351
|
Abstract
Natural killer (NK) cells express an array of germ-line encoded receptors that are capable of triggering cytotoxicity. NK cells tend to express many members of a given family of signalling molecules. The presence of many activating receptors and many members of a given family of signalling molecules can enable NK cells to detect different kinds of target cells, and to mount different kinds of responses. This contributes also to the robustness of NK cells responses; cytotoxic functions of NK cells often remain unaffected in the absence of selected signalling molecules. NK cells express many MHC-I-specific inhibitory receptors. Signals from MHC-I-specific inhibitory receptors tightly control NK cell cytotoxicity and, paradoxically, maintain NK cells in a state of proper responsiveness. This review provides a brief overview of the events that underlie NK cell activation, and how signals from inhibitory receptors intercept NK cell activation to prevent inappropriate triggering of cytotoxicity.
Collapse
Affiliation(s)
- Santosh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
| |
Collapse
|
352
|
Dhar P, Wu JD. NKG2D and its ligands in cancer. Curr Opin Immunol 2018; 51:55-61. [PMID: 29525346 PMCID: PMC6145810 DOI: 10.1016/j.coi.2018.02.004] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/02/2018] [Accepted: 02/23/2018] [Indexed: 01/12/2023]
Abstract
NKG2D is an activating immune receptor expressed by NK and effector T cells. Induced expression of NKG2D ligand on tumor cell surface during oncogenic insults renders cancer cells susceptible to immune destruction. In advanced human cancers, tumor cells shed NKG2D ligand to produce an immune soluble form as a means of immune evasion. Soluble NKG2D ligands have been associated with poor clinical prognosis in cancer patients. Harnessing NKG2D pathway is considered a viable avenue in cancer immunotherapy over recent years. In this review, we will discuss the progress and perspectives.
Collapse
Affiliation(s)
- Payal Dhar
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago IL60611, United States; Driskill Graduate Program in Life Sciences, Feinberg School of Medicine, Chicago, Northwestern University, Chicago IL60611, United States
| | - Jennifer D Wu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago IL60611, United States; Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago IL60611, United States; Robert Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL60611, United States.
| |
Collapse
|
353
|
Ferrari de Andrade L, Tay RE, Pan D, Luoma AM, Ito Y, Badrinath S, Tsoucas D, Franz B, May KF, Harvey CJ, Kobold S, Pyrdol JW, Yoon C, Yuan GC, Hodi FS, Dranoff G, Wucherpfennig KW. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity. Science 2018; 359:1537-1542. [PMID: 29599246 PMCID: PMC6626532 DOI: 10.1126/science.aao0505] [Citation(s) in RCA: 333] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 12/18/2017] [Accepted: 02/01/2018] [Indexed: 12/15/2022]
Abstract
MICA and MICB are expressed by many human cancers as a result of cellular stress, and can tag cells for elimination by cytotoxic lymphocytes through natural killer group 2D (NKG2D) receptor activation. However, tumors evade this immune recognition pathway through proteolytic shedding of MICA and MICB proteins. We rationally designed antibodies targeting the MICA α3 domain, the site of proteolytic shedding, and found that these antibodies prevented loss of cell surface MICA and MICB by human cancer cells. These antibodies inhibited tumor growth in multiple fully immunocompetent mouse models and reduced human melanoma metastases in a humanized mouse model. Antitumor immunity was mediated mainly by natural killer (NK) cells through activation of NKG2D and CD16 Fc receptors. This approach prevents the loss of important immunostimulatory ligands by human cancers and reactivates antitumor immunity.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/immunology
- Antibodies, Blocking/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Histocompatibility Antigens Class I/chemistry
- Histocompatibility Antigens Class I/immunology
- Humans
- Immunocompetence
- Killer Cells, Natural/immunology
- Ligands
- Melanoma/immunology
- Melanoma/pathology
- Melanoma/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- NK Cell Lectin-Like Receptor Subfamily K/immunology
- Neoplasm Metastasis
- Protein Domains/immunology
- Receptors, IgG/immunology
Collapse
Affiliation(s)
- Lucas Ferrari de Andrade
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Rong En Tay
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Deng Pan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Adrienne M Luoma
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Yoshinaga Ito
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Soumya Badrinath
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Daphne Tsoucas
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Bettina Franz
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Kenneth F May
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Christopher J Harvey
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Sebastian Kobold
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Jason W Pyrdol
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Charles Yoon
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
- Department of Surgery, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
354
|
Affiliation(s)
- Adelheid Cerwenka
- Centre for Biomedicine and Medical Technology (CBTM) and European Center for Angioscience (ECAS), Heidelberg University, Medical Faculty Mannheim, Ludolf-Krehl-Strasse 13–17, D-68167 Mannheim, Germany
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
355
|
Zöller T, Wittenbrink M, Hoffmeister M, Steinle A. Cutting an NKG2D Ligand Short: Cellular Processing of the Peculiar Human NKG2D Ligand ULBP4. Front Immunol 2018; 9:620. [PMID: 29651291 PMCID: PMC5884875 DOI: 10.3389/fimmu.2018.00620] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Stress-induced cell surface expression of MHC class I-related glycoproteins of the MIC and ULBP families allows for immune recognition of dangerous "self cells" by human cytotoxic lymphocytes via the NKG2D receptor. With two MIC molecules (MICA and MICB) and six ULBP molecules (ULBP1-6), there are a total of eight human NKG2D ligands (NKG2DL). Since the discovery of the NKG2D-NKG2DL system, the cause for both redundancy and diversity of NKG2DL has been a major and ongoing matter of debate. NKG2DL diversity has been attributed, among others, to the selective pressure by viral immunoevasins, to diverse regulation of expression, to differential tissue expression as well as to variations in receptor interactions. Here, we critically review the current state of knowledge on the poorly studied human NKG2DL ULBP4. Summarizing available facts and previous studies, we picture ULBP4 as a peculiar ULBP family member distinct from other ULBP family members by various aspects. In addition, we provide novel experimental evidence suggesting that cellular processing gives rise to mature ULBP4 glycoproteins different to previous reports. Finally, we report on the proteolytic release of soluble ULBP4 and discuss these results in the light of known mechanisms for generation of soluble NKG2DL.
Collapse
Affiliation(s)
- Tobias Zöller
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Mareike Wittenbrink
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Meike Hoffmeister
- Institute of Biochemistry II, Goethe University Frankfurt am Main, Frankfurt am Main, Germany.,Brandenburg Medical School (MHB) Theodor Fontane, Institute of Biochemistry, Neuruppin, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
356
|
Stabile H, Fionda C, Santoni A, Gismondi A. Impact of bone marrow-derived signals on NK cell development and functional maturation. Cytokine Growth Factor Rev 2018; 42:13-19. [PMID: 29622473 DOI: 10.1016/j.cytogfr.2018.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/18/2018] [Accepted: 03/19/2018] [Indexed: 12/28/2022]
Abstract
Natural killer (NK) cells are cytotoxic members of type I innate lymphocytes (ILC1) with a prominent role in anti-tumor and anti-viral immune responses. Despite the increasing insight into NK cell biology, the steps and stages leading to mature circulating NK cells require further investigation. Natural killer cell development and functional maturation are complex and multi-stage processes that occur predominantly in the bone marrow (BM) and originate from haematopoietic stem cells CD34+ (HSC). Within the BM, NK cell precursor (NKP) and NK cell development intermediates reside in specialized niches that are characterized by particular cellular components that provide signals required for their maturation. These signals consist of soluble factors or direct cellular-contact interactions mediated by cytokines and growth factors with complementary, as well as overlapping roles in distinct developmental steps. Emerging evidence highlights the plasticity of the early phase of NK cell development, and the capacity of different signal combinations to redirect precursor lineage commitment through other innate cell populations. Here, we summarize the role of signals known to guide NK cell differentiation with a particular focus on the cytokines and the receptor/ligand pairs playing a critical role in these processes. A comprehensive understanding of the mechanisms underlying NK cell development will elucidate their roles in pathological conditions and will improve protocols for NK cell therapeutic application.
