301
|
Natural killer cells involved in tumour immune escape of hepatocellular carcinomar. Int Immunopharmacol 2019; 73:10-16. [PMID: 31078921 DOI: 10.1016/j.intimp.2019.04.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/21/2019] [Accepted: 04/25/2019] [Indexed: 02/08/2023]
Abstract
Natural killer cells are the first line of host immune surveillance and play major roles in the defence against infection and tumours. Hepatic NK cells exhibit unique phenotypic and functional characteristics compared to circulating and spleen NK cells, such as higher levels of cytolytic activity and cytotoxicity mediators against tumour cells. However, the activities of NK cells may be reversed during tumour progression. Recent studies demonstrated that hepatic NK cells were exhausted in hepatocellular carcinoma (HCC) and exhibited impaired cytolytic activity and decreased production of effector cytokines. The present review discusses current knowledge on the role of exhausted NK cells in promoting HCC development and the mechanisms contributing to tumour immune escape, including an imbalance of activating and inhibitory receptors on NK cells, abnormal receptor-ligand interaction, and cross-talk with immune cells and other stromal cells in the tumour environment. We provide a fundamental basis for further study of innate immunity in tumour progression and serve the purpose of exploring new HCC treatment strategies.
Collapse
|
302
|
Sivori S, Vacca P, Del Zotto G, Munari E, Mingari MC, Moretta L. Human NK cells: surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol 2019; 16:430-441. [PMID: 30778167 PMCID: PMC6474200 DOI: 10.1038/s41423-019-0206-4] [Citation(s) in RCA: 348] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 01/22/2019] [Indexed: 12/29/2022] Open
Abstract
NK cells play important roles in innate defenses against viruses and in the control of tumor growth and metastasis. The regulation/induction of NK cell function is mediated by an array of activating or inhibitory surface receptors. In humans, major activating receptors involved in target cell killing are the natural cytotoxicity receptors (NCRs) and NKG2D. Activating receptors recognize ligands that are overexpressed or expressed de novo upon cell stress, viral infection, or tumor transformation. The HLA-class I-specific inhibitory receptors, including KIRs recognizing HLA-class I allotypic determinants and CD94/NKG2A recognizing the class-Ib HLA-E, constitute a fail-safe mechanism to avoid unwanted NK-mediated damage to healthy cells. Other receptors such as PD-1, primarily expressed by activated T lymphocytes, are important inhibitory checkpoints of immune responses that ensure T-cell tolerance. PD-1 also may be expressed by NK cells in cancer patients. Since PD-1 ligand (PD-L1) may be expressed by different tumors, PD-1/PD-L1 interactions inactivate both T and NK cells. Thus, the reliable evaluation of PD-L1 expression in tumors has become a major issue to select patients who may benefit from therapy with mAbs disrupting PD-1/PD-L1 interactions. Recently, NKG2A was revealed to be an important checkpoint controlling both NK and T-cell activation. Since most tumors express HLA-E, mAbs targeting NKG2A has been used alone or in combination with other therapeutic mAbs targeting PD-1 or tumor antigens (e.g., EGFR), with encouraging results. The translational value of NK cells and their receptors is evidenced by the extraordinary therapeutic success of haploidentical HSCT to cure otherwise fatal high-risk leukemias.
Collapse
Affiliation(s)
- Simona Sivori
- Department of Experimental Medicine (DIMES) and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Paola Vacca
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Genny Del Zotto
- Department of Research and Diagnostics, Istituto G. Gaslini, Genoa, Italy
| | - Enrico Munari
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Department of Pathology, Sacro Cuore Don Calabria, Negrar, VR, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES) and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino Genova, Genoa, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.
| |
Collapse
|
303
|
Parodi M, Favoreel H, Candiano G, Gaggero S, Sivori S, Mingari MC, Moretta L, Vitale M, Cantoni C. NKp44-NKp44 Ligand Interactions in the Regulation of Natural Killer Cells and Other Innate Lymphoid Cells in Humans. Front Immunol 2019; 10:719. [PMID: 31024551 PMCID: PMC6465645 DOI: 10.3389/fimmu.2019.00719] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
Natural Killer (NK) cells are potent cytotoxic cells belonging to the family of Innate Lymphoid Cells (ILCs). Their most characterized effector functions are directed to the control of aberrant cells in the body, including both transformed and virus-infected cells. NK cell-mediated recognition of abnormal cells primarily occurs through receptor-ligand interactions, involving an array of inhibitory and activating NK receptors and different types of ligands expressed on target cells. While most of the receptors have become known over many years, their respective ligands were only defined later and their impressive complexity has only recently become evident. NKp44, a member of Natural Cytotoxicity Receptors (NCRs), is an activating receptor playing a crucial role in most functions exerted by activated NK cells and also by other NKp44+ immune cells. The large and heterogeneous panel of NKp44 ligands (NKp44L) now includes surface expressed glycoproteins and proteoglycans, nuclear proteins that can be exposed outside the cell, and molecules that can be either released in the extracellular space or carried in extracellular vesicles. Recent findings have extended our knowledge on the nature of NKp44L to soluble plasma glycoproteins, such as secreted growth factors or extracellular matrix (ECM)-derived glycoproteins. NKp44L are induced upon tumor transformation or viral infection but may also be expressed in normal cells and tissues. In addition, NKp44-NKp44L interactions are involved in the crosstalk between NK cells and different innate and adaptive immune cell types. NKp44 expression in different ILCs located in tissues further extends the potential role of NKp44-NKp44L interactions.
Collapse
Affiliation(s)
- Monica Parodi
- Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Herman Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Silvia Gaggero
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Simona Sivori
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Maria Cristina Mingari
- Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Massimo Vitale
- Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
304
|
Gimeno L, Martínez-Banaclocha H, Bernardo MV, Bolarin JM, Marín L, López-Hernández R, López-Alvarez MR, Moya-Quiles MR, Muro M, Frias-Iniesta JF, Martínez-Escribano J, Alvarez-López MR, Minguela A, Campillo JA. NKG2D Polymorphism in Melanoma Patients from Southeastern Spain. Cancers (Basel) 2019; 11:cancers11040438. [PMID: 30925758 PMCID: PMC6521179 DOI: 10.3390/cancers11040438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 11/17/2022] Open
Abstract
Background: Natural killer (NK) and CD8+ T cells are involved in the immune response against melanoma. C-Type lectin-like NK cell receptors are located in the Natural Killer Complex (NKC) region 12p13.2-p12.3 and play a critical role in regulating the activity of NK and CD8+ T cells. An association between polymorphisms in the NKC region, including the NKG2D gene and NKG2A promoter, and the risk of cancer has been previously described. The aim of this study was to analyze the association of polymorphisms in the NKC region with cutaneous melanoma in patients from southeastern Spain. Methods: Seven single-nucleotide polymorphisms (SNPs) in the NKG2D gene (NKC3,4,7,9,10,11,12), and one SNP in the NKG2A promoter (NKC17) were genotyped by a TaqMan 5′ Nuclease Assay in 233 melanoma patients and 200 matched healthy controls. Results: A linkage disequilibrium analysis of the SNPs performed in the NKC region revealed two blocks of haplotypes (Hb-1 and Hb-2) with 14 and seven different haplotype subtypes, respectively. The third most frequent haplotype from the block Hb-2—NK3 (CAT haplotype)—was significantly more frequent on melanoma patients than on healthy controls (p = 0.00009, Pc = 0.0006). No further associations were found when NKC SNPs were considered independently. Conclusions: Our results suggest an association between NKG2D polymorphisms and the risk of cutaneous malignant melanoma.
Collapse
Affiliation(s)
- Lourdes Gimeno
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | | | - M Victoria Bernardo
- Department of Pharmacy, Faculty of Health Sciences, Universidad Católica de San Antonio de Murcia (UCAM), 30107 Murcia, Spain.
| | - José Miguel Bolarin
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - Luis Marín
- Immunology Section, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain.
| | - Ruth López-Hernández
- Immunology Section, Hospital Universitario de Gran Canaria Doctor Negrín, 35010 Las Palmas de Gran Canaria, Spain.
| | - M Rocío López-Alvarez
- Center for Preventive Medicine, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk CB8 7UU, UK.
| | - M Rosa Moya-Quiles
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - Manuel Muro
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - José Francisco Frias-Iniesta
- Dermatology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - Jorge Martínez-Escribano
- Dermatology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - M Rocío Alvarez-López
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - Alfredo Minguela
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| | - José Antonio Campillo
- Immunology Service, Hospital Clínico Universitario Virgen de la Arrixaca (HCUVA), Instituto Murciano de Investigación Biomédica (IMIB), 30120 Murcia, Spain.
| |
Collapse
|
305
|
Wagner AK, Alici E, Lowdell MW. Characterization of human natural killer cells for therapeutic use. Cytotherapy 2019; 21:315-326. [PMID: 30910383 DOI: 10.1016/j.jcyt.2018.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/04/2018] [Indexed: 11/25/2022]
Abstract
As a part of the innate immune system, natural killer (NK) cells are cytotoxic lymphocytes that can exert cytotoxic activity against infected or transformed cells. Furthermore, due to their expression of a functional Fc receptor, they have also been eluded as a major effector fraction in antibody-dependent cellular cytotoxicity. These characteristics have led to multiple efforts to use them for adoptive immunotherapy against various malignancies. There are now at least 70 clinical trials testing the safety and efficacy of NK cell products around the world in early-phase clinical trials. NK cells are also being tested in the context of tumor retargeting via chimeric antigen receptors, other genetic modification strategies, as well as tumor-specific activation strategies such as bispecific engagers with or without cytokine stimulations. One advantage of the use of NK cells for adoptive immunotherapy is their potential to overcome HLA barriers. This has led to a plethora of sources, such as cord blood hematopoietic stem cells and induced pluripotent stem cells, which can generate comparatively high cytotoxic NK cells to peripheral blood counterparts. However, the variety of the sources has led to a heterogeneity in the characterization of the final infusion product. Therefore, in this review, we will discuss a comparative assessment strategy, from characterization of NK cells at collection to final product release by various phenotypic and functional assays, in an effort to predict potency of the cellular product.
