201
|
Xuan XY, Zhang JF, Hu GM, Li QR, Liu PP, Du Y. Upregulated expression of NKG2D and its ligands give potential therapeutic targets for patients with thymoma. Cancer Gene Ther 2015; 22:368-74. [PMID: 26113176 DOI: 10.1038/cgt.2015.29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 02/06/2023]
Abstract
The activating receptor NKG2D (natural killer group 2, member D) of natural killer (NK) cells promotes tumor immune surveillance by targeting ligands selectively induced on cancer cells, and thus having an important role in antitumor immune response. Because these ligands are not widely expressed on healthy adult tissue, NKG2D ligands may present as useful target for immunotherapeutic approaches in cancer. In this study, to elucidate the role of NKG2D-NKG2D ligand interaction in thymoma tissues and to evaluate the potential role of NKG2D ligands as therapeutic target for thymoma, we examined the expression of NKG2D and its specific ligands: MICA (major histocompatibility complex class I chain-related protein A), MICB (major histocompatibility complex class I chain-related protein B) and ULBP (UL16-binding protein) in 36 thymomas (6 subtype A, 6 subtype AB, 8 subtype B1, 5 subtype B2, 6 subtype B3 and 5 subtype C), 15 thymic atrophy and 8 thymic hyperplasia by immunohistochemistry and reverse transcription-real-time-PCR methods. We demonstrated that both mRNA and protein levels of NKG2D, MICA, MICB and ULBP were upregulated in six types of thymomas compared with those in atrophic thymus or proliferating thymus. Furthermore, the NKG2D ligands were found to be frequently coexpressed on thymoma cells. Furthermore, the expression of MICA, MICB and ULBP in subtype C was higher compared with those in subtype A, AB, B1, B2 and B3. Thus, we concluded that high expressions of NKG2D, MICA, MICB and ULBP1 were shown in patients with thymoma, and this may enhance the recognition function of NK cells to eliminate tumor cells. MICA, MICB and ULBP presented an attractive target for thymoma therapy. The abnormal expression of NKG2D, MICA, MICB and ULBP1 can provide us with evidence of the occurrence of thymoma and could also be used as a target in the treatment of thymoma.
Collapse
Affiliation(s)
- X Y Xuan
- Department of Immunology and Microbiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - J F Zhang
- Department of Laboratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - G M Hu
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Q R Li
- Department of Immunology and Microbiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - P P Liu
- Department of Immunology and Microbiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Y Du
- Department of Immunology and Microbiology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
202
|
A Peptide Mimetic of 5-Acetylneuraminic Acid-Galactose Binds with High Avidity to Siglecs and NKG2D. PLoS One 2015; 10:e0130532. [PMID: 26110603 PMCID: PMC4482410 DOI: 10.1371/journal.pone.0130532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 05/21/2015] [Indexed: 01/01/2023] Open
Abstract
We previously identified several peptide sequences that mimicked the terminal sugars of complex glycans. Using plant lectins as analogs of lectin-type cell-surface receptors, a tetravalent form of a peptide with the sequence NPSHPLSG, designated svH1C, bound with high avidity to lectins specific for glycans with terminal 5-acetylneuraminic acid (Neu5Ac)-galactose (Gal)/N-acetylgalactosamine (GalNAc) sequences. In this report, we show by circular dichroism and NMR spectra that svH1C lacks an ordered structure and thus interacts with binding sites from a flexible conformation. The peptide binds with high avidity to several recombinant human siglec receptors that bind preferentially to Neu5Ac(α2,3)Gal, Neu5Ac(α2,6)GalNAc or Neu5Ac(α2,8)Neu5Ac ligands. In addition, the peptide bound the receptor NKG2D, which contains a lectin-like domain that binds Neu5Ac(α2,3)Gal. The peptide bound to these receptors with a KD in the range of 0.6 to 1 μM. Binding to these receptors was inhibited by the glycoprotein fetuin, which contains multiple glycans that terminate in Neu5Ac(α2,3)Gal or Neu5Ac(α2,6)Gal, and by sialyllactose. Binding of svH1C was not detected with CLEC9a, CLEC10a or DC-SIGN, which are lectin-type receptors specific for other sugars. Incubation of neuraminidase-treated human peripheral blood mononuclear cells with svH1C resulted in binding of the peptide to a subset of the CD14+ monocyte population. Tyrosine phosphorylation of siglecs decreased dramatically when peripheral blood mononuclear cells were treated with 100 nM svH1C. Subcutaneous, alternate-day injections of svH1C into mice induced several-fold increases in populations of several types of immune cells in the peritoneal cavity. These results support the conclusion that svH1C mimics Neu5Ac-containing sequences and interacts with cell-surface receptors with avidities sufficient to induce biological responses at low concentrations. The attenuation of inhibitory receptors suggests that svH1C has characteristics of a checkpoint inhibitor.
Collapse
|
203
|
Zingoni A, Cecere F, Vulpis E, Fionda C, Molfetta R, Soriani A, Petrucci MT, Ricciardi MR, Fuerst D, Amendola MG, Mytilineos J, Cerboni C, Paolini R, Cippitelli M, Santoni A. Genotoxic Stress Induces Senescence-Associated ADAM10-Dependent Release of NKG2D MIC Ligands in Multiple Myeloma Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:736-48. [PMID: 26071561 DOI: 10.4049/jimmunol.1402643] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 05/06/2015] [Indexed: 01/10/2023]
Abstract
Genotoxic stress can promote antitumor NK cell responses by upregulating the surface expression of activating ligands on cancer cells. Moreover, a number of studies suggested a role for soluble NK group 2D ligands in the impairment of NK cell tumor recognition and killing. We investigated whether genotoxic stress could promote the release of NK group 2D ligands (MHC class I-related chain [MIC]A and MICB), as well as the molecular mechanisms underlying this event in human multiple myeloma (MM) cells. Our results show that genotoxic agents used in the therapy of MM (i.e., doxorubicin and melphalan) selectively affect the shedding of MIC molecules that are sensitive to proteolytic cleavage, whereas the release of the short MICA*008 allele, which is frequent in the white population, is not perturbed. In addition, we found that a disintegrin and metalloproteinase 10 expression is upregulated upon chemotherapeutic treatment both in patient-derived CD138(+)/CD38(+) plasma cells and in several MM cell lines, and we demonstrate a crucial role for this sheddase in the proteolytic cleavage of MIC by means of silencing and pharmacological inhibition. Interestingly, the drug-induced upregulation of a disintegrin and metalloproteinase 10 on MM cells is associated with a senescent phenotype and requires generation of reactive oxygen species. Moreover, the combined use of chemotherapeutic drugs and metalloproteinase inhibitors enhances NK cell-mediated recognition of MM cells, preserving MIC molecules on the cell surface and suggesting that targeting of metalloproteinases in conjunction with chemotherapy could be exploited for NK cell-based immunotherapeutic approaches, thus contributing to avoid the escape of malignant cells from stress-elicited immune responses.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy;
| | - Francesca Cecere
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Elisabetta Vulpis
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Maria Teresa Petrucci
- Department of Cellular Biotechnology and Hematology, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Maria Rosaria Ricciardi
- Department of Cellular Biotechnology and Hematology, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Daniel Fuerst
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm of the German Red Cross Blood Transfusion Service, Baden Wuerttemberg-Hessen, 89081 Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, 89081 Ulm, Germany; and
| | | | - Joannis Mytilineos
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm of the German Red Cross Blood Transfusion Service, Baden Wuerttemberg-Hessen, 89081 Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, 89081 Ulm, Germany; and
| | - Cristina Cerboni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Angela Santoni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy; Institute Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
204
|
Cox ST, Laza-Briviesca R, Pearson H, Soria B, Gibson D, Gomez S, Madrigal JA, Saudemont A. Umbilical cord blood plasma contains soluble NKG2D ligands that mediate loss of natural killer cell function and cytotoxicity. Eur J Immunol 2015; 45:2324-34. [PMID: 25991034 DOI: 10.1002/eji.201444990] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 04/27/2015] [Accepted: 05/18/2015] [Indexed: 12/20/2022]
Abstract
NK cells play a key role in innate elimination of virally infected or neoplastic cells but they can be circumvented by immunoevasive mechanisms enabling viral spread or tumor progression. Engagement of the NKG2D activating receptor with soluble forms of its ligand is one such mechanism of inducing NK cell hyporesponsiveness. Interestingly, this immunoevasive strategy among others is described at the maternal-fetal interface where tolerance of the semi-allogeneic fetus is required to allow successful human pregnancy. Understanding of maternal-fetal tolerance is increasing but mechanisms preventing alloreactivity of fetal immune cells against the maternal host are less well understood. The study of umbilical cord blood has enabled insight of the fetal immune system, which appears immature and inert. We have found that soluble NKG2D ligands (sNKG2DLs) are present in cord blood plasma (CBP) and associate with adult NK cell hyporesponsiveness demonstrated by reduced CD107a expression and secretion of IFN-γ upon stimulation. The capacity of NK cells to kill K562 cells or proliferate was also reduced by incubation with CBP; however, physical removal of sNKG2DL from CBP restored K562 lytic function and NKG2D expression. Therefore, our results strongly suggest sNKG2DLs are expressed in CBP as a mechanism of fetal-maternal tolerance in human pregnancy.
