101
|
Lazarova M, Steinle A. The NKG2D axis: an emerging target in cancer immunotherapy. Expert Opin Ther Targets 2019; 23:281-294. [DOI: 10.1080/14728222.2019.1580693] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Mariya Lazarova
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
102
|
Abumaree MH, Alshehri NA, Almotery A, Al Subayyil AM, Bahattab E, Abomaray FM, Khatlani T, Kalionis B, Jawdat D, El-Muzaini MF, Al Jumah MA, AlAskar AS. Preconditioning human natural killer cells with chorionic villous mesenchymal stem cells stimulates their expression of inflammatory and anti-tumor molecules. Stem Cell Res Ther 2019; 10:50. [PMID: 30728068 PMCID: PMC6366106 DOI: 10.1186/s13287-019-1153-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/22/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
Background Mesenchymal stem cells derived from the chorionic villi of human placentae (pMSCs) produce a unique array of mediators that regulate the essential cellular functions of their target cells. These properties make pMSCs attractive candidates for cell-based therapy. Here, we examined the effects of culturing human natural killer (NK) cells with pMSCs on NK cell functions. Methods pMSCs were cultured with IL-2-activated and non-activated NK cells. NK cell proliferation and cytolytic activities were monitored. NK cell expression of receptors mediating their cytolytic activity against pMSCs, and the mechanisms underlying this effect on pMSCs, were also investigated. Results Our findings show that IL-2-activated NK cells, but not freshly isolated NK cells, efficiently lyse pMSCs and that this response might involve the activating NK cell receptor CD69. Interestingly, although pMSCs expressed HLA class I molecules, they were nevertheless lysed by NK cells, suggesting that HLA class I antigens do not play a significant role in protecting pMSCs from NK cell cytolytic activity. Co-culturing NK cells with pMSCs also inhibited NK cell expression of receptors, including CD69, NKpG2D, CD94, and NKp30, although these co-cultured NK cells were not inhibited in lysing cancer cells in vitro. Importantly, co-cultured NK cells significantly increased their production of molecules with anti-tumor effects. Conclusions These findings suggest that pMSCs might have potential applications in cancer therapy.
Collapse
Affiliation(s)
- M H Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia. .,College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, 11481, Mail Code 3124, Riyadh, Saudi Arabia.
| | - N A Alshehri
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia
| | - A Almotery
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, P.O. Box 3660, 11481, Mail Code, Riyadh, 3124, Saudi Arabia
| | - A M Al Subayyil
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia
| | - E Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - F M Abomaray
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, 14186, Stockholm, Sweden.,Center for Hematology and Regenerative Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - T Khatlani
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia
| | - B Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - D Jawdat
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia
| | - M F El-Muzaini
- Department of Obstetrics and Gynaecology, King Abdulaziz Medical City, Minstry of National Guard Health Affairs, P.O. Box 3660, 11481, Mail Code, Riyadh, 3124, Saudi Arabia
| | - M A Al Jumah
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia
| | - A S AlAskar
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code 1515, Riyadh, Saudi Arabia.,College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, 11481, Mail Code, Riyadh, 3124, Saudi Arabia.,Adult Hematology and Stem Cell Transplantation, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, 11426, Mail Code, Riyadh, 1515, Saudi Arabia
| |
Collapse
|
103
|
Kloess S, Kretschmer A, Stahl L, Fricke S, Koehl U. CAR-Expressing Natural Killer Cells for Cancer Retargeting. Transfus Med Hemother 2019; 46:4-13. [PMID: 31244577 DOI: 10.1159/000495771] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/23/2018] [Indexed: 12/15/2022] Open
Abstract
Since the approval in 2017 and the outstanding success of Kymriah® and Yescarta®, the number of clinical trials investigating the safety and efficacy of chimeric antigen receptor-modified autologous T cells has been constantly rising. Currently, more than 200 clinical trials are listed on clinicaltrial.gov. In contrast to CAR-T cells, natural killer (NK) cells can be used from allogeneic donors as an "off the shelf product" and provide alternative candidates for cancer retargeting. This review summarises preclinical results of CAR-engineered NK cells using both primary human NK cells and the cell line NK-92, and provides an overview about the first clinical CAR-NK cell studies targeting haematological malignancies and solid tumours, respectively.
Collapse
Affiliation(s)
- Stephan Kloess
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute for Cellular Therapeutics, ATMP-GMPDU, Hannover Medical School, Hannover, Germany
| | - Anna Kretschmer
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Lilly Stahl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Clinical Immunology, Faculty of Medicine, University Leipzig, Leipzig, Germany.,Institute for Cellular Therapeutics, ATMP-GMPDU, Hannover Medical School, Hannover, Germany
| |
Collapse
|
104
|
Increase of Tumor Infiltrating γδ T-cells in Pancreatic Ductal Adenocarcinoma Through Remodeling of the Extracellular Matrix by a Hyaluronan Synthesis Suppressor, 4-Methylumbelliferone. Pancreas 2019; 48:292-298. [PMID: 30589828 DOI: 10.1097/mpa.0000000000001211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Desmoplastic changes of extracellular matrix (ECM) containing large amounts of hyaluronan (HA) are of interest in chemo- and immunoresistance of pancreatic ductal adenocarcinoma (PDAC). The goal of this study was to evaluate the effects of 4-methylumbelliferone (MU), a selective inhibitor of HA, on ECM and to examine how MU affects adoptive immunotherapy. METHODS The effect of MU on cell proliferation, HA synthesis and formation of ECM were investigated in four PDAC cell lines. In addition, the cytotoxicity of γδ T-cell-rich peripheral blood mononuclear cells (PBMCs) collected from healthy donors and stimulated with zoledronate and interleukin-2 was examined in the presence of MU. The amount of HA and tumor-infiltrating lymphocytes were also investigated in mice xenograft models. RESULTS In vitro, 1.0 mM MU inhibited cell proliferation by 45-70% and HA synthesis by 55-80% in all four PDAC cell lines, and enhanced γδ T-cell-rich PBMC-mediated cytotoxicity against PDAC cells. In vivo, MU reduced intratumoral HA and promoted infiltration of inoculated γδ T-cells into tumor tissue, and consequently suppressed tumor growth. CONCLUSIONS 4-methylumbelliferone may be an effective immunosensitizer against PDAC through induction of structural changes in the ECM.
Collapse
|
105
|
Tremblay-McLean A, Coenraads S, Kiani Z, Dupuy FP, Bernard NF. Expression of ligands for activating natural killer cell receptors on cell lines commonly used to assess natural killer cell function. BMC Immunol 2019; 20:8. [PMID: 30696399 PMCID: PMC6352444 DOI: 10.1186/s12865-018-0272-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Natural killer cell responses to virally-infected or transformed cells depend on the integration of signals received through inhibitory and activating natural killer cell receptors. Human Leukocyte Antigen null cells are used in vitro to stimulate natural killer cell activation through missing-self mechanisms. On the other hand, CEM.NKr.CCR5 cells are used to stimulate natural killer cells in an antibody dependent manner since they are resistant to direct killing by natural killer cells. Both K562 and 721.221 cell lines lack surface major histocompatibility compatibility complex class Ia ligands for inhibitory natural killer cell receptors. Previous work comparing natural killer cell stimulation by K562 and 721.221 found that they stimulated different frequencies of natural killer cell functional subsets. We hypothesized that natural killer cell function following K562, 721.221 or CEM.NKr.CCR5 stimulation reflected differences in the expression of ligands for activating natural killer cell receptors. RESULTS K562 expressed a higher intensity of ligands for Natural Killer G2D and the Natural Cytotoxicity Receptors, which are implicated in triggering natural killer cell cytotoxicity. 721.221 cells expressed a greater number of ligands for activating natural killer cell receptors. 721.221 expressed cluster of differentiation 48, 80 and 86 with a higher mean fluorescence intensity than did K562. The only ligands for activating receptor that were detected on CEM.NKr.CCR5 cells at a high intensity were cluster of differentiation 48, and intercellular adhesion molecule-2. CONCLUSIONS The ligands expressed by K562 engage natural killer cell receptors that induce cytolysis. This is consistent with the elevated contribution that the cluster of differentiation 107a function makes to total K562 induced natural killer cell functionality compared to 721.221 cells. The ligands expressed on 721.221 cells can engage a larger number of activating natural killer cell receptors, which may explain their ability to activate a larger frequency of these cells to become functional and secrete cytokines. The few ligands for activating natural killer cell receptors expressed by CEM.NKr.CCR5 may reduce their ability to activate natural killer cells in an antibody independent manner explaining their relative resistance to direct natural killer cell cytotoxicity.
Collapse
Affiliation(s)
- Alexandra Tremblay-McLean
- Research Institute of the McGill University Health Center, Glen Site, 1001 Décarie Boulevard, Block E, Rm EM3.3238, Montréal, Québec, H4A 3J1, Canada.,Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Sita Coenraads
- Research Institute of the McGill University Health Center, Glen Site, 1001 Décarie Boulevard, Block E, Rm EM3.3238, Montréal, Québec, H4A 3J1, Canada
| | - Zahra Kiani
- Research Institute of the McGill University Health Center, Glen Site, 1001 Décarie Boulevard, Block E, Rm EM3.3238, Montréal, Québec, H4A 3J1, Canada.,Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Franck P Dupuy
- Research Institute of the McGill University Health Center, Glen Site, 1001 Décarie Boulevard, Block E, Rm EM3.3238, Montréal, Québec, H4A 3J1, Canada
| | - Nicole F Bernard
- Research Institute of the McGill University Health Center, Glen Site, 1001 Décarie Boulevard, Block E, Rm EM3.3238, Montréal, Québec, H4A 3J1, Canada. .,Division of Experimental Medicine, McGill University, Montréal, Québec, Canada. .,Chronic Viral Illness Service, McGill University Health Centre, Montréal, Québec, Canada. .,Division of Clinical Immunology, McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
106
|
Trinh TL, Kandell WM, Donatelli SS, Tu N, Tejera MM, Gilvary DL, Eksioglu EA, Burnette A, Adams WA, Liu J, Teer JK, Djeu JY, Coppola D, Wei S. Immune evasion by TGFβ-induced miR-183 repression of MICA/B expression in human lung tumor cells. Oncoimmunology 2019; 8:e1557372. [PMID: 30906652 PMCID: PMC6422376 DOI: 10.1080/2162402x.2018.1557372] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 11/07/2018] [Accepted: 12/01/2018] [Indexed: 01/02/2023] Open
Abstract
Immune escape is a hallmark of cancer. In human lung cancer, we have identified a unique microRNA (miR)-based pathway employed by tumor cells to repress detection by immune cells via the NKG2D-MICA/B receptor-ligand system. MICA/B is readily induced by cell transformation and serves as a danger signal and ligand to alert NK and activated CD8+ T cells. However, immunohistochemical analysis indicated that human lung adenocarcinoma and squamous cell carcinoma specimens express little MICA/B while high levels of miR-183 were detected in both tumor types in a TCGA database. Human lung tumor cell lines confirmed the reverse relationship in expression of MICA/B and miR-183. Importantly, a miR-183 binding site was identified on the 3'untranslated region (UTR) of both MICA and MICB, suggesting its role in MICA/B regulation. Luciferase reporter constructs bearing the 3'UTR of MICA or MICB in 293 cells supported the function of miR-183 in repressing MICA/B expression. Additionally, anti-sense miR-183 transfection into H1355 or H1299 tumor cells caused the upregulation of MICA/B. Abundant miR-183 expression in tumor cells was traced to transforming growth factor-beta (TGFβ), as evidenced by antisense TGFβ transfection into H1355 or H1299 tumor cells which subsequently lost miR-183 expression accompanied by MICA/B upregulation. Most significantly, anti-sense miR-183 transfected tumor cells became more sensitive to lysis by activated CD8+ T cells that express high levels of NKG2D. Thus, high miR-183 triggered by TGFβ expressed in lung tumor cells can target MICA/B expression to circumvent detection by NKG2D on immune cells.