Collapse
Affiliation(s)
- Helena Stabile
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Institute Pasteur -Italia, 00161 Rome, Italy; IRCCS, Neuromed, Pozzilli, 86077 IS, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; Eleonora Lorillard Spencer Cenci Foundation, Italy
| |
Collapse
|
357
|
Transcriptomic signatures of NK cells suggest impaired responsiveness in HIV-1 infection and increased activity post-vaccination. Nat Commun 2018; 9:1212. [PMID: 29572470 PMCID: PMC5865158 DOI: 10.1038/s41467-018-03618-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/28/2018] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells limit viral replication by direct recognition of infected cells, antibody-dependent cellular cytotoxicity (ADCC), and releasing cytokines. Although growing evidence supports NK cell antiviral immunity in HIV-1 infection, further knowledge of their response is necessary. Here we show that NK cells responding to models of direct cell recognition, ADCC, and cytokine activation have unique transcriptional fingerprints. Compared with healthy volunteers, individuals with chronic HIV-1 infection have higher expression of genes commonly associated with activation, and lower expression of genes associated with direct cell recognition and cytokine stimulation in their NK cells. By contrast, NK cell transcriptional profiles of individuals receiving a modified vaccinia Ankara (MVA) vectored HIV-1 vaccine show upregulation of genes associated with direct cell recognition. These findings demonstrate that targeted transcriptional profiling provides a sensitive assessment of NK cell activity, which helps understand how NK cells respond to viral infections and vaccination. Natural killer (NK) cells are important for eliminating cells under stress or infected by virus, and may have a function in anti-HIV immunity. Here the authors show that different NK-activating stimuli induce distinct transcriptional fingerprints in human NK cells that are analogous to changes caused by HIV vaccination or chronic infection.
Collapse
|
358
|
Zekri ARN, El Deeb S, Bahnassy AA, Badr AM, Abdellateif MS, Esmat G, Salama H, Mohanad M, El-dien AE, Rabah S, Abd Elkader A. Role of relevant immune-modulators and cytokines in hepatocellular carcinoma and premalignant hepatic lesions. World J Gastroenterol 2018; 24:1228-1238. [PMID: 29568203 PMCID: PMC5859225 DOI: 10.3748/wjg.v24.i11.1228] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 12/24/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the levels of different immune modulators in patients with hepatocellular carcinoma (HCC), in relation to other hepatic diseases.
METHODS Eighty-eight patients were included in the current study and represented patients with HCC (20), liver cirrhosis (28) and chronic hepatitis (CH; 25), and normal controls (NC; 15). Peripheral blood was isolated for immunophenotyping of active myeloid dendritic cells (mDCs; CD1c and CD40), mature inactive myeloid cells (CD1c and HLA), active plasmacytoid cells (pDCs; CD303 and CD40), mature inactive pDCs (CD30 and HLA), active natural killer (NK) cells (CD56 and CD161), active NK cells (CD56 and CD314) and inactive NK cells (CD56 and CD158) was done by flow cytometry. Serum levels of interleukin (IL)-2, IL-10, IL-12, IL-1β, interferon (IFN)-α, IFN-γ and tumor necrosis factor (TNF)-αR2 were assessed by ELISA.
RESULTS Active mDCs (CD1C+/CD40+) and inactive mDCs (CD1c+/HLA+) were significantly decreased in HCC patients in relation to NC (P < 0.001). CD40+ expression on active pDCs was decreased in HCC patients (P < 0.001), and its level was not significantly changed among other groups. Inactive pDCs (CD303+/HLA+), inactive NKs (CD56+/CD158+) and active NKs (CD56+/CD161+) were not statistically changed among the four groups studied; however, the latter was increased in CH (P < 0.05). NKG2D was statistically decreased in HCC, CH and cirrhosis (P < 0.001), and it was not expressed in 63% (12/20) of HCC patients. There was significant decrease of IL-2, IFN-α and IFN-γ (P < 0.001), and a significant increase in IL-10, IL-1β, and TNF-αR2 (P <0.01, P < 0.001 and P < 0.001; respectively) in HCC patients. There was inverted correlation between IL-12 and IL-1β in HCC (r = -0.565, P < 0.01), with a strong correlation between pDCs (CD303+/CD40+) and NKs (CD56+/CD161+; r = 0.512, P < 0.05) as well as inactive mDCs (CD1c+/HLA+) and inactive NK cells (CD56+/CD158+; r = 0.945, P < 0.001).
CONCLUSION NKG2D, CD40, IL-2 and IL-10 are important modulators in the development and progression of HCC.
Collapse
Affiliation(s)
- Abdel-Rahman N Zekri
- Molecular Virology and Immunology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Somaya El Deeb
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Abeer A Bahnassy
- Department of Pathology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Abeer M Badr
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Gamal Esmat
- Department of Hepatology and Tropical Medicine, Faculty of Medicine, Cairo University, Cairo 11441, Egypt
| | - Hosny Salama
- Department of Hepatology and Tropical Medicine, Faculty of Medicine, Cairo University, Cairo 11441, Egypt
| | - Marwa Mohanad
- Department of Biochemistry, Misr University for Science and Technology, 6th October 12945, Giza Governorate, Egypt
| | - Ahmed Esam El-dien
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Shimaa Rabah
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Assmaa Abd Elkader
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
359
|
Schönrich G, Raftery MJ. CD1-Restricted T Cells During Persistent Virus Infections: "Sympathy for the Devil". Front Immunol 2018; 9:545. [PMID: 29616036 PMCID: PMC5868415 DOI: 10.3389/fimmu.2018.00545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/02/2018] [Indexed: 12/12/2022] Open
Abstract
Some of the clinically most important viruses persist in the human host after acute infection. In this situation, the host immune system and the viral pathogen attempt to establish an equilibrium. At best, overt disease is avoided. This attempt may fail, however, resulting in eventual loss of viral control or inadequate immune regulation. Consequently, direct virus-induced tissue damage or immunopathology may occur. The cluster of differentiation 1 (CD1) family of non-classical major histocompatibility complex class I molecules are known to present hydrophobic, primarily lipid antigens. There is ample evidence that both CD1-dependent and CD1-independent mechanisms activate CD1-restricted T cells during persistent virus infections. Sophisticated viral mechanisms subvert these immune responses and help the pathogens to avoid clearance from the host organism. CD1-restricted T cells are not only crucial for the antiviral host defense but may also contribute to tissue damage. This review highlights the two edged role of CD1-restricted T cells in persistent virus infections and summarizes the viral immune evasion mechanisms that target these fascinating immune cells.