Collapse
Affiliation(s)
- Arnika K Wagner
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mark W Lowdell
- Department of Haematology, Cancer Institute, University College London, London, UK.
| |
Collapse
|
306
|
Giuliani E, Desimio MG, Doria M. Hexamethylene bisacetamide impairs NK cell-mediated clearance of acute T lymphoblastic leukemia cells and HIV-1-infected T cells that exit viral latency. Sci Rep 2019; 9:4373. [PMID: 30867508 PMCID: PMC6416400 DOI: 10.1038/s41598-019-40760-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/19/2019] [Indexed: 12/27/2022] Open
Abstract
The hexamethylene bisacetamide (HMBA) anticancer drug was dismissed due to limited efficacy in leukemic patients but it may re-enter into the clinics in HIV-1 eradication strategies because of its recently disclosed capacity to reactivate latent virus. Here, we investigated the impact of HMBA on the cytotoxicity of natural killer (NK) cells against acute T lymphoblastic leukemia (T-ALL) cells or HIV-1-infected T cells that exit from latency. We show that in T-ALL cells HMBA upmodulated MICB and ULBP2 ligands for the NKG2D activating receptor. In a primary CD4+ T cell-based latency model, HMBA did not reactivate HIV-1, yet enhanced ULBP2 expression on cells harboring virus reactivated by prostratin (PRO). However, HMBA reduced the expression of NKG2D and its DAP10 adaptor in NK cells, hence impairing NKG2D-mediated cytotoxicity and DAP10-dependent response to IL-15 stimulation. Alongside, HMBA dampened killing of T-ALL targets by IL-15-activated NK cells and impaired NK cell-mediated clearance of PRO-reactivated HIV-1+ cells. Overall, our results demonstrate a dominant detrimental effect of HMBA on the NKG2D pathway that crucially controls NK cell-mediated killing of tumors and virus-infected cells, providing one possible explanation for poor clinical outcome in HMBA-treated cancer patients and raising concerns for future therapeutic application of this drug.
Collapse
Affiliation(s)
- Erica Giuliani
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Maria Giovanna Desimio
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Margherita Doria
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy.
| |
Collapse
|
307
|
Abstract
Natural killer (NK) cells have evolved to complement T and B cells in host defense against pathogens and cancer. They recognize infected cells and tumors using a sophisticated array of activating, costimulatory, and inhibitory receptors that are expressed on NK cell subsets to create extensive functional diversity. NK cells can be targeted to kill with exquisite antigen specificity by antibody-dependent cellular cytotoxicity. NK and T cells share many of the costimulatory and inhibitory receptors that are currently under evaluation in the clinic for cancer immunotherapy. As with T cells, genetic engineering is being employed to modify NK cells to specifically target them to tumors and to enhance their effector functions. As the selective pressures exerted by immunotherapies to augment CD8+T cell responses may result in loss of MHC class I, NK cells may provide an important fail-safe to eliminate these tumors by their capacity to eliminate tumors that are “missing self.”
Collapse
Affiliation(s)
- Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California 94143, USA
- The Parker Institute for Cancer Immunotherapy, San Francisco, California 94143, USA
| |
Collapse
|
308
|
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
|
309
|
Nayyar G, Chu Y, Cairo MS. Overcoming Resistance to Natural Killer Cell Based Immunotherapies for Solid Tumors. Front Oncol 2019; 9:51. [PMID: 30805309 PMCID: PMC6378304 DOI: 10.3389/fonc.2019.00051] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/18/2019] [Indexed: 12/22/2022] Open
Abstract
Despite advances in the diagnostic and therapeutic modalities, the prognosis of several solid tumor malignancies remains poor. Different factors associated with solid tumors including a varied genetic signature, complex molecular signaling pathways, defective cross talk between the tumor cells and immune cells, hypoxic and immunosuppressive effects of tumor microenvironment result in a treatment resistant and metastatic phenotype. Over the past several years, immunotherapy has emerged as an attractive therapeutic option against multiple malignancies. The unique ability of natural killer (NK) cells to target cancer cells without antigen specificity makes them an ideal candidate for use against solid tumors. However, the outcomes of adoptive NK cell infusions into patients with solid tumors have been disappointing. Extensive studies have been done to investigate different strategies to improve the NK cell function, trafficking and tumor targeting. Use of cytokines and cytokine analogs has been well described and utilized to enhance the proliferation, stimulation and persistence of NK cells. Other techniques like blocking the human leukocyte antigen-killer cell receptors (KIR) interactions with anti-KIR monoclonal antibodies, preventing CD16 receptor shedding, increasing the expression of activating NK cell receptors like NKG2D, and use of immunocytokines and immune checkpoint inhibitors can enhance NK cell mediated cytotoxicity. Using genetically modified NK cells with chimeric antigen receptors and bispecific and trispecific NK cell engagers, NK cells can be effectively redirected to the tumor cells improving their cytotoxic potential. In this review, we have described these strategies and highlighted the need to further optimize these strategies to improve the clinical outcome of NK cell based immunotherapy against solid tumors.
Collapse
Affiliation(s)
- Gaurav Nayyar
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, United States.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, United States.,Department of Medicine, New York Medical College, Valhalla, NY, United States.,Department of Pathology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
310
|
Legroux L, Moratalla AC, Laurent C, Deblois G, Verstraeten SL, Arbour N. NKG2D and Its Ligand MULT1 Contribute to Disease Progression in a Mouse Model of Multiple Sclerosis. Front Immunol 2019; 10:154. [PMID: 30787931 PMCID: PMC6372829 DOI: 10.3389/fimmu.2019.00154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
NKG2D is an activating receptor expressed on the surface of immune cells including subsets of T lymphocytes. NKG2D binds multiple ligands (NKG2DL) whose expression are differentially triggered in a cell type and stress specific manner. The NKG2D-NKG2DL interaction has been involved in autoimmune disorders but its role in animal models of multiple sclerosis (MS) remains incompletely resolved. Here we show that NKG2D and its ligand MULT1 contribute to the pathobiology of experimental autoimmune encephalomyelitis (EAE). MULT1 protein levels are increased in the central nervous system (CNS) at EAE disease peak; soluble MULT1 is elevated in the cerebrospinal fluid of both active and passive EAE. We establish that such soluble MULT1 enhances effector functions (e.g., IFNγ production) of activated CD8 T lymphocytes from wild type but not from NKG2D-deficient (Klrk1−/−) mice in vitro. The adoptive transfer of activated T lymphocytes from wild type donors induced a significantly reduced EAE disease in Klrk1−/− compared to wild type (Klrk1+/+) recipients. Characterization of T lymphocytes infiltrating the CNS of recipient mice shows that donor (CD45.1) rather than endogenous (CD45.2) CD4 T cells are the main producers of key cytokines (IFNγ, GM-CSF). In contrast, infiltrating CD8 T lymphocytes include mainly endogenous (CD45.2) cells exhibiting effector properties (NKG2D, granzyme B and IFNγ). Our data support the notion that endogenous CD8 T cells contribute to passive EAE pathobiology in a NKG2D-dependent manner. Collectively, our results point to the deleterious role of NKG2D and its MULT1 in the pathobiology of a MS mouse model.
Collapse
Affiliation(s)
- Laurine Legroux
- Department of Neurosciences Université de Montréal, Montreal, QC, Canada
| | | | - Cyril Laurent
- Department of Neurosciences Université de Montréal, Montreal, QC, Canada
| | - Gabrielle Deblois
- Department of Neurosciences Université de Montréal, Montreal, QC, Canada
| | | | - Nathalie Arbour
- Department of Neurosciences Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
311
|
Aiello A, Accardi G, Candore G, Caruso C, Colomba C, Di Bona D, Duro G, Gambino CM, Ligotti ME, Pandey JP. Role of Immunogenetics in the Outcome of HCMV Infection: Implications for Ageing. Int J Mol Sci 2019; 20:ijms20030685. [PMID: 30764515 PMCID: PMC6386818 DOI: 10.3390/ijms20030685] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/30/2022] Open
Abstract
The outcome of host-virus interactions is determined by a number of factors, some related to the virus, others to the host, such as environmental factors and genetic factors. Therefore, different individuals vary in their relative susceptibility to infections. Human cytomegalovirus (HCMV) is an important pathogen from a clinical point of view, as it causes significant morbidity and mortality in immunosuppressed or immunosenescent individuals, such as the transplanted patients and the elderly, respectively. It is, therefore, important to understand the mechanisms of virus infection control. In this review, we discuss recent advances in the immunobiology of HCMV-host interactions, with particular emphasis on the immunogenetic aspects (human leukocyte antigens, HLA; killer cell immunoglobulin-like receptors, KIRs; immunoglobulin genetic markers, GM allotypes) to elucidate the mechanisms underlying the complex host-virus interaction that determine various outcomes of HCMV infection. The results, which show the role of humoral and cellular immunity in the control of infection by HCMV, would be valuable in directing efforts to reduce HCMV spurred health complications in the transplanted patients and in the elderly, including immunosenescence. In addition, concerning GM allotypes, it is intriguing that, in a Southern Italian population, alleles associated with the risk of developing HCMV symptomatic infection are negatively associated with longevity.
Collapse
Affiliation(s)
- Anna Aiello
- Sezione di Patologia Generale, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Giulia Accardi
- Sezione di Patologia Generale, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Giuseppina Candore
- Sezione di Patologia Generale, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Calogero Caruso
- Sezione di Patologia Generale, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Claudia Colomba
- Dipartimento di Scienze per la Promozione della Salute e Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", Università di Palermo, Via del Vespro 129, 90127 Palermo, Italy.
| | - Danilo Di Bona
- Dipartimento dell'Emergenza e dei Trapianti d'Organo, Università di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy.
| | - Giovanni Duro
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy.
| | - Caterina Maria Gambino
- Sezione di Patologia Generale, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Mattia Emanuela Ligotti
- Sezione di Patologia Generale, Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC 29425, USA.
| |
Collapse
|
312
|
Cancer Exosomes as Conveyors of Stress-Induced Molecules: New Players in the Modulation of NK Cell Response. Int J Mol Sci 2019; 20:ijms20030611. [PMID: 30708970 PMCID: PMC6387166 DOI: 10.3390/ijms20030611] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells that play a pivotal role in tumor surveillance. Exosomes are nanovesicles released into the extracellular environment via the endosomal vesicle pathway and represent an important mode of intercellular communication. The ability of anticancer chemotherapy to enhance the immunogenic potential of malignant cells mainly relies on the establishment of the immunogenic cell death (ICD) and the release of damage-associated molecular patterns (DAMPs). Moreover, the activation of the DNA damage response (DDR) and the induction of senescence represent two crucial modalities aimed at promoting the clearance of drug-treated tumor cells by NK cells. Emerging evidence has shown that stress stimuli provoke an increased release of exosome secretion. Remarkably, tumor-derived exosomes (Tex) produced in response to stress carry distinct type of DAMPs that activate innate immune cell populations. Moreover, stress-induced ligands for the activating receptor NKG2D are transported by this class of nanovesicles. Here, we will discuss how Tex interact with NK cells and provide insight into their potential role in response to chemotherapy-induced stress stimuli. The capability of some "danger signals" carried by exosomes that indirectly affect the NK cell activity in the tumor microenvironment will be also addressed.