Collapse
Affiliation(s)
- Thu Le Trinh
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Wendy M Kandell
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
| | | | - Nhan Tu
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Melba M Tejera
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Erika A Eksioglu
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Alexis Burnette
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - William A Adams
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jinhong Liu
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jamie K Teer
- Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Julie Y Djeu
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Sheng Wei
- Departments of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
107
|
Barrow AD, Colonna M. Exploiting NK Cell Surveillance Pathways for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11010055. [PMID: 30626155 PMCID: PMC6356551 DOI: 10.3390/cancers11010055] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/28/2018] [Accepted: 01/03/2019] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells can evoke potent anti-tumour activity. This function is largely mediated through a battery of specialised cell-surface receptors which probe the tissue microenvironment for changes in surface and secretory phenotypes that may alert to the presence of infection or malignancy. These receptors have the potential to arouse the robust cytotoxic and cytokine-secreting functions of NK cells and so must be tightly regulated to prevent autoimmunity. However, such functions also hold great promise for clinical intervention. In this review, we highlight some of the latest breakthroughs in fundamental NK cell receptor biology that have illuminated our understanding of the molecular strategies NK cells employ to perceive malignant cells from normal healthy cells. Moreover, we highlight how these sophisticated tumour recognition strategies are being harnessed for cancer immunotherapies in the clinic.
Collapse
Affiliation(s)
- Alexander David Barrow
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia.
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
108
|
Le Saux G, Bar-Hanin N, Edri A, Hadad U, Porgador A, Schvartzman M. Nanoscale Mechanosensing of Natural Killer Cells is Revealed by Antigen-Functionalized Nanowires. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805954. [PMID: 30488552 DOI: 10.1002/adma.201805954] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/06/2018] [Indexed: 06/09/2023]
Abstract
Cells sense their environment by transducing mechanical stimuli into biochemical signals. Commonly used tools to study cell mechanosensing provide limited spatial and force resolution. Here, a novel nanowire-based platform for monitoring cell forces is reported. Nanowires are functionalized with ligands for cell immunoreceptors, and they are used to explore the mechanosensitivity of natural killer (NK) cells. In particular, it is found that NK cells apply centripetal forces to nanowires, and that the nanowires stimulate cell contraction. Based on the nanowire deformation, it is calculated that cells apply forces of down to 10 pN, which is the smallest value demonstrated so far by microstructured platforms for cell spreading. Furthermore, the roles of: i) nanowire topography and ii) activating ligands in the cell immune function are studied and it is found that only their combination produces enhanced population of activated NK cells. Thus, a mechanosensing mechanism of NK cells is proposed, by which they integrate biochemical and mechanical stimuli into a decision-making machinery analogous to the AND logic gate, whose output is the immune activation. This work reveals unprecedented mechanical aspects of NK cell immune function and introduces an innovative nanomaterial for studying cellular mechanics with unparalleled spatial and mechanical resolution.
Collapse
Affiliation(s)
- Guillaume Le Saux
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Netanel Bar-Hanin
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Uzi Hadad
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| |
Collapse
|
109
|
Luo D, Dong XW, Yan B, Liu M, Xue TH, Liu H, You JH, Li F, Wang ZL, Chen ZN. MG132 selectively upregulates MICB through the DNA damage response pathway in A549 cells. Mol Med Rep 2018; 19:213-220. [PMID: 30483783 PMCID: PMC6297755 DOI: 10.3892/mmr.2018.9676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
Natural killer (NK) cells recognize stress-activated NK group 2, member D (NKG2D) ligands in tumors. In the present study, the expression levels of NKG2D ligands were examined in four lung cancer cell lines (A549, PLA801D, NCI-H157 and NCI-H520). In the A549 cells, the expression of MHC class I polypeptiderelated sequence (MIC)A/B and UL16 binding protein (ULBP)1 was weak, the expression of ULBP2 was typical, and neither ULBP3 nor ULBP4 were expressed. The mechanism underlying the regulatory effect of a cancer treatment agent on the expression of NKG2D ligands was investigated using the proteasome inhibitor MG132. Following treatment for 8 h with MG132, the transcription levels of MICB and ULBP1 were upregulated 10.62- and 11.09-fold, respectively, and the expression levels of MICB and ULBP1 were increased by 68.18 and 23.65%, respectively. Notably, MICB exhibited significant time-dependent change. MG132 increased the transcription of MICB by acting at a site in the 480-bp MICB upstream promoter. The activity of the MICB promoter was upregulated 1.77-fold following treatment with MG132. MG132 treatment improved the cytotoxicity of NK cells, which was partially blocked by an antibody targeting NKG2D, and more specifically the MICB molecule. The expression of MICB induced by MG132 was inhibited by KU-55933 [ataxia telangiectasia mutated (ATM) kinase inhibitor], wortmannin (phosphoinositide 3 kinase inhibitor) and caffeine (ATM/ATM-Rad3-related inhibitor). The phosphorylation of checkpoint kinase 2 (Chk2), an event associated with DNA damage, was observed following treatment with MG132. These results indicated that MG132 selectively upregulates the expression of MICB in A549 cells, and increases the NKG2D-mediated cytotoxicity of NK cells. The regulatory effect of MG132 may be associated with the activation of Chk2, an event associated with DNA damage. The combination of MG132 with NK cell immunotherapy may have a synergistic effect that improves the therapeutic effect of lung cancer treatment.
Collapse
Affiliation(s)
- Dan Luo
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Xi-Wen Dong
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Bing Yan
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Mei Liu
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Tian-Hui Xue
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Hui Liu
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Jun-Hao You
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Fang Li
- Department of Oncology, Hainan Branch of General Hospital of PLA, Sanya, Hainan 572013, P.R. China
| | - Zi-Ling Wang
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Zhi-Nan Chen
- College of Life Science and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| |
Collapse
|
110
|
Kajikawa M, Ose T, Fukunaga Y, Okabe Y, Matsumoto N, Yonezawa K, Shimizu N, Kollnberger S, Kasahara M, Maenaka K. Structure of MHC class I-like MILL2 reveals heparan-sulfate binding and interdomain flexibility. Nat Commun 2018; 9:4330. [PMID: 30337538 PMCID: PMC6193965 DOI: 10.1038/s41467-018-06797-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 09/27/2018] [Indexed: 12/23/2022] Open
Abstract
The MILL family, composed of MILL1 and MILL2, is a group of nonclassical MHC class I molecules that occur in some orders of mammals. It has been reported that mouse MILL2 is involved in wound healing; however, the molecular mechanisms remain unknown. Here, we determine the crystal structure of MILL2 at 2.15 Å resolution, revealing an organization similar to classical MHC class I. However, the α1-α2 domains are not tightly fixed on the α3-β2m domains, indicating unusual interdomain flexibility. The groove between the two helices in the α1-α2 domains is too narrow to permit ligand binding. Notably, an unusual basic patch on the α3 domain is involved in the binding to heparan sulfate which is essential for MILL2 interactions with fibroblasts. These findings suggest that MILL2 has a unique structural architecture and physiological role, with binding to heparan sulfate proteoglycans on fibroblasts possibly regulating cellular recruitment in biological events. The MILL (MHC-I-like located near the leukocyte receptor complex) family is a group of related nonclassical MHC-I molecules. Here the authors present the crystal structure of MILL2, which reveals an unusual interdomain flexibility, and show that MILL2 binds heparan sulfate on the surface of fibroblasts through a basic patch.
Collapse
Affiliation(s)
- Mizuho Kajikawa
- Laboratory of Microbiology, Showa Pharmaceutical University, Machida, Tokyo, 190-8543, Japan.,Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Toyoyuki Ose
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yuko Fukunaga
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yuki Okabe
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.,Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Naoki Matsumoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Kento Yonezawa
- Photon Factory, High Energy Accelerator Research Organization, Tsukuba, Ibaraki, 305-0801, Japan
| | - Nobutaka Shimizu
- Photon Factory, High Energy Accelerator Research Organization, Tsukuba, Ibaraki, 305-0801, Japan
| | - Simon Kollnberger
- Cardiff Institute of Infection & Immunity, University of Cardiff, Henry Wellcome Building, Heath Park, Cardiff, CF14 4XN, UK
| | - Masanori Kasahara
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Katsumi Maenaka
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan. .,Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
111
|
Fong JJ, Tsai CM, Saha S, Nizet V, Varki A, Bui JD. Siglec-7 engagement by GBS β-protein suppresses pyroptotic cell death of natural killer cells. Proc Natl Acad Sci U S A 2018; 115:10410-10415. [PMID: 30254166 PMCID: PMC6187154 DOI: 10.1073/pnas.1804108115] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and destroy abnormal host cells, such as tumor cells or those infected by viral pathogens. To safely accomplish these functions, NK cells display activating receptors that detect stress molecules or viral ligands displayed at the cell surface, balanced by inhibitory receptors that bind to self-molecules. To date, such activating and inhibitory receptors on NK cells are not known to recognize bacterial determinants. Moreover, NK cell responses to direct interactions with extracellular bacteria are poorly explored. In this study, we observed the human neonatal pathogen group B Streptococcus (GBS) can directly engage human NK cells. The interaction was mediated through the B6N segment of streptococcal β-protein, binding to the inhibitory receptor Siglec-7 via its amino-terminal V-set domain. Unlike classical Siglec binding, the interaction is also independent of its sialic acid recognition property. In contrast to WT GBS, mutants lacking β-protein induced efficient pyroptosis of NK cells through the NLRP3 inflammasome, with production and secretion of the proinflammatory cytokine IL-1β and dissemination of the cytotoxic molecule granzyme B. We postulate that GBS evolved β-protein engagement of inhibitory human Siglec-7 to suppress the pyroptotic response of NK cells and thereby block recruitment of a broader innate immune response, i.e., by "silencing the sentinel."