Collapse
Affiliation(s)
- Steven T Cox
- The Anthony Nolan Research Institute, The Royal Free Hospital, Hampstead, London, UK
| | - Raquel Laza-Briviesca
- The Anthony Nolan Research Institute, The Royal Free Hospital, Hampstead, London, UK
| | - Hayley Pearson
- The Anthony Nolan Research Institute, The Royal Free Hospital, Hampstead, London, UK
| | - Bernat Soria
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Fundación Progreso y Salud, Seville, Spain.,Biomedical Research Network on Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Gibson
- Cell Therapy Centre, Nottingham Trent University, Nottingham, UK
| | - Susana Gomez
- Cell Therapy Centre, Nottingham Trent University, Nottingham, UK
| | - J Alejandro Madrigal
- The Anthony Nolan Research Institute, The Royal Free Hospital, Hampstead, London, UK.,Cell Therapy Centre, Nottingham Trent University, Nottingham, UK.,UCL Cancer Institute, Royal Free Campus, London, UK
| | - Aurore Saudemont
- The Anthony Nolan Research Institute, The Royal Free Hospital, Hampstead, London, UK.,UCL Cancer Institute, Royal Free Campus, London, UK
| |
Collapse
|
205
|
Yang H, Bi Y, Han F, Lu Y, Wang J, Zhang Z, Liu G. Myeloid-derived suppressor cells in immunity and autoimmunity. Expert Rev Clin Immunol 2015; 11:911-9. [DOI: 10.1586/1744666x.2015.1052794] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
206
|
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
|
207
|
Uhlenbrock F, van Andel E, Andresen L, Skov S. A conserved WW domain-like motif regulates invariant chain-dependent cell-surface transport of the NKG2D ligand ULBP2. Mol Immunol 2015; 66:418-27. [PMID: 25983110 DOI: 10.1016/j.molimm.2015.04.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 04/16/2015] [Accepted: 04/22/2015] [Indexed: 11/16/2022]
Abstract
Malignant cells expressing NKG2D ligands on their cell surface can be directly sensed and killed by NKG2D-bearing lymphocytes. To ensure this immune recognition, accumulating evidence suggests that NKG2D ligands are trafficed via alternative pathways to the cell surface. We have previously shown that the NKG2D ligand ULBP2 traffics over an invariant chain (Ii)-dependent pathway to the cell surface. This study set out to elucidate how Ii regulates ULBP2 cell-surface transport: We discovered conserved tryptophan (Trp) residues in the primary protein sequence of ULBP1-6 but not in the related MICA/B. Substitution of Trp to alanine resulted in cell-surface inhibition of ULBP2 in different cancer cell lines. Moreover, the mutated ULBP2 constructs were retained and not degraded inside the cell, indicating a crucial role of this conserved Trp-motif in trafficking. Finally, overexpression of Ii increased surface expression of wt ULBP2 while Trp-mutants could not be expressed, proposing that this Trp-motif is required for an Ii-dependent cell-surface transport of ULBP2. Aberrant soluble ULBP2 is immunosuppressive. Thus, targeting a distinct protein module on the ULBP2 sequence could counteract this abnormal expression of ULBP2.
Collapse
Affiliation(s)
- Franziska Uhlenbrock
- Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, Stigbøjlen 7, DK-1870 Frederiksberg, Denmark.
| | - Esther van Andel
- Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, Stigbøjlen 7, DK-1870 Frederiksberg, Denmark
| | - Lars Andresen
- Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, Stigbøjlen 7, DK-1870 Frederiksberg, Denmark
| | - Søren Skov
- Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, Stigbøjlen 7, DK-1870 Frederiksberg, Denmark.
| |
Collapse
|
208
|
Chu PS, Ebinuma H, Nakamoto N, Sugiyama K, Usui S, Wakayama Y, Taniki N, Yamaguchi A, Shiba S, Yamagishi Y, Wakita T, Hibi T, Saito H, Kanai T. Genotype-Associated Differential NKG2D Expression on CD56+CD3+ Lymphocytes Predicts Response to Pegylated-Interferon/Ribavirin Therapy in Chronic Hepatitis C. PLoS One 2015; 10:e0125664. [PMID: 25965701 PMCID: PMC4428701 DOI: 10.1371/journal.pone.0125664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 03/16/2015] [Indexed: 12/13/2022] Open
Abstract
Hepatitis C virus (HCV) genotype 1 infections are significantly more difficult to eradicate with PEG-IFN/ribavirin therapy, compared to HCV genotype 2. The aim of this work is to investigate the difference of immunological impairments underlying this phenomenon. Pre-treatment NKG2D expression on peripheral CD56+CD3+ lymphocytes and CD56+CD3- NK cells from cases of chronic hepatitis C were analyzed and assessed by treatment effect. Two strains of HCV were used to co-incubate with immune cells in vitro. NKG2D expression on peripheral CD56+CD3+ lymphocytes, but not NK cells, was significantly impaired in genotype 1 infection, compared to genotype 2. When peripheral blood mononuclear cells from healthy donors were co-incubated with TNS2J1, a genotype 1b/2a chimera strain, or with JFH1, a genotype 2a strain, genotype-specific decrease of NKG2D on CD56+CD3+ lymphocytes, but not NK cells, was observed. Pre-treatment NKG2D expression on peripheral CD56+CD3+ lymphocytes significantly correlated with reduction in serum HCV RNA levels from week 0 to week 4, and predicted treatment response. Ex vivo stimulation of peripheral CD56+CD3+ lymphocytes showed NKG2D expression-correlated IFN-γ production. In conclusion, Decreased NKG2D expression on CD56+CD3+ lymphocytes in chronic HCV genotype 1 infection predicts inferior treatment response to PEG-IFN/ribavirin therapy compared to genotype 2.
Collapse
Affiliation(s)
- Po-sung Chu
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Hirotoshi Ebinuma
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Nobuhiro Nakamoto
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Kazuo Sugiyama
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Shingo Usui
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Yuko Wakayama
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Nobuhito Taniki
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Akihiro Yamaguchi
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Shunsuke Shiba
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Yoshiyuki Yamagishi
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato Institute Hospital, Kitasato University, Tokyo, Japan
| | - Hidetsugu Saito
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
- Division of Pharmacotherapeutics, School of Pharmacy, Keio University, Tokyo, Japan
| | - Takanori Kanai
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
209
|
Ullrich N, Heinemann A, Nilewski E, Scheffrahn I, Klode J, Scherag A, Schadendorf D, Singer BB, Helfrich I. CEACAM1-3S Drives Melanoma Cells into NK Cell-Mediated Cytolysis and Enhances Patient Survival. Cancer Res 2015; 75:1897-907. [PMID: 25744717 DOI: 10.1158/0008-5472.can-14-1752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 02/17/2015] [Indexed: 11/16/2022]
Abstract
CEACAM1 is a widely expressed multifunctional cell-cell adhesion protein reported to serve as a poor prognosis marker in melanoma patients. In this study, we examine the functional and clinical contributions of the four splice isoforms of CEACAM1. Specifically, we present in vitro and in vivo evidence that they affect melanoma progression and immune surveillance in a negative or positive manner that is isoform specific in action. In contrast with isoforms CEACAM1-4S and CEACAM1-4L, expression of isoforms CEACAM1-3S and CEACAM1-3L is induced during disease progression shown to correlate with clinical stage. Unexpectedly, overall survival was prolonged in patients with advanced melanomas expressing CEACAM1-3S. The favorable effects of CEACAM1-3S related to enhanced immunogenicity, which was mediated by cell surface upregulation of NKG2D receptor ligands, thereby sensitizing melanoma cells to lysis by natural killer cells. Conversely, CEACAM1-4L downregulated cell surface levels of the NKG2D ligands MICA and ULBP2 by enhanced shedding, thereby promoting malignant character. Overall, our results define the splice isoform-specific immunomodulatory and cell biologic functions of CEACAM1 in melanoma pathogenesis.
Collapse
Affiliation(s)
- Nico Ullrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Anja Heinemann
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Elena Nilewski
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Inka Scheffrahn
- Institute for Gastroenterology and Hepatology, Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Joachim Klode
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - André Scherag
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Bernhard B Singer
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany.
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
210
|
Ribeiro ST, Ribot JC, Silva-Santos B. Five Layers of Receptor Signaling in γδ T-Cell Differentiation and Activation. Front Immunol 2015; 6:15. [PMID: 25674089 PMCID: PMC4306313 DOI: 10.3389/fimmu.2015.00015] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
The contributions of γδ T-cells to immunity to infection or tumors critically depend on their activation and differentiation into effectors capable of secreting cytokines and killing infected or transformed cells. These processes are molecularly controlled by surface receptors that capture key extracellular cues and convey downstream intracellular signals that regulate γδ T-cell physiology. The understanding of how environmental signals are integrated by γδ T-cells is critical for their manipulation in clinical settings. Here, we discuss how different classes of surface receptors impact on human and murine γδ T-cell differentiation, activation, and expansion. In particular, we review the role of five receptor types: the T-cell receptor (TCR), costimulatory receptors, cytokine receptors, NK receptors, and inhibitory receptors. Some of the key players are the costimulatory receptors CD27 and CD28, which differentially impact on pro-inflammatory subsets of γδ T-cells; the cytokine receptors IL-2R, IL-7R, and IL-15R, which drive functional differentiation and expansion of γδ T-cells; the NK receptor NKG2D and its contribution to γδ T-cell cytotoxicity; and the inhibitory receptors PD-1 and BTLA that control γδ T-cell homeostasis. We discuss these and other receptors in the context of a five-step model of receptor signaling in γδ T-cell differentiation and activation, and discuss its implications for the manipulation of γδ T-cells in immunotherapy.
Collapse
Affiliation(s)
- Sérgio T Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa , Lisboa , Portugal
| | - Julie C Ribot
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa , Lisboa , Portugal
| | - Bruno Silva-Santos
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa , Lisboa , Portugal
| |
Collapse
|
211
|
Euler Z, Alter G. Exploring the potential of monoclonal antibody therapeutics for HIV-1 eradication. AIDS Res Hum Retroviruses 2015; 31:13-24. [PMID: 25385703 DOI: 10.1089/aid.2014.0235] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The HIV field has seen an increased interest in novel cure strategies. In particular, new latency reversal agents are in development to reverse latency to flush the virus out of its hiding place. Combining these efforts with immunotherapeutic approaches may not only drive the virus out of latency, but allow for the rapid elimination of these infected cells in a "shock and kill" approach. Beyond cell-based approaches, growing interest lies in the potential use of functionally enhanced "killer" monoclonal therapeutics to purge the reservoir. Here we discuss prospects for a monoclonal therapeutic-based "shock and kill" strategy that may lead to the permanent elimination of replication-competent virus, making a functional cure a reality for all patients afflicted with HIV worldwide.
Collapse
Affiliation(s)
- Zelda Euler
- Ragon Institute of MGH, MIT, and Harvard University , Cambridge, Massachusetts
| | | |
Collapse
|
212
|
Luoma AM, Castro CD, Adams EJ. γδ T cell surveillance via CD1 molecules. Trends Immunol 2014; 35:613-621. [PMID: 25283967 PMCID: PMC4383740 DOI: 10.1016/j.it.2014.09.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 12/15/2022]
Abstract
γδ T cells are a prominent epithelial-resident lymphocyte population, possessing multi-functional capacities in the repair of host tissue, pathogen clearance, and tumor surveillance. Although three decades have now passed since their discovery, the nature of γδ T cell receptor (TCR)-mediated ligand recognition remains poorly defined. Recent studies have provided structural insight into this recognition, demonstrating that γδ T cells survey both CD1 and the presented lipid, and in some cases are exquisitely lipid specific. We review these findings here, examining the molecular basis for and the functional relevance of this interaction. We discuss potential implications on the notion that non-classical major histocompatibility complex (MHC) molecules may function as important restricting elements of γδ TCR specificity, and on our understanding of γδ T cell activation and function.