Collapse
Affiliation(s)
- Günther Schönrich
- Berlin Institute of Health, Institute of Virology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin J Raftery
- Berlin Institute of Health, Institute of Virology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
360
|
Zingoni A, Molfetta R, Fionda C, Soriani A, Paolini R, Cippitelli M, Cerboni C, Santoni A. NKG2D and Its Ligands: "One for All, All for One". Front Immunol 2018; 9:476. [PMID: 29662484 PMCID: PMC5890157 DOI: 10.3389/fimmu.2018.00476] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/22/2018] [Indexed: 01/30/2023] Open
Abstract
The activating receptor NKG2D is peculiar in its capability to bind to numerous and highly diversified MHC class I-like self-molecules. These ligands are poorly expressed on normal cells but can be induced on damaged, transformed or infected cells, with the final NKG2D ligand expression resulting from multiple levels of regulation. Although redundant molecular mechanisms can converge in the regulation of all NKG2D ligands, different stimuli can induce specific cellular responses, leading to the expression of one or few ligands. A large body of evidence demonstrates that NK cell activation can be triggered by different NKG2D ligands, often expressed on the same cell, suggesting a functional redundancy of these molecules. However, since a number of evasion mechanisms can reduce membrane expression of these molecules both on virus-infected and tumor cells, the co-expression of different ligands and/or the presence of allelic forms of the same ligand guarantee NKG2D activation in various stressful conditions and cell contexts. Noteworthy, NKG2D ligands can differ in their ability to down-modulate NKG2D membrane expression in human NK cells supporting the idea that NKG2D transduces different signals upon binding various ligands. Moreover, whether proteolytically shed and exosome-associated soluble NKG2D ligands share with their membrane-bound counterparts the same ability to induce NKG2D-mediated signaling is still a matter of debate. Here, we will review recent studies on the NKG2D/NKG2D ligand biology to summarize and discuss the redundancy and/or diversity in ligand expression, regulation, and receptor specificity.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristina Cerboni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
361
|
Han S, Ding S, Miao X, Lin Z, Lu G, Xiao W, Ding Y, Qian L, Zhang Y, Jia X, Zhu G, Gong W. TGF-β1 expression in regulatory NK1.1 -CD4 +NKG2D + T cells dependents on the PI3K-p85α/JNK, NF-κB and STAT3 pathways. Am J Cancer Res 2018; 8:489-501. [PMID: 29637003 PMCID: PMC5883098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 02/25/2018] [Indexed: 06/08/2023] Open
Abstract
NK1.1-CD4+NKG2D+ cells exert their immune-regulatory function in tumor as an unconventional regulatory T cell subset through the production of TGF-β1; however, the molecular mechanisms involving with the activation of nuclear factors for TGF-β1 transcription remain unclear. Here we determined that the PI3K-p85α subunit was specifically activated in NK1.1-CD4+NKG2D+ cells following an 8-hour stimulation by sRAE-1 or α-CD3/sRAE-1, subsequently leading to the activation of PI3K-p110, Akt, and JNK. On the contrary, α-CD3/α-CD28 stimulation did not induce the activation of PI3K-p85 and JNK. Consequently, activation of the nuclear transcription factor AP-1 as a consequence of JNK activation regulated TGF-β1 expression in NK1.1-CD4+NKG2D+ cells. Furthermore, activation of NF-κB in NK1.1-CD4+NKG2D+ cells resulted from both protein kinase C activation downstream of TCR/CD3 signaling and PI3K activation induced by NKG2D engagement. The STAT3-Y705 phosphorylation, as activated by PI3K, under stimulations of the sRAE-1 or α-CD3/sRAE-1 also contributed to the TGF-β1 expression in NK1.1-CD4+NKG2D+ cells. Moreover, ChIP assay confirmed that STAT3 was capable of binding with the promoter regions of TGF-β1. In conclusion, our data showed that the TGF-β1 transcription in NK1.1-CD4+NKG2D+ cells induced by sRAE-1 or α-CD3/sRAE-1 was involved with the AP-1, NF-κB, and STAT3 signaling pathways; therefore, regulation of AP-1, NF-κB, and STAT3 activation may play important roles in the development and function of NK1.1-CD4+NKG2D+ cells.
Collapse
Affiliation(s)
- Sen Han
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Shizhen Ding
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Xin Miao
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Zhijie Lin
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Guotao Lu
- Department of Gastroenterology, The Affiliated Hospital, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Weiming Xiao
- Department of Gastroenterology, The Affiliated Hospital, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Yanbing Ding
- Department of Gastroenterology, The Affiliated Hospital, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Li Qian
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
| | - Yu Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesP. R. China
| | - Xiaoqin Jia
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesP. R. China
| | - Guoqiang Zhu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesP. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou UniversityYangzhou 225009, P. R. China
| | - Weijuan Gong
- Department of Immunology, School of Medicine, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
- Department of Gastroenterology, The Affiliated Hospital, Yangzhou UniversityYangzhou, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile DiseasesP. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesP. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou UniversityYangzhou 225009, P. R. China
| |
Collapse
|
362
|
Dukovska D, Fernández-Soto D, Valés-Gómez M, Reyburn HT. NKG2H-Expressing T Cells Negatively Regulate Immune Responses. Front Immunol 2018; 9:390. [PMID: 29545803 PMCID: PMC5837990 DOI: 10.3389/fimmu.2018.00390] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/12/2018] [Indexed: 01/03/2023] Open
Abstract
The biology and function of NKG2H receptor, unlike the better characterized members of the NKG2 family NKG2A, NKG2C, and NKG2D, remains largely unclear. Here, we show that NKG2H is able to associate with the signaling adapter molecules DAP12 and DAP10 suggesting that this receptor can signal for cell activation. Using a recently described NKG2H-specific monoclonal antibody (mAb), we have characterized the expression and function of lymphocytes that express this receptor. NKG2H is expressed at the cell surface of a small percentage of peripheral blood mononuclear cell (PBMC) and is found more frequently on T cells, rather than NK cells. Moreover, although NKG2H is likely to trigger activation, co-cross-linking of this receptor with an NKG2H-specific mAb led to decreased T cell activation and proliferation in polyclonal PBMC cultures stimulated by anti-CD3 mAbs. This negative regulatory activity was seen only after cross-linking with NKG2H, but not NKG2A- or NKG2C-specific monoclonal antibodies. The mechanism underlying this negative effect is as yet unclear, but did not depend on the release of soluble factors or recognition of MHC class I molecules. These observations raise the intriguing possibility that NKG2H may be a novel marker for T cells able to negatively regulate T cell responses.
Collapse
Affiliation(s)
- Daniela Dukovska
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| | - Daniel Fernández-Soto
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| | - Mar Valés-Gómez
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| | - Hugh T Reyburn
- Department of Immunology and Oncology, National Centre for Biotechnology, CSIC, Madrid, Spain
| |
Collapse
|
363
|
Harrer DC, Dörrie J, Schaft N. Chimeric Antigen Receptors in Different Cell Types: New Vehicles Join the Race. Hum Gene Ther 2018; 29:547-558. [PMID: 29320890 DOI: 10.1089/hum.2017.236] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adoptive cellular therapy has evolved into a powerful force in the battle against cancer, holding promise for curative responses in patients with advanced and refractory tumors. Autologous T cells, reprogrammed to target malignant cells via the expression of a chimeric antigen receptor (CAR) represent the frontrunner in this approach. Tremendous clinical regressions have been achieved using CAR-T cells against a variety of cancers both in numerous preclinical studies and in several clinical trials, most notably against acute lymphoblastic leukemia, and resulted in a very recent United States Food and Drug Administration approval of the first CAR-T-cell therapy. In most studies CARs are transferred to conventional αβT cells. Nevertheless, transferring a CAR into different cell types, such as γδT cells, natural killer cells, natural killer T cells, and myeloid cells has yet received relatively little attention, although these cell types possess unique features that may aid in surmounting some of the hurdles CAR-T-cell therapy currently faces. This review focuses on CAR therapy using effectors beyond conventional αβT cells and discusses those strategies against the backdrop of developing a safe, powerful, and durable cancer therapy.
Collapse
Affiliation(s)
- Dennis C Harrer
- 1 Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Jan Dörrie
- 1 Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Niels Schaft
- 1 Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| |
Collapse
|
364
|
Wang D, Gu X, Liu X, Wei S, Wang B, Fang M. NK cells inhibit anti-Mycobacterium bovis BCG T cell responses and aggravate pulmonary inflammation in a direct lung infection mouse model. Cell Microbiol 2018; 20:e12833. [PMID: 29447423 DOI: 10.1111/cmi.12833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/22/2018] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Tuberculosis remains a threat to public health. The major problem for curing this disease is latent infection, of which the underlying mechanisms are still not fully understood. Previous studies indicate that natural killer (NK) cells do not play a role in inhibiting the growth of Mycobacterium tuberculosis in the lung, and recent studies have revealed that NK cells regulate the adaptive immunity during mycobacterial infection. By using a mouse model of direct lung infection with Mycobacterium bovis bacillus Calmette-Guerin (BCG), we found that the presence of NK cells postponed the priming and activation of T cells after BCG infection. In addition, depletion of NK cells before infection alleviated pulmonary pathology. Further studies showed that NK cells lysed BCG-infected macrophages in an NKG2D dependent manner. Thus, NK cells did not play a direct role in control BCG, but aggravated the pulmonary inflammation and impaired anti-BCG T cell immunity, likely through killing BCG-infected macrophages. Our results may have important implications for the design of immune therapy to treat tuberculosis.