Collapse
Affiliation(s)
- Jerry J Fong
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Chih-Ming Tsai
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sudeshna Saha
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Victor Nizet
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Skaggs School of Pharmacy and Pharmaceutical Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ajit Varki
- Glycobiology Research and Training Center, School of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Jack D Bui
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
112
|
Azzawi S, Penzi LR, Senna MM. Immune Privilege Collapse and Alopecia Development: Is Stress a Factor. Skin Appendage Disord 2018; 4:236-244. [PMID: 30410890 PMCID: PMC6219219 DOI: 10.1159/000485080] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/07/2017] [Indexed: 01/05/2023] Open
Abstract
Hair is a defining mammalian feature that serves as a hallmark of human communication. Given the critical significance of hair in social, religious, and political contexts, it is important to understand factors that play a role in hair loss disorders. The hair follicle is an immune privileged site, and mounting evidence suggests that the collapse of immune privilege contributes to the pathogenesis of autoimmune hair loss disorders, including alopecia areata and lichen planopilaris. This review comprehensively appraises the current literature to shed light on mechanisms for immune privilege collapse, and examines the role of neurogenic stress in triggering this process.
Collapse
Affiliation(s)
| | - Lauren R. Penzi
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| | - Maryanne M. Senna
- Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
113
|
Schmiedel D, Mandelboim O. NKG2D Ligands-Critical Targets for Cancer Immune Escape and Therapy. Front Immunol 2018; 9:2040. [PMID: 30254634 PMCID: PMC6141707 DOI: 10.3389/fimmu.2018.02040] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022] Open
Abstract
DNA damage, oncogene activation and excessive proliferation, chromatin modulations or oxidative stress are all important hallmarks of cancer. Interestingly, all of these abnormalities also induce a cellular stress response. By upregulating “stress-induced ligands,” damaged or transformed cells can be recognized by immune cells and cleared. The human genome encodes eight functional “stress-induced ligands”: MICA, MICB, and ULBP1-6. All of them are recognized by a single receptor, NKG2D, which is expressed on natural killer (NK) cells, cytotoxic T cells and other T cell subsets. The NKG2D ligand/NKG2D-axis is well-recognized as an important mediator of anti-tumor activity; however, patient data about the role of NKG2D ligands in immune surveillance and escape appears conflicting. As these ligands are often actively transcribed, tumor cells are urged to manipulate the expression of these ligands on post-transcriptional or post-translational level. Although our knowledge on the regulation of NKG2D ligand expression remains fragmentary, research of the past years revealed multiple cellular mechanisms that are adopted by tumor cells to reduce the expression of “stress-induced ligands” and therefore escape immune recognition. Here, we review the post-transcriptional and post-translational mechanisms by which NKG2D ligands are modulated in cancer cells and their impact on patient prognosis.We discuss controversies and approaches to apply our understanding of the NKG2D ligand/NKG2D-axis for cancer therapy.
Collapse
Affiliation(s)
- Dominik Schmiedel
- 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, Jerusalem, Israel
| | - 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, Jerusalem, Israel
| |
Collapse
|
114
|
Tao K, He M, Tao F, Xu G, Ye M, Zheng Y, Li Y. Development of NKG2D-based chimeric antigen receptor-T cells for gastric cancer treatment. Cancer Chemother Pharmacol 2018; 82:815-827. [PMID: 30132099 DOI: 10.1007/s00280-018-3670-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022]
Abstract
Gastric cancer is the third leading cause of cancer-related mortalities worldwide and mostly incurable. It remains an urgent need for novel strategies in the management of patients with advanced gastric cancer. Chimeric antigen receptor (CAR) T therapy has shown unprecedented clinical success in hematological malignancies and potential utility is going on various solid tumors like gastric cancer. In this study, a broad expression of NKG2D ligands was observed in gastric cancer cell lines, making them suitable targets for gastric cancer therapy. T cells were engineered with an NKG2D-based second-generation CAR and the resulting NKG2D-CAR-T cells showed significantly increased cytolytic activity against gastric cancer compared to untransduced T cells. In vivo, these cells can significantly suppressed the growth of established gastric cancer xenografts. Besides, cisplatin was shown to upregulate NKG2D ligand expression in gastric cancer cells and enhance the susceptibility to NKG2D-CAR-T-cell-mediated cytotoxicity. In conclusion, NKG2D-based CAR-T cells have potent in vivo and in vitro anti-tumor activities against gastric cancer and could be a new paradigm for patients with gastric cancer, either used alone or combined with chemotherapy.
Collapse
Affiliation(s)
- Kelong Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Meng He
- Department of Respiratory Medicine, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, People's Republic of China
| | - Feng Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Guangen Xu
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Minfeng Ye
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, People's Republic of China
| | - Yaoqing Li
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, People's Republic of China.
| |
Collapse
|
115
|
Keydar Y, Le Saux G, Pandey A, Avishay E, Bar-Hanin N, Esti T, Bhingardive V, Hadad U, Porgador A, Schvartzman M. Natural killer cells' immune response requires a minimal nanoscale distribution of activating antigens. NANOSCALE 2018; 10:14651-14659. [PMID: 30033475 DOI: 10.1039/c8nr04038a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
NK cells recognize cancer and viral cells by binding their activating receptors to antigens presenting on the membrane of target cells. Although the activation mechanism of NK cells is a subject of extensive research today, the role of the composition and spatial distribution of activating ligands in NK cell cytotoxicity is barely understood. In this work, we engineered a nanochip whose surface was patterned with matrices of antigens for NKG2D activating receptors. These matrices mimicked the spatial order of the surface of antigen presenting cells with molecular resolution. Using this chip, we elucidated the effect of the antigen spatial distribution on the NK cell spreading and immune activation. We found that the spatial distribution of the ligand within the 100 nm length-scale provides the minimal conditions for NKG2D regulated cell spreading. Furthermore, we found that the immune activation of NK cells requires the same minimal spatial distribution of activating ligands. Above this threshold, both spreading and activation plateaued, confirming that these two cell functions work hand in hand. Our study provides an important insight on the spatial mechanism of the cytotoxic activity of NK cells. This insight opens the way to rationally designed antitumor therapies that harness NK cytotoxicity.
Collapse
Affiliation(s)
- Yossi Keydar
- Department of Materials Engineering, Ben Gurion University of the Negev, Beer Sheva 84105, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Chan WK, Kang S, Youssef Y, Glankler EN, Barrett ER, Carter AM, Ahmed EH, Prasad A, Chen L, Zhang J, Benson DM, Caligiuri MA, Yu J. A CS1-NKG2D Bispecific Antibody Collectively Activates Cytolytic Immune Cells against Multiple Myeloma. Cancer Immunol Res 2018; 6:776-787. [PMID: 29769244 DOI: 10.1158/2326-6066.cir-17-0649] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/28/2018] [Accepted: 05/11/2018] [Indexed: 12/20/2022]
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy of plasma cells, with an estimated 30,000 new cases diagnosed each year in the United States, signifying the need for new therapeutic approaches. We hypothesized that targeting MM using a bispecific antibody (biAb) to simultaneously engage both innate and adaptive cytolytic immune cells could present potent antitumor activity. We engineered a biAb by fusing an anti-CS1 single-chain variable fragment (scFv) and an anti-NKG2D scFv (CS1-NKG2D biAb). Although NKG2D is a potent activation receptor ubiquitously expressed on mostly cytolytic immune cells including NK cells, CD8+ T cells, γδ T cells, and NKT cells, the CS1 tumor-associated antigen on MM represents a promising target. CS1-NKG2D biAb engaged human MM cell lines and NKG2D+ immune cells, forming immune synapses. In effector cells, CS1-NKG2D biAb triggered the phosphorylation of AKT, a downstream protein kinase of the activated NKG2D-DAP10 complex. The EC50 values of CS1-NKG2D biAb for CS1high and for CS1low MM cell lines with effector PBMCs were 10-12 and 10-9 mol/L, respectively. CS1-NKG2D biAb acted through multiple types of immune cells, and this induced cytotoxicity was both CS1- and NKG2D-specific. In vivo, survival was significantly prolonged using CS1-NKG2D biAb in a xenograft NOD-SCIDIL2γc-/- (NSG) mouse model engrafted with both human PBMCs and MM cell lines. Collectively, we demonstrated that the CS1-NKG2D biAb facilitated an enhanced immune synapse between CS1+ MM cells and NKG2D+ cytolytic innate and antigen-specific effector cells, which, in turn, activated these immune cells for improved clearance of MM. Cancer Immunol Res; 6(7); 776-87. ©2018 AACR.
Collapse
Affiliation(s)
- Wing Keung Chan
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Siwen Kang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Youssef Youssef
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Erin N Glankler
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Emma R Barrett
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Alex M Carter
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Elshafa H Ahmed
- Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Aman Prasad
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Luxi Chen
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Don M Benson
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio. .,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio. .,Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| |
Collapse
|
117
|
Silva-Santos B, Strid J. Working in "NK Mode": Natural Killer Group 2 Member D and Natural Cytotoxicity Receptors in Stress-Surveillance by γδ T Cells. Front Immunol 2018; 9:851. [PMID: 29740448 PMCID: PMC5928212 DOI: 10.3389/fimmu.2018.00851] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/06/2018] [Indexed: 01/26/2023] Open
Abstract
Natural killer cell receptors (NKRs) are germline-encoded transmembrane proteins that regulate the activation and homeostasis of NK cells as well as other lymphocytes. For γδ T cells, NKRs play critical roles in discriminating stressed (transformed or infected) cells from their healthy counterparts, as proposed in the “lymphoid stress-surveillance” theory. Whereas the main physiologic role is seemingly fulfilled by natural killer group 2 member D, constitutively expressed by γδ T cells, enhancement of their therapeutic potential may rely on natural cytotoxicity receptors (NCRs), like NKp30 or NKp44, that can be induced selectively on human Vδ1+ T cells. Here, we review the contributions of NCRs, NKG2D, and their multiple ligands, to γδ T cell biology in mouse and human.