Collapse
Affiliation(s)
- Adrienne M Luoma
- Committee on Immunology and Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Caitlin D Castro
- Committee on Immunology and Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Erin J Adams
- Committee on Immunology and Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
213
|
Witherden DA, Ramirez K, Havran WL. Multiple Receptor-Ligand Interactions Direct Tissue-Resident γδ T Cell Activation. Front Immunol 2014; 5:602. [PMID: 25505467 PMCID: PMC4241470 DOI: 10.3389/fimmu.2014.00602] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/07/2014] [Indexed: 11/13/2022] Open
Abstract
γδ T cells represent a major T cell population in epithelial tissues, such as skin, intestine, and lung, where they function in maintenance of the epithelium and provide a crucial first line defense against environmental and pathogenic insults. Despite their importance, the molecular mechanisms directing their activation and function have remained elusive. Epithelial-resident γδ T cells function through constant communication with neighboring cells, either via direct cell-to-cell contact or cell-to-matrix interactions. These intimate relationships allow γδ T cells to facilitate the maintenance of epithelial homeostasis, tissue repair following injury, inflammation, and protection from malignancy. Recent studies have identified a number of molecules involved in these complex interactions, under both homeostatic conditions, as well as following perturbation of these barrier tissues. These interactions are crucial to the timely production of cytokines, chemokines, growth factors, and extracellular matrix proteins for restoration of homeostasis. In this review, we discuss recent advances in understanding the mechanisms directing epithelial-T cell crosstalk and the distinct roles played by individual receptor-ligand pairs of cell surface molecules in this process.
Collapse
Affiliation(s)
- Deborah A Witherden
- Department of Immunology and Microbial Science, The Scripps Research Institute , La Jolla, CA , USA
| | - Kevin Ramirez
- Department of Immunology and Microbial Science, The Scripps Research Institute , La Jolla, CA , USA
| | - Wendy L Havran
- Department of Immunology and Microbial Science, The Scripps Research Institute , La Jolla, CA , USA
| |
Collapse
|
214
|
Classical and non-classical MHC I molecule manipulation by human cytomegalovirus: so many targets—but how many arrows in the quiver? Cell Mol Immunol 2014; 12:139-53. [PMID: 25418469 PMCID: PMC4654289 DOI: 10.1038/cmi.2014.105] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/26/2014] [Accepted: 09/26/2014] [Indexed: 02/07/2023] Open
Abstract
Major mechanisms for the recognition of pathogens by immune cells have evolved to employ classical and non-classical major histocompatibility complex class I (MHC I) molecules. Classical MHC I molecules present antigenic peptide ligands on infected cells to CD8+ T cells, whereas a key function for non-classical MHC I molecules is to mediate inhibitory or activating stimuli in natural killer (NK) cells. The structural diversity of MHC I puts immense pressure on persisting viruses, including cytomegaloviruses. The very large coding capacity of the human cytomegalovirus allows it to express a whole arsenal of immunoevasive factors assigned to individual MHC class I targets. This review summarizes achievements from more than two decades of intense research on how human cytomegalovirus manipulates MHC I molecules and escapes elimination by the immune system.
Collapse
|
215
|
Pyzik M, Dumaine AA, Charbonneau B, Fodil-Cornu N, Jonjic S, Vidal SM. Viral MHC Class I–like Molecule Allows Evasion of NK Cell Effector Responses In Vivo. THE JOURNAL OF IMMUNOLOGY 2014; 193:6061-9. [DOI: 10.4049/jimmunol.1401386] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
216
|
Poggi A, Zocchi MR. γδ T Lymphocytes as a First Line of Immune Defense: Old and New Ways of Antigen Recognition and Implications for Cancer Immunotherapy. Front Immunol 2014; 5:575. [PMID: 25426121 PMCID: PMC4226920 DOI: 10.3389/fimmu.2014.00575] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/27/2014] [Indexed: 01/11/2023] Open
Abstract
Among γδT cells, the Vδ1 subset, resident in epithelial tissues, is implied in the defense against viruses, fungi, and certain hematological malignancies, while the circulating Vδ2 subpopulation mainly respond to mycobacteria and solid tumors. Both subsets can be activated by stress-induced molecules (MIC-A, MIC-B, ULBPs) to produce pro-inflammatory cytokines and lytic enzymes and destroy bacteria or damaged cells. γδT lymphocytes can also recognize lipids, as those associated to M. tuberculosis, presented by the CD1 molecule, or phosphoantigens (P-Ag), either autologous, which accumulates in virus-infected cells, or microbial produced by prokaryotes and parasites. In cancer cells, P-Ag accumulate due to alterations in the mevalonate pathway; recently, butyrophilin 3A1 has been shown to be the presenting molecule for P-Ag. Of interest, aminobisphosphonates indirectly activate Vδ2 T cells inducing the accumulation of P-Ag. Based on these data, γδT lymphocytes are attractive effectors for cancer immunotherapy. However, the results obtained in clinical trials so far have been disappointing: this review will focus on the possible reasons of this failure as well as on suggestions for implementation of the therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Poggi
- Unit of Molecular Oncology and Angiogenesis, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, IRCCS San Raffaele, Milan, Italy
| |
Collapse
|
217
|
Lu X, Zhu A, Cai X, Jia Z, Han W, Ma L, Zhou M, Qian K, Cen L, Chen B. Role of NKG2D in cytokine-induced killer cells against multiple myeloma cells. Cancer Biol Ther 2014; 13:623-9. [DOI: 10.4161/cbt.19850] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
218
|
Spear P, Barber A, Sentman CL. Collaboration of chimeric antigen receptor (CAR)-expressing T cells and host T cells for optimal elimination of established ovarian tumors. Oncoimmunology 2014; 2:e23564. [PMID: 23734311 PMCID: PMC3654581 DOI: 10.4161/onci.23564] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/07/2013] [Accepted: 01/09/2013] [Indexed: 12/24/2022] Open
Abstract
Conditioning strategies that deplete host lymphocytes have been shown to enhance clinical responses to some adoptive T-cell therapies. However, host T cells are capable of eliminating tumor cells upon the relief of immunosuppression, indicating that lymphodepletion prior to T-cell transfer may reduce optimal tumor protection elicited by cell treatments that are capable of shaping host immunity. In this study, we show that adoptively transferred T cells bearing a chimeric antigen receptor (CAR) harness endogenous T cells for optimal tumor elimination and the development of a tumor-specific memory T cell response. Mice bearing ID8 ovarian cancer cells were treated with T cells transduced with a NKG2D-based CAR. CAR-expressing T cells increased the number of host CD4+ and CD8+ T cells at the tumor site in a CXCR3-dependent manner and increased the number of antigen-specific host CD4+ T cells in the tumor and draining lymph nodes. In addition, the administration of CAR-expressing T cells increased antigen presentation to CD4+ T cells, and this increase was dependent on interferon γ and granulocyte-macrophage colony-stimulating factor produced by the former. Host CD4+ T cells were sufficient for optimal tumor protection mediated by NKG2D CAR-expressing T cells, but they were not necessary if CD4+ T cells were adoptively co-transferred. However, host CD4+ T cells were essential for the development of an antigen-specific memory T-cell response to tumor cells. Moreover, optimal tumor elimination as orchestrated by NKG2D CAR-expressing T cells was dependent on host CD8+ T cells. These results demonstrate that adoptively transferred T cells recruit and activate endogenous T-cell immunity to enhance the elimination of tumor cells and the development of tumor-specific memory responses.
Collapse
Affiliation(s)
- Paul Spear
- Department of Microbiology & Immunology; The Geisel School of Medicine at Dartmouth; Lebanon, NH USA
| | | | | |
Collapse
|
219
|
Adlercreutz EH, Weile C, Larsen J, Engkilde K, Agardh D, Buschard K, Antvorskov JC. A gluten-free diet lowers NKG2D and ligand expression in BALB/c and non-obese diabetic (NOD) mice. Clin Exp Immunol 2014; 177:391-403. [PMID: 24673402 PMCID: PMC4226590 DOI: 10.1111/cei.12340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/22/2022] Open
Abstract
The interplay between diet and immune parameters which could affect type 1 diabetes (T1D) pathogenesis is not sufficiently clarified. Intestinal up-regulation of the activating receptor natural killer group 2D (NKG2D) (CD314) and its ligands is a hallmark of coeliac disease. However, the direct effect of gluten on NKG2D expression is not known. We studied, by fluorescence activated cell sorter (lymphoid tissues) and reverse transcription–quantitative polymerase chain reaction (intestine and pancreatic islets), if a gluten-free diet (GF diet) from 4 weeks of age or a gluten-free diet introduced in breeding pairs (SGF diet), induced changes in NKG2D expression on DX5+(CD49b) natural killer (NK) cells, CD8+ T cells and in intestinal and islet levels of NKG2D and ligands in BALB/c and non-obese diabetic (NOD) mice. Gluten-free NOD mice had lower insulitis (P < 0·0001); reduced expression of NKG2D on DX5+ NK cells in spleen and auricular lymph nodes (P < 0·05); and on CD8+ T cells in pancreas-associated lymph nodes (P = 0·04). Moreover, the level of CD71 on DX5+ NK cells and CD8+ T cells (P < 0·005) was markedly reduced. GF and SGF mice had reduced expression of NKG2D and DX5 mRNA in intestine (P < 0·05). Differences in intestinal mRNA expression were found in mice at 8, 13 and 20 weeks. Intestinal expression of NKG2D ligands was reduced in SGF mice with lower expression of all ligands. In isolated islets, a SGF diet induced a higher expression of specific NKG2D ligands. Our data show that a gluten-free diet reduces the level of NKG2D and the expression of NKG2D ligands. These immunological changes may contribute to the lower T1D incidence associated with a gluten-free diet.