Collapse
|
313
|
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
|
314
|
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
|
315
|
Ebrahim AA, Salem RM, El Fallah AA, Younis ET. Serum Interleukin-15 is a Marker of Alopecia Areata Severity. Int J Trichology 2019; 11:26-30. [PMID: 30820130 PMCID: PMC6385510 DOI: 10.4103/ijt.ijt_80_18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Interleukin-15 (IL-15) is a cytokine that is involved in many inflammatory and autoimmune diseases. Although alopecia areata (AA) is an autoimmune disease, serum levels of IL-15 have not been studied well in AA patients. Aim of the Work: We aims at evaluating the serum levels of IL-15 in active AA. Subject and Methods: This case-control study included 40 AA patients and 40 apparently healthy matched controls. Written informed consents were obtained from all the participants. The scalp was examined to assess sites, number, and size of alopecia patches, and the severity of AA lesions was assessed using the Severity of Alopecia Tool score (SALT score) which determine the percentage of hair loss in the scalp. The body was carefully examined to detect any alopecia patches in any hairy area. Nail examination was carried out to detect any nail involvement. Serum IL-15 levels were measured using an ELISA kits. Results: Serum levels of IL-15 in patients were significantly higher than those in the control group (P < 0.001). Serum levels in alopecia totalis were significantly higher than those with one or two patches, and serum levels in patients with both scalp and body involvement were significantly elevated than the levels of patients with either scalp or body involvement. There was a statistically significant positive correlation between SALT score and serum levels of IL-15 (P < 0.001). Conclusion: Serum IL-15 may be a marker of AA severity.
Collapse
Affiliation(s)
- Adel Ali Ebrahim
- Department of Dermatology, Faculty of Medicine, Benha University, Qalubia, Egypt
| | - Rehab Mohammed Salem
- Department of Dermatology, Faculty of Medicine, Benha University, Qalubia, Egypt
| | - Asmaa Adel El Fallah
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Benha University, Qalubia, Egypt
| | | |
Collapse
|
316
|
Breman E, Demoulin B, Agaugué S, Mauën S, Michaux A, Springuel L, Houssa J, Huberty F, Jacques-Hespel C, Marchand C, Marijsse J, Nguyen T, Ramelot N, Violle B, Daro D, De Waele P, Gilham DE, Steenwinckel V. Overcoming Target Driven Fratricide for T Cell Therapy. Front Immunol 2018; 9:2940. [PMID: 30619300 PMCID: PMC6299907 DOI: 10.3389/fimmu.2018.02940] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Abstract
Chimeric Antigen Receptor (CAR) T cells expressing the fusion of the NKG2D protein with CD3ζ (NKG2D-CAR T Cells) acquire a specificity for stress-induced ligands expressed on hematological and solid cancers. However, these stress ligands are also transiently expressed by activated T cells implying that NKG2D-based T cells may undergo self-killing (fratricide) during cell manufacturing or during the freeze thaw cycle prior to infusion in patients. To avoid target-driven fratricide and enable the production of NKG2D-CAR T cells for clinical application, two distinct approaches were investigated. The first focused upon the inclusion of a Phosphoinositol-3-Kinase inhibitor (LY294002) into the production process. A second strategy involved the inclusion of antibody blockade of NKG2D itself. Both processes impacted T cell fratricide, albeit at different levels with the antibody process being the most effective in terms of cell yield. While both approaches generated comparable NKG2D-CAR T cells, there were subtle differences, for example in differentiation status, that were fine-tuned through the phasing of the inhibitor and antibody during culture in order to generate a highly potent NKG2D-CAR T cell product. By means of targeted inhibition of NKG2D expression or generic inhibition of enzyme function, target-driven CAR T fratricide can be overcome. These strategies have been incorporated into on-going clinical trials to enable a highly efficient and reproducible manufacturing process for NKG2D-CAR T cells.
Collapse
|
317
|
Sun L, Chen L, Li H. Checkpoint-modulating immunotherapies in tumor treatment: Targets, drugs, and mechanisms. Int Immunopharmacol 2018; 67:160-175. [PMID: 30553199 DOI: 10.1016/j.intimp.2018.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/20/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023]
Abstract
Tumor immunotherapy, as a new treatment of cancer, has been developing on the basis of tumor immunology. Tumor immunotherapy stimulates and enhances the function of immune system in human bodies, in order to control and kill tumor cells. It is often used as an adjuvant therapy combined with surgery, chemotherapy, radiotherapy and other conventional methods. Cancer immunotherapies involve cells, antibodies and cytokines, etc. Some immunotherapies are widely used to activate the immune system, while some others precisely target at different tumor antigens. With the development of tumor immunotherapy, immune regulation activities of small molecules and biological agents have been gradually becoming a hot research area these years. In this review, we summarize the therapeutic targets, drugs, biologics, and their mechanisms in tumor immunotherapies.
Collapse
Affiliation(s)
- Lu Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
318
|
Cell-based immunotherapy approaches for multiple myeloma. Br J Cancer 2018; 120:38-44. [PMID: 30518815 PMCID: PMC6325139 DOI: 10.1038/s41416-018-0346-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/31/2022] Open
Abstract
Despite the arrival of novel therapies, multiple myeloma (MM) remains incurable and new treatment options are needed. Chimeric antigen receptor (CAR) T cells are genetically modified T cells that express a CAR directed against specific tumour antigens. CAR T cells are able to kill target tumour cells and may result in long-lasting immune responses in vivo. The rapid development of CAR technologies has led to clinical trials in haematological cancers including MM, and CAR T cells might evolve into a standard treatment in the next few years. Only small patient cohorts with relapsed or refractory disease have so far been investigated, but promising preliminary results with high response rates have been obtained in phase I clinical trials with B cell maturation antigen (BCMA), CD19, CD38 and κ-light-chain CAR T cells. Additional preclinical studies on CD38 and SLAMF7-CAR T cells in MM treatment yielded preclinical results that merit further investigation. Beyond the T cell approach, recent studies have focussed on CAR natural killer (NK) cells in order to increase the reactivity of these effector cells. Finally, to investigate the targeting of intracellular antigens, cellular therapies based on engineered T cell receptors (TCRs) are in development. In this review, we discuss results from preclinical and early-phase clinical trials testing the feasibility and safety of CAR T cell administration in MM, as well as early studies into approaches that utilise CAR NK cell and genetically modified TCRs.
Collapse
|
319
|
Haeryfar SMM, Shaler CR, Rudak PT. Mucosa-associated invariant T cells in malignancies: a faithful friend or formidable foe? Cancer Immunol Immunother 2018; 67:1885-1896. [PMID: 29470597 PMCID: PMC11028145 DOI: 10.1007/s00262-018-2132-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/13/2018] [Indexed: 12/12/2022]
Abstract
Mucosa-associated invariant T (MAIT) cells are a subset of innate-like T lymphocytes known for their ability to respond to MHC-related protein 1 (MR1)-restricted stimuli and select cytokine signals. They are abundant in humans and especially enriched in mucosal layers, common sites of neoplastic transformation. MAIT cells have been found within primary and metastatic tumors. However, whether they promote malignancy or contribute to anticancer immunity is unclear. On the one hand, MAIT cells produce IL-17A in certain locations and under certain circumstances, which could in turn facilitate neoangiogenesis, intratumoral accumulation of immunosuppressive cell populations, and cancer progression. On the other hand, they can express a potent arsenal of cytotoxic effector molecules, NKG2D and IFN-γ, all of which have established roles in cancer immune surveillance. In this review, we highlight MAIT cells' characteristics as they might pertain to cancer initiation, progression, or control. We discuss recent findings, including our own, that link MAIT cells to cancer, with a focus on colorectal carcinoma, as well as some of the outstanding questions in this active area of research. Finally, we provide a hypothetical picture in which MAIT cells constitute attractive targets in cancer immunotherapy.
Collapse
Affiliation(s)
- S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, N6A 5C1, Canada.
- Centre for Human Immunology, Western University, London, ON, Canada.
- Lawson Health Research Institute, London, ON, Canada.
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON, Canada.
| | - Christopher R Shaler
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| | - Patrick T Rudak
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| |
Collapse
|
320
|
Gao L, Jiang Y, Wang Y, Qu X, Li L, Lou X, Wang Y, Guo H, Liu Y. Male asymptomatic hyperuricemia patients display a lower number of NKG2D+ NK cells before and after a low-purine diet. Medicine (Baltimore) 2018; 97:e13668. [PMID: 30558070 PMCID: PMC6320027 DOI: 10.1097/md.0000000000013668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Aberrant activation of the immune system has been reported in asymptomatic hyperuricemia (HUA) patients. However, very few studies have elucidated the role of natural killer (NK) cells in this disease. METHODS In this study, we evaluated the relationship between NK cells and HUA in 16 control subjects and 20 patients, who were all on a low-purine diet. We analyzed the number of circulating NK cells, its subsets, interferon-γ, and CD107 NK cells, by flow cytometry, before and after 4 and 24 weeks of diet control. We also assessed the potential association of the NK cells with clinical measures. RESULTS The patients consistently had a lower number of NKG2D NK cells before and after low-purine diet, even the serum uric acid (SUA) levels <7 mg/dL after diet control. Moreover, a lower number of NK cells and a higher number of CD107a NK cells were observed on recruitment. Low-purine diet was benefit on the improvement of the SUA levels, body mass index (BMI), and the number and functions of NK cells. Furthermore, the number of CD3CD56 NK cells and NKG2D NK cells negatively correlated with the BMI before and after diet control. CONCLUSION The consistent lower number of NKG2D NK cells and correlated with BMI before and after low-purine diet may be involved in the occurrence and development of HUA.