Collapse
Affiliation(s)
- Bruno Silva-Santos
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Jessica Strid
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
118
|
Stojanovic A, Correia MP, Cerwenka A. The NKG2D/NKG2DL Axis in the Crosstalk Between Lymphoid and Myeloid Cells in Health and Disease. Front Immunol 2018; 9:827. [PMID: 29740438 PMCID: PMC5924773 DOI: 10.3389/fimmu.2018.00827] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
Natural killer group 2, member D (NKG2D) receptor is a type II transmembrane protein expressed by both innate and adaptive immune cells, including natural killer (NK) cells, CD8+ T cells, invariant NKT cells, γδ T cells, and some CD4+ T cells under certain pathological conditions. NKG2D is an activating NK receptor that induces cytotoxicity and production of cytokines by effector cells and supports their proliferation and survival upon engagement with its ligands. In both innate and T cell populations, NKG2D can costimulate responses induced by other receptors, such as TCR in T cells or NKp46 in NK cells. NKG2D ligands (NKG2DLs) are remarkably diverse. Initially, NKG2DL expression was typically attributed to stressed, infected, or transformed cells, thus signaling “dysregulated-self.” However, many reports indicated their expression under homeostatic conditions, usually in the context of cell activation and/or proliferation. Myeloid cells, including macrophages and dendritic cells (DCs), are among the first cells sensing and responding to pathogens and tissue damage. By secreting a plethora of soluble mediators, by presenting antigens to T cells and by expressing costimulatory molecules, myeloid cells play vital roles in inducing and supporting responses of other immune cells in lymphoid organs and tissues. When activated, both macrophages and DCs upregulate NKG2DLs, thereby enabling them with additional mechanisms for regulating lymphocyte responses. In this review, we will focus on the expression of NKG2D by innate and adaptive lymphocytes, the regulation of NKG2DL expression on myeloid cells, and the contribution of the NKG2D/NKG2DL axis to the crosstalk of myeloid cells with NKG2D-expressing lymphocytes. In addition, we will highlight pathophysiological conditions associated with NKG2D/NKG2DL dysregulation and discuss the putative involvement of the NKG2D/NKG2DL axis in the lymphocyte/myeloid cell crosstalk in these diseases.
Collapse
Affiliation(s)
- Ana Stojanovic
- Innate Immunity (D080), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Immunobiochemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Margareta P Correia
- Innate Immunity (D080), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Immunobiochemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adelheid Cerwenka
- Innate Immunity (D080), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Immunobiochemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
119
|
Abumaree MH, Bahattab E, Alsadoun A, Al Dosaimani A, Abomaray FM, Khatlani T, Kalionis B, El-Muzaini MF, Alawad AO, AlAskar AS. Characterization of the interaction between human decidua parietalis mesenchymal stem/stromal cells and natural killer cells. Stem Cell Res Ther 2018; 9:102. [PMID: 29650045 PMCID: PMC5898063 DOI: 10.1186/s13287-018-0844-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Background Human decidua parietalis mesenchymal stem/multipotent stromal cells (DPMSCs) have unique phenotypic and functional properties that make them promising candidates for cell-based therapy. Here, we investigated DPMSC interaction with natural killer (NK) cells, and the effects of this interaction on NK cell phenotypic characteristics and functional activities. Methods DPMSCs isolated from the decidua parietalis of human fetal membranes were cultured with interleukin (IL)-2-activated and IL-2-unactivated NK cells isolated from healthy human peripheral blood. NK cell proliferation and cytolytic activities were then examined using functional assays. NK cell expression of receptors mediating the cytolytic activity against DPMSCs, and the mechanism underlying this effect on DPMSCs, were also examined using flow cytometry and light microscopy, respectively. Results DPMSCs stimulated IL-2-induced proliferation of resting NK cells and the proliferation of activated NK cells. Moreover, IL-2-activated NK cells, but not freshly isolated NK cells, efficiently lysed DPMSCs. The induction of this NK cell cytolytic activity against DPMSCs was mediated by the activating NK cell receptors NKG2D, CD69, NKp30, and NKp44. However, DPMSCs showed a direct induction of NK cell cytolytic activity through CD69. We also found that DPMSCs expressed the ligands for these activating NK cell receptors including Nectin-2, ULBP-2, MICA, and MICB. Although DPMSCs expressed HLA class I molecules, they were susceptible to lysis by NK cells, suggesting that HLA class I antigens do not play a significant role in NK cell cytolytic action. In addition, DPMSCs did not inhibit NK cell cytolytic activity against cancer cells. Importantly, DPMSCs significantly increased NK expression of inflammatory molecules with anticancer activities. Conclusions We conclude that DPMSCs have potential for therapeutic application in cancer therapy, but not in transplantation or immunological diseases.
Collapse
Affiliation(s)
- M H Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia. .,College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia.
| | - E Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - A Alsadoun
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - A Al Dosaimani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia
| | - F M Abomaray
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, 14186, Stockholm, Sweden.,Center for Hematology and Regenerative Medicine, Karolinska Institutet, 14186, Stockholm, Sweden
| | - T Khatlani
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia
| | - B Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, VIC, 3052, Australia
| | - M F El-Muzaini
- Department of Obstetrics and Gynaecology, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia
| | - A O Alawad
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh, 11442, Saudi Arabia
| | - A S AlAskar
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia.,College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 3660, Mail Code 3124, Riyadh, 11481, Saudi Arabia.,Adult Hematology and Stem Cell Transplantation, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Mail Code 1515, Riyadh, 11426, Saudi Arabia
| |
Collapse
|
120
|
Lin D, Hiron TK, O'Callaghan CA. Intragenic transcriptional interference regulates the human immune ligand MICA. EMBO J 2018; 37:embj.201797138. [PMID: 29643123 DOI: 10.15252/embj.201797138] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/27/2022] Open
Abstract
Many human genes have tandem promoters driving overlapping transcription, but the value of this distributed promoter configuration is generally unclear. Here we show that MICA, a gene encoding a ligand for the activating immune receptor NKG2D, contains a conserved upstream promoter that expresses a noncoding transcript. Transcription from the upstream promoter represses the downstream standard promoter activity in cis through transcriptional interference. The effect of transcriptional interference depends on the strength of transcription from the upstream promoter and can be described quantitatively by a simple reciprocal repressor function. Transcriptional interference coincides with recruitment at the standard downstream promoter of the FACT histone chaperone complex, which is involved in nucleosomal remodelling during transcription. The mechanism is invoked in the regulation of MICA expression by the physiological inputs interferon-γ and interleukin-4 that act on the upstream promoter. Genome-wide analysis indicates that transcriptional interference between tandem intragenic promoters may constitute a general mechanism with widespread importance in human transcriptional regulation.
Collapse
Affiliation(s)
- Da Lin
- Nuffield Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Thomas K Hiron
- Nuffield Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christopher A O'Callaghan
- Nuffield Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
121
|
Abstract
Natural killer (NK) cells express an array of germ-line encoded receptors that are capable of triggering cytotoxicity. NK cells tend to express many members of a given family of signalling molecules. The presence of many activating receptors and many members of a given family of signalling molecules can enable NK cells to detect different kinds of target cells, and to mount different kinds of responses. This contributes also to the robustness of NK cells responses; cytotoxic functions of NK cells often remain unaffected in the absence of selected signalling molecules. NK cells express many MHC-I-specific inhibitory receptors. Signals from MHC-I-specific inhibitory receptors tightly control NK cell cytotoxicity and, paradoxically, maintain NK cells in a state of proper responsiveness. This review provides a brief overview of the events that underlie NK cell activation, and how signals from inhibitory receptors intercept NK cell activation to prevent inappropriate triggering of cytotoxicity.
Collapse
Affiliation(s)
- Santosh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
| |
Collapse
|
122
|
Bálint Š, Lopes FB, Davis DM. A nanoscale reorganization of the IL-15 receptor is triggered by NKG2D in a ligand-dependent manner. Sci Signal 2018; 11:11/525/eaal3606. [PMID: 29636390 DOI: 10.1126/scisignal.aal3606] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Natural killer group 2D (NKG2D), an activating receptor on natural killer (NK) cells and a subset of T cells, recognizes stress-inducible proteins, including MICA and ULBP2, which are present on infected or transformed cells. Whether each NKG2D ligand (NKG2DL) has a distinct biological role is not clear. Using superresolution microscopy, we found that NKG2D is constitutively arranged in nanoclusters at the surface of human primary NK cells. Nanoclusters of NKG2D became smaller upon ligation with MICA but became larger upon activation by ULBP2. In addition, ULBP2 induced the reorganization of nanoclusters of the cytokine receptor subunit for both interleukin-2 (IL-2) and IL-15 (IL-2/IL-15Rβ), such that these cytokine receptor subunits coalesced with nanoclusters of NKG2D. Functionally, the response of NK cells activated by ULBP2 was augmented by an interaction between ULBP2-bound NKG2D and IL-15R ligated by IL-15 (trans-presented by IL-15Rα-coated surfaces). These data suggest that NKG2DLs are not equivalent in their capacity to activate NKG2D and establish a previously unknown paradigm in how ligand-induced changes to the nanoscale organization of the cell surface can affect immune responses.
Collapse
Affiliation(s)
- Štefan Bálint
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Filipa B Lopes
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, The University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK.
| |
Collapse
|
123
|
Dhar P, Wu JD. NKG2D and its ligands in cancer. Curr Opin Immunol 2018; 51:55-61. [PMID: 29525346 PMCID: PMC6145810 DOI: 10.1016/j.coi.2018.02.004] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/02/2018] [Accepted: 02/23/2018] [Indexed: 01/12/2023]
Abstract
NKG2D is an activating immune receptor expressed by NK and effector T cells. Induced expression of NKG2D ligand on tumor cell surface during oncogenic insults renders cancer cells susceptible to immune destruction. In advanced human cancers, tumor cells shed NKG2D ligand to produce an immune soluble form as a means of immune evasion. Soluble NKG2D ligands have been associated with poor clinical prognosis in cancer patients. Harnessing NKG2D pathway is considered a viable avenue in cancer immunotherapy over recent years. In this review, we will discuss the progress and perspectives.