Collapse
Affiliation(s)
- E H Adlercreutz
- Diabetes and Celiac Disease Unit, Lund University, Malmö, Sweden
| | | | | | | | | | | | | |
Collapse
|
220
|
Pawlick R, Gala-Lopez B, Pepper AR, McCall M, Ziff O, Shapiro AMJ. The combination of anti-NKG2D and CTLA-4 Ig therapy prolongs islet allograft survival in a murine model. Am J Transplant 2014; 14:2367-74. [PMID: 25179027 DOI: 10.1111/ajt.12838] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/05/2014] [Accepted: 05/08/2014] [Indexed: 01/25/2023]
Abstract
Islet transplantation is an effective means of treating severe type 1 diabetes in patients with life-threatening hypoglycemia. Improvements in glycemic control with correction of HbA1C enhance quality of life irrespective of insulin independence. By antagonizing the Natural Killer Group 2, member D (NKG2D) receptor expression on NK and CD8+ T cells, in combination with blocking CTLA-4 binding sites, we demonstrate a significant delay of graft rejection in islet allotransplant. Anti-NKG2D combined with CTLA-4 Ig (n = 15) results in prolonged allograft survival, with 84.6 ± 10% of the recipients displaying insulin independence compared to controls (n = 10, p < 0.001). The effect of combination therapy on graft survival is superior to treatments alone (CTLA-4 Ig vs. combination p = 0.024, anti-NKG2D vs. combination p < 0.001) indicating an interaction between these pathways. In addition, combination treatment also improves glucose tolerance when compared to controls (n = 10, p = 0.018). Histologically, NKG2D+ cells were significantly decreased within the allograft after 7 days of combination treatment (n = 6, p = 0.029). T cell proliferation was significantly reduced with anti-NKG2D therapy and CD8+ T cell daughter fractions were also significantly decreased with mAb and combination treatment when measured by in vitro mixed lymphocyte reaction (n = 5, p = 0.015, p = 0.005 and p = 0.048). These results demonstrate that inhibition of NKG2D receptors and costimulatory pathways enhance islet allograft survival.
Collapse
Affiliation(s)
- R Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | |
Collapse
|
221
|
Mostafavi S, Ortiz-Lopez A, Bogue MA, Hattori K, Pop C, Koller D, Mathis D, Benoist C. Variation and genetic control of gene expression in primary immunocytes across inbred mouse strains. THE JOURNAL OF IMMUNOLOGY 2014; 193:4485-96. [PMID: 25267973 DOI: 10.4049/jimmunol.1401280] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To determine the breadth and underpinning of changes in immunocyte gene expression due to genetic variation in mice, we performed, as part of the Immunological Genome Project, gene expression profiling for CD4(+) T cells and neutrophils purified from 39 inbred strains of the Mouse Phenome Database. Considering both cell types, a large number of transcripts showed significant variation across the inbred strains, with 22% of the transcriptome varying by 2-fold or more. These included 119 loci with apparent complete loss of function, where the corresponding transcript was not expressed in some of the strains, representing a useful resource of "natural knockouts." We identified 1222 cis-expression quantitative trait loci (cis-eQTL) that control some of this variation. Most (60%) cis-eQTLs were shared between T cells and neutrophils, but a significant portion uniquely impacted one of the cell types, suggesting cell type-specific regulatory mechanisms. Using a conditional regression algorithm, we predicted regulatory interactions between transcription factors and potential targets, and we demonstrated that these predictions overlap with regulatory interactions inferred from transcriptional changes during immunocyte differentiation. Finally, comparison of these and parallel data from CD4(+) T cells of healthy humans demonstrated intriguing similarities in variability of a gene's expression: the most variable genes tended to be the same in both species, and there was an overlap in genes subject to strong cis-acting genetic variants. We speculate that this "conservation of variation" reflects a differential constraint on intraspecies variation in expression levels of different genes, either through lower pressure for some genes, or by favoring variability for others.
Collapse
Affiliation(s)
- Sara Mostafavi
- Department of Computer Science, Stanford University, Stanford, CA 94305
| | - Adriana Ortiz-Lopez
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; and
| | | | - Kimie Hattori
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; and
| | - Cristina Pop
- Department of Computer Science, Stanford University, Stanford, CA 94305
| | - Daphne Koller
- Department of Computer Science, Stanford University, Stanford, CA 94305
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; and
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; and
| | | |
Collapse
|
222
|
Telcian AG, Zdrenghea MT, Caramori G, Laza-Stanca V, Message SD, Kebadze T, Kon OM, Groh V, Papi A, Johnston SL, Mallia P, Stanciu LA. Soluble major histocompatibility complex class I-related chain B molecules are increased and correlate with clinical outcomes during rhinovirus infection in healthy subjects. Chest 2014; 146:32-40. [PMID: 24556715 PMCID: PMC4077410 DOI: 10.1378/chest.13-2247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND: Surface major histocompatibility complex class I-related chain (MIC) A and B molecules are increased by IL-15 and have a role in the activation of natural killer group 2 member D-positive natural killer and CD8 T cells. MICA and MICB also exist in soluble forms (sMICA and sMICB). Rhinoviruses (RVs) are the major cause of asthma exacerbations, and IL-15 levels are decreased in the airways of subjects with asthma. The role of MIC molecules in immune responses in the lung has not been studied. Here, we determine the relationship between MICA and MICB and RV infection in vitro in respiratory epithelial cells and in vivo in healthy subjects and subjects with asthma. METHODS: Surface MICA and MICB, as well as sMICA and sMICB, in respiratory epithelial cells were measured in vitro in response to RV infection and exposure to IL-15. Levels of sMICA and sMICB in serum, sputum, and BAL were measured and correlated with blood and bronchoalveolar immune cells in healthy subjects and subjects with asthma before and during RV infection. RESULTS: RV increased MICA and MICB in vitro in epithelial cells. Exogenous IL-15 upregulated sMICB levels in RV-infected epithelial cells. Levels of sMICB molecules in serum were increased in healthy subjects compared with subjects with stable asthma. Following RV infection, airway levels of sMIC are upregulated, and there are positive correlations between sputum MICB levels and the percentage of bronchoalveolar natural killer cells in healthy subjects but not subjects with asthma. CONCLUSIONS: RV infection induces MIC molecules in respiratory epithelial cells in vitro and in vivo. Induction of MICB molecules is impaired in subjects with asthma, suggesting these molecules may have a role in the antiviral immune response to RV infections.
Collapse
Affiliation(s)
- Aurica G Telcian
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England
| | - Mihnea T Zdrenghea
- Medical Research Council, London, England; Asthma UK Centre in Allergic Mechanisms of Asthma, London, England; The Oncology Institute "Prof. Dr. Ion Chiricuţă" Cluj-Napoca and Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania.
| | - Gaetano Caramori
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF), Sezione di Malattie dell'Apparato Respiratorio, Università degli Studi di Ferrara, Ferrara, Italy
| | - Vasile Laza-Stanca
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England
| | - Simon D Message
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Imperial College Healthcare NHS Trust, London, England
| | - Tatiana Kebadze
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England
| | - Onn M Kon
- Imperial College Healthcare NHS Trust, London, England
| | | | - Alberto Papi
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF), Sezione di Malattie dell'Apparato Respiratorio, Università degli Studi di Ferrara, Ferrara, Italy
| | - Sebastian L Johnston
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Imperial College Healthcare NHS Trust, London, England
| | - Patrick Mallia
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Imperial College Healthcare NHS Trust, London, England
| | - Luminita A Stanciu
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Medical Research Council, London, England; Asthma UK Centre in Allergic Mechanisms of Asthma, London, England; The Oncology Institute "Prof. Dr. Ion Chiricuţă" Cluj-Napoca and Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
223
|
Hagemann-Jensen M, Uhlenbrock F, Kehlet S, Andresen L, Gabel-Jensen C, Ellgaard L, Gammelgaard B, Skov S. The selenium metabolite methylselenol regulates the expression of ligands that trigger immune activation through the lymphocyte receptor NKG2D. J Biol Chem 2014; 289:31576-90. [PMID: 25258323 DOI: 10.1074/jbc.m114.591537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
For decades, selenium research has been focused on the identification of active metabolites, which are crucial for selenium chemoprevention of cancer. In this context, the metabolite methylselenol (CH3SeH) is known for its action to selectively kill transformed cells through mechanisms that include increased formation of reactive oxygen species, induction of DNA damage, triggering of apoptosis, and inhibition of angiogenesis. Here we reveal that CH3SeH modulates the cell surface expression of NKG2D ligands. The expression of NKG2D ligands is induced by stress-associated pathways that occur early during malignant transformation and enable the recognition and elimination of tumors by activating the lymphocyte receptor NKG2D. CH3SeH regulated NKG2D ligands both on the transcriptional and the posttranscriptional levels. CH3SeH induced the transcription of MHC class I polypeptide-related sequence MICA/B and ULBP2 mRNA. However, the induction of cell surface expression was restricted to the ligands MICA/B. Remarkably, our studies showed that CH3SeH inhibited ULBP2 surface transport through inhibition of the autophagic transport pathway. Finally, we identified extracellular calcium as being essential for CH3SeH regulation of NKG2D ligands. A balanced cell surface expression of NKG2D ligands is considered to be an innate barrier against tumor development. Therefore, our work indicates that the application of selenium compounds that are metabolized to CH3SeH could improve NKG2D-based immune therapy.
