG2D signals also via the DAP12-Syk/ZAP70 pathway that triggers both cytotoxicity and cytokine production. It remains controversial whether NKG2D on DETCs is a primary activating receptor or functions only as a costimulatory receptor, and signaling pathways initiated by NKG2D ligation in DETCs have not been analyzed. We show that stimulation of short-term DETC lines with recombinant NKG2D ligands triggers degranulation (exocytosis of cytotoxic granules) via the PI3K-dependent signaling pathway, but does not induce cytokine production or Syk/ZAP70 activation. Coengagement of TCR or Syk/ZAP70 signaling was not crucial for DETC-mediated killing of NKG2D ligand-expressing target cells. Thus, NKG2D can function as a coactivating stress receptor that directly triggers cytotoxicity in DETCs, at least after priming, via the PI3K-dependent, Syk/ZAP70-independent signaling pathway.
Collapse
|
248
|
Holder KA, Stapleton SN, Gallant ME, Russell RS, Grant MD. Hepatitis C virus-infected cells downregulate NKp30 and inhibit ex vivo NK cell functions. THE JOURNAL OF IMMUNOLOGY 2013; 191:3308-18. [PMID: 23960237 DOI: 10.4049/jimmunol.1300164] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepatitis C virus (HCV) successfully evades the immune system and establishes chronic infection in ∼80% of cases. Immune evasion may involve modulating NK cell functions. Therefore, we developed a short-term assay to assess immediate effects of HCV-infected cells on ex vivo NK cytotoxicity and cytokine production. Natural cytotoxicity, Ab-dependent cell-mediated cytotoxicity, IFN-γ production, and TNF-α production were all significantly inhibited by short-term direct exposure to HCV-infected hepatoma-derived Huh-7.5 cells. Inhibition required cell-to-cell contact and increased together with multiplicity of infection and HCV protein levels. Blocking potential interaction between HCV E2 and NK CD81 did not abrogate NK cell inhibition mediated by HCV-infected cells. We observed no change in expression levels of NKG2D, NKG2A, NKp46, or CD16 on NK cells exposed to HCV-infected Huh-7.5 cells for 5 h or of human histocompatibility-linked leukocyte Ag E on HCV-infected compared with uninfected Huh-7.5 cells. Inhibition of ex vivo NK functions did correspond with reduced surface expression of the natural cytotoxicity receptor NKp30, and downregulation of NKp30 was functionally reflected in reduced anti-NKp30 redirected lysis of P815 cells. Infection of Huh-7.5 cells with HCV JFH1(T) increased surface binding of an NKp30-IgG1 Fcγ fusion protein, suggesting upregulation of an antagonistic NKp30 ligand on HCV-infected cells. Our assay demonstrates rapid inhibition of critical NK cell functions by HCV-infected cells. Similar localized effects in vivo may contribute to establishment of chronic HCV infection and associated phenotypic and functional changes in the NK population.
Collapse
Affiliation(s)
- Kayla A Holder
- Division of BioMedical Sciences, Immunology and Infectious Diseases Program, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland A1B 3V6, Canada
| | | | | | | | | |
Collapse
|
249
|
Jarduli LR, Sell AM, Reis PG, Sippert EÂ, Ayo CM, Mazini PS, Alves HV, Teixeira JJV, Visentainer JEL. Role of HLA, KIR, MICA, and cytokines genes in leprosy. BIOMED RESEARCH INTERNATIONAL 2013; 2013:989837. [PMID: 23936864 PMCID: PMC3722889 DOI: 10.1155/2013/989837] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/16/2013] [Accepted: 06/05/2013] [Indexed: 01/08/2023]
Abstract
Many genes including HLA, KIR, and MICA genes, as well as polymorphisms in cytokines have been investigated for their role in infectious disease. HLA alleles may influence not only susceptibility or resistance to leprosy, but also the course of the disease. Some combinations of HLA and KIR may result in negative as well as positive interactions between NK cells and infected host cells with M. leprae, resulting in activation or inhibition of NK cells and, consequently, in death of bacillus. In addition, studies have demonstrated the influence of MICA genes in the pathogenesis of leprosy. Specifically, they may play a role in the interaction between NK cells and infected cells. Finally, pro- and anti-inflammatory cytokines have been influencing the clinical course of leprosy. Data from a wide variety of sources support the existence of genetic factors influencing the leprosy pathogenesis. These sources include twin studies, segregation analyses, family-based linkage and association studies, candidate gene association studies, and, most recently, genome-wide association studies (GWAS). The purpose of this brief review was to highlight the importance of some immune response genes and their correlation with the clinical forms of leprosy, as well as their implications for disease resistance and susceptibility.
Collapse
Affiliation(s)
- Luciana Ribeiro Jarduli
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Ana Maria Sell
- Basic Health Sciences Department, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Pâmela Guimarães Reis
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Emília Ângela Sippert
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Christiane Maria Ayo
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Priscila Saamara Mazini
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Hugo Vicentin Alves
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | - Jorge Juarez Vieira Teixeira
- Program of Biosciences Applied to Pharmacy, Department of Clinical Analysis and Biomedicine, Maringa State University, Avenida Colombo 5790, 87020-900 Maringá, PR, Brazil
| | | |
Collapse
|
250
|
Characterization of 3′untranslated region (3′UTR) of the MICB gene. Hum Immunol 2013; 74:746-50. [DOI: 10.1016/j.humimm.2013.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/28/2012] [Accepted: 01/24/2013] [Indexed: 01/27/2023]
|