Collapse
Affiliation(s)
- Lichao Gao
- The School of Public Health
- Department of Endocrinology of The First Hospital, Jilin University
| | - Yanfang Jiang
- Genetic Diagnosis Center
- Key Laboratory of Zoonoses Research, Ministry of Education, The First Hospital of Jilin University, Changchun
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yichen Wang
- Department of Endocrinology of The First Hospital, Jilin University
| | - Xiaozhang Qu
- Department of Endocrinology of The First Hospital, Jilin University
| | - Lei Li
- Department of Endocrinology of The First Hospital, Jilin University
| | - Xiaoqian Lou
- Department of Endocrinology of The First Hospital, Jilin University
| | - Ye Wang
- Department of Endocrinology of The First Hospital, Jilin University
| | - Hui Guo
- Department of Endocrinology of The First Hospital, Jilin University
| | - Ya Liu
- The School of Public Health
| |
Collapse
|
321
|
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
|
322
|
Baumeister SH, Murad J, Werner L, Daley H, Trebeden-Negre H, Gicobi JK, Schmucker A, Reder J, Sentman CL, Gilham DE, Lehmann FF, Galinsky I, DiPietro H, Cummings K, Munshi NC, Stone RM, Neuberg DS, Soiffer R, Dranoff G, Ritz J, Nikiforow S. Phase I Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol Res 2018; 7:100-112. [PMID: 30396908 DOI: 10.1158/2326-6066.cir-18-0307] [Citation(s) in RCA: 247] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/02/2018] [Accepted: 10/29/2018] [Indexed: 11/16/2022]
Abstract
NKG2D ligands are widely expressed in solid and hematologic malignancies but absent or poorly expressed on healthy tissues. We conducted a phase I dose-escalation study to evaluate the safety and feasibility of a single infusion of NKG2D-chimeric antigen receptor (CAR) T cells, without lymphodepleting conditioning in subjects with acute myeloid leukemia/myelodysplastic syndrome or relapsed/refractory multiple myeloma. Autologous T cells were transfected with a γ-retroviral vector encoding a CAR fusing human NKG2D with the CD3ζ signaling domain. Four dose levels (1 × 106-3 × 107 total viable T cells) were evaluated. Twelve subjects were infused [7 acute myeloid leukemia (AML) and 5 multiple myeloma]. NKG2D-CAR products demonstrated a median 75% vector-driven NKG2D expression on CD3+ T cells. No dose-limiting toxicities, cytokine release syndrome, or CAR T cell-related neurotoxicity was observed. No significant autoimmune reactions were noted, and none of the ≥ grade 3 adverse events were attributable to NKG2D-CAR T cells. At the single injection of low cell doses used in this trial, no objective tumor responses were observed. However, hematologic parameters transiently improved in one subject with AML at the highest dose, and cases of disease stability without further therapy or on subsequent treatments were noted. At 24 hours, the cytokine RANTES increased a median of 1.9-fold among all subjects and 5.8-fold among six AML patients. Consistent with preclinical studies, NKG2D-CAR T cell-expansion and persistence were limited. Manufactured NKG2D-CAR T cells exhibited functional activity against autologous tumor cells in vitro, but modifications to enhance CAR T-cell expansion and target density may be needed to boost clinical activity.
Collapse
Affiliation(s)
- Susanne H Baumeister
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Pediatric Hematology-Oncology Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Joana Murad
- Celdara Medical, LLC, Lebanon, New Hampshire
| | - Lillian Werner
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Heather Daley
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Helene Trebeden-Negre
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joanina K Gicobi
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jake Reder
- Celdara Medical, LLC, Lebanon, New Hampshire
| | | | | | | | - Ilene Galinsky
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Heidi DiPietro
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kristen Cummings
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nikhil C Munshi
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Richard M Stone
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Donna S Neuberg
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Glenn Dranoff
- Novartis BioMedical Institutes, Cambridge, Massachusetts
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
323
|
Zhao R, Cheng L, Jiang Z, Wei X, Li B, Wu Q, Wang S, Lin S, Long Y, Zhang X, Wu Y, Du X, Pei D, Liu P, Li Y, Cui S, Yao Y, Li P. DNAX-activating protein 10 co-stimulation enhances the anti-tumor efficacy of chimeric antigen receptor T cells. Oncoimmunology 2018; 8:e1509173. [PMID: 30546945 PMCID: PMC6287795 DOI: 10.1080/2162402x.2018.1509173] [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: 03/08/2018] [Revised: 07/19/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell immunotherapies have shown remarkable efficacy in treating multiple types of hematological malignancies but are not sufficiently effective at treating solid tumors. NKG2D is a strong activating receptor for NK cells and a co-stimulatory receptor for T cells. NKG2D signal transduction depends on DNAX-activating protein 10 (DAP10). Here, we introduced the cytoplasmic domain of DAP10 into the second-generation CARs M28z and G28z to generate M28z10 and G28z10, which target mesothelin (MSLN) and glypican 3 (GPC3), respectively. T cells expressing M28z10 or G28z10 showed enhanced and prolonged effector function against MSLN+ lung cancer or GPC3+ hepatocellular carcinoma cell lines in culture and secreted elevated levels of cytokines, including IL-2, IFN-γ, granzyme B, and GM-CSF. In addition, M28z10 CAR-T cells showed greater anti-tumor activity than those expressing M28z in both A549 cell line xenografts and human lung cancer patient-derived xenografts (PDX). Similarly, G28z10 exhibited higher efficacy in causing tumor regression than did G28z in hepatocellular carcinoma PDX. Therefore, our results show that DAP10 signaling contributes to the function of CAR-T cells in both lung cancer and hepatocellular carcinoma and can enhance the efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Ruocong Zhao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lin Cheng
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhiwu Jiang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xinru Wei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Baiheng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiting Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Suna Wang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Simiao Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Youguo Long
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xuchao Zhang
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin Du
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, Stem Cell and Regenerative Medicine Centre, University of Hong Kong, Hong Kong, China
| | - Yangqiu Li
- Institute of Hematology, Medical College, Jinan University, Guangzhou, China
| | - Shuzhong Cui
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Yao Yao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Peng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
324
|
Sarhan M, Land WG, Tonnus W, Hugo CP, Linkermann A. Origin and Consequences of Necroinflammation. Physiol Rev 2018; 98:727-780. [PMID: 29465288 DOI: 10.1152/physrev.00041.2016] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When cells undergo necrotic cell death in either physiological or pathophysiological settings in vivo, they release highly immunogenic intracellular molecules and organelles into the interstitium and thereby represent the strongest known trigger of the immune system. With our increasing understanding of necrosis as a regulated and genetically determined process (RN, regulated necrosis), necrosis and necroinflammation can be pharmacologically prevented. This review discusses our current knowledge about signaling pathways of necrotic cell death as the origin of necroinflammation. Multiple pathways of RN such as necroptosis, ferroptosis, and pyroptosis have been evolutionary conserved most likely because of their differences in immunogenicity. As the consequence of necrosis, however, all necrotic cells release damage associated molecular patterns (DAMPs) that have been extensively investigated over the last two decades. Analysis of necroinflammation allows characterizing specific signatures for each particular pathway of cell death. While all RN-pathways share the release of DAMPs in general, most of them actively regulate the immune system by the additional expression and/or maturation of either pro- or anti-inflammatory cytokines/chemokines. In addition, DAMPs have been demonstrated to modulate the process of regeneration. For the purpose of better understanding of necroinflammation, we introduce a novel classification of DAMPs in this review to help detect the relative contribution of each RN-pathway to certain physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Walter G Land
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Wulf Tonnus
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Christian P Hugo
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Andreas Linkermann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
325
|
de Alcântara BBR, Cruz FM, Fonseca FLA, da Costa Aguiar Alves B, Perez MM, Varela P, Pesquero JB, de Iracema Gomes Cubero D, De Melo Sette CV, Del Giglio A. Chemotherapy-induced fatigue is associated with changes in gene expression in the peripheral blood mononuclear cell fraction of patients with locoregional breast cancer. Support Care Cancer 2018; 27:2479-2486. [DOI: 10.1007/s00520-018-4519-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/16/2018] [Indexed: 01/27/2023]
|
326
|
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
|
327
|
Bonacini M, Soriano A, Zerbini A, Calò E, Cimino L, Muratore F, Fontana L, Braglia L, Parmeggiani M, Salvarani C, Croci S. Higher Frequencies of Lymphocytes Expressing the Natural Killer Group 2D Receptor in Patients With Behçet Disease. Front Immunol 2018; 9:2157. [PMID: 30319620 PMCID: PMC6167483 DOI: 10.3389/fimmu.2018.02157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/31/2018] [Indexed: 12/31/2022] Open
Abstract
Behçet disease (BD) is an inflammatory systemic disease with a fluctuating course, which can affect the skin, eyes, central nervous system, musculoskeletal, gastrointestinal, and vascular systems. No laboratory tests are currently available for the diagnosis of BD and monitoring disease activity. Moreover there is a lack of knowledge on BD pathogenesis. This study focused on circulating Natural Killer (NK), NKT and T cells evaluated as CD3neg CD56pos, CD3pos CD56pos, and CD3pos CD56neg. Peripheral blood mononuclear cells (PBMCs) were collected from 38 BD patients and 20 healthy controls (HC). The frequencies of NK, NKT, and T cells expressing CD16, CD69, NKG2D, Nkp30, Nkp46, and NKG2A were assessed by flow cytometry. Cytotoxic potential of NK cells was evaluated by flow cytometry as the percentage of cells expressing the degranulation marker CD107a after incubation with K562 cells. The levels of 27 cytokines were determined in plasma with a multiplex bead-based assay. Higher percentages of NK, NKT, and T cells expressing NKG2D were detected in PBMCs of BD patients than HC. ROC curve analysis showed that the evaluation of NKG2Dpos NK, NKT, and T cell percentages discriminated between BD patients and HC. Moreover, there was a positive correlation between the BD Current Activity Form (BDCAF) scores and the frequencies of NKG2Dpos NK and NKT cells. A higher frequency of NK cells expressing CD107a was induced in PBMCs from BD patients than HC after incubation with K562 cells. Concentrations of IL-5, IL-6, IL-10, IL-13, IP-10, and MIP-1β were higher in plasma of BD patients than HC. Monitoring the frequencies of NKG2Dpos lymphocytes could help the clinicians in BD patients management. In addition, the increased expression of NKG2D in BD patients is likely involved in disease pathogenesis.