Collapse
Affiliation(s)
| | | | - Stephanie Kehlet
- From the Laboratory of Immunology, Department of Veterinary Disease Biology
| | - Lars Andresen
- From the Laboratory of Immunology, Department of Veterinary Disease Biology
| | | | - Lars Ellgaard
- Department of Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Copenhagen, Denmark
| | | | - Søren Skov
- From the Laboratory of Immunology, Department of Veterinary Disease Biology,
| |
Collapse
|
224
|
Chatzinikolaou G, Karakasilioti I, Garinis GA. DNA damage and innate immunity: links and trade-offs. Trends Immunol 2014; 35:429-35. [DOI: 10.1016/j.it.2014.06.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 12/20/2022]
|
225
|
Wang B, Wang Q, Wang Z, Jiang J, Yu SC, Ping YF, Yang J, Xu SL, Ye XZ, Xu C, Yang L, Qian C, Wang JM, Cui YH, Zhang X, Bian XW. Metastatic consequences of immune escape from NK cell cytotoxicity by human breast cancer stem cells. Cancer Res 2014; 74:5746-57. [PMID: 25164008 DOI: 10.1158/0008-5472.can-13-2563] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Breast cancer stem-like cells (BCSC) are crucial for metastasis but the underlying mechanisms remain elusive. Here, we report that tumor-infiltrating natural killer (NK) cells failed to limit metastasis and were not associated with improved therapeutic outcome of BCSC-rich breast cancer. Primary BCSCs were resistant to cytotoxicity mediated by autologous/allogeneic NK cells due to reduced expression of MICA and MICB, two ligands for the stimulatory NK cell receptor NKG2D. Furthermore, the downregulation of MICA/MICB in BCSCs was mediated by aberrantly expressed oncogenic miR20a, which promoted the resistance of BCSC to NK cell cytotoxicity and resultant lung metastasis. The breast cancer cell differentiation-inducing agent, all-trans retinoic acid, restored the miR20a-MICA/MICB axis and sensitized BCSC to NK cell-mediated killing, thereby reducing immune escape-associated BCSC metastasis. Together, our findings reveal a novel mechanism for immune escape of human BCSC and identify the miR20a-MICA/MICB signaling axis as a therapeutic target to limit metastatic breast cancer.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Qiang Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Zhe Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jun Jiang
- Breast Surgery Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jing Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Sen-Lin Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xian-Zong Ye
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Chuan Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Lang Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| |
Collapse
|
226
|
The regulatory effect of UL-16 binding protein-3 expression on the cytotoxicity of NK cells in cancer patients. Sci Rep 2014; 4:6138. [PMID: 25138242 PMCID: PMC4138521 DOI: 10.1038/srep06138] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 07/31/2014] [Indexed: 12/19/2022] Open
Abstract
The activating immunoreceptor NKG2D (natural killer group 2, member D) and its ligands play important roles in the innate and adaptive immune responses. UL16-binding protein 3 (ULBP3), an NKG2D ligand, is overexpressed on certain epithelial tumor cells. In this study, we investigated the effect of ULBP3 expression on the cytotoxic activity of natural killer (NK) cells. ULBP3 were measured by flow cytometry analysis, immunohistochemistry, and time-resolved fluoroimmunoassay. The cytotoxicity of NK cells was determined with the lactate dehydrogenase release assay. We found that ULBP3 was overexpressed on tumor cell lines and tumor tissues. Serum from cancer patients, but not from healthy donors, contained elevated levels of soluble ULBP3 (sULBP3). Importantly, high expression of ULBP3 on the cell surface of tumor cells augmented NKG2D-mediated NK cell cytotoxicity. However, low levels of sULBP3 (<15 ng/ml) weakened the cytotoxicity of NK cells by decreasing NKG2D expression on NK cells. Further analysis showed that serum samples from most cancer patients (>70%) contained the low level of sULBP3. Our results demonstrate that tumor cells express surface and soluble ULBP3, which regulate NK cell activity. Thus, ULBP3 is a potential therapeutic target for improving the immune response against cancer.
Collapse
|
227
|
Immune evasion mediated by tumor-derived lactate dehydrogenase induction of NKG2D ligands on myeloid cells in glioblastoma patients. Proc Natl Acad Sci U S A 2014; 111:12823-8. [PMID: 25136121 DOI: 10.1073/pnas.1413933111] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Myeloid cells are key regulators of the tumor microenvironment, governing local immune responses. Here we report that tumor-infiltrating myeloid cells and circulating monocytes in patients with glioblastoma multiforme (GBM) express ligands for activating the Natural killer group 2, member D (NKG2D) receptor, which cause down-regulation of NKG2D on natural killer (NK) cells. Tumor-infiltrating NK cells isolated from GBM patients fail to lyse NKG2D ligand-expressing tumor cells. We demonstrate that lactate dehydrogenase (LDH) isoform 5 secreted by glioblastoma cells induces NKG2D ligands on monocytes isolated from healthy individuals. Furthermore, sera from GBM patients contain elevated amounts of LDH, which correlate with expression of NKG2D ligands on their autologous circulating monocytes. NKG2D ligands also are present on circulating monocytes isolated from patients with breast, prostate, and hepatitis C virus-induced hepatocellular carcinomas. Together, these findings reveal a previously unidentified immune evasion strategy whereby tumors produce soluble factors that induce NKG2D ligands on myeloid cells, subverting antitumor immune responses.
Collapse
|
228
|
Zhou YF, Xu LX, Huang LY, Guo F, Zhang F, He XY, Yuan YZ, Yao WY. Combined detection of serum UL16-binding protein 2 and macrophage inhibitory cytokine-1 improves early diagnosis and prognostic prediction of pancreatic cancer. Oncol Lett 2014; 8:2096-2102. [PMID: 25295097 PMCID: PMC4186586 DOI: 10.3892/ol.2014.2429] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 07/08/2014] [Indexed: 01/03/2023] Open
Abstract
Pancreatic cancer (PC) is the fourth leading cause of cancer-related mortality in the United States. There is no effective serum biomarker for the early diagnosis of PC at present. Although serum UL16-binding protein 2 (ULBP2) and macrophage inhibitory cytokine-1 (MIC-1) levels are reported to be elevated in PC patients, the diagnostic and prognostic value of ULBP2 and MIC-1 alone or in combination remains unknown. The aim of the present case-control study was to compare the diagnostic value of ULBP2, MIC-1 and carbohydrate antigen 19-9 (CA19-9) in 359 serum samples, consisting of 152 cases of PC, 20 cases of pre-pancreatic cancer, 91 cases of chronic pancreatitis (CP) and 96 normal controls (NC). All patients were followed up for a median of 2 years. It was found that the serum levels of ULBP2, MIC-1 and CA19-9 were significantly higher in the PC patients compared with those in the NC group. In distinguishing PC from the CP, the highest sensitivity and specificity were ULBP2 (0.878) and CA19-9 (0.816), respectively. The area under the receiver operating characteristic curve of ULBP2 was 0.923, which was the highest of the three biomarkers. MIC-1 was the optimal choice for the diagnosis of early-stage PC (area under the curve, 0.831). Overall, MIC-1 in combination with ULBP2 improved the diagnostic accuracy in differentiating PC from CP and NC. In addition, a higher level of MIC-1 was correlated with a poorer prognosis, as calculated by the Kaplan-Meier test (P=0.039). Patients with serum MIC-1 levels of ≥1,932 ng/ml had a median survival time of 15.62±2.44 months (mean ± standard deviation) vs. 18.66±2.43 months in patients with a lower level of MIC-1. Overall, combined detection of serum MIC-1 and ULBP2 improved the diagnostic accuracy in differentiating PC from CP and NC, and serum MIC-1 level alone was a predictor of survival in the patients with PC.
Collapse
Affiliation(s)
- Yu-Fen Zhou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Ling-Xiao Xu
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Li-Ya Huang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, P.R. China
| | - Fang Guo
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Fan Zhang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Xiang-Yi He
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Yao-Zong Yuan
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Wei-Yan Yao
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
229
|
Whitman E, Barber A. NKG2D receptor activation of NF-κB enhances inflammatory cytokine production in murine effector CD8(+) T cells. Mol Immunol 2014; 63:268-78. [PMID: 25089028 DOI: 10.1016/j.molimm.2014.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/10/2014] [Accepted: 07/14/2014] [Indexed: 01/03/2023]
Abstract
To induce strong immune responses, naïve CD8(+) T cells require stimulation through the TCR and costimulatory receptors. However, the biological effect of activating costimulatory receptors on effector T cells is still unclear. One costimulatory receptor that is likely to be engaged at the target site is NKG2D. This activating receptor is expressed on human and murine CD8(+) T cells with its ligands expressed on the majority of tumor cells and during some infections. In order to determine how activation of costimulatory receptors alters effector CD8(+) T cell functions, this study compared the activation of the NF-κB signaling pathway by two costimulatory receptors, CD28 and NKG2D. Compared to CD28 costimulation, activation of murine effector CD8(+) T cells through CD3 and NKG2D receptors enhanced activation of NF-κB as shown by increased phosphorylation of IKKα, IκBα, and NF-κB and IκBα degradation. NKG2D costimulation also increased activation, nuclear translocation, and DNA binding of NF-κB p65/p50 dimers. Activation of the NF-κB pathway also lead to increased gene expression and secretion of pro-inflammatory cytokines, including IFNα and IFNγ, and decreased gene expression and secretion of anti-inflammatory cytokines, including IL-10 and CCL2. Altered NF-κB activation also increased expression of the effector molecules TNFα, lymphotoxins α and β, and Fas ligand, and increased tumor cell killing through FasL. These data show that compared to CD28 costimulation, activation through the NKG2D receptor leads to the differential activation of the NF-κB signaling pathway and potentially enhances the anti-tumor and anti-viral functions of effector CD8(+) T cells.
Collapse
Affiliation(s)
- Emily Whitman
- Department of Biological and Environmental Sciences, Longwood University, Chichester Science Center 305A, 201 High Street, Farmville, VA 23909, USA
| | - Amorette Barber
- Department of Biological and Environmental Sciences, Longwood University, Chichester Science Center 305A, 201 High Street, Farmville, VA 23909, USA.
| |
Collapse
|
230
|
Dalessandri T, Strid J. Beneficial autoimmunity at body surfaces - immune surveillance and rapid type 2 immunity regulate tissue homeostasis and cancer. Front Immunol 2014; 5:347. [PMID: 25101088 PMCID: PMC4105846 DOI: 10.3389/fimmu.2014.00347] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/08/2014] [Indexed: 12/27/2022] Open
Abstract
Epithelial cells (ECs) line body surface tissues and provide a physicochemical barrier to the external environment. Frequent microbial and non-microbial challenges such as those imposed by mechanical disruption, injury or exposure to noxious environmental substances including chemicals, carcinogens, ultraviolet-irradiation, or toxins cause activation of ECs with release of cytokines and chemokines as well as alterations in the expression of cell-surface ligands. Such display of epithelial stress is rapidly sensed by tissue-resident immunocytes, which can directly interact with self-moieties on ECs and initiate both local and systemic immune responses. ECs are thus key drivers of immune surveillance at body surface tissues. However, ECs have a propensity to drive type 2 immunity (rather than type 1) upon non-invasive challenge or stress – a type of immunity whose regulation and function still remain enigmatic. Here, we review the induction and possible role of type 2 immunity in epithelial tissues and propose that rapid immune surveillance and type 2 immunity are key regulators of tissue homeostasis and carcinogenesis.