Collapse
Affiliation(s)
- Payal Dhar
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago IL60611, United States; Driskill Graduate Program in Life Sciences, Feinberg School of Medicine, Chicago, Northwestern University, Chicago IL60611, United States
| | - Jennifer D Wu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago IL60611, United States; Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago IL60611, United States; Robert Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL60611, United States.
| |
Collapse
|
124
|
Lin C, Zhang J. Reformation in chimeric antigen receptor based cancer immunotherapy: Redirecting natural killer cell. Biochim Biophys Acta Rev Cancer 2018; 1869:200-215. [DOI: 10.1016/j.bbcan.2018.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/20/2018] [Indexed: 01/05/2023]
|
125
|
Affiliation(s)
- Adelheid Cerwenka
- Centre for Biomedicine and Medical Technology (CBTM) and European Center for Angioscience (ECAS), Heidelberg University, Medical Faculty Mannheim, Ludolf-Krehl-Strasse 13–17, D-68167 Mannheim, Germany
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
126
|
Zöller T, Wittenbrink M, Hoffmeister M, Steinle A. Cutting an NKG2D Ligand Short: Cellular Processing of the Peculiar Human NKG2D Ligand ULBP4. Front Immunol 2018; 9:620. [PMID: 29651291 PMCID: PMC5884875 DOI: 10.3389/fimmu.2018.00620] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Stress-induced cell surface expression of MHC class I-related glycoproteins of the MIC and ULBP families allows for immune recognition of dangerous "self cells" by human cytotoxic lymphocytes via the NKG2D receptor. With two MIC molecules (MICA and MICB) and six ULBP molecules (ULBP1-6), there are a total of eight human NKG2D ligands (NKG2DL). Since the discovery of the NKG2D-NKG2DL system, the cause for both redundancy and diversity of NKG2DL has been a major and ongoing matter of debate. NKG2DL diversity has been attributed, among others, to the selective pressure by viral immunoevasins, to diverse regulation of expression, to differential tissue expression as well as to variations in receptor interactions. Here, we critically review the current state of knowledge on the poorly studied human NKG2DL ULBP4. Summarizing available facts and previous studies, we picture ULBP4 as a peculiar ULBP family member distinct from other ULBP family members by various aspects. In addition, we provide novel experimental evidence suggesting that cellular processing gives rise to mature ULBP4 glycoproteins different to previous reports. Finally, we report on the proteolytic release of soluble ULBP4 and discuss these results in the light of known mechanisms for generation of soluble NKG2DL.
Collapse
Affiliation(s)
- Tobias Zöller
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Mareike Wittenbrink
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Meike Hoffmeister
- Institute of Biochemistry II, Goethe University Frankfurt am Main, Frankfurt am Main, Germany.,Brandenburg Medical School (MHB) Theodor Fontane, Institute of Biochemistry, Neuruppin, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
127
|
Zingoni A, Molfetta R, Fionda C, Soriani A, Paolini R, Cippitelli M, Cerboni C, Santoni A. NKG2D and Its Ligands: "One for All, All for One". Front Immunol 2018; 9:476. [PMID: 29662484 PMCID: PMC5890157 DOI: 10.3389/fimmu.2018.00476] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/22/2018] [Indexed: 01/30/2023] Open
Abstract
The activating receptor NKG2D is peculiar in its capability to bind to numerous and highly diversified MHC class I-like self-molecules. These ligands are poorly expressed on normal cells but can be induced on damaged, transformed or infected cells, with the final NKG2D ligand expression resulting from multiple levels of regulation. Although redundant molecular mechanisms can converge in the regulation of all NKG2D ligands, different stimuli can induce specific cellular responses, leading to the expression of one or few ligands. A large body of evidence demonstrates that NK cell activation can be triggered by different NKG2D ligands, often expressed on the same cell, suggesting a functional redundancy of these molecules. However, since a number of evasion mechanisms can reduce membrane expression of these molecules both on virus-infected and tumor cells, the co-expression of different ligands and/or the presence of allelic forms of the same ligand guarantee NKG2D activation in various stressful conditions and cell contexts. Noteworthy, NKG2D ligands can differ in their ability to down-modulate NKG2D membrane expression in human NK cells supporting the idea that NKG2D transduces different signals upon binding various ligands. Moreover, whether proteolytically shed and exosome-associated soluble NKG2D ligands share with their membrane-bound counterparts the same ability to induce NKG2D-mediated signaling is still a matter of debate. Here, we will review recent studies on the NKG2D/NKG2D ligand biology to summarize and discuss the redundancy and/or diversity in ligand expression, regulation, and receptor specificity.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristina Cerboni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
128
|
Mace EM. Phosphoinositide-3-Kinase Signaling in Human Natural Killer Cells: New Insights from Primary Immunodeficiency. Front Immunol 2018; 9:445. [PMID: 29563913 PMCID: PMC5845875 DOI: 10.3389/fimmu.2018.00445] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Human natural killer (NK) cells play a critical role in the control of viral infections and malignancy. Their importance in human health and disease is illustrated by severe viral infections in patients with primary immunodeficiencies that affect NK cell function and/or development. The recent identification of patients with phosphoinositide-3-kinase (PI3K)-signaling pathway mutations that can cause primary immunodeficiency provides valuable insight into the role that PI3K signaling plays in human NK cell maturation and lytic function. There is a rich literature that demonstrates a requirement for PI3K in multiple key aspects of NK cell biology, including development/maturation, homing, priming, and function. Here, I briefly review these previous studies and place them in context with recent findings from the study of primary immunodeficiency patients, particularly those with hyperactivating mutations in PI3Kδ signaling.
Collapse
Affiliation(s)
- Emily M Mace
- Department of Pediatrics, Baylor College of Medicine, Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
129
|
Dukovska D, Fernández-Soto D, Valés-Gómez M, Reyburn HT. NKG2H-Expressing T Cells Negatively Regulate Immune Responses. Front Immunol 2018; 9:390. [PMID: 29545803 PMCID: PMC5837990 DOI: 10.3389/fimmu.2018.00390] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/12/2018] [Indexed: 01/03/2023] Open
Abstract
The biology and function of NKG2H receptor, unlike the better characterized members of the NKG2 family NKG2A, NKG2C, and NKG2D, remains largely unclear. Here, we show that NKG2H is able to associate with the signaling adapter molecules DAP12 and DAP10 suggesting that this receptor can signal for cell activation. Using a recently described NKG2H-specific monoclonal antibody (mAb), we have characterized the expression and function of lymphocytes that express this receptor. NKG2H is expressed at the cell surface of a small percentage of peripheral blood mononuclear cell (PBMC) and is found more frequently on T cells, rather than NK cells. Moreover, although NKG2H is likely to trigger activation, co-cross-linking of this receptor with an NKG2H-specific mAb led to decreased T cell activation and proliferation in polyclonal PBMC cultures stimulated by anti-CD3 mAbs. This negative regulatory activity was seen only after cross-linking with NKG2H, but not NKG2A- or NKG2C-specific monoclonal antibodies. The mechanism underlying this negative effect is as yet unclear, but did not depend on the release of soluble factors or recognition of MHC class I molecules. These observations raise the intriguing possibility that NKG2H may be a novel marker for T cells able to negatively regulate T cell responses.
Collapse
Affiliation(s)
- Daniela Dukovska
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| | - Daniel Fernández-Soto
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| | - Mar Valés-Gómez
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| | - Hugh T Reyburn
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| |
Collapse
|
130
|
Ke M, Wang H, Zhou Y, Li J, Liu Y, Zhang M, Dou J, Xi T, Shen B, Zhou C. SEP enhanced the antitumor activity of 5-fluorouracil by up-regulating NKG2D/MICA and reversed immune suppression via inhibiting ROS and caspase-3 in mice. Oncotarget 2018; 7:49509-49526. [PMID: 27385218 PMCID: PMC5226525 DOI: 10.18632/oncotarget.10375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 06/16/2016] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy and immunotherapy are the main remedies used in cancer treatment. Because immunotherapy can not only reduce the toxicity of chemotherapeutics but also enhance antitumor effects in vivo, combining these two therapies is a trend that continues to gain more attention in clinic. SEP, a polysaccharide isolated from Strongylocentrotus nudus egg, has been reported to display antitumor activity by stimulating immune cells, including NK and T cells, via TLR2 and TLR4. In the present study, the synergistic effect between SEP and 5-fluorouracil (5-FU), a traditional cytotoxic drug, in vitro and in vivo was investigated. The results obtained indicated that SEP alone stimulated NK-92 cytotoxicity and coordinated with 5-FU to augment the cytotoxicity of NK-92 cells against HepG-2 or A549 cells in vitro. SEP promoted NK-92 activity by stimulating NKG2D and its downstream DAP10/PI3K/Erk signaling pathway. Additionally, 5-FU could increase MICA expression on HepG-2 or A549 cells and prevent membrane MICA from shedding as soluble MICA, which were abrogated in the tumor cells transfected with ADAM 10 overexpression plasmid. Moreover, in H22- or Lewis lung cancer (LLC)-bearing mouse models, SEP reversed 5-FU-induced atrophy and apoptosis in both the spleen and bone marrow in vivo by suppressing ROS generation and caspase-3 activation. All of these results highlight the potential for the combination of SEP and 5-FU in cancer therapy in the future.
Collapse
Affiliation(s)
- Mengyun Ke
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.,Research Institute of Advanced Surgical Techniques and Engineering of Xi'an Jiaotong University, Regenerative Medicine and Surgery Engineering Research Center of Shaanxi Province, First Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, Xi'an, 710061, PR China
| | - Hui Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Yiran Zhou
- Department of General Surgery, Rui Jin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai JiaoTong University, Shanghai, 200025, PR China
| | - Jingwen Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Min Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Jie Dou
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Tao Xi
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Baiyong Shen
- Department of General Surgery, Rui Jin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai JiaoTong University, Shanghai, 200025, PR China
| | - Changlin Zhou
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| |
Collapse
|
131
|
García-Martínez E, Smith M, Buqué A, Aranda F, de la Peña FA, Ivars A, Cánovas MS, Conesa MAV, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulation with recombinant cytokines for cancer therapy. Oncoimmunology 2018; 7:e1433982. [PMID: 29872569 PMCID: PMC5980390 DOI: 10.1080/2162402x.2018.1433982] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 12/15/2022] Open
Abstract
Cytokines regulate virtually aspects of innate and adaptive immunity, including the initiation, execution and extinction of tumor-targeting immune responses. Over the past three decades, the possibility of using recombinant cytokines as a means to elicit or boost clinically relevant anticancer immune responses has attracted considerable attention. However, only three cytokines have been approved so far by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, namely, recombinant interleukin (IL)-2 and two variants of recombinant interferon alpha 2 (IFN-α2a and IFN-α2b). Moreover, the use of these cytokines in the clinics is steadily decreasing, mostly as a consequence of: (1) the elevated pleiotropism of IL-2, IFN-α2a and IFN-α2b, resulting in multiple unwarranted effects; and (2) the development of highly effective immunostimulatory therapeutics, such as immune checkpoint blockers. Despite this and other obstacles, research in the field continues as alternative cytokines with restricted effects on specific cell populations are being evaluated. Here, we summarize research preclinical and clinical developments on the use of recombinant cytokines for immunostimulation in cancer patients.