Collapse
Affiliation(s)
- Dongfang Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiuling Gu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoman Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Songtao Wei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Bin Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,International College, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
365
|
Trembath AP, Markiewicz MA. More than Decoration: Roles for Natural Killer Group 2 Member D Ligand Expression by Immune Cells. Front Immunol 2018; 9:231. [PMID: 29483917 PMCID: PMC5816059 DOI: 10.3389/fimmu.2018.00231] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/26/2018] [Indexed: 01/16/2023] Open
Abstract
The activating immune receptor natural killer group 2 member D (NKG2D), which is expressed by natural killer cells and T cell subsets, recognizes a number of ligands expressed by "stressed" or damaged cells. NKG2D has been extensively studied for its role in tumor immunosurveillance and antiviral immunity. To date, the majority of studies have focused on NKG2D-mediated killing of target cells expressing NKG2D ligands. However, with a number of reports describing expression of NKG2D ligands by cells that are not generally considered stressed, it is becoming clear that some healthy cells also express NKG2D ligands. Expression of these ligands by cells within the skin, intestinal epithelium, and the immune system suggests other immune functions for NKG2D ligand expression in addition to its canonical role as a "kill me" signal. How NKG2D ligands function in this capacity is just now starting to be unraveled. In this review, we examine the expression of NKG2D ligands by immune cells and discuss current literature describing the effects of this expression on immunity and immune regulation.
Collapse
Affiliation(s)
- Andrew P. Trembath
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, MO, United States
| | - Mary A. Markiewicz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, MO, United States
| |
Collapse
|
366
|
Espinoza JL, Minami M. Sensing Bacterial-Induced DNA Damaging Effects via Natural Killer Group 2 Member D Immune Receptor: From Dysbiosis to Autoimmunity and Carcinogenesis. Front Immunol 2018; 9:52. [PMID: 29422899 PMCID: PMC5788971 DOI: 10.3389/fimmu.2018.00052] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/09/2018] [Indexed: 12/23/2022] Open
Abstract
The human genome is constantly exposed to exogenous and endogenous DNA damaging factors that frequently cause DNA damages. Unless repaired, damaged DNA can result in deleterious mutations capable of causing malignant transformation. Accordingly, cells have developed an advanced and effective surveillance system, the DNA damage response (DDR) pathway, which maintains genetic integrity. In addition to well-defined outcomes, such as cell cycle arrest, apoptosis, and senescence, another consequence of DDR activation is the induction of natural killer group 2 member D ligands (NKG2D-Ls) on the surface of stressed cells. Consequently, NKG2D-Ls-expressing cells are recognized and eliminated by NKG2D receptor-expressing immune cells, including NK cells, and various subsets of T-cells. Recent pieces of evidence indicate that commensal microbial imbalance (known as dysbiosis) can trigger DDR activation in host cells, which may result in sustained inflammatory responses. Therefore, dysbiosis can be seen as an important source of DNA damage agents that may be partially responsible for the overexpression of NKG2D-Ls on intestinal epithelial cells that is frequently observed in patients with inflammatory bowel disease and other disorders associated with altered human microbiota, including the development of colorectal cancer. In this article, we discuss recent evidence that appears to link an altered human microbiota with autoimmunity and carcinogenesis via the activation of DDR signals and the induction of NKG2D-Ls in stressed cells.
Collapse
Affiliation(s)
- J Luis Espinoza
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Mika Minami
- Faculty of Medicine, Kindai University, Higashi-osaka, Japan
| |
Collapse
|
367
|
Guan Y, Li W, Hou Z, Han Q, Lan P, Zhang J, Tian Z, Zhang C. HBV suppresses expression of MICA/B on hepatoma cells through up-regulation of transcription factors GATA2 and GATA3 to escape from NK cell surveillance. Oncotarget 2018; 7:56107-56119. [PMID: 27528231 PMCID: PMC5302899 DOI: 10.18632/oncotarget.11271] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
Decreased expression of NKG2D ligands on HBV-infected human hepatoma cells impairs NK cells lysis. However, which components of HBV exert this effect and the precise mechanisms need to be further investigated. In the present study, we observed that the HBx and HBc genes significantly down-regulated MICA expression. Through analysis with the chromatin immunoprecipitation assay, we found that HBV infection promotes the expression of transcription factors GATA-2 and GATA-3, which specifically suppressed MICA/B expression by directly binding to the promoter region of MICA/B. HBx protein, acting as a co-regulator, forms a tripolymer with GATA2 and GATA3, thus promotes the GATA-2 or GATA-3-mediated of MICA/B suppression. HBc protein inhibits MICA/B expression via directly binding to the CpG island in the MICA/B promoter. Thus, our study identified the novel role of transcription factors GATA-2 and GATA-3 in suppressing MICA/B expression and clarified the mechanisms of HBx and HBc in downregulation of MICA/B expression. These findings provide novel mechanisms for the contribution of HBV to hepatoma cells escape from NK cell surveillance.
Collapse
Affiliation(s)
- Yun Guan
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Weiqun Li
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Zhaohua Hou
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Qiuju Han
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Peixiang Lan
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Jian Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Cai Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
368
|
Endogenous DAMPs, Category I: Constitutively Expressed, Native Molecules (Cat. I DAMPs). DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7122936 DOI: 10.1007/978-3-319-78655-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This chapter provides the reader with a collection of endogenous DAMPs in terms of constitutively expressed native molecules. The first class of this category refers to DAMPs, which are passively released from necrotic cells, and includes the most prominent subclasses of high mobility group box I and heat shock proteins. Further subclasses of DAMPs that are passively released from necrotic cells include S100 proteins, nucleic acids, histones, pro-forms of interleukin-1-family members, mitochondria-derived N-formylated peptides, F-actin, and heme. A particular subclass of these passively released DAMPs are molecules, which indirectly activate the inflammasome, including adenosine-5′-triphosphate, monosodium urate crystals, cholesterol crystals, some lipolytic species, and beta-amyloid. All these passively released DAMPs are characterized by their capability to promote necroinflammatory responses. The second class of this Category I refers to molecules, which are exposed on the surface of stressed cells. They include the subclass of phagocytosis-facilitating molecules such as calreticulin, as well as the subclass of MHC-I-related molecules such as MHC-I-related molecule A and B. These DAMPs are capable of inducing the activation of innate lymphoid cells and unconventional T cells. One of these DAMPs, the major histocompatibility complex I-related molecule A, is shown to act as a bona fide transplantation antigen. In sum, the endogenous constitutively expressed native molecules represent an impressive category of DAMPs with extraordinary properties, which play a critical role in the pathogenesis of many human diseases.