Collapse
Affiliation(s)
- Tim Dalessandri
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London , London , UK
| | - Jessica Strid
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London , London , UK
| |
Collapse
|
231
|
Uhlenbrock F, Hagemann-Jensen M, Kehlet S, Andresen L, Pastorekova S, Skov S. The NKG2D ligand ULBP2 is specifically regulated through an invariant chain-dependent endosomal pathway. THE JOURNAL OF IMMUNOLOGY 2014; 193:1654-65. [PMID: 25024379 DOI: 10.4049/jimmunol.1303275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Soluble ULBP2 is a marker for poor prognosis in several types of cancer. In this study we demonstrate that both soluble and cell surface-bound ULBP2 is transported via a so far unrecognized endosomal pathway. ULBP2 surface expression, but not MICA/B, could specifically be targeted and retained by affecting endosomal/lysosomal integrity and protein kinase C activity. The invariant chain was further essential for endosomal transport of ULBP2. This novel pathway was identified through screening experiments by which methylselenic acid was found to possess notable NKG2D ligand regulatory properties. The protein kinase C inhibitor methylselenic acid induced MICA/B surface expression but dominantly blocked ULBP2 surface transport. Remarkably, by targeting this novel pathway we could specifically block the production of soluble ULBP2 from different, primary melanomas. Our findings strongly suggest that the endosomal transport pathway constitutes a novel therapeutic target for ULBP2-producing tumors.
Collapse
Affiliation(s)
- Franziska Uhlenbrock
- Section for Experimental Animal Models, Laboratory of Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; and
| | - Michael Hagemann-Jensen
- Section for Experimental Animal Models, Laboratory of Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; and
| | - Stephanie Kehlet
- Section for Experimental Animal Models, Laboratory of Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; and
| | - Lars Andresen
- Section for Experimental Animal Models, Laboratory of Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; and
| | - Silvia Pastorekova
- Department of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, 845 05 Bratislava, Slovak Republic
| | - Søren Skov
- Section for Experimental Animal Models, Laboratory of Immunology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark; and
| |
Collapse
|
232
|
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
|
233
|
Nachmani D, Gutschner T, Reches A, Diederichs S, Mandelboim O. RNA-binding proteins regulate the expression of the immune activating ligand MICB. Nat Commun 2014; 5:4186. [PMID: 24924487 DOI: 10.1038/ncomms5186] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/22/2014] [Indexed: 12/28/2022] Open
Abstract
The recognition of stress-induced ligands by the activating receptor NKG2D expressed on cytotoxic lymphocytes is crucial for the prevention and containment of various diseases and is also one of the best-studied examples of how danger is sensed by the immune system. Still, however, the mechanisms leading to the expression of the NKG2D ligands are far from being completely understood. Here, we use an unbiased and systematic RNA pull-down approach combined with mass spectrometry to identify six RNA-binding proteins (RBPs) that bind and regulate the expression of MICB, one of the major stress-induced ligands of NKG2D. We further demonstrate that at least two of the identified RBPs function during genotoxic stress. Our data provide insights into stress recognition and hopefully open new therapeutic venues.
Collapse
Affiliation(s)
- Daphna Nachmani
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School, 91120 Jerusalem, Israel
| | - Tony Gutschner
- Helmholtz-University-Group "Molecular RNA Biology & Cancer", German Cancer Research Center DKFZ and Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Adi Reches
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School, 91120 Jerusalem, Israel
| | - Sven Diederichs
- Helmholtz-University-Group "Molecular RNA Biology & Cancer", German Cancer Research Center DKFZ and Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel Canada of the Faculty of Medicine, The Hebrew University Hadassah Medical School, 91120 Jerusalem, Israel
| |
Collapse
|
234
|
Baldwin CL, Hsu H, Chen C, Palmer M, McGill J, Waters WR, Telfer JC. The role of bovine γδ T cells and their WC1 co-receptor in response to bacterial pathogens and promoting vaccine efficacy: A model for cattle and humans. Vet Immunol Immunopathol 2014; 159:144-55. [DOI: 10.1016/j.vetimm.2014.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
235
|
Mellergaard M, Skovbakke SL, Schneider CL, Lauridsen F, Andresen L, Jensen H, Skov S. N-glycosylation of asparagine 8 regulates surface expression of major histocompatibility complex class I chain-related protein A (MICA) alleles dependent on threonine 24. J Biol Chem 2014; 289:20078-91. [PMID: 24872415 DOI: 10.1074/jbc.m114.573238] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
NKG2D is an activating receptor expressed on several types of human lymphocytes. NKG2D ligands can be induced upon cell stress and are frequently targeted post-translationally in infected or transformed cells to avoid immune recognition. Virus infection and inflammation alter protein N-glycosylation, and we have previously shown that changes in cellular N-glycosylation are involved in regulation of NKG2D ligand surface expression. The specific mode of regulation through N-glycosylation is, however, unknown. Here we investigated whether direct N-glycosylation of the NKG2D ligand MICA itself is critical for cell surface expression and sought to identify the essential residues. We found that a single N-glycosylation site (Asn(8)) was important for MICA018 surface expression. The frequently expressed MICA allele 008, with an altered transmembrane and intracellular domain, was not affected by mutation of this N-glycosylation site. Mutational analysis revealed that a single amino acid (Thr(24)) in the extracellular domain of MICA018 was essential for the N-glycosylation dependence, whereas the intracellular domain was not involved. The HHV7 immunoevasin, U21, was found to inhibit MICA018 surface expression by affecting N-glycosylation, and the retention was rescued by T24A substitution. Our study reveals N-glycosylation as an allele-specific regulatory mechanism important for regulation of surface expression of MICA018, and we pinpoint the residues essential for this N-glycosylation dependence. In addition, we show that this regulatory mechanism of MICA surface expression is likely targeted during different pathological conditions.
Collapse
Affiliation(s)
- Maiken Mellergaard
- From the Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark and
| | - Sarah Line Skovbakke
- From the Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark and
| | | | - Felicia Lauridsen
- From the Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark and
| | - Lars Andresen
- From the Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark and
| | - Helle Jensen
- From the Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark and
| | - Søren Skov
- From the Laboratory of Immunology, Section for Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark and
| |
Collapse
|
236
|
Watanabe M, Kudo Y, Kawano M, Nakayama M, Nakamura K, Kameda M, Ebara M, Sato T, Nakamura M, Omine K, Kametani Y, Suzuki R, Ogasawara K. NKG2D functions as an activating receptor on natural killer cells in the common marmoset (Callithrix jacchus). Int Immunol 2014; 26:597-606. [PMID: 24860119 DOI: 10.1093/intimm/dxu053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The natural killer group 2 membrane D (NKG2D) receptor is an NK-activating receptor that plays an important role in host defense against tumors and viral infections. Although the marmoset is an important and reliable animal model, especially for the study of human-specific viral infections, functional characterization of NKG2D on marmoset NK cells has not previously been conducted. In the present study, we investigated a subpopulation of marmoset NK cells that express NKG2D and exhibit cytolytic potential. On the basis of their CD16 and CD56 expression patterns, marmoset NK cells can be classified into three subpopulations: CD16(+) CD56(-), CD16(-) CD56(+) and CD16(-) CD56(-) cells. NKG2D expression on marmoset CD16(+) CD56(-) and CD16(-) CD56(+) splenocytes was confirmed using an NKG2D ligand composed of an MHC class I chain-related molecule A (MICA)-Fc fusion protein. When marmoset splenocytes were cultured with IL-2 for 4 days, NKG2D expression was retained on CD16(+) CD56(-) and CD16(-) CD56(+). In addition, CD16(+) CD56(+) cells within the marmoset NK population appeared which expressed NKG2D after IL-2 stimulation. IL-2-activated marmoset NK cells showed strong cytolytic activity against K562 target cells and target cells stably expressing MICA. Further, the cytolytic activity of marmoset splenocytes was significantly reduced after addition of MICA-Fc fusion protein. Thus, NKG2D functions as an activating receptor on marmoset NK cells that possesses cytotoxic potential, and phenotypic profiles of marmoset NK cell subpopulations are similar to those seen in humans.
Collapse
Affiliation(s)
- Masamichi Watanabe
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Yohei Kudo
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Mitsuko Kawano
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Masafumi Nakayama
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kyohei Nakamura
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Mai Kameda
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Masamune Ebara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Takeki Sato
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Marina Nakamura
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kaito Omine
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Yoshie Kametani
- Department of Immunology, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Sagamihara 252-0315, Japan
| | - Kouetsu Ogasawara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
237
|
Lin Z, Wang C, Xia H, Liu W, Xiao W, Qian L, Jia X, Ding Y, Ji M, Gong W. CD4(+) NKG2D(+) T cells induce NKG2D down-regulation in natural killer cells in CD86-RAE-1ε transgenic mice. Immunology 2014; 141:401-15. [PMID: 24708417 DOI: 10.1111/imm.12203] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 10/23/2013] [Accepted: 10/23/2013] [Indexed: 11/30/2022] Open
Abstract
The binding of NKG2D to its ligands strengthens the cross-talk between natural killer (NK) cells and dendritic cells, particularly at early stages, before the initiation of the adaptive immune response. We found that retinoic acid early transcript-1ε (RAE-1ε), one of the ligands of NKG2D, was persistently expressed on antigen-presenting cells in a transgenic mouse model (pCD86-RAE-1ε). By contrast, NKG2D expression on NK cells, NKG2D-dependent cytotoxicity and tumour rejection, and dextran sodium sulphate-induced colitis were all down-regulated in this mouse model. The down-regulation of NKG2D on NK cells was reversed by stimulation with poly (I:C). The ectopic expression of RAE-1ε on dendritic cells maintained NKG2D expression levels and stimulated the activity of NK cells ex vivo, but the higher frequency of CD4(+) NKG2D(+) T cells in transgenic mice led to the down-regulation of NKG2D on NK cells in vivo. Hence, high levels of RAE-1ε expression on antigen-presenting cells would be expected to induce the down-regulation of NK cell activation by a regulatory T-cell subset.