Collapse
Affiliation(s)
- Elena García-Martínez
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Melody Smith
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System, IDIBAPS, Barcelona, Spain
| | | | - Alejandra Ivars
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Manuel Sanchez Cánovas
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | | | - Jitka Fucikova
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Université Paris Descartes/Paris V, France
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
132
|
Jin F, Lin H, Gao S, Hu Z, Zuo S, Sun L, Jin C, Li W, Yang Y. The anti-tumor role of NK cells in vivo pre-activated and re-stimulated by interleukins in acute lymphoblastic leukemia. Oncotarget 2018; 7:79187-79202. [PMID: 27816971 PMCID: PMC5346707 DOI: 10.18632/oncotarget.13007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 10/26/2016] [Indexed: 01/20/2023] Open
Abstract
Although natural killer cells (NK cells) were traditionally classified as members of the innate immune system, NK cells have recently been found also to be an important player in the adaptive immune systems. In this context, in vitro activation of NK cells by cytokines leads to generation of NK cells with memory-like properties characterized by increased interferon-γ (IFNγ) production. However, it remains to be defined whether these memory-like NK cells exist in vivo after cytokine activation. Furthermore, it is also unclear whether such memory-like NK cells induced in vivo by cytokines could have effective anti-leukemia response. To address these issues, we used an in vivo pre-activation and re-stimulation system that was able to produce NK cells with increased IFNγ secretion. It was found that after in vivo pre-activation and re-stimulation with interleukins (ILs), NK cells retained a state to produce increased amount of IFNγ. Of note, whereas this intrinsic capacity of enhanced IFNγ production after in vivo IL pre-activation and re-stimulation could be transferred to the next generation of NK cells and was associated with prolonged survival of the mice with acute lymphoid leukemia. Moreover, the anti-leukemia activity of these memory-like NK cells was associated with IFNγ production and up-regulation of NK cells activation receptor-NK Group 2 member D (NKG2D). Together, these findings argue strongly that in vivo IL pre-activation and re-stimulation is capable to induce memory-like NK cells as observed previously in vitro, which are effective against acute lymphoblastic leukemia, likely via NKG2D-dependent IFNγ production, in intact animals.
Collapse
Affiliation(s)
- Fengyan Jin
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Hai Lin
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Sujun Gao
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Zheng Hu
- Institute of Translational Medicine, The First Bethune Hospital of Jilin University, Changchun, China
| | - Song Zuo
- Institute of Translational Medicine, The First Bethune Hospital of Jilin University, Changchun, China
| | - Liguang Sun
- Institute of Translational Medicine, The First Bethune Hospital of Jilin University, Changchun, China
| | - Chunhui Jin
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, China
| | - Yanping Yang
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, China
| |
Collapse
|
133
|
Trembath AP, Markiewicz MA. More than Decoration: Roles for Natural Killer Group 2 Member D Ligand Expression by Immune Cells. Front Immunol 2018; 9:231. [PMID: 29483917 PMCID: PMC5816059 DOI: 10.3389/fimmu.2018.00231] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/26/2018] [Indexed: 01/16/2023] Open
Abstract
The activating immune receptor natural killer group 2 member D (NKG2D), which is expressed by natural killer cells and T cell subsets, recognizes a number of ligands expressed by "stressed" or damaged cells. NKG2D has been extensively studied for its role in tumor immunosurveillance and antiviral immunity. To date, the majority of studies have focused on NKG2D-mediated killing of target cells expressing NKG2D ligands. However, with a number of reports describing expression of NKG2D ligands by cells that are not generally considered stressed, it is becoming clear that some healthy cells also express NKG2D ligands. Expression of these ligands by cells within the skin, intestinal epithelium, and the immune system suggests other immune functions for NKG2D ligand expression in addition to its canonical role as a "kill me" signal. How NKG2D ligands function in this capacity is just now starting to be unraveled. In this review, we examine the expression of NKG2D ligands by immune cells and discuss current literature describing the effects of this expression on immunity and immune regulation.
Collapse
Affiliation(s)
- Andrew P. Trembath
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, MO, United States
| | - Mary A. Markiewicz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, MO, United States
| |
Collapse
|
134
|
Parry HM, Stevens T, Oldreive C, Zadran B, McSkeane T, Rudzki Z, Paneesha S, Chadwick C, Stankovic T, Pratt G, Zuo J, Moss P. NK cell function is markedly impaired in patients with chronic lymphocytic leukaemia but is preserved in patients with small lymphocytic lymphoma. Oncotarget 2018; 7:68513-68526. [PMID: 27655680 PMCID: PMC5356570 DOI: 10.18632/oncotarget.12097] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/12/2016] [Indexed: 01/09/2023] Open
Abstract
Chronic lymphocytic leukemia (B-CLL) and small lymphocytic lymphoma (SLL) are part of the same disease classification but are defined by differential distribution of tumor cells. B-CLL is characterized by significant immune suppression and dysregulation but this is not typical of patients with SLL. Natural killer cells (NK) are important mediators of immune function but have been poorly studied in patients with B-CLL/SLL. Here we report for the first time the NK cell phenotype and function in patients with B-CLL and SLL alongside their transcriptional profile. We show for the first time impaired B-CLL NK cell function in a xenograft model with reduced activating receptor expression including NKG2D, DNAM-1 and NCRs in-vitro. Importantly, we show these functional differences are associated with transcriptional downregulation of cytotoxic pathway genes, including activating receptors, adhesion molecules, cytotoxic molecules and intracellular signalling molecules, which remain intact in patients with SLL. In conclusion, NK cell function is markedly influenced by the anatomical site of the tumor in patients with B-CLL/SLL and lymphocytosis leads to marked impairment of NK cell activity. These observations have implications for treatment protocols which seek to preserve immune function by limiting the exposure of NK cells to tumor cells within the peripheral circulation.
Collapse
Affiliation(s)
- Helen M Parry
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK
| | - Tom Stevens
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK
| | - Ceri Oldreive
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Science, University of Birmingham, B15 2TT, UK
| | - Bassier Zadran
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK
| | - Tina McSkeane
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Science, University of Birmingham, B15 2TT, UK
| | - Zbigniew Rudzki
- Department of Haematology, Birmingham Heartlands Hospital, Birmingham, B9 5SS, UK
| | - Shankara Paneesha
- Department of Haematology, Birmingham Heartlands Hospital, Birmingham, B9 5SS, UK
| | | | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Science, University of Birmingham, B15 2TT, UK
| | - Guy Pratt
- Department of Haematology, Birmingham Heartlands Hospital, Birmingham, B9 5SS, UK
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK
| |
Collapse
|
135
|
Prajapati K, Perez C, Rojas LBP, Burke B, Guevara-Patino JA. Functions of NKG2D in CD8 + T cells: an opportunity for immunotherapy. Cell Mol Immunol 2018; 15:470-479. [PMID: 29400704 DOI: 10.1038/cmi.2017.161] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/30/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022] Open
Abstract
Natural killer group 2 member D (NKG2D) is a type II transmembrane receptor. NKG2D is present on NK cells in both mice and humans, whereas it is constitutively expressed on CD8+ T cells in humans but only expressed upon T-cell activation in mice. NKG2D is a promiscuous receptor that recognizes stress-induced surface ligands. In NK cells, NKG2D signaling is sufficient to unleash the killing response; in CD8+ T cells, this requires concurrent activation of the T-cell receptor (TCR). In this case, the function of NKG2D is to authenticate the recognition of a stressed target and enhance TCR signaling. CD28 has been established as an archetype provider of costimulation during T-cell priming. It has become apparent, however, that signals from other costimulatory receptors, such as NKG2D, are required for optimal T-cell function outside the priming phase. This review will focus on the similarities and differences between NKG2D and CD28; less well-described characteristics of NKG2D, such as the potential role of NKG2D in CD8+ T-cell memory formation, cancer immunity and autoimmunity; and the opportunities for targeting NKG2D in immunotherapy.
Collapse
Affiliation(s)
- Kushal Prajapati
- Loyola University Chicago, Oncology Institute, 60153, Maywood, IL, USA
| | - Cynthia Perez
- Loyola University Chicago, Oncology Institute, 60153, Maywood, IL, USA
| | | | - Brianna Burke
- Loyola University Chicago, Oncology Institute, 60153, Maywood, IL, USA
| | | |
Collapse
|
136
|
Najar M, Fayyad-Kazan M, Meuleman N, Bron D, Fayyad-Kazan H, Lagneaux L. Immunological impact of Wharton's Jelly mesenchymal stromal cells and natural killer cell co-culture. Mol Cell Biochem 2018; 447:111-124. [PMID: 29380244 DOI: 10.1007/s11010-018-3297-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/23/2018] [Indexed: 12/20/2022]
Abstract
Due to their easier isolation, multilineage potential, and immunomodulatory capacity, Wharton's Jelly-derived mesenchymal stromal cells (WJ-MSCs) exhibit promising efficacy in the field of regenerative medicine and immunotherapy. Characterization of WJ-MSCs-natural killer (NK) cells crosstalk is required for ameliorating the medicinal value of WJ-MSCs. Here, we revealed that the outcome of WJ-MSCs-NK cells crosstalk varied according to the type of cytokines (IL-2, IL-12, IL-15 and IL-21) utilized to activate NK cells. Differently activated NK cells exerted distinct cytotoxicities against WJ-MSCs causing their probable death. Cell surface ligands (CD112, CD155, ULPB-3) and receptors (LAIR, CD226, CD314, CD335, CD336 and CD337) governing the interaction between NK cells and their targets, exhibited altered expression profiles following the co-culture with WJ-MSCs. Although partly inhibited NK cell proliferation, WJ-MSCs enhanced activated NK-cell-mediated secretion of IFN-γ and TNF-α. Moreover, WJ-MSCs reinforced NK cells' degranulation as well as secretion of perforin and granzymes. On the other hand, WJ-MSCs displayed only slight increase in ROS generation but significant decrease in A1 and C1 serpins expression following co-culture with activated NK cells. Altogether, our results highlight that WJ-MSCs-NK cells interaction may affect both cell type features and, therefore, their therapeutic properties.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Mohammad Fayyad-Kazan
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium.
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
137
|
Lee JC, Lee KM, Ahn YO, Suh B, Heo DS. A possible mechanism of impaired NK cytotoxicity in cancer patients: Down-regulation of DAP10 by TGF-β1. TUMORI JOURNAL 2018; 97:350-7. [DOI: 10.1177/030089161109700316] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Elevated TGF-β1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. However, the molecular mechanism of immunosuppression by TGF-β1 is not yet clarified. Methods IL-2-activated human NK cells were cultured with TGF-β1. Protein levels of NKG2D and DAP10 were examined by FACS or immunoblot analyses. Real-time RT-PCR was performed to quantify the transcription levels. MAPK inhibitors were used to investigate intracellular signaling. Results TGF-β1 down-regulated total and surface NKG2D, which was partially dependent on transcriptional regulation. TGF-β1 treatment of human NK cells resulted in significant changes in both transcriptional and translational levels of DAP10. Moreover, treatment with bafilomycin A1 or folimycin restored total NKG2D levels in TGF-β1-treated NK cells. The impaired NKG2D down-modulation by TGF-β1 was not associated with activation of the MAPK signaling pathway. Conclusions TGF-β1 down-modulates surface NKG2D expression by controlling the transcriptional and translational levels of DAP10.