Collapse
|
369
|
Thompson TW, Kim AB, Li PJ, Wang J, Jackson BT, Huang KTH, Zhang L, Raulet DH. Endothelial cells express NKG2D ligands and desensitize antitumor NK responses. eLife 2017; 6:30881. [PMID: 29231815 PMCID: PMC5792093 DOI: 10.7554/elife.30881] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/11/2017] [Indexed: 12/15/2022] Open
Abstract
Natural Killer (NK) cells confer protection from tumors and infections by releasing cytotoxic granules and pro-inflammatory cytokines upon recognition of diseased cells. The responsiveness of NK cells to acute stimulation is dynamically tuned by steady-state receptor-ligand interactions of an NK cell with its cellular environment. Here, we demonstrate that in healthy WT mice the NK activating receptor NKG2D is engaged in vivo by one of its ligands, RAE-1ε, which is expressed constitutively by lymph node endothelial cells and highly induced on tumor-associated endothelium. This interaction causes internalization of NKG2D from the NK cell surface and transmits an NK-intrinsic signal that desensitizes NK cell responses globally to acute stimulation, resulting in impaired NK antitumor responses in vivo. White blood cells called “natural killer cells” are part of the first line of immune defense. Often called NK cells for short, one job of these cells is to help prevent cancer by killing tumor cells. If an NK cell spots a tumor cell, it must become energized so that it can deliver the killing blow, which comes in the form of a packet of cell-killing “cytotoxic” granules. Yet tumor cells look very similar to healthy cells, and NK cells must be able to tell the difference to be effective. Molecules on the outer surface of the NK cell control how the cell recognizes tumors, and deliver the signals the cell needs to become energized. One of these surface molecules is called NKG2D. It interacts with “partner” molecules found on the surface of cancer cells and tells the NK cell to attack. These partner molecules are not usually found on healthy cells, helping the immune system to tell the difference. After NKG2D interacts with its partner molecules, it moves inside the NK cell. This makes the cell less able to become energized. If the NK cells do not encounter any partner molecules in healthy mice, blocking the interactions should have no effect on NKG2D levels. But now, Thompson et al. find that blocking one of these interactions increased the levels of NKG2D on the surface of NK cells in healthy mice. Further experiments revealed that NK cells in mice constantly encounter an NKG2D partner molecule called RAE-1ε. A search for the source of RAE-1ε in healthy mice pointed to blood vessels inside the lymph nodes. NK cells pass through theses organs as part of their normal path around the body. Thompson et al. also saw that NK cells from healthy mice were less responsive than NK cells from mutant mice that lacked RAE-1ε. As a result of their encounters with RAE-1ε in healthy mice, the NK cells were less able to kill tumor cells. Blocking the interaction between NKG2D and RAE-1ε in mice re-energized their NK cells. More cells were able to enter tumors in these mice and the cells became better at killing tumors. Together these findings increase the current understanding of the biological processes that control NK cells. Further research may lead to new treatments for diseases like cancer. But first, scientists need to find out whether NK cells behave in the same way in humans as they do in mice. If so, developing ways to block the interaction could re-energize human NK cells to better kill cancer cells.
Collapse
Affiliation(s)
- Thornton W Thompson
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - Alexander Byungsuk Kim
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - P Jonathan Li
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - Jiaxi Wang
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - Benjamin T Jackson
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - Kristen Ting Hui Huang
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - Lily Zhang
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| | - David H Raulet
- Department of Molecular and Cell Biology, Cancer Research Laboratory, University of California, Berkeley, United States
| |
Collapse
|
370
|
Ren X, Xie W, Wang Y, Xu M, Liu F, Tang M, Li C, Wang M, Zhang J. VEGFR2-targeted fusion antibody improved NK cell-mediated immunosurveillance against K562 cells. Immunol Res 2017; 64:1060-70. [PMID: 27154226 DOI: 10.1007/s12026-016-8800-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MHC class I polypeptide-related sequence A (MICA), which is normally expressed on cancer cells, activates NK cells via NK group 2-member D pathway. However, some cancer cells escape NK-mediated immune surveillance by shedding membrane MICA causing immune suppression. To address this issue, we designed an antibody-MICA fusion targeting tumor-specific antigen (vascular endothelial growth factor receptor 2, VEGFR2) based on our patented antibody (mAb04) against VEGFR2. In vitro results demonstrate that the fusion antibody retains both the antineoplastic and the immunomodulatory activity of mAb04. Further, we revealed that it enhanced NK-mediated immunosurveillance against K562 cells through increasing degranulation and cytokine production of NK cells. The overall data suggest our new fusion protein provides a promising approach for cancer-targeted immunotherapy and has prospects for potential application of chronic myeloid leukemia.
Collapse
Affiliation(s)
- Xueyan Ren
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Wei Xie
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Youfu Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Menghuai Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Fang Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Mingying Tang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Chenchen Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China.
| | - Juan Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 154#, Tong Jia Xiang 24, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
371
|
Bazett M, Costa AM, Bosiljcic M, Anderson RM, Alexander MP, Wong SWY, Dhanji S, Chen JM, Pankovich J, Lam S, Sutcliffe S, Gunn H, Kalyan S, Mullins DW. Harnessing innate lung anti-cancer effector functions with a novel bacterial-derived immunotherapy. Oncoimmunology 2017; 7:e1398875. [PMID: 29399400 PMCID: PMC5790356 DOI: 10.1080/2162402x.2017.1398875] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/16/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023] Open
Abstract
Acute infection is known to induce strong anti-tumor immune responses, but clinical translation has been hindered by the lack of an effective strategy to safely and consistently provoke a therapeutic response. These limitations are overcome with a novel treatment approach involving repeated subcutaneous delivery of a Klebsiella-derived investigational immunotherapeutic, QBKPN. In preclinical models of lung cancer, QBKPN administration consistently showed anti-cancer efficacy, which was dependent on Klebsiella pre-exposure, but was independent of adaptive immunity. Rather, QBKPN induced anti-tumor innate immunity that required NK cells and NKG2D engagement. QBKPN increased NK cells and macrophages in the lungs, altered macrophage polarization, and augmented the production of cytotoxic molecules. An exploratory trial in patients with non-small cell lung cancer demonstrated QBKPN was well tolerated, safe, and induced peripheral immune changes suggestive of macrophage polarization and reduction of PD-1 and PD-L1 expression on leukocytes. These data demonstrate preclinical efficacy, and clinical safety and tolerability, for this cancer immunotherapy strategy that exploits innate anti-tumor immune mechanisms.
Collapse
Affiliation(s)
- Mark Bazett
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada
| | - Amanda M Costa
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | | | - Matthew P Alexander
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Stephanie W Y Wong
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Salim Dhanji
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada
| | | | | | - Stephen Lam
- BC Cancer Research Center, Vancouver, BC, Canada
| | | | - Hal Gunn
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada
| | - Shirin Kalyan
- Qu Biologics Inc., Vancouver, BC, V5 T 4T5, Canada.,Department of Medicine, Division of Endocrinology, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - David W Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Medical Education, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
372
|
Enk J, Levi A, Weisblum Y, Yamin R, Charpak-Amikam Y, Wolf DG, Mandelboim O. HSV1 MicroRNA Modulation of GPI Anchoring and Downstream Immune Evasion. Cell Rep 2017; 17:949-956. [PMID: 27760325 PMCID: PMC5081403 DOI: 10.1016/j.celrep.2016.09.077] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 06/28/2016] [Accepted: 09/22/2016] [Indexed: 12/24/2022] Open
Abstract
Herpes simplex virus 1 (HSV1) is a ubiquitous human pathogen that utilizes variable mechanisms to evade immune surveillance. The glycosylphosphatidylinositol (GPI) anchoring pathway is a multistep process in which a myriad of different proteins are covalently attached to a GPI moiety to be presented on the cell surface. Among the different GPI-anchored proteins there are many with immunological importance. We present evidence that the HSV1-encoded miR H8 directly targets PIGT, a member of the protein complex that covalently attaches proteins to GPI in the final step of GPI anchoring. This results in a membrane down-modulation of several different immune-related, GPI-anchored proteins, including ligands for natural killer-activating receptors and the prominent viral restriction factor tetherin. Thus, we suggest that by utilizing just one of dozens of miRNAs encoded by HSV1, the virus can counteract the host immune response at several key points. HSV1 miR H8 targets PIGT of the GPI anchoring pathway Expression of the anti-viral protein tetherin is reduced and viral spread enhanced Expression of GPI-anchored activating NK cell ligands is reduced Recognition and elimination by NK cells decrease
Collapse
Affiliation(s)
- Jonatan Enk
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Assi Levi
- Photodermatosis Clinic and Laser Unit, Dermatology Department, Rabin Medical Center, Petah Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yiska Weisblum
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; Department of Biochemistry and the Chanock Center for Virology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Rachel Yamin
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Yoav Charpak-Amikam
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Dana G Wolf
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Ofer Mandelboim
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel.