Collapse
Affiliation(s)
- Martina Bonacini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandra Soriano
- Unit of Rheumatology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Campus Bio-Medico, University of Rome, Rome, Italy
| | - Alessandro Zerbini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Eleonora Calò
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Luca Cimino
- Unit of Ocular Immunology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Muratore
- Unit of Rheumatology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,University of Modena and Reggio Emilia, Modena, Italy
| | - Luigi Fontana
- Unit of Ophtalmology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Luca Braglia
- Research and Statistics Infrastructure, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Maria Parmeggiani
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Salvarani
- Unit of Rheumatology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Croci
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
328
|
Pasanen A, Karjalainen MK, Kummola L, Waage J, Bønnelykke K, Ruotsalainen M, Piippo-Savolainen E, Goksör E, Nuolivirta K, Chawes B, Vissing N, Bisgaard H, Jartti T, Wennergren G, Junttila I, Hallman M, Korppi M, Rämet M. NKG2D gene variation and susceptibility to viral bronchiolitis in childhood. Pediatr Res 2018; 84:451-457. [PMID: 29967528 DOI: 10.1038/s41390-018-0086-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Genetic factors associated with bronchiolitis are inadequately characterized. We therefore inspected a selected subpopulation of our previous genome-wide association study (GWAS) of bronchiolitis for overlap with known quantitative trait loci (QTLs) to identify susceptibility loci that potentially affect mRNA and protein levels. METHODS GWAS included a Finnish-Swedish case-control population (n = 187), matched for age and site. We integrated GWAS variants (p < 10-4) with QTL data. We subsequently verified allele-specific expression of identified QTLs by flow cytometry. Association of the resulting candidate loci with bronchiolitis was tested in three additional cohorts from Finland and Denmark (n = 1201). RESULTS Bronchiolitis-susceptibility variant rs10772271 resided within QTLs previously associated with NKG2D (NK group 2, member D) mRNA and protein levels. Flow cytometric analysis confirmed the association with protein level in NK cells. The GWAS susceptibility allele (A) of rs10772271 (odds ratio [OR] = 2.34) corresponded with decreased NKG2D expression. The allele was nominally associated with bronchiolitis in one Finnish replicate (OR = 1.50), and the other showed directional consistency (OR = 1.43). No association was detected in Danish population CONCLUSIONS: The bronchiolitis GWAS susceptibility allele was linked to decreased NKG2D expression in the QTL data and in our expression analysis. We propose that reduced NKG2D expression predisposes infants to severe bronchiolitis.
Collapse
Affiliation(s)
- Anu Pasanen
- PEDEGO Research Unit, Medical Research Center Oulu, and Department of Children and Adolescents, University of Oulu, Oulu University Hospital, Oulu, Finland.
| | - Minna K Karjalainen
- PEDEGO Research Unit, Medical Research Center Oulu, and Department of Children and Adolescents, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Laura Kummola
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Johannes Waage
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marja Ruotsalainen
- Department of Pediatrics, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Eija Piippo-Savolainen
- Department of Pediatrics, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Emma Goksör
- Department of Pediatrics, University of Gothenburg, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Kirsi Nuolivirta
- Department of Pediatrics, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nadja Vissing
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tuomas Jartti
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Göran Wennergren
- Department of Pediatrics, University of Gothenburg, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Ilkka Junttila
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere, Finland
| | - Mikko Hallman
- PEDEGO Research Unit, Medical Research Center Oulu, and Department of Children and Adolescents, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Matti Korppi
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Mika Rämet
- PEDEGO Research Unit, Medical Research Center Oulu, and Department of Children and Adolescents, University of Oulu, Oulu University Hospital, Oulu, Finland.,BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| |
Collapse
|
329
|
Nixon BG, Li MO. Tissue-Resident Cytolytic Innate Lymphocytes in Cancer. THE JOURNAL OF IMMUNOLOGY 2018; 200:408-414. [PMID: 29311382 DOI: 10.4049/jimmunol.1701124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/19/2017] [Indexed: 12/24/2022]
Abstract
Innate lymphoid cells (ILCs) are critical components of tissues in the body, providing a first line of defense against challenges to host integrity. In contrast to strictly cytokine-producing helper ILCs, resident innate lymphocyte populations with cytolytic potential have been identified in multiple tissues in both mouse and human. These cells express the transcription factor Tbet, NK cell receptors, granzymes, perforin, and death receptors, and can directly kill tumor cells. Signals in the tumor microenvironment may promote this response, including the cytokine IL-15 and stress-associated ligands for activating NK receptors. Although there is evidence that these cells are tissue and tumor resident, their lineage remains unclear. Whether they are derived from the NK or helper ILC lineages or represent a third differentiation pathway remains to be determined. A better understanding of their lineage will help clarify their regulation and function in the context of antitumor immunity.
Collapse
Affiliation(s)
- Briana G Nixon
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and.,Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and
| |
Collapse
|
330
|
Zou Y, Luo W, Guo J, Luo Q, Deng M, Lu Z, Fang Y, Zhang CC. NK cell-mediated anti-leukemia cytotoxicity is enhanced using a NKG2D ligand MICA and anti-CD20 scfv chimeric protein. Eur J Immunol 2018; 48:1750-1763. [PMID: 30063799 DOI: 10.1002/eji.201847550] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 07/06/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023]
Abstract
NK cells are important innate cytotoxic lymphocytes that have potential in treatment of leukemia. Engagement of NKG2D receptor on NK cells enhances the target cytotoxicity. Here, we produced a fusion protein consisting of the extracellular domain of the NKG2D ligand MICA and the anti-CD20 single-chain variable fragment (scfv). This recombinant protein is capable of binding both NK cells and CD20+ tumor cells. Using a human NKG2D reporter cell system we developed, we showed that this fusion protein could decorate CD20+ tumor cells with MICA extracellular domain and activate NK through NKG2D. We further demonstrated that this protein could specifically induce the ability of a NK cell line (NKL) and primary NK cells to lyse CD20+ leukemia cells. Moreover, we found that downregulation of surface HLA class I expression in the target cells improved NKL-mediated killing. Our results demonstrated that this recombinant protein specifically lyses leukemia cells by NK cells, which may lead to development of a novel strategy for treating leukemia and other tumors.
Collapse
Affiliation(s)
- Yizhou Zou
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Weiguang Luo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Jing Guo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qizhi Luo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Mi Deng
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Zhigang Lu
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Yi Fang
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Cheng Cheng Zhang
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| |
Collapse
|
331
|
The Human Cytomegalovirus Protein UL148A Downregulates the NK Cell-Activating Ligand MICA To Avoid NK Cell Attack. J Virol 2018; 92:JVI.00162-18. [PMID: 29950412 DOI: 10.1128/jvi.00162-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/18/2018] [Indexed: 01/19/2023] Open
Abstract
Natural killer (NK) cells are lymphocytes of the innate immune system capable of killing hazardous cells, including virally infected cells. NK cell-mediated killing is triggered by activating receptors. Prominent among these is the activating receptor NKG2D, which binds several stress-induced ligands, among them major histocompatibility complex (MHC) class I-related chain A (MICA). Most of the human population is persistently infected with human cytomegalovirus (HCMV), a virus which employs multiple immune evasion mechanisms, many of which target NK cell responses. HCMV infection is mostly asymptomatic, but in congenitally infected neonates and in immunosuppressed patients it can lead to serious complications and mortality. Here we discovered that an HCMV protein named UL148A whose role was hitherto unknown is required for evasion of NK cells. We demonstrate that UL148A-deficient HCMV strains are impaired in their ability to downregulate MICA expression. We further show that when expressed by itself, UL148A is not sufficient for MICA targeting, but rather acts in concert with an unknown viral factor. Using inhibitors of different cellular degradation pathways, we show that UL148A targets MICA for lysosomal degradation. Finally, we show that UL148A-mediated MICA downregulation hampers NK cell-mediated killing of HCMV-infected cells. Discovering the full repertoire of HCMV immune evasion mechanisms will lead to a better understanding of the ability of HCMV to persist in the host and may also promote the development of new vaccines and drugs against HCMV.IMPORTANCE Human cytomegalovirus (HCMV) is a ubiquitous pathogen which is usually asymptomatic but that can cause serious complications and mortality in congenital infections and in immunosuppressed patients. One of the difficulties in developing novel vaccines and treatments for HCMV is its remarkable ability to evade our immune system. In particular, HCMV directs significant efforts to thwarting cells of the innate immune system known as natural killer (NK) cells. These cells are crucial for successful control of HCMV infection, and yet our understanding of the mechanisms which HCMV utilizes to elude NK cells is partial at best. In the present study, we discovered that a protein encoded by HCMV which had no known function is important for preventing NK cells from killing HCMV-infected cells. This knowledge can be used in the future for designing more-efficient HCMV vaccines and for formulating novel therapies targeting this virus.
Collapse
|
332
|
Sheppard S, Ferry A, Guedes J, Guerra N. The Paradoxical Role of NKG2D in Cancer Immunity. Front Immunol 2018; 9:1808. [PMID: 30150983 PMCID: PMC6099450 DOI: 10.3389/fimmu.2018.01808] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
The activating receptor NKG2D and its ligands are recognized as a potent immune axis that controls tumor growth and microbial infections. With regards to cancer surveillance, various studies have demonstrated the antitumor function mediated by NKG2D on natural killer cells and on conventional and unconventional T cells. The use of NKG2D-deficient mice established the importance of NKG2D in delaying tumor development in transgenic mouse models of cancer. However, we recently demonstrated an unexpected, flip side to this coin, the ability for NKG2D to contribute to tumor growth in a model of inflammation-driven liver cancer. With a focus on the liver, here, we review current knowledge of NKG2D-mediated tumor surveillance and discuss evidence supporting a dual role for NKG2D in cancer immunity. We postulate that in certain advanced cancers, expression of ligands for NKG2D can drive cancer progression rather than rejection. We propose that the nature of the microenvironment within and surrounding tumors impacts the outcome of NKG2D activation. In a form of autoimmune attack, NKG2D promotes tissue damage, mostly in the inflamed tissue adjacent to the tumor, facilitating tumor progression while being ineffective at rejecting transformed cells in the tumor bed.
Collapse
Affiliation(s)
- Sam Sheppard
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Memorial Sloan Kettering Cancer Center, Zuckerman Research Center, New York, NY, United States
| | - Amir Ferry
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Joana Guedes
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
333
|
Johnson JK, Miller JS. Current strategies exploiting NK-cell therapy to treat haematologic malignancies. Int J Immunogenet 2018; 45:237-246. [PMID: 30009514 DOI: 10.1111/iji.12387] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/10/2018] [Accepted: 06/07/2018] [Indexed: 12/30/2022]
Abstract
Natural killer (NK) cells recognize targets that have been changed via malignant transformation or infection. Previously, NK cells were thought to be short-lived, but we now know that NK cells can be long-lived and remember past exposures in response to CMV. NK cells use a plethora of activating and inhibitory receptors to recognize these changes and attack targets, but tumour cells often evade NK cells. Therefore, major efforts are being made to hone in on NK cell antitumour properties in immunotherapy. In the clinical setting, haploidentical NK cells can be adoptively transferred to help treat cancer. To expand NK cells in vivo and enhance tumour targeting, IL-15 is being tested in combination with a glycogen synthase kinase (GSK) 3 inhibitor (CHIR99021), an inhibitor that has been shown to expand mature, highly functional NK cells capable of killing multiple tumour targets. One major limitation to NK cell therapy is lack of specificity. To address this concern, bispecific or trispecific engagers that target NK cells to the tumour and an ADAM17 inhibitor that prevents CD16 shedding after NK cell activation are being tested. Additionally, monoclonal antibodies are being designed to redirect the inhibitory signals that limit NK cell functionality. Further understanding of the biology of NK cells will inform strategies to exploit NK cells for therapeutic purposes.