Collapse
Affiliation(s)
- Zhijie Lin
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Ovarian tumor-associated microRNA-20a decreases natural killer cell cytotoxicity by downregulating MICA/B expression. Cell Mol Immunol 2014; 11:495-502. [PMID: 24813230 DOI: 10.1038/cmi.2014.30] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding regulatory RNAs, and changes in miRNAs are involved in tumor origin and progression. Studies have shown that miR-20a is overexpressed in human ovarian cancer tissues and that this miRNA enhances long-term cellular proliferation and invasion capabilities. In this study, a positive correlation between serum miR-20a expression and ovarian cancer stage was observed. We found that miR-20a binds directly to the 3'-untranslated region of MICA/B mRNA, resulting in its degradation and reducing its protein levels on the plasma membrane. Reduction of membrane-bound MICA/B proteins, which are ligands of the natural killer group 2 member D (NKG2D) receptor found on natural killer (NK) cells, γδ(+) T cells and CD8(+) T cells, allows tumor cells to evade immune-mediated killing. Notably, antagonizing miR-20a action enhanced the NKG2D-mediated killing of tumor cells in both in vitro and in vivo models of tumors. Taken together, our data indicate that increased levels of miR-20a in tumor cells may indirectly suppress NK cell cytotoxicity by downregulating MICA/B expression. These data provide a potential link between metastasis capability and immune escape of tumor cells from NK cells.
Collapse
|
239
|
Beltrán D, López-Vergès S. NK Cells during Dengue Disease and Their Recognition of Dengue Virus-Infected cells. Front Immunol 2014; 5:192. [PMID: 24829565 PMCID: PMC4017149 DOI: 10.3389/fimmu.2014.00192] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/18/2014] [Indexed: 12/13/2022] Open
Abstract
The innate immune response, in addition to the B- and T-cell response, plays a role in protection against dengue virus (DENV) infection and the degree of disease severity. Early activation of natural killer (NK) cells and type-I interferon-dependent immunity may be important in limiting viral replication during the early stages of DENV infection and thus reducing subsequent pathogenesis. NK cells may also produce cytokines that reduce inflammation and tissue injury. On the other hand, NK cells are also capable of inducing liver injury at early-time points of DENV infection. In vitro, NK cells can kill antibody-coated DENV-infected cells through antibody-dependent cell-mediated cytotoxicity. In addition, NK cells may directly recognize DENV-infected cells through their activating receptors, although the increase in HLA class I expression may allow infected cells to escape the NK response. Recently, genome-wide association studies have shown an association between MICB and MICA, which encode ligands of the activating NK receptor NKG2D, and dengue disease outcome. This review focuses on recognition of DENV-infected cells by NK cells and on the regulation of expression of NK cell ligands by DENV.
Collapse
Affiliation(s)
- Davis Beltrán
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies , Panama City , Panama ; Institute for Scientific Research and Technology Services (INDICASAT-AIP) , Panama City , Panama ; Department of Biotechnology, Acharya Nagarjuna University , Guntur , India
| | - Sandra López-Vergès
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies , Panama City , Panama
| |
Collapse
|
240
|
Poggi A, Musso A, Dapino I, Zocchi MR. Mechanisms of tumor escape from immune system: role of mesenchymal stromal cells. Immunol Lett 2014; 159:55-72. [PMID: 24657523 DOI: 10.1016/j.imlet.2014.03.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/01/2014] [Accepted: 03/05/2014] [Indexed: 12/12/2022]
Abstract
Tumor microenvironment represents the site where the tumor tries to survive and escape from immune system-mediated recognition. Indeed, to proliferate tumor cells can divert the immune response inducing the generation of myeloid derived suppressor cells and regulatory T cells which can limit the efficiency of effector antitumor lymphocytes in eliminating neoplastic cells. Many components of the tumor microenvironment can serve as a double sword for the tumor and the host. Several types of fibroblast-like cells, which herein we define mesenchymal stromal cells (MSC), secrete extracellular matrix components and surrounding the tumor mass can limit the expansion of the tumor. On the other hand, MSC can interfere with the immune recognition of tumor cells producing immunoregulatory cytokines as transforming growth factor (TGF)ß, releasing soluble ligands of the activating receptors expressed on cytolytic effector cells as decoy molecules, affecting the correct interaction among lymphocytes and tumor cells. MSC can also serve as target for the same anti-tumor effector lymphocytes or simply impede the interaction between these lymphocytes and neoplastic cells. Thus, several evidences point out the role of MSC, both in epithelial solid tumors and hematological malignancies, in regulating tumor cell growth and immune response. Herein, we review these evidences and suggest that MSC can be a suitable target for a more efficient anti-tumor therapy.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy.
| | - Alessandra Musso
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy
| | - Irene Dapino
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, Istituto Scientifico San Raffaele Milan, 20132 Milan, Italy
| |
Collapse
|
241
|
Schlecker E, Fiegler N, Arnold A, Altevogt P, Rose-John S, Moldenhauer G, Sucker A, Paschen A, von Strandmann EP, Textor S, Cerwenka A. Metalloprotease-Mediated Tumor Cell Shedding of B7-H6, the Ligand of the Natural Killer Cell–Activating Receptor NKp30. Cancer Res 2014; 74:3429-40. [DOI: 10.1158/0008-5472.can-13-3017] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
242
|
Abstract
Radiation therapy is currently one of the most widely utilized treatment strategies in the clinical management of cancer. Classically, radiation therapy was developed as an anticancer treatment on the basis of its capacity to induce DNA double strand breaks in exposed cancer cells, ultimately resulting in tumor cell death. Recently, our understanding of radiation effects has expanded widely in terms of the consequences of radiation-induced tumor cell death and the pertinent cells, signaling pathways, and molecular sensors that modify the tumor response to radiation. It is now well accepted that inflammation plays a complex dual role in promoting or inhibiting tumor growth. The capacity of inflammatory responses to alter the tumor response to radiation therapy, and vice versa, is now the subject of intense scientific and clinical investigation. Herein, we review the concepts regarding the immunostimulatory properties of radiation therapy with particular focus on the effects of radiation therapy on the tumor microenvironment.
Collapse
|
243
|
Yang F, Liu M, Yang F, Zhang X, Yang B, Guo C, Huang J, Che J, Yan Y, Zheng J. Matrix metallopeptidase 2 (MMP2) mediates MHC class I polypeptide-related sequence A (MICA) shedding in renal cell carcinoma. Actas Urol Esp 2014; 38:172-8. [PMID: 24461475 DOI: 10.1016/j.acuro.2013.09.015] [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: 04/16/2013] [Revised: 08/08/2013] [Accepted: 09/08/2013] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The MHC class i chain-related molecule A (MICA) is a ligand for the natural killer group 2, member D (NKG2D) immunoreceptor activation. The engagement of tumor cell surface MICA to NKG2D stimulates the NK and T cell antitumor immunity. Shedding of MICA by tumor cells facilitates tumor immune evasion, which might partially contribute to tumor progression. MATERIAL AND METHODS Inmunohistochemistry was performed on both normal and neoplastic renal tissue. Human renal carcinoma cell lines 786-0 and ACHIN were transfected and target sequences to silence human MMP2 by shRNA expression were established. The degree of MICA shedding was measured and quantitative real-time PCR and Western-blot analysis were performed. RESULTS The membrane type matrix metalloproteinase 2 (MMP2) mediated the MICA shedding, which was blocked by suppression of MMP2 expression. Concomitantly, MMP2 over-expression enhanced the MICA shedding, indicating that MMP2 was involved in the renal cell carcinoma-associated proteolytic release of soluble MICA (sMICA), which facilitated the tumor immune escape. CONCLUSIONS These findings suggested that MMP2 might be a new potential target for tumor immune therapy. Elucidation of the mechanisms by which tumors shed MICA could be of a great importance for cancer treatment in order to reinforce the NK and T cell antitumor immunity.
Collapse
|
244
|
Shifrin N, Raulet DH, Ardolino M. NK cell self tolerance, responsiveness and missing self recognition. Semin Immunol 2014; 26:138-44. [PMID: 24629893 PMCID: PMC3984600 DOI: 10.1016/j.smim.2014.02.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 02/12/2014] [Indexed: 01/01/2023]
Abstract
Natural killer (NK) cells represent a first line of defense against pathogens and tumor cells. The activation of NK cells is regulated by the integration of signals deriving from activating and inhibitory receptors expressed on their surface. However, different NK cells respond differently to the same stimulus, be it target cells or agents that crosslink activating receptors. The processes that determine the level of NK cell responsiveness have been referred to collectively as NK cell education. NK cell education plays an important role in steady state conditions, where potentially auto-reactive NK cells are rendered tolerant to the surrounding environment. According to the "tuning" concept, the responsiveness of each NK cell is quantitatively adjusted to ensure self tolerance while at the same time ensuring useful reactivity against potential threats. MHC-specific inhibitory receptors displayed by NK cells play a major role in tuning NK cell responsiveness, but recent studies indicate that signaling from activating receptors is also important, suggesting that the critical determinant is an integrated signal from both types of receptors. An important and still unresolved question is whether NK cell education involves interactions with a specific cell population in the environment. Whether hematopoietic and/or non-hematopoietic cells play a role is still under debate. Recent results demonstrated that NK cell tuning exhibits plasticity in steady state conditions, meaning that it can be re-set if the MHC environment changes. Other evidence suggests, however, that inflammatory conditions accompanying infections may favor high responsiveness, indicating that inflammatory agents can over-ride the natural tendency of NK cells to adjust to the steady state environment. These findings raise many questions such as whether viruses and tumor cells manipulate NK cell responsiveness to evade immune-recognition. As knowledge of the underlying processes grows, the possibility of modulating NK cell responsiveness for therapeutic purposes is becoming increasingly attractive, and is now under serious investigation in clinical studies.
Collapse
Affiliation(s)
- Nataliya Shifrin
- Department of Molecular and Cell Biology and Cancer Research Laboratory, Division of Immunology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - David H Raulet
- Department of Molecular and Cell Biology and Cancer Research Laboratory, Division of Immunology, University of California at Berkeley, Berkeley, CA 94720, USA.
| | - Michele Ardolino
- Department of Molecular and Cell Biology and Cancer Research Laboratory, Division of Immunology, University of California at Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
245
|
El-Gazzar A, Groh V, Spies T. Immunobiology and conflicting roles of the human NKG2D lymphocyte receptor and its ligands in cancer. THE JOURNAL OF IMMUNOLOGY 2014; 191:1509-15. [PMID: 23913973 DOI: 10.4049/jimmunol.1301071] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancers adopt diverse strategies to safeguard their survival, which often involve blinding or incapacitating the immune response, thereby gaining battleground advantage against the host. In immune responses against cancer, an important stimulatory lymphocyte receptor is NKG2D because the tumor-associated expression of its ligands promotes destruction of malignant cells. However, with advanced human cancers profound changes unfold wherein NKG2D and its ligands are targeted or exploited for immune evasion and suppression. This negative imprinting on the immune system may be accompanied by another functional state wherein cancer cells coopt expression of NKG2D to complement the presence of its ligands for self-stimulation of tumor growth and presumably malignant progression. This review emphasizes these conflicting functional dynamics at the immunity-cancer biology interface in humans, within an overview of the immunobiology of NKG2D and mechanisms underlying the regulation of its ligands in cancer, with reference to instructive clinical observations and translational approaches.