| |
Collapse
|
373
|
Subramanian N, Wu Z, Reister F, Sampaio KL, Frascaroli G, Cicin-Sain L, Mertens T. Naïve T cells are activated by autologous HCMV-infected endothelial cells through NKG2D and can control HCMV transmission in vitro. J Gen Virol 2017; 98:3068-3085. [PMID: 29165229 DOI: 10.1099/jgv.0.000976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Apart from classical antigen-presenting cells (APCs) like dendritic cells and macrophages, there are semiprofessional APCs such as endothelial cells (ECs) and Langerhans' cells. Human cytomegalovirus (HCMV) infects a wide range of cell types including the ECs which are involved in the trafficking and homing of T cells. By investigating the interaction of naïve T cells obtained from HCMV-seronegative umbilical cord blood with autologous HCMV-infected human umbilical vein ECs (HUVECs), we could show that the activation of naïve T cells occurred after 1 day of peripheral blood mononuclear cell (PBMC) exposure to HCMV-infected HUVECs. The percentage of activated T cells increased over time and the activation of naïve T cells was not induced by either autologous uninfected HUVECs or by autologous HCMV-infected fibroblasts. The activation of T cells occurred also when purified T cells were co-cultured with HCMV-infected HUVECs. In addition, in most of the donors only CD8+ T cells were activated, when the purified T cells were exposed to HCMV-infected HUVECs. The activation of naïve T cells was inhibited when the NKG2D receptor was blocked on the surface of T cells and among the different NKG2D ligands, we identified two ligands (ULBP4 and MICA) on HCMV-infected HUVECs which might be the interaction partners of the NKG2D receptor. Using a functional cell culture assay, we could show that these activated naïve T cells specifically inhibited HCMV transmission. Altogether, we identified a novel specific activation mechanism of naïve T cells from the umbilical cord by HCMV-infected autologous HUVECs through interaction with NKG2D.
Collapse
Affiliation(s)
| | - Zeguang Wu
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Frank Reister
- Gynecology and Obstetrics Clinics, Ulm University Hospital, Ulm, Germany
| | | | - Giada Frascaroli
- Institute of Virology, Ulm University Medical Center, Ulm, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Location Hannover-Braunschweig, Germany
| | - Thomas Mertens
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
374
|
Lonez C, Verma B, Hendlisz A, Aftimos P, Awada A, Van Den Neste E, Catala G, Machiels JPH, Piette F, Brayer JB, Sallman DA, Kerre T, Odunsi K, Davila ML, Gilham DE, Lehmann FF. Study protocol for THINK: a multinational open-label phase I study to assess the safety and clinical activity of multiple administrations of NKR-2 in patients with different metastatic tumour types. BMJ Open 2017; 7:e017075. [PMID: 29133316 PMCID: PMC5695348 DOI: 10.1136/bmjopen-2017-017075] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION NKR-2 are autologous T cells genetically modified to express a chimeric antigen receptor (CAR) comprising a fusion of the natural killer group 2D (NKG2D) receptor with the CD3ζ signalling domain, which associates with the adaptor molecule DNAX-activating protein of 10 kDa (DAP10) to provide co-stimulatory signal upon ligand binding. NKG2D binds eight different ligands expressed on the cell surface of many tumour cells and which are normally absent on non-neoplastic cells. In preclinical studies, NKR-2 demonstrated long-term antitumour activity towards a breadth of tumour indications, with maximum efficacy observed after multiple NKR-2 administrations. Importantly, NKR-2 targeted tumour cells and tumour neovasculature and the local tumour immunosuppressive microenvironment and this mechanism of action of NKR-2 was established in the absence of preconditioning. METHODS AND ANALYSIS This open-label phase I study will assess the safety and clinical activity of NKR-2 treatment administered three times, with a 2-week interval between each administration in different tumour types. The study will contain two consecutive segments: a dose escalation phase followed by an expansion phase. The dose escalation study involves two arms, one in solid tumours (five specific indications) and one in haematological tumours (two specific indications) and will include three dose levels in each arm: 3×108, 1×109 and 3×109 NKR-2 per injection. On the identification of the recommended dose in the first segment, based on dose-limiting toxicity occurrences, the study will expand to seven different cohorts examining the seven different tumour types separately. Clinical responses will be determined according to standard Response Evaluation Criteria In Solid Tumors (RECIST) criteria for solid tumours or international working group response criteria in haematological tumours. ETHICS APPROVAL AND DISSEMINATION Ethical approval has been obtained at all sites. Written informed consent will be taken from all participants. The results of this study will be disseminated through presentation at international scientific conferences and reported in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER NCT03018405, EudraCT 2016-003312-12; Pre-result.
Collapse
Affiliation(s)
| | | | - Alain Hendlisz
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Aftimos
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ahmad Awada
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Van Den Neste
- Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Gaetan Catala
- Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | - Fanny Piette
- International Drug Development Institute, Louvain-la-Neuve, Belgium
| | | | | | | | - Kunle Odunsi
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Marco L Davila
- H. Lee Moffitt Cancer Center, Tampa, Florida, USA
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | | |
Collapse
|
375
|
Hong CS, Sharma P, Yerneni SS, Simms P, Jackson EK, Whiteside TL, Boyiadzis M. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci Rep 2017; 7:14684. [PMID: 29089618 PMCID: PMC5666018 DOI: 10.1038/s41598-017-14661-w] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/13/2017] [Indexed: 12/31/2022] Open
Abstract
Exosomes, small (30–150 nm) extracellular vesicles (EVs) isolated from plasma of patients with acute myeloid leukemia (AML) carry leukemia-associated antigens and multiple inhibitory molecules. Circulating exosomes can deliver suppressive cargos to immune recipient cells, inhibiting anti-tumor activities. Pre-therapy plasma of refractory/relapsed AML patients contains elevated levels of immunosuppressive exosomes which interfere with anti-leukemia functions of activated immune cells. We show that exosomes isolated from pre-therapy plasma of the AML patients receiving adoptive NK-92 cell therapy block anti-leukemia cytotoxicity of NK-92 cells and other NK-92 cell functions. NK-92 cells do not internalize AML exosomes. Instead, signaling via surface receptors expressed on NK-92 cells, AML exosomes simultaneously deliver multiple inhibitory ligands to the cognate receptors. The signals are processed downstream and activate multiple suppressive pathways in NK-92 cells. AML exosomes reprogram NK-92 cells, interfering with their anti-leukemia functions and reducing the therapeutic potential of adoptive cell transfers. Plasma-derived exosomes interfere with immune cells used for adoptive cell therapy and may limit expected therapeutic benefits of adoptive cell therapy.
Collapse
Affiliation(s)
- Chang-Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Priyanka Sharma
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Patricia Simms
- FACS Core Facility, Loyola University School of Medicine, Maywood, IL, 60153, USA
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Michael Boyiadzis
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Medicine, Division of hematology-Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
376
|
Duggan MC, Campbell AR, McMichael EL, Opheim KS, Levine KM, Bhave N, Culbertson MC, Noel T, Yu L, Carson WE. Co-stimulation of the fc receptor and interleukin-12 receptor on human natural killer cells leads to increased expression of cd25. Oncoimmunology 2017; 7:e1381813. [PMID: 29308301 DOI: 10.1080/2162402x.2017.1381813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/31/2017] [Accepted: 09/14/2017] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells serve a critical role in the immune response against microbes and developing tumors. We have demonstrated that NK cells produce stimulatory cytokines (e.g., IFN-γ) in response to potent stimulation via immobilized IgG (to engage Fc receptors) and interleukin (IL)-12. CD25 is a component of the high-affinity IL-2R, which promotes NK cell activation in response to low doses of IL-2 such as those released by activated T cells. We hypothesized that stimulation of NK cells via IgG and IL-12 would enhance CD25 expression and promote NK cell anti-tumor activity in response to low-dose IL-2. It was confirmed that this dual stimulation strategy significantly enhanced NK cell CD25 expression compared to unstimulated cells or cells treated with IgG or IL-12 alone. Dual stimulated NK cells also were more responsive to low-dose IL-2. Dual stimulated NK cells subsequently treated with low-dose IL-2 (10 pg/mL) displayed enhanced intracellular signaling as indicated by increased pSTAT5 levels. IFN-γ production and cytotoxicity against K562 cells by NK cells stimulated with low-dose IL-2 was comparable to that of cells treated with high-dose IL-2 (10 ng/mL). Importantly, cells isolated from head and neck cancer patients receiving the mAb cetuximab and IL-12 on a clinical trial displayed increased CD25 expression following combination therapy compared to baseline. Altogether, these findings suggest that FcR and IL-12R co-stimulation induces expression of the high-affinity IL-2R and promotes NK cell anti-tumor activity.