Collapse
Affiliation(s)
- June-Chul Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Mi Lee
- Department of Biochemistry and Division of Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Yong-Oon Ahn
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Beomseok Suh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dae Seog Heo
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
138
|
Mehta RS, Randolph B, Daher M, Rezvani K. NK cell therapy for hematologic malignancies. Int J Hematol 2018; 107:262-270. [PMID: 29383623 DOI: 10.1007/s12185-018-2407-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022]
Abstract
Natural killer (NK) cells are part of the innate immune system and represent the first line of defense against infections and tumors. In contrast to T cells, NK cells do not require prior antigen sensitization to induce cytotoxicity and do not cause graft-versus-host disease. These, along with other advantages, make NK cells an attractive candidate for adoptive cellular therapy. Herein, we describe the mechanisms of NK cell cytotoxicity, which is governed by an intricate balance between various activating and inhibitory receptors, including the killer cell immunoglobulin-like receptors (KIRs). We illustrate the advantages of NK alloreactivity as demonstrated in various types of hematopoietic stem cell transplants (HSCT), such as haploidentical, human leukocyte antigen-matched related or unrelated donor and umbilical cord blood transplant. We elaborate on different models used to predict NK cell alloreactivity in these studies, which are either based on the absence of the ligands for inhibitory KIRs, presence of activating NK cell receptors and KIR genes content in donors, or a combination of these. We will review clinical studies demonstrating anti-tumor efficacy of NK cells used either as a stand-alone immunotherapy or as an adjunct to HSCT and novel genetic engineering strategies to improve the anti-tumor activity of NK cells.
Collapse
Affiliation(s)
- Rohtesh S Mehta
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Brion Randolph
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - May Daher
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplant and Cellular Therapy, University of Texas M. D. Anderson Cancer Center, Unit 0423, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| |
Collapse
|
139
|
Iwaszko M, Świerkot J, Kolossa K, Jeka S, Wiland P, Bogunia-Kubik K. Influence of NKG2D Genetic Variants on Response to Anti-TNF Agents in Patients with Rheumatoid Arthritis. Genes (Basel) 2018; 9:genes9020064. [PMID: 29370129 PMCID: PMC5852560 DOI: 10.3390/genes9020064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
A natural killer group 2 member D (NKG2D) acts as a powerful activating and co-stimulatory receptor on immune effector cells including NK and T cells. Disruptions within the NKG2D signalling pathway may trigger an exacerbated immune response and promote autoimmune reactions. The objective of the study was to evaluate a plausible role of polymorphisms within the NKG2D gene as a predictor of how effective anti-tumor necrosis factor (TNF) therapy is in rheumatoid arthritis (RA) patients. A total of 280 RA patients receiving anti-TNF therapy were genotyped for NKG2D rs2255336 (A > G), rs1049174 (C > G), and rs1154831 (C > A). Clinical response was evaluated according to the European League against Rheumatism (EULAR) criteria at the 12th and 24th week. Both the NKG2D rs225336 and rs1049174 polymorphisms were significantly associated with efficacy of TNF inhibitors. Inefficient therapy was more frequently observed in patients with rs2255336 GG or rs1049174 CC genotype as compared to other genotypes (p-value = 0.003 and p-value = 0.004, respectively). The presence of the rs2255336 G or the rs1049174 C allele correlated with a worse EULAR response (p-value = 0.002, p-value = 0.031, respectively). Moreover, patients carrying the rs2255336 or rs1049174 heterozygous genotype achieved better EULAR responses than patients with homozygous genotypes (p-value = 0.010 and p-value = 0.002, respectively). Data from the present study provides evidence that NKG2D polymorphisms may affect response to anti-TNF inhibitors in RA patients.
Collapse
Affiliation(s)
- Milena Iwaszko
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland.
| | - Jerzy Świerkot
- Department of Rheumatology and Internal Medicine, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland.
| | - Katarzyna Kolossa
- Clinical Department of Rheumatology and Connective Tissue Diseases, Hospital University Number 2 Jana Biziela, Ujejskiego 75, 85-168 Bydgoszcz, Poland.
| | - Sławomir Jeka
- Clinical Department of Rheumatology and Connective Tissue Diseases, Hospital University Number 2 Jana Biziela, Ujejskiego 75, 85-168 Bydgoszcz, Poland.
| | - Piotr Wiland
- Department of Rheumatology and Internal Medicine, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland.
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland.
- Department of Internal, Occupational Diseases, Hypertension and Clinical Oncology, Wrocław Medical University, Borowska 213, 50-556 Wrocław, Poland.
| |
Collapse
|
140
|
Najar M, Fayyad-Kazan M, Meuleman N, Bron D, Fayyad-Kazan H, Lagneaux L. Immunomodulatory effects of foreskin mesenchymal stromal cells on natural killer cells. J Cell Physiol 2018; 233:5243-5254. [PMID: 29194614 DOI: 10.1002/jcp.26305] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
Abstract
Foreskin-mesenchymal stromal cells (FSK-MSCs) are immune-privileged thus making them valuable immunotherapeutic cell product. Characterization of the relationship between FSK-MSCs and natural killer (NK) cells is essential to improve cell-based therapy. In the present study, we studied for the first time FSK-MSCs-NK interaction and showed that the result of such cross talk was robustly dependent on the type of cytokines (IL-2, IL-12, IL-15, and IL-21) employed to activate NK cells. Distinctly activated-NK cells showed uneven cytotoxicity against FSK-MSCs, triggering their death in fine. The expression of different cell-surface ligands (CD112, CD155, ULPB-3) and receptors (LAIR, KIRs) ensuring such interaction was altered following co-culture of both populations. Despite their partial negative effect on NK cell proliferation, FSK-MSCs boosted the capacity of activated NK-cells to secrete IFN-γ and TNF-α. Moreover, FSK-MSCs enhanced degranulation of NK cells, reinforced secretion of perforin and granzymes, while only modestly increased ROS production. On the other hand, FSK-MSCs-mediated expression of C1 and B9 serpins was significantly lowered in the presence of activated NK cells. Altogether, our results highlight major immunological changes following FSK-MSCs-NK interaction. Understanding these outcomes will therefore enhance the value of the therapeutic strategy.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Mohammad Fayyad-Kazan
- Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, Gosselies, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, Bruxelles, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, Bruxelles, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
141
|
Najar M, Fayyad-Kazan M, Meuleman N, Bron D, Fayyad-Kazan H, Lagneaux L. Mesenchymal stromal cells of the bone marrow and natural killer cells: cell interactions and cross modulation. J Cell Commun Signal 2018; 12:673-688. [PMID: 29350342 DOI: 10.1007/s12079-018-0448-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are multipotent progenitor cells that have shown promise for several different therapeutic applications. As they are able to modulate the function of several types of immune cells, BM-MSCs are highly important in the field of cell-based immunotherapy. Understanding BM-MSC-natural killer (NK) cell interactions is crucial for improving their therapeutic efficiency. Here, we observed that the type of NK cell-activating cytokine (e.g., IL-2, IL-12, IL-15 and IL-21) strongly influenced the outcomes of their interactions with BM-MSCs. The expression patterns of the ligands (CD112, CD155, ULPB-3) and receptors (LAIR, NCR) mediating the cross-talk between BM-MSCs and NK cells were critically modulated following co-culture. BM-MSCs partially impaired NK cell proliferation but up-regulated their secretion of IFN-γ and TNF-α. As they are cytotoxic, activated NK cells induced the killing of BM-MSCs. Indeed, BM-MSCs triggered the degranulation of NK cells and increased their release of perforin and granzymes. Interestingly, activated NK cells induced ROS generation within BM-MSCs that caused their decreased viability and reduced expression of serpin B9. Collectively, our observations reveal that BM-MSC-NK cell interactions may impact the immunobiology of both cell types. The therapeutic potential of BM-MSCs will be significantly improved once these issues are well characterized.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Mohammad Fayyad-Kazan
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium.
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
142
|
Benson DM, Caligiuri MA. Natural Killer Cell Immunity. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00022-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
143
|
NARUSE TK, AKARI H, MATANO T, KIMURA A. Diversity of ULBP5 in Old-World monkeys (Cercopithecidae) and divergence of the ULBP gene family in primates. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2018; 94:441-453. [PMID: 30541969 PMCID: PMC6374140 DOI: 10.2183/pjab.94.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/10/2018] [Indexed: 06/09/2023]
Abstract
Non-human primates such as rhesus macaque and cynomolgus macaque are important animals for medical research. These species are classified as Old-World monkeys (Cercopithecidae), in which the immune-related genome structure is characterized by gene duplications. In the present study, we investigated polymorphisms in two genes for ULBP5 encoding ligands for NKG2D. We found 18 and 11 ULBP5.1 alleles and 11 and 13 ULBP5.2 alleles in rhesus macaques and cynomolgus macaques, respectively. In addition, phylogenetic analyses revealed that ULBP5.2 diverged from a branch of ULBP5.1. These data suggested that human ULBP genes diverged from an ancestral gene of ULBP2-ULBP5 and that ULBP6/RAET1L, specifically identified in human, diverged from an ancestral ULBP2 by a recent gene duplication after the diversification of homininae (human and other higher great apes), which were consistent with the findings in our previous analysis of ULBP2 genes in rhesus and cynomolgus macaques.
Collapse
Affiliation(s)
- Taeko K. NARUSE
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirofumi AKARI
- Primate Research Institute, Kyoto University, Inuyama, Japan
| | - Tetsuro MATANO
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akinori KIMURA
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
144
|
McCarthy MT, Moncayo G, Hiron TK, Jakobsen NA, Valli A, Soga T, Adam J, O'Callaghan CA. Purine nucleotide metabolism regulates expression of the human immune ligand MICA. J Biol Chem 2017; 293:3913-3924. [PMID: 29279329 DOI: 10.1074/jbc.m117.809459] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/12/2017] [Indexed: 12/31/2022] Open
Abstract
Expression of the cell-surface glycoprotein MHC class I polypeptide-related sequence A (MICA) is induced in dangerous, abnormal, or "stressed" cells, including cancer cells, virus-infected cells, and rapidly proliferating cells. MICA is recognized by the activating immune cell receptor natural killer group 2D (NKG2D), providing a mechanism by which immune cells can identify and potentially eliminate pathological cells. Immune recognition through NKG2D is implicated in cancer, atherosclerosis, transplant rejection, and inflammatory diseases, such as rheumatoid arthritis. Despite the wide range of potential therapeutic applications of MICA manipulation, the factors that control MICA expression are unclear. Here we use metabolic interventions and metabolomic analyses to show that the transition from quiescent cellular metabolism to a "Warburg" or biosynthetic metabolic state induces MICA expression. Specifically, we show that glucose transport into the cell and active glycolytic metabolism are necessary to up-regulate MICA expression. Active purine synthesis is necessary to support this effect of glucose, and increases in purine nucleotide levels are sufficient to induce MICA expression. Metabolic induction of MICA expression directly influences NKG2D-dependent cytotoxicity by immune cells. These findings support a model of MICA regulation whereby the purine metabolic activity of individual cells is reflected by cell-surface MICA expression and is the subject of surveillance by NKG2D receptor-expressing immune cells.