Collapse
Affiliation(s)
- Jenna K Johnson
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, Masonic Cancer Center, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
- Medical Scientist Training Program, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, Masonic Cancer Center, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| |
Collapse
|
334
|
Gaggero S, Bruschi M, Petretto A, Parodi M, Del Zotto G, Lavarello C, Prato C, Santucci L, Barbuto A, Bottino C, Candiano G, Moretta A, Vitale M, Moretta L, Cantoni C. Nidogen-1 is a novel extracellular ligand for the NKp44 activating receptor. Oncoimmunology 2018; 7:e1470730. [PMID: 30228939 PMCID: PMC6140582 DOI: 10.1080/2162402x.2018.1470730] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/09/2018] [Accepted: 04/25/2018] [Indexed: 11/25/2022] Open
Abstract
The release of soluble ligands of activating Natural Killer (NK) cell receptors may represent a regulatory mechanism of NK cell function both in physiologic and in pathologic conditions. Here, we identified the extracellular matrix protein Nidogen-1 (NID1) as a ligand of NKp44, an important activating receptor expressed by activated NK cells. When released as soluble molecule, NID1 regulates NK cell function by modulating NKp44-induced IFN-γ production or cytotoxicity. In particular, it also modulates IFN-γ production induced by Platelet-Derived Growth Factor (PDGF)-DD following NKp44 engagement. We also show that NID1 may be present at the cell surface. In this form or when bound to a solid support (bNID1), NID1 fails to induce NK cell cytotoxicity or cytokine release. However, analysis by mass spectrometry revealed that exposure to bNID1 can induce in human NK cells relevant changes in the proteomic profiles suggesting an effect on different biological processes.
Collapse
Affiliation(s)
- Silvia Gaggero
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Maurizio Bruschi
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea Petretto
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Monica Parodi
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Genny Del Zotto
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Chiara Lavarello
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Carola Prato
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Laura Santucci
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alessandra Barbuto
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Cristina Bottino
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giovanni Candiano
- Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alessandro Moretta
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Massimo Vitale
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo Moretta
- Immunology area, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Dipartimento dei Laboratori di Ricerca, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
335
|
Lin Z, Han S, Qian X, Hu C, Xiao W, Qian L, Zhang Y, Ding Y, Jia X, Zhu G, Gong W. Regulatory NK1.1 -CD4 +NKG2D + subset induced by NKG2DL + cells promotes tumor evasion in mice. Cancer Immunol Immunother 2018; 67:1159-1173. [PMID: 29802426 PMCID: PMC11028319 DOI: 10.1007/s00262-018-2172-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 05/12/2018] [Indexed: 12/24/2022]
Abstract
Regulatory T cells play critical roles in self-tolerance and tumor evasion. CD4+NKG2D+ cells with regulatory activity are present in patients with NKG2DL+ tumors and juvenile systemic lupus erythematosus. We previously showed that TGF-β-producing CD4+NKG2D+ T cells are present in pCD86-Rae-1ε transgenic mice. Here, we performed both ex vivo and in vivo studies on pCD86-Rae-1ε transgenic mice and an MC38 tumor-bearing mouse model and show that NK1.1-CD4+NKG2D+ T cells have regulatory activity in pCD86-Rae-1ε transgenic mice. Furthermore, this T-cell subset was induced in mice transplanted with NKG2DL+ tumor cells and produced TGF-β and FasL, and secreted low amounts of IFN-γ. This T-cell subset downregulated the function of effector T cells and dendritic cells, which were abolished by anti-TGF-β antibody. In vivo, adoptive transfer of NK1.1-CD4+NKG2D+ T cells promoted TGF-β-dependent tumor growth in mice. We further found that ex vivo induction of NK1.1-CD4+NKG2D+ T cells was dependent on both anti-CD3 and NKG2DL stimulation. Furthermore, regulatory NK1.1-CD4+NKG2D+ T cells did not express Foxp3 or CD25 and expressed intermediate levels of T-bet. Western-blotting showed that STAT3 signaling was activated in NK1.1-CD4+NKG2D+ T cells of MC38 tumor-bearing and pCD86-Rae-1ε transgenic mice. In conclusion, we describe a regulatory NK1.1-CD4+NKG2D+ T-cell population, different from other regulatory T cells and abnormally elevated in pCD86-Rae-1ε transgenic and MC38 tumor-bearing mice.
Collapse
Affiliation(s)
- Zhijie Lin
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, 11 Huaihai Road, Yangzhou, 225001, People's Republic of China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 E. Wenhui Road, Yangzhou, 225009, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| | - Sen Han
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, 11 Huaihai Road, Yangzhou, 225001, People's Republic of China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, People's Republic of China
| | - Xingxing Qian
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, 11 Huaihai Road, Yangzhou, 225001, People's Republic of China
| | - Chunxia Hu
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, 11 Huaihai Road, Yangzhou, 225001, People's Republic of China
| | - Weiming Xiao
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, People's Republic of China
| | - Li Qian
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, 11 Huaihai Road, Yangzhou, 225001, People's Republic of China
| | - Yu Zhang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 E. Wenhui Road, Yangzhou, 225009, People's Republic of China
| | - Yanbing Ding
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, People's Republic of China
| | - Xiaoqin Jia
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, People's Republic of China
| | - Guoqiang Zhu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 E. Wenhui Road, Yangzhou, 225009, People's Republic of China.
| | - Weijuan Gong
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, 11 Huaihai Road, Yangzhou, 225001, People's Republic of China.
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, People's Republic of China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, People's Republic of China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 E. Wenhui Road, Yangzhou, 225009, People's Republic of China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China.
| |
Collapse
|
336
|
Hosomi S, Grootjans J, Huang YH, Kaser A, Blumberg RS. New Insights Into the Regulation of Natural-Killer Group 2 Member D (NKG2D) and NKG2D-Ligands: Endoplasmic Reticulum Stress and CEA-Related Cell Adhesion Molecule 1. Front Immunol 2018; 9:1324. [PMID: 29973929 PMCID: PMC6020765 DOI: 10.3389/fimmu.2018.01324] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/28/2018] [Indexed: 01/02/2023] Open
Abstract
Natural-killer group 2 member D (NKG2D) is a well-characterized activating receptor expressed by natural killer (NK) cells, NKT cells, activated CD8+ T cells, subsets of γδ+ T cells, and innate-like T cells. NKG2D recognizes multiple ligands (NKG2D-ligands) to mount an innate immune response against stressed, transformed, or infected cells. NKG2D-ligand surface expression is tightly restricted on healthy cells through transcriptional and post-transcriptional mechanisms, while transformed or infected cells express the ligands as a danger signal. Recent studies have revealed that unfolded protein response pathways during endoplasmic reticulum (ER) stress result in upregulation of ULBP-related protein via the protein kinase RNA-like ER kinase-activating factor 4-C/EBP homologous protein (PERK-ATF4-CHOP) pathway, which can be linked to the pathogenesis of autoimmune diseases. Transformed cells, however, possess mechanisms to escape NKG2D-mediated immune surveillance, such as upregulation of carcinoembryonic antigen (CEA)-related cell adhesion molecule 1 (CEACAM1), a negative regulator of NKG2D-ligands. In this review, we discuss mechanisms of NKG2D-ligand regulation, with a focus on newly discovered mechanisms that promote NKG2D-ligand expression on epithelial cells, including ER stress, and mechanisms that suppress NKG2D-ligand-mediated killing of cancer cells, namely by co-expression of CEACAM1.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Joep Grootjans
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Yu-Hwa Huang
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
337
|
Baek IC, Shin DH, Choi EJ, Kim HJ, Yoon JH, Cho BS, Kim YJ, Lee S, Min WS, Kim HJ, Kim TG. Association of MICA and MICB polymorphisms with the susceptibility of leukemia in Korean patients. Blood Cancer J 2018; 8:58. [PMID: 29895953 PMCID: PMC5997647 DOI: 10.1038/s41408-018-0092-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 04/19/2018] [Accepted: 05/08/2018] [Indexed: 02/05/2023] Open
Affiliation(s)
- In-Cheol Baek
- Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong-Hwan Shin
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun-Jeong Choi
- Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyoung-Jae Kim
- Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae-Ho Yoon
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung-Sik Cho
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoo-Jin Kim
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok Lee
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Sung Min
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Tai-Gyu Kim
- Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Korea.
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
338
|
Rakic M, Persic V, Kehler T, Bastiancic AL, Rosovic I, Laskarin G, Sotosek Tokmadzic V. Possible role of circulating endothelial cells in patients after acute myocardial infarction. Med Hypotheses 2018; 117:42-46. [PMID: 30077195 DOI: 10.1016/j.mehy.2018.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 06/05/2018] [Indexed: 01/16/2023]
Abstract
Acute myocardial infarction (AMI) occurs as a result of insufficient myocardial perfusion leading to cell necrosis. This is most commonly due to the obstruction of the coronary artery by ruptured atherosclerotic plaque and thrombosis. Damaged ischemic and necrotic myocardial cells release pro-inflammatory substances in tissue and plasma, leading to a systemic inflammatory response. Profound systemic inflammatory response during ischemia/reperfusion injury causes disruption of endothelial glycocalyx and detachment of endothelial cells that express von Willebrant factor (vWF). We hypothesize that circulating vWF+ endothelial cells could act as antigen presenting cells which interact with T and NK cells directly, by cell to cell contact and indirectly by cytokine and chemokine secretion, leading to the immune response towards inflammation. Analyzing the frequency, phenotype and pro-inflammatory substances produced in circulating vWF positive (+) cells in patients with AMI could be beneficial to determine the severity of the pro-inflammatory response, according to the level of endothelial dysfunction in the early period of AMI. To evaluate these hypotheses, we suggest to determine frequency, phenotype, and ability of cytokine/chemokine production in circulating vWF+ endothelial cells by simultaneous surface and intracellular cell staining, and flow cytometry analysis. Secretion of pro-inflammatory cytokines and chemokines, pro-atherogenic substances and the components of glycocalyx might be measured in supernatants of magnetically separated or sorted vWF+ endothelial cells, as well as in the serum of a patient with acute AMI by enzyme linked-immunoassay tests. The interaction of increasing concentrations of isolated circulating vWF+ endothelial cells and cognate T and NK cells might be investigated by lymphocyte proliferation rate, cytotoxic mediators' expression, and cytokine production. If our hypothesis is correct, characterization of circulating vWF+ endothelial cells could grant us greater insight into their role in pathophysiology of AMI and the degree of myocardial damage.