Collapse
Affiliation(s)
- Megan C Duggan
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Amanda R Campbell
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH.,Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH
| | - Elizabeth L McMichael
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Kallan S Opheim
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Kala M Levine
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Neela Bhave
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Michelle C Culbertson
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Tiffany Noel
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, OH
| | - W E Carson
- Comprehensive Cancer Center, The Arthur G. James Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH.,Department of Surgery, The Ohio State University, Columbus, OH
| |
Collapse
|
377
|
Anti-NKG2D mAb: A New Treatment for Crohn's Disease? Int J Mol Sci 2017; 18:ijms18091997. [PMID: 28926962 PMCID: PMC5618646 DOI: 10.3390/ijms18091997] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 01/09/2023] Open
Abstract
Crohn’s disease (CD) and ulcerative colitis (UC) are immunologically-mediated, debilitating conditions resulting from destructive inflammation of the gastrointestinal tract. The pathogenesis of IBD is incompletely understood, but is considered to be the result of an abnormal immune response with a wide range of cell types and proteins involved. Natural Killer Group 2D (NKG2D) is an activating receptor constitutively expressed on human Natural Killer (NK), γδ T, mucosal-associated invariant T (MAIT), CD56+ T, and CD8+ T cells. Activation of NKG2D triggers cellular proliferation, cytokine production, and target cell killing. Research into the NKG2D mechanism of action has primarily been focused on cancer and viral infections where cytotoxicity evasion is a concern. In human inflammatory bowel disease (IBD) this system is less characterized, but the ligands have been shown to be highly expressed during intestinal inflammation and the following receptor activation may contribute to tissue degeneration. A recent phase II clinical trial showed that an antibody against NKG2D induced clinical remission of CD in some patients, suggesting NKG2D and its ligands to be of importance in the pathogenesis of CD. This review will describe the receptor and its ligands in intestinal tissues and the clinical potential of blocking NKG2D in Crohn’s disease.
Collapse
|
378
|
Abstract
PURPOSE OF REVIEW This review article discusses current knowledge on natural killer (NK) cells in asthma. RECENT FINDINGS It is now well accepted that NK cell activities go beyond cancer immune surveillance and antiviral defense. Recent reports indicate that NK cells are activated in response to allergens in vivo. NK cells promote allergic sensitization, type-2 immune response, development of eosinophilic inflammation, and airway hyperresponsiveness. NK cells are activated by respiratory syncytial virus and other respiratory viruses. When infection occurs in the setting of active allergic inflammation, NK cells augment its magnitude and contribute to asthma exacerbations. Proasthma activities of NK cells can be programmed during embryogenesis through maternal exposure to environmental pollutants. Prenatally programmed NK cells produce type-2 and type-3 cytokines and mediate asthma predisposition. NK cells can also act as asthma antagonists. NK cells contribute to the resolution of inflammation through suppression of antigen-specific CD4+ T cells and type-3 immunity. When viral infection occurs in naïve mice prior to allergic sensitization, NK cells antagonize type-2 immunity and prevent development of asthma. SUMMARY NK cells are nonredundant participants of allergic inflammation. The environmental context determines whether NK cells act as protagonists or antagonists.
Collapse
Affiliation(s)
- Magdalena M Gorska
- aNational Jewish Health, Denver bUniversity of Colorado, Aurora, Colorado, USA
| |
Collapse
|
379
|
Sharma N, Trinidad CV, Trembath AP, Markiewicz MA. NKG2D Signaling between Human NK Cells Enhances TACE-Mediated TNF-α Release. THE JOURNAL OF IMMUNOLOGY 2017; 199:2865-2872. [PMID: 28893955 DOI: 10.4049/jimmunol.1700647] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/15/2017] [Indexed: 12/22/2022]
Abstract
NK group 2 member D (NKG2D) is a strong NK cell-activating receptor, with engagement by ligands triggering granule release and cytokine production. The function of NKG2D signaling in NK cells has largely been studied in the context of engagement of the receptor by ligands expressed on the surface of target cells. We report that upon activation with IL-12, IL-15, and IL-18 human NK cells express NKG2D ligands of the UL16 binding protein family on the cell surface. NKG2D-ligand interaction between cytokine-stimulated NK cells increases the activity of the metalloprotease TNF-α-converting enzyme. This enhanced TNF-α-converting enzyme activity significantly increases the release of TNF-α and UL16 binding protein from the surface of the NK cells. These results demonstrate that NKG2D signaling is critical for maximal TNF-α release by NK cells. Further, they demonstrate a role for NKG2D-ligand interaction via homotypic NK cell contact in NK cell effector function.
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Camille V Trinidad
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Andrew P Trembath
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
380
|
Chung BK, Karlsen TH, Folseraas T. Cholangiocytes in the pathogenesis of primary sclerosing cholangitis and development of cholangiocarcinoma. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1390-1400. [PMID: 28844951 DOI: 10.1016/j.bbadis.2017.08.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/16/2017] [Accepted: 08/21/2017] [Indexed: 12/15/2022]
Abstract
Primary sclerosing cholangitis (PSC) is an idiopathic cholangiopathy strongly associated with inflammatory bowel disease (IBD) and characterized by cholestasis, chronic immune infiltration and progressive fibrosis of the intrahepatic and extrahepatic bile ducts. PSC confers a high risk of cholangiocarcinoma (CCA) with PSC-CCA representing the leading cause of PSC-associated mortality. PSC-CCA is derived from cholangiocytes and associated progenitor cells - a heterogeneous group of dynamic epithelial cells lining the biliary tree that modulate the composition and volume of bile production by the liver. Infection, inflammation and cholestasis can trigger cholangiocyte activation leading to an increased expression of adhesion and antigen-presenting molecules as well as the release of various inflammatory and fibrogenic mediators. As a result, activated cholangiocytes engage in a myriad of cellular processes, including hepatocellular proliferation, apoptosis, angiogenesis and fibrosis. Cholangiocytes can also regulate the recruitment of immune cells, mesenchymal cells, and endothelial cells that participate in tissue repair and destruction in settings of persistent inflammation. In PSC, the role of cholangiocytes and the mechanisms governing their transformation to PSC-CCA are unclear however localization of disease suggests that cholangiocytes are a key target and potential regulator of hepatobiliary immunity, fibrogenesis and tumorigenesis. Herein, we summarize mechanisms of cholangiocyte activation in PSC and highlight new insights into disease pathways that may contribute to the development of PSC-CCA. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Collapse
Affiliation(s)
- Brian K Chung
- Centre for Liver Research and NIHR Birmingham Inflammation Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| | - Tom Hemming Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trine Folseraas
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
381
|
Vyas M, Reinartz S, Hoffmann N, Reiners KS, Lieber S, Jansen JM, Wagner U, Müller R, von Strandmann EP. Soluble NKG2D ligands in the ovarian cancer microenvironment are associated with an adverse clinical outcome and decreased memory effector T cells independent of NKG2D downregulation. Oncoimmunology 2017; 6:e1339854. [PMID: 28932639 PMCID: PMC5599084 DOI: 10.1080/2162402x.2017.1339854] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
The immune receptor NKG2D is predominantly expressed on NK cells and T cell subsets and confers anti-tumor activity. According to the current paradigm, immune surveillance is counteracted by soluble ligands shed into the microenvironment, which down-regulate NKG2D receptor expression. Here, we analyzed the clinical significance of the soluble NKG2D ligands sMICA and sULBP2 in the malignancy-associated ascites of ovarian cancer. We show that high levels of sMICA and sULBP2 in ascites were associated with a poor prognosis. Ascites inhibited the activation of normal NK cells, which, in contrast to the prevailing notion, was not associated with decreased NKG2D expression. Of note, an inverse correlation of soluble NKG2D ligands with effector memory T cells and a direct correlation with pro-tumorigenic CD163+CD206+ macrophages was observed. Thus, the role of soluble NKG2D ligands within the ovarian cancer microenvironment is more complex than anticipated and does not exclusively function via NKG2D downregulation.