Collapse
Affiliation(s)
- Michael T McCarthy
- From the Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Gerard Moncayo
- From the Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Thomas K Hiron
- From the Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Niels A Jakobsen
- From the Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Alessandro Valli
- the Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom, and
| | - Tomoyoshi Soga
- the Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Julie Adam
- From the Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom.,the Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom, and
| | - Christopher A O'Callaghan
- From the Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom,
| |
Collapse
|
145
|
Pugh JL, Nemat-Gorgani N, Norman PJ, Guethlein LA, Parham P. Human NK Cells Downregulate Zap70 and Syk in Response to Prolonged Activation or DNA Damage. THE JOURNAL OF IMMUNOLOGY 2017; 200:1146-1158. [PMID: 29263215 DOI: 10.4049/jimmunol.1700542] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 11/15/2017] [Indexed: 01/28/2023]
Abstract
The extent of NK cell activity during the innate immune response affects downstream immune functions and, ultimately, the outcome of infectious or malignant disease. However, the mechanisms that terminate human NK cell responses have yet to be defined. When activation receptors expressed on NK cell surfaces bind to ligands on diseased cells, they initiate a signal that is propagated by a number of intracellular kinases, including Zap70 and Syk, eventually leading to NK cell activation. We assayed Zap70 and Syk content in NK cells from healthy human donors and identified a subset of NK cells with unusually low levels of these two kinases. We found that this Zap70lowSyklow subset consisted of NK cells expressing a range of surface markers, including CD56hi and CD56low NK cells. Upon in vitro stimulation with target cells, Zap70lowSyklow NK cells failed to produce IFN-γ and lysed target cells at one third the capacity of Zap70hiSykhi NK cells. We determined two independent in vitro conditions that induce the Zap70lowSyklow phenotype in NK cells: continuous stimulation with activation beads and DNA damage. The expression of inhibitory receptors, including NKG2A and inhibitory killer Ig-like receptors (KIRs), was negatively correlated with the Zap70lowSyklow phenotype. Moreover, expression of multiple KIRs reduced the likelihood of Zap70 downregulation during continuous activation, regardless of whether NK cells had been educated through KIR-HLA interactions in vivo. Our findings show that human NK cells are able to terminate their functional activity without the aid of other immune cells through the downregulation of activation kinases.
Collapse
Affiliation(s)
- Jason L Pugh
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Neda Nemat-Gorgani
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Paul J Norman
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Lisbeth A Guethlein
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Peter Parham
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and .,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
146
|
Tissue regeneration: The crosstalk between mesenchymal stem cells and immune response. Cell Immunol 2017; 326:86-93. [PMID: 29221689 DOI: 10.1016/j.cellimm.2017.11.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 11/18/2017] [Accepted: 11/18/2017] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) exist in almost all tissues with the capability to differentiate into several different cell types and hold great promise in tissue repairs in a cell replacement manner. The study of the bidirectional regulation between MSCs and immune response has ushered an age of rethinking of tissue regeneration in the process of stem cell-based tissue repairs. By sensing damaged signals, both endogenous and exogenous MSCs migrate to the damaged site where they involve in the reconstitution of the immune microenvironment and empower tissue stem/progenitor cells and other resident cells, whereby facilitate tissue repairs. This MSC-based therapeutic manner is conferred as cell empowerment. In this process, MSCs have been found to exert extensive immunosuppression on both innate and adaptive immune response, while such regulation needs to be licensed by inflammation. More importantly, the immunoregulation of MSCs is highly plastic, especially in the context of pathological microenvironment. Understanding the immunoregulatory properties of MSCs is necessary for appropriate application of MSCs. Here we review the current studies on the crosstalk of MSCs and immune response in disease pathogenesis and therapy.
Collapse
|
147
|
Trembath AP, Sharma N, Raju S, Polić B, Markiewicz MA. A Protective Role for NKG2D-H60a Interaction via Homotypic T Cell Contact in Nonobese Diabetic Autoimmune Diabetes Pathogenesis. Immunohorizons 2017; 1:198-212. [PMID: 29497709 PMCID: PMC5828234 DOI: 10.4049/immunohorizons.1700011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The NK group 2 member D (NKG2D) immune receptor is implicated in both human and mouse autoimmune diabetes. However, the significance of NKG2D in diabetes pathogenesis has been unclear due to conflicting reports as to the importance of this receptor in the NOD mouse model. In this study we demonstrate that NKG2D expression affects NOD diabetes development by at least two previously undescribed, and opposing, mechanisms. First, we demonstrate that the NKG2D ligand H60a is induced on activated NOD T cells, and that NKG2D-H60a interaction during CD8+ T cell differentiation into CTLs generally decreases the subsequent CTL effector cytokine response. This corresponds to an increase in diabetes development in NKG2D-deficient compared with wild-type NOD mice under microbiota-depleted conditions. Second, we demonstrate that NKG2D promotes NOD diabetes development through interaction with the microbiota. Together these findings reveal a previously undescribed role for NKG2D ligand expression by activated T cells in CTL development. Further, they demonstrate that NKG2D has both diabetogenic and antidiabetogenic roles in NOD diabetes development.
Collapse
Affiliation(s)
- Andrew P Trembath
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Neekun Sharma
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Saravanan Raju
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Bojan Polić
- Department of Histology and Embryology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
148
|
Koch C, Kim Y, Zöller T, Born C, Steinle A. Chronic NKG2D Engagement In Vivo Differentially Impacts NK Cell Responsiveness by Activating NK Receptors. Front Immunol 2017; 8:1466. [PMID: 29163533 PMCID: PMC5675847 DOI: 10.3389/fimmu.2017.01466] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/19/2017] [Indexed: 11/25/2022] Open
Abstract
Immunosuppression is a typical hallmark of cancer and frequently includes perturbations of the NKG2D tumor recognition system as well as impaired signaling by other activating NK cell receptors. Several in vitro studies suggested that sustained engagement of the NKG2D receptor, as it is occurring in the tumor microenvironment, not only impairs expression and function of NKG2D but also impacts signaling by other activating NK receptors. Here, we made use of a transgenic mouse model of ubiquitous NKG2D ligand expression (H2-Kb-MICA mice) to investigate consequences of chronic NKG2D engagement in vivo for functional responsiveness by other activating NK receptors such as NKp46 and Ly49D. Unexpectedly, we found no evidence for an impairment of NKp46 expression and function in H2-Kb-MICA mice, as anticipated from previous in vitro experiments. However, we observed a marked downregulation and dysfunction of the activating receptor Ly49D in activated NK cells from H2-Kb-MICA mice. Ly49D shares the adaptor proteins DAP10 and DAP12 with NKG2D possibly explaining the collateral impairment of Ly49D function in situations of chronic NKG2D engagement. Altogether, our results demonstrate that persistent engagement of NKG2D in vivo, as often observed in tumors, can selectively impair functions of unrelated NK receptors and thereby compromise NK responsiveness to third-party antigens.
Collapse
Affiliation(s)
- Christine Koch
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany.,Department of Internal Medicine I, Division of Gastroenterology and Hepatology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Younghoon Kim
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Tobias Zöller
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Christina Born
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
149
|
Raulet DH, Marcus A, Coscoy L. Dysregulated cellular functions and cell stress pathways provide critical cues for activating and targeting natural killer cells to transformed and infected cells. Immunol Rev 2017; 280:93-101. [PMID: 29027233 PMCID: PMC5687887 DOI: 10.1111/imr.12600] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Natural killer (NK) cells recognize and kill cancer cells and infected cells by engaging cell surface ligands that are induced preferentially or exclusively on these cells. These ligands are recognized by activating receptors on NK cells, such as NKG2D. In addition to activation by cell surface ligands, the acquisition of optimal effector activity by NK cells is driven in vivo by cytokines and other signals. This review addresses a developing theme in NK cell biology: that NK-activating ligands on cells, and the provision of cytokines and other signals that drive high effector function in NK cells, are driven by abnormalities that arise from transformation or the infected state. The pathways include genomic damage, which causes self DNA to be exposed in the cytosol of affected cells, where it activates the DNA sensor cGAS. The resulting signaling induces NKG2D ligands and also mobilizes NK cell activation. Other key pathways that regulate NKG2D ligands include PI-3 kinase activation, histone acetylation, and the integrated stress response. This review summarizes the roles of these pathways and their relevance in both viral infections and cancer.
Collapse
Affiliation(s)
- David H Raulet
- Department of Molecular and Cell Biology, Cancer Research Laboratory, Immunotherapy and Vaccine Research Initiative, University of California, Berkeley, Berkeley, CA, USA
| | - Assaf Marcus
- Department of Molecular and Cell Biology, Cancer Research Laboratory, Immunotherapy and Vaccine Research Initiative, University of California, Berkeley, Berkeley, CA, USA
| | - Laurent Coscoy
- Department of Molecular and Cell Biology, Cancer Research Laboratory, Immunotherapy and Vaccine Research Initiative, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
150
|
Anti-NKG2D mAb: A New Treatment for Crohn's Disease? Int J Mol Sci 2017; 18:ijms18091997. [PMID: 28926962 PMCID: PMC5618646 DOI: 10.3390/ijms18091997] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 01/09/2023] Open
Abstract
Crohn’s disease (CD) and ulcerative colitis (UC) are immunologically-mediated, debilitating conditions resulting from destructive inflammation of the gastrointestinal tract. The pathogenesis of IBD is incompletely understood, but is considered to be the result of an abnormal immune response with a wide range of cell types and proteins involved. Natural Killer Group 2D (NKG2D) is an activating receptor constitutively expressed on human Natural Killer (NK), γδ T, mucosal-associated invariant T (MAIT), CD56+ T, and CD8+ T cells. Activation of NKG2D triggers cellular proliferation, cytokine production, and target cell killing. Research into the NKG2D mechanism of action has primarily been focused on cancer and viral infections where cytotoxicity evasion is a concern. In human inflammatory bowel disease (IBD) this system is less characterized, but the ligands have been shown to be highly expressed during intestinal inflammation and the following receptor activation may contribute to tissue degeneration. A recent phase II clinical trial showed that an antibody against NKG2D induced clinical remission of CD in some patients, suggesting NKG2D and its ligands to be of importance in the pathogenesis of CD. This review will describe the receptor and its ligands in intestinal tissues and the clinical potential of blocking NKG2D in Crohn’s disease.
Collapse
|