Collapse
Affiliation(s)
- Marijana Rakic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Viktor Persic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia; Department of Medical Rehabilitation, Medical Faculty, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia
| | - Tatjana Kehler
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Ana Lanca Bastiancic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Ivan Rosovic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Gordana Laskarin
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia; Department of Physiology and Immunology, Medical Faculty University of Rijeka, B.Branchetta 20, 51000 Rijeka, Croatia
| | - Vlatka Sotosek Tokmadzic
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
339
|
Abouelghar A, Hasnah R, Taouk G, Saad M, Karam M. Prognostic values of the mRNA expression of natural killer receptor ligands and their association with clinicopathological features in breast cancer patients. Oncotarget 2018; 9:27171-27196. [PMID: 29930758 PMCID: PMC6007477 DOI: 10.18632/oncotarget.25506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells are lymphocytes of the innate immune system that have potent cytotoxic activity against tumor cells. NK cell recognition and activity towards cancer cells are regulated by an integrated interplay between numerous inhibitory and activating receptors acting in concert to eliminate tumor cells expressing cognate ligands. Despite strong evidence supporting the role of NK cells in breast cancer (BC) control, BC still develops and progresses to form large tumors and metastases. A major mechanism of BC escape from NK immunity is the alteration of the expression of NK receptor ligands. The aim of this study was to determine whether NK receptor ligands' mRNA expression might influence prognosis in BC patients and whether these effects differ by molecular subtypes and clinicopathological features. METHODS We used the KM plotter platform to analyze the correlation between mRNA expression of 32 NK receptor ligands and relapse-free survival (RFS) and overall survival (OS) in 3951 and 1402 BC patients, respectively. The association with tumor subtypes and clinicopathological features was determined. BC samples were split into high and low expression groups according to the best cutoff value and the two patient cohorts were compared by Kaplan-Meier survival plots. The hazard ratios with 95% confidence intervals and log rank P values were calculated and FDR-adjusted for multiple testing correction. The data was considered to be statistically significant when FDR-adjusted P value < 0.05. RESULTS High mRNA expression of around 80% of ligands for NK activating and inhibitory receptors associated with better RFS, which correlated with longer OS for only about half of the NK-activating ligands but for most NK-inhibitory ligands. Also, five NK-activating ligands correlated with worse prognosis. These prognostic values were differentially associated with the BC clinical criteria. In addition, the favorable prognostic influence of NK-activating ligands' upregulation, as a whole, was mainly significantly associated with HER2-positive and basal-like subtypes, lymph node positive phenotype, and high-grade tumors. CONCLUSIONS NK receptor ligands appear to play an important role in defining BC patient prognosis. Identification of a group of patients with worse prognosis expressing high levels of NK-activating ligands and low levels of NK-inhibitory ligands makes them ideal potential candidates for NK-based immunotherapy to eliminate residual tumor cells, prevent relapse and improve patient survival.
Collapse
Affiliation(s)
- Ali Abouelghar
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Reem Hasnah
- Department of Biological Sciences, Carnegie Mellon University in Qatar, Doha, Qatar
| | - Ghina Taouk
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Mohamad Saad
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Manale Karam
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| |
Collapse
|
340
|
Babic M, Romagnani C. The Role of Natural Killer Group 2, Member D in Chronic Inflammation and Autoimmunity. Front Immunol 2018; 9:1219. [PMID: 29910814 PMCID: PMC5992374 DOI: 10.3389/fimmu.2018.01219] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/15/2018] [Indexed: 01/06/2023] Open
Abstract
Current medicine and medical science puts great effort into elucidating the basis of chronicity and finding appropriate treatments for inflammatory diseases; however, the mechanisms driving aberrant immune responses are mostly unknown and deserve further study. Of particular interest is the identification of checkpoints that regulate the function and differentiation of pro-inflammatory cells during pathogenesis, along with means of their modulation for therapeutic purposes. Natural killer group 2, member D (NKG2D) is a potent activator of the immune system, known as a sensor for “induced-self” ligands, i.e., cellular danger signals that, in the context of chronic inflammation and autoimmunity, can be presented by cells being exposed to an inflammatory cytokine milieu, endoplasmic reticulum stress, or cell death. Engagement by such ligands can be translated by NKG2D into activation or co-stimulation of NK cells and different subsets of T cells, respectively, thus contributing to the regulation of the inflammatory response. In this review, we discuss the current knowledge on the contribution of the NKG2D–NKG2DL signaling axis during intestinal inflammation, type 1 diabetes, multiple sclerosis, and rheumatoid arthritis, where the role of NKG2D has been associated either by aberrant expression of the receptor and its ligands and/or by functional data in corresponding mouse models.
Collapse
Affiliation(s)
- Marina Babic
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany.,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Center (DRFZ), Leibniz Association, Berlin, Germany.,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
341
|
Sato S, Weaver AM. Extracellular vesicles: important collaborators in cancer progression. Essays Biochem 2018; 62:149-163. [PMID: 29666212 PMCID: PMC6377252 DOI: 10.1042/ebc20170080] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are membrane vesicles that are released from cells and mediate cell-cell communication. EVs carry protein, lipid, and nucleic acid cargoes that interact with recipient cells to alter their phenotypes. Evidence is accumulating that tumor-derived EVs can play important roles in all steps of cancer progression. Here, we review recent studies reporting critical roles for EVs in four major areas of cancer progression: promotion of cancer invasiveness and motility, enhancement of angiogenesis and vessel permeability, conditioning premetastatic niches, and immune suppression.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| |
Collapse
|
342
|
Easom NJW, Stegmann KA, Swadling L, Pallett LJ, Burton AR, Odera D, Schmidt N, Huang WC, Fusai G, Davidson B, Maini MK. IL-15 Overcomes Hepatocellular Carcinoma-Induced NK Cell Dysfunction. Front Immunol 2018; 9:1009. [PMID: 29867983 PMCID: PMC5954038 DOI: 10.3389/fimmu.2018.01009] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/23/2018] [Indexed: 12/23/2022] Open
Abstract
NK cells have potent antitumor capacity. They are enriched in the human liver, with a large subset specialized for tissue-residence. The potential for liver-resident versus liver-infiltrating NK cells to populate, and exert antitumor functions in, human liver tumors has not been studied. We examined liver-resident and liver-infiltrating NK cells directly ex vivo from human hepatocellular carcinomas (HCCs) and liver colorectal (CRC) metastases, compared with matched uninvolved liver tissue. We found that NK cells were highly prevalent in both HCC and liver CRC metastases, although at lower frequencies than unaffected liver. Up to 79% of intratumoral NK cells had the CXCR6+CD69+ liver-resident phenotype. Direct ex vivo staining showed that liver-resident NK cells had increased NKG2D expression compared to their non-resident counterparts, but both subsets had NKG2D downregulation within liver tumors compared to uninvolved liver. Proliferation of intratumoral NK cells (identified by Ki67) was selectively impaired in those with the most marked NKG2D downregulation. Human liver tumor NK cells were functionally impaired, with reduced capacity for cytotoxicity and production of cytokines, even when compared to the hypo-functional tissue-resident NK cells in unaffected liver. Coculture of human liver NK cells with the human hepatoma cell line PLC/PRF/5, or with autologous HCC, recapitulated the defects observed in NK cells extracted from tumors, with downmodulation of NKG2D, cytokine production, and target cell cytotoxicity. Transwells and conditioned media confirmed a requirement for cell contact with PLC/PRF/5 to impose NK cell inhibition. IL-15 was able to recover antitumor functionality in NK cells inhibited by in vitro exposure to HCC cell lines or extracted directly from HCC. In summary, our data suggest that the impaired antitumor function of local NK cells reflects a combination of the tolerogenic features inherent to liver-resident NK cells together with additional contact-dependent inhibition imposed by HCC itself. The demonstration that IL-15 can recover hepatic NK cell function following tumor exposure supports its inclusion in immunotherapy strategies.
Collapse
Affiliation(s)
- Nicholas J W Easom
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Kerstin A Stegmann
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Laura J Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Alice R Burton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Dennis Odera
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Nathalie Schmidt
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Wei-Chen Huang
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Giuseppe Fusai
- Centre for Digestive Diseases, Institute of Liver and Digestive Health, University College London, London, United Kingdom
| | - Brian Davidson
- Centre for Digestive Diseases, Institute of Liver and Digestive Health, University College London, London, United Kingdom
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
343
|
Rohn H, Tomoya Michita R, Schwich E, Dolff S, Gäckler A, Trilling M, Le-Trilling VTK, Wilde B, Korth J, Heinemann FM, Horn PA, Kribben A, Witzke O, Rebmann V. The Donor Major Histocompatibility Complex Class I Chain-Related Molecule A Allele rs2596538 G Predicts Cytomegalovirus Viremia in Kidney Transplant Recipients. Front Immunol 2018; 9:917. [PMID: 29867932 PMCID: PMC5953334 DOI: 10.3389/fimmu.2018.00917] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/13/2018] [Indexed: 01/14/2023] Open
Abstract
The interaction of major histocompatibility complex class I chain-related protein A (MICA) and its cognate activating receptor natural killer (NK) group 2 member D (NKG2D) receptor plays a significant role in viral immune control. In the context of kidney transplantation (KTx), cytomegalovirus (CMV) frequently causes severe complications. Hypothesizing that functional polymorphisms of the MICA/NKG2D axis might affect antiviral NK and T cell responses to CMV, we explored the association of the MICA-129 Met/Val single nucleotide polymorphism (SNP) (affecting the binding affinity of MICA with the NKG2D receptor), the MICA rs2596538 G/A SNP (influencing MICA transcription), and the NKG2D rs1049174 G/C SNP (determining the cytotoxic potential of effector cells) with the clinical outcome of CMV during the first year after KTx in a cohort of 181 kidney donor-recipients pairs. Univariate analyses identified the donor MICA rs2596538 G allele status as a protective prognostic determinant for CMV disease. In addition to the well-known prognostic factors CMV high-risk sero-status of patients and the application of lymphocyte-depleting drugs, the donor MICA rs2596538 G allele carrier status was confirmed by multivariate analyses as novel-independent factor predicting the development of CMV infection/disease during the first year after KTx. The results of our study emphasize the clinical importance of the MICA/NKG2D axis in CMV control in KTx and point out that the potential MICA transcription in the donor allograft is of clinically relevant importance for CMV immune control in this allogeneic situation. Furthermore, they provide substantial evidence that the donor MICA rs2596538 G allele carrier status is a promising genetic marker predicting CMV viremia after KTx. Thus, in the kidney transplant setting, donor MICA rs2596538 G may help to allow the future development of personal CMV approaches within a genetically predisposed patient cohort.