Collapse
Affiliation(s)
- Ahmed El-Gazzar
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | |
Collapse
|
246
|
Abstract
UL16 binding proteins (ULBPs) are a family of cell surface proteins that are present in transformed and stressed cells and ligands for NKG2D. Soluble NKG2D ligands have been found in sera from cancer patients with their protein concentrations correlated with poor cancer prognosis. Here we show, for the first time, that human tumor cells lost their surface expression of ULBP2, but not ULBP1 and ULBP3, during NK cell-mediated cytolysis. In contrast to spontaneous shedding of NKG2D ligands, NK cytolysis-mediated shedding of ULBP2 was linked to target cell apoptosis, although both resulted from metalloproteinase cleavages. Inhibition of ULBP2 shedding by a metalloproteinase inhibitor BB-94 lead to reduced NK cell-mediated cytotoxicity and cytokine production. These results illustrate a regulation of NK cell effector functions through cytolysis-induced NKG2D ligand shedding. Consequently, compounds inhibiting NKG2D ligand shedding may offer therapeutic means to reduce excessive pathogenic NK cell activities.
Collapse
Affiliation(s)
- Ruipeng Wang
- Structural Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Peter D. Sun
- Structural Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
247
|
Li W, Tian Y, Li Z, Gao J, Shi W, Zhu J, Zhang D. Ex vivo converted double negative T cells suppress activated B cells. Int Immunopharmacol 2014; 20:164-9. [PMID: 24613134 DOI: 10.1016/j.intimp.2014.02.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/21/2014] [Accepted: 02/24/2014] [Indexed: 02/06/2023]
Abstract
Although the ability of endogenous CD4(-)CD8(-) double negative (DN) T cells to suppress B cells has been documented, the extent to which ex vivo converted DN T cells suppress B cells activity is still being explored. The aim of this study was to determine whether and what extent ex vivo converted CD4(-)CD8(-) DN T cells suppress B cell activation and antibody production. We found that ex vivo converted DN T cells suppressed proliferation of activated B cells in a perforin and cell-cell contact dependent manner. In addition, ex vivo converted DN T cells significantly inhibited the production of IgG by stimulated B cells. This study provides evidence that ex vivo converted CD4(-)CD8(-) double negative T cells can down-regulate immune responses by suppressing B cell proliferation and IgG production, and supports efforts to develop ex vivo DN T cell therapies.
Collapse
Affiliation(s)
- WenXia Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yue Tian
- Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95, Yongan Road, Xicheng District, Beijing 100050, China
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Wen Shi
- Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95, Yongan Road, Xicheng District, Beijing 100050, China
| | - JiYe Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, NO. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China.
| | - Dong Zhang
- Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95, Yongan Road, Xicheng District, Beijing 100050, China.
| |
Collapse
|
248
|
Crosby EJ, Goldschmidt MH, Wherry EJ, Scott P. Engagement of NKG2D on bystander memory CD8 T cells promotes increased immunopathology following Leishmania major infection. PLoS Pathog 2014; 10:e1003970. [PMID: 24586170 PMCID: PMC3937277 DOI: 10.1371/journal.ppat.1003970] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/17/2014] [Indexed: 02/07/2023] Open
Abstract
One of the hallmarks of adaptive immunity is the development of a long-term pathogen specific memory response. While persistent memory T cells certainly impact the immune response during a secondary challenge, their role in unrelated infections is less clear. To address this issue, we utilized lymphocytic choriomeningitis virus (LCMV) and Listeria monocytogenes immune mice to investigate whether bystander memory T cells influence Leishmania major infection. Despite similar parasite burdens, LCMV and Listeria immune mice exhibited a significant increase in leishmanial lesion size compared to mice infected with L. major alone. This increased lesion size was due to a severe inflammatory response, consisting not only of monocytes and neutrophils, but also significantly more CD8 T cells. Many of the CD8 T cells were LCMV specific and expressed gzmB and NKG2D, but unexpectedly expressed very little IFN-γ. Moreover, if CD8 T cells were depleted in LCMV immune mice prior to challenge with L. major, the increase in lesion size was lost. Strikingly, treating with NKG2D blocking antibodies abrogated the increased immunopathology observed in LCMV immune mice, showing that NKG2D engagement on LCMV specific memory CD8 T cells was required for the observed phenotype. These results indicate that bystander memory CD8 T cells can participate in an unrelated immune response and induce immunopathology through an NKG2D dependent mechanism without providing increased protection. Cutaneous leishmaniasis has a wide spectrum of clinical presentations, from mild self-healing lesions to severe chronic infections. Differences in each individual's response are related to pathogen dose and the genetic and physiological status of the host, but exactly what causes the broad spectrum of disease is not well understood. Here we show that previous infection with a viral or bacterial pathogen led to increased immunopathology associated with L. major infection. This increase in immunopathology was not associated with any changes in parasite control and was characterized by an exaggerated inflammatory infiltrate into the site of infection. Ultimately, this increase in immunopathology was dependent on the presence of memory CD8 T cells from the previous infection and their expression of the NK cell receptor NKG2D, as depletion of these cells prior to infection with L. major or blockade of this receptor during infection ameliorated the disease. Our work suggests that the immunological history of a patient may be playing an underlying role in the pathology associated with leishmania infection and could be an important consideration for the understanding and treatment of this and other human diseases. This work also identifies the NKG2D pathway as a potential new target for therapeutic intervention.
Collapse
Affiliation(s)
- Erika J. Crosby
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael H. Goldschmidt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - E. John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
249
|
Son CH, Keum JH, Yang K, Nam J, Kim MJ, Kim SH, Kang CD, Oh SO, Kim CD, Park YS, Bae J. Synergistic enhancement of NK cell-mediated cytotoxicity by combination of histone deacetylase inhibitor and ionizing radiation. Radiat Oncol 2014; 9:49. [PMID: 24512718 PMCID: PMC3923253 DOI: 10.1186/1748-717x-9-49] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 12/06/2013] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The overexpression of histone deacetylase (HDAC) and a subsequent decrease in the acetylation levels of nuclear histones are frequently observed in cancer cells. Generally it was accepted that the deacetylation of histones suppressed expression of the attached genes. Therefore, it has been suggested that HDAC might contribute to the survival of cancer cells by altering the NKG2D ligands transcripts. By the way, the translational regulation of NKG2D ligands remains unclear in cancer cells. It appears the modulation of this unclear mechanism could enhance NKG2D ligand expressions and the susceptibility of cancer cells to NK cells. Previously, it was reported that irradiation can increase the surface expressions of NKG2D ligands on several cancer cell types without increasing the levels of NKG2D ligand transcripts via ataxia telangiectasia mutated and ataxia telangiectasia and Rad3 related (ATM-ATR) pathway, and suggested that radiation therapy might be used to increase the translation of NKG2D ligands. METHODS Two NSCLC cell lines, that is, A549 and NCI-H23 cells, were used to investigate the combined effects of ionizing radiation and HDAC inhibitors on the expressions of five NKG2D ligands. The mRNA expressions of the NKG2D ligands were quantitated by multiplex reverse transcription-PCR. Surface protein expressions were measured by flow cytometry, and the susceptibilities of cancer cells to NK cells were assayed by time-resolved fluorometry using the DELFIA® EuTDA cytotoxicity kit and by flow cytometry. RESULTS The expressions of NKG2D ligands were found to be regulated at the transcription and translation levels. Ionizing radiation and HDAC inhibitors in combination synergistically increased the expressions of NKG2D ligands. Furthermore, treatment with ATM-ATR inhibitors efficiently blocked the increased translations of NKG2D ligands induced by ionizing radiation but did not block the increased ligand translations induced by HDAC inhibitors. The study confirms that increased NKG2D ligand levels by ionizing radiation and HDAC inhibitors could synergistically enhance the susceptibilities of cancer cells to NK-92 cells. CONCLUSIONS This study suggests that the expressions of NKG2D ligands are regulated in a complex manner at the multilevel of gene expression, and that their expressions can be induced by combinatorial treatments in lung cancer cells.
Collapse
Affiliation(s)
- Cheol-Hun Son
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, South Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Sckisel GD, Tietze JK, Zamora AE, Hsiao HH, Priest SO, Wilkins DEC, Lanier LL, Blazar BR, Baumgarth N, Murphy WJ. Influenza infection results in local expansion of memory CD8(+) T cells with antigen non-specific phenotype and function. Clin Exp Immunol 2014; 175:79-91. [PMID: 23937663 DOI: 10.1111/cei.12186] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2013] [Indexed: 12/30/2022] Open
Abstract
Primary viral infections induce activation of CD8(+) T cells responsible for effective resistance. We sought to characterize the nature of the CD8(+) T cell expansion observed after primary viral infection with influenza. Infection of naive mice with different strains of influenza resulted in the rapid expansion of memory CD8(+) T cells exhibiting a unique bystander phenotype with significant up-regulation of natural killer group 2D (NKG2D), but not CD25, on the CD44(high) CD8(+) T cells, suggesting an antigen non-specific phenotype. We further confirmed the non-specificity of this phenotype on ovalbumin-specific (OT-I) CD8(+) T cells, which are not specific to influenza. These non-specific CD8(+) T cells also displayed increased lytic capabilities and were observed primarily in the lung. Thus, influenza infection was shown to induce a rapid, antigen non-specific memory T cell expansion which is restricted to the specific site of inflammation. In contrast, CD8(+) T cells of a similar phenotype could be observed in other organs following administration of systemic agonistic anti-CD40 and interleukin-2 immunotherapy, demonstrating that bystander expansion in multiple sites is possible depending on whether the nature of activation is either acute or systemic. Finally, intranasal blockade of NKG2D resulted in a significant increase in viral replication early during the course of infection, suggesting that NKG2D is a critical mediator of anti-influenza responses prior to the initiation of adaptive immunity. These results characterize further the local bystander expansion of tissue-resident, memory CD8(+) T cells which, due to their early induction, may play an important NKG2D-mediated, antigen non-specific role during the early stages of viral infection.
Collapse
Affiliation(s)
- Gail D Sckisel
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Graduate Group in Immunology, University of California, Davis, Davis, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|