Collapse
Affiliation(s)
- Hana Rohn
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Rafael Tomoya Michita
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Esther Schwich
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Dolff
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Anja Gäckler
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Johannes Korth
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Falko M Heinemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Vera Rebmann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
344
|
Monteiro A, Cruto C, Rosado P, Martinho A, Rosado L, Fonseca M, Paiva A. Characterization of circulating gamma-delta T cells in relapsing vs remission multiple sclerosis. J Neuroimmunol 2018; 318:65-71. [DOI: 10.1016/j.jneuroim.2018.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 12/23/2022]
|
345
|
Celyad's novel CAR T-cell therapy for solid malignancies. Curr Res Transl Med 2018; 66:53-56. [DOI: 10.1016/j.retram.2018.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/16/2018] [Indexed: 12/30/2022]
|
346
|
Zingoni A, Vulpis E, Cecere F, Amendola MG, Fuerst D, Saribekyan T, Achour A, Sandalova T, Nardone I, Peri A, Soriani A, Fionda C, Mariggiò E, Petrucci MT, Ricciardi MR, Mytilineos J, Cippitelli M, Cerboni C, Santoni A. MICA-129 Dimorphism and Soluble MICA Are Associated With the Progression of Multiple Myeloma. Front Immunol 2018; 9:926. [PMID: 29765374 PMCID: PMC5938351 DOI: 10.3389/fimmu.2018.00926] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/13/2018] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells are immune innate effectors playing a pivotal role in the immunosurveillance of multiple myeloma (MM) since they are able to directly recognize and kill MM cells. In this regard, among activating receptors expressed by NK cells, NKG2D represents an important receptor for the recognition of MM cells, being its ligands expressed by tumor cells, and being able to trigger NK cell cytotoxicity. The MHC class I-related molecule A (MICA) is one of the NKG2D ligands; it is encoded by highly polymorphic genes and exists as membrane-bound and soluble isoforms. Soluble MICA (sMICA) is overexpressed in the serum of MM patients, and its levels correlate with tumor progression. Interestingly, a methionine (Met) to valine (Val) substitution at position 129 of the α2 heavy chain domain classifies the MICA alleles into strong (MICA-129Met) and weak (MICA-129Val) binders to NKG2D receptor. We addressed whether the genetic polymorphisms in the MICA-129 alleles could affect MICA release during MM progression. The frequencies of Val/Val, Val/Met, and Met/Met MICA-129 genotypes in a cohort of 137 MM patients were 36, 43, and 22%, respectively. Interestingly, patients characterized by a Val/Val genotype exhibited the highest levels of sMICA in the sera. In addition, analysis of the frequencies of MICA-129 genotypes among different MM disease states revealed that Val/Val patients had a significant higher frequency of relapse. Interestingly, NKG2D was downmodulated in NK cells derived from MICA-129Met/Met MM patients. Results obtained by structural modeling analysis suggested that the Met to Val dimorphism could affect the capacity of MICA to form an optimal template for NKG2D recognition. In conclusion, our findings indicate that the MICA-129Val/Val variant is associated with significantly higher levels of sMICA and the progression of MM, strongly suggesting that the usage of soluble MICA as prognostic marker has to be definitely combined with the patient MICA genotype.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Elisabetta Vulpis
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Francesca Cecere
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Maria G Amendola
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Daniel Fuerst
- German Red Cross Blood Donor Services, Baden-Wuerttemberg-Hessia, Ulm, Germany
| | - Taron Saribekyan
- German Red Cross Blood Donor Services, Baden-Wuerttemberg-Hessia, Ulm, Germany
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Ilaria Nardone
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Agnese Peri
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Elena Mariggiò
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy
| | - Maria T Petrucci
- Department of Cellular Biotechnologies and Hematology, "Sapienza" University of Rome, Rome, Italy
| | - Maria R Ricciardi
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Joannis Mytilineos
- German Red Cross Blood Donor Services, Baden-Wuerttemberg-Hessia, Ulm, Germany
| | - Marco Cippitelli
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Cristina Cerboni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Istituto Pasteur Italia-Cenci Bolognetti Fondazione, Rome, Italy
| |
Collapse
|
347
|
Lee YS, Heo W, Nam J, Jeung YH, Bae J. The combination of ionizing radiation and proteasomal inhibition by bortezomib enhances the expression of NKG2D ligands in multiple myeloma cells. JOURNAL OF RADIATION RESEARCH 2018; 59:245-252. [PMID: 29518205 PMCID: PMC5967576 DOI: 10.1093/jrr/rry005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 12/27/2017] [Indexed: 06/10/2023]
Abstract
Bortezomib, which is a potent proteasome inhibitor, has been used as a first-line drugs to treat multiple myeloma for a few decades, and radiotherapy has frequently been applied to manage acute bone lesions in the patients. Therefore, it was necessary to investigate what the benefits might be if the two therapies were applied simultaneously in the treatment of multiple myeloma. Since it was known that radiotherapy and proteasome inhibitors could increase the expression of NKG2D ligands through induction of protein synthesis and suppression of protein degradation of NKG2D ligands, respectively, we supposed that the combined treatment might further enhance the expression of NKG2D ligands. In this study, we analyzed the expression level of NKG2D ligands using multiplex PCR and flow cytometry after treatment of IM-9 and RPMI-8226 myeloma cells with bortezomib and ionizing radiation; we then assayed the susceptibility to NK-92 cells. Although the expression of only some kinds of NKG2D ligands were increased by treatment with bortezomib alone, five kinds of NKG2D ligands that we assayed were further induced at the surface protein level after combined treatment with ionizing radiation and bortezomib. Furthermore, combined treatment made myeloma cells more susceptible to NK-92 cells, compared with treatment with bortezomib alone. In conclusion, the combination therapy of ionizing radiation plus the proteasome inhibitor bortezomib is a promising therapeutical strategy for enhancing NK cell-mediated anticancer immune responses.
Collapse
Affiliation(s)
- Young Shin Lee
- Department of Biochemistry, Pusan National University School of Medicine, Busandaehakro-49, Mulgeum-eup Yangsan-si Gyeongsangnam-do, 50612, South Korea
- PNU BK21 Plus Biomedical Science Education Center, Pusan National University School of Medicine, Busandaehakro-49, Mulgeum-eup Yangsan-si Gyeongsangnam-do 50612, South Korea
| | - Woong Heo
- Department of Biochemistry, Pusan National University School of Medicine, Busandaehakro-49, Mulgeum-eup Yangsan-si Gyeongsangnam-do, 50612, South Korea
- PNU BK21 Plus Biomedical Science Education Center, Pusan National University School of Medicine, Busandaehakro-49, Mulgeum-eup Yangsan-si Gyeongsangnam-do 50612, South Korea
| | - Jiho Nam
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Geumo-ro 20, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 50612, South Korea
| | - Young Hwa Jeung
- Department of Cogno-Mechatronics Engineering, Pusan National University, 2, Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, South Korea
| | - Jaeho Bae
- Department of Biochemistry, Pusan National University School of Medicine, Busandaehakro-49, Mulgeum-eup Yangsan-si Gyeongsangnam-do, 50612, South Korea
- PNU BK21 Plus Biomedical Science Education Center, Pusan National University School of Medicine, Busandaehakro-49, Mulgeum-eup Yangsan-si Gyeongsangnam-do 50612, South Korea
| |
Collapse
|
348
|
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
|
349
|
Xu LJ, Ma Q, Zhu J, Li J, Xue BX, Gao J, Sun CY, Zang YC, Zhou YB, Yang DR, Shan YX. Combined inhibition of JAK1,2/Stat3‑PD‑L1 signaling pathway suppresses the immune escape of castration‑resistant prostate cancer to NK cells in hypoxia. Mol Med Rep 2018; 17:8111-8120. [PMID: 29693186 PMCID: PMC5983983 DOI: 10.3892/mmr.2018.8905] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Castration‑resistant prostate cancer (CRPC) is difficult to treat in current clinical practice. Hypoxia is an important feature of the CRPC microenvironment and is closely associated with the progress of CRPC invasion. However, no research has been performed on the immune escape of CRPC from NK cells. The present study focused on this subject. Firstly, when the CRPC cell lines C4‑2 and CWR22Rv1 were induced by hypoxia, the expression of the UL16 binding protein (ULBP) ligand family of natural killer (NK) group 2D (NKG2D; ULBP‑1, ULBP‑2 and ULBP‑3) and MHC class I chain‑related proteins A and B (MICA/MICB) decreased. NKG2D is the main activating receptor of NK cells. Tumor cells were then co‑cultured with NK cells to conduct NK cell‑mediated cytotoxicity experiments, which revealed the decreased immune cytolytic activity of NK cells on hypoxia‑induced CRPC cells. In exploring the mechanism behind this observation, an increase in programmed death‑ligand 1 (PD‑L1) expression in CRPC cells induced by hypoxia was observed, while the addition of PD‑L1 antibody effectively reversed the expression of NKG2D ligand and enhanced the cytotoxic effect of NK cells on CRPC cells. In the process of exploring the upstream regulatory factors of PD‑L1, inhibition of the Janus kinase (JAK)1,2/signal transducer and activator of transcription 3 (Stat3) signaling pathway decreased the expression of PD‑L1 in CRPC cells. Finally, it was observed that combined inhibition of JAK1,2/PD‑L1 or Stat3/PD‑L1 was more effective than inhibition of a single pathway in enhancing the immune cytolytic activity of NK cells. Taking these results together, it is thought that combined inhibition of the JAK1,2/PD‑L1 and Stat3/PD‑L1 signaling pathways may enhance the immune cytolytic activity of NK cells toward hypoxia‑induced CRPC cells, which is expected to provide novel ideas and targets for the immunotherapy of CRPC.
Collapse
Affiliation(s)
- Li-Jun Xu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Qi Ma
- Department of Ultrasound, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jin Zhu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jian Li
- First Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Bo-Xin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Jie Gao
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Chuan-Yang Sun
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ya-Chen Zang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Yi-Bin Zhou
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Dong-Rong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Yu-Xi Shan
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
350
|
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
|