Collapse
Affiliation(s)
- Maulik Vyas
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecologic Oncology and Endocrinology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Nathalie Hoffmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany.,Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Katrin S Reiners
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Julia M Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
382
|
Jensen H, Potempa M, Gotthardt D, Lanier LL. Cutting Edge: IL-2-Induced Expression of the Amino Acid Transporters SLC1A5 and CD98 Is a Prerequisite for NKG2D-Mediated Activation of Human NK Cells. THE JOURNAL OF IMMUNOLOGY 2017; 199:1967-1972. [PMID: 28784848 DOI: 10.4049/jimmunol.1700497] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/20/2017] [Indexed: 01/03/2023]
Abstract
Priming of human NK cells with IL-2 is necessary to render them functionally competent upon NKG2D engagement. We examined the underlying mechanisms that control NKG2D responsiveness in NK cells and found that IL-2 upregulates expression of the amino acid transporters SLC1A5 and CD98. Using specific inhibitors to block SLC1A5 and CD98 function, we found that production of IFN-γ and degranulation by CD56bright and CD56dim NK cells following NKG2D stimulation were dependent on both transporters. IL-2 priming increased the activity of mTORC1, and inhibition of mTORC1 abrogated the ability of the IL-2-primed NK cells to produce IFN-γ in response to NKG2D-mediated stimulation. This study identifies a series of IL-2-induced cellular changes that regulates the NKG2D responsiveness in human NK cells.
Collapse
Affiliation(s)
- Helle Jensen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143; and
| | - Marc Potempa
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143; and
| | - Dagmar Gotthardt
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143; and.,Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143; and .,Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143
| |
Collapse
|
383
|
Berraondo P, Minute L, Ajona D, Corrales L, Melero I, Pio R. Innate immune mediators in cancer: between defense and resistance. Immunol Rev 2017; 274:290-306. [PMID: 27782320 DOI: 10.1111/imr.12464] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic inflammation in the tumor microenvironment and evasion of the antitumor effector immune response are two of the emerging hallmarks required for oncogenesis and cancer progression. The innate immune system not only plays a critical role in perpetuating these tumor-promoting hallmarks but also in developing antitumor adaptive immune responses. Thus, understanding the dual role of the innate system in cancer immunology is required for the design of combined immunotherapy strategies able to tackle established tumors. Here, we review recent advances in the understanding of the role of cell populations and soluble components of the innate immune system in cancer, with a focus on complement, the adapter molecule Stimulator of Interferon Genes, natural killer cells, myeloid cells, and B cells.
Collapse
Affiliation(s)
- Pedro Berraondo
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Luna Minute
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Daniel Ajona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain.,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Ruben Pio
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain. .,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain. .,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
384
|
Molfetta R, Quatrini L, Santoni A, Paolini R. Regulation of NKG2D-Dependent NK Cell Functions: The Yin and the Yang of Receptor Endocytosis. Int J Mol Sci 2017; 18:ijms18081677. [PMID: 28767057 PMCID: PMC5578067 DOI: 10.3390/ijms18081677] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 07/27/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022] Open
Abstract
Natural-killer receptor group 2, member D (NKG2D) is a well characterized natural killer (NK) cell activating receptor that recognizes several ligands poorly expressed on healthy cells but up-regulated upon stressing stimuli in the context of cancer or viral infection. Although NKG2D ligands represent danger signals that render target cells more susceptible to NK cell lysis, accumulating evidence demonstrates that persistent exposure to ligand-expressing cells causes the decrease of NKG2D surface expression leading to a functional impairment of NKG2D-dependent NK cell functions. Upon ligand binding, NKG2D is internalized from the plasma membrane and sorted to lysosomes for degradation. However, receptor endocytosis is not only a mechanism of receptor clearance from the cell surface, but is also required for the proper activation of signalling events leading to the functional program of NK cells. This review is aimed at providing a summary of current literature relevant to the molecular mechanisms leading to NKG2D down-modulation with particular emphasis given to the role of NKG2D endocytosis in both receptor degradation and signal propagation. Examples of chronic ligand-induced down-regulation of NK cell activating receptors other than NKG2D, including natural cytotoxicity receptors (NCRs), DNAX accessory molecule-1 (DNAM1) and CD16, will be also discussed.
Collapse
Affiliation(s)
- Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy.
| | - Linda Quatrini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy.
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France.
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy.
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy.
| |
Collapse
|
385
|
Mirjačić Martinović KM, Vuletić AM, Lj. Babović N, Džodić RR, Konjević GM, Jurišić VB. Attenuated in vitro effects of IFN-α, IL-2 and IL-12 on functional and receptor characteristics of peripheral blood lymphocytes in metastatic melanoma patients. Cytokine 2017; 96:30-40. [DOI: 10.1016/j.cyto.2017.02.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 02/20/2017] [Accepted: 02/24/2017] [Indexed: 11/24/2022]
|
386
|
Nabekura T, Gotthardt D, Niizuma K, Trsan T, Jenus T, Jonjic S, Lanier LL. Cutting Edge: NKG2D Signaling Enhances NK Cell Responses but Alone Is Insufficient To Drive Expansion during Mouse Cytomegalovirus Infection. THE JOURNAL OF IMMUNOLOGY 2017; 199:1567-1571. [PMID: 28760883 DOI: 10.4049/jimmunol.1700799] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/12/2017] [Indexed: 12/25/2022]
Abstract
NK cells play a critical role in host defense against viruses. In this study, we investigated the role of NKG2D in the expansion of NK cells after mouse CMV (MCMV) infection. Wild-type and NKG2D-deficient (Klrk1-/- ) Ly49H+ NK cells proliferated robustly when infected with MCMV strains engineered to allow expression of NKG2D ligands, which enhanced the response of wild-type NK cells. Naive NK cells exclusively express NKG2D-L, which pairs only with DAP10, whereas NKG2D-S expressed by activated NK cells pairs with DAP10 and DAP12, similar to Ly49H. However, NKG2D alone was unable to drive robust expansion of Ly49H- NK cells when mice were infected with these MCMV strains, likely because NKG2D-S was only transiently expressed postinfection. These findings demonstrate that NKG2D augments Ly49H-dependent proliferation of NK cells; however, NKG2D signaling alone is inadequate for expansion of NK cells, likely due to only transient expression of the NKG2D-DAP12 complex.
Collapse
Affiliation(s)
- Tsukasa Nabekura
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143.,Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143.,Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Dagmar Gotthardt
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143.,Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143
| | - Kouta Niizuma
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki 305-8575, Japan; and
| | - Tihana Trsan
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Tina Jenus
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143; .,Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143
| |
Collapse
|
387
|
Hosomi S, Grootjans J, Tschurtschenthaler M, Krupka N, Matute JD, Flak MB, Martinez-Naves E, Gomez Del Moral M, Glickman JN, Ohira M, Lanier LL, Kaser A, Blumberg R. Intestinal epithelial cell endoplasmic reticulum stress promotes MULT1 up-regulation and NKG2D-mediated inflammation. J Exp Med 2017; 214:2985-2997. [PMID: 28747426 PMCID: PMC5626394 DOI: 10.1084/jem.20162041] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 05/25/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022] Open
Abstract
Hosomi et al. show that intestinal epithelial cell–specific deletion of X-box–binding protein 1, an unfolded protein response–related transcription factor, results in CHOP-dependent increased expression of specific natural killer group 2 member D (NKG2D) ligands. This activates NKG2D-expressing intraepithelial group 1 ILCs and promotes small intestinal inflammation. Endoplasmic reticulum (ER) stress is commonly observed in intestinal epithelial cells (IECs) and can, if excessive, cause spontaneous intestinal inflammation as shown by mice with IEC-specific deletion of X-box–binding protein 1 (Xbp1), an unfolded protein response–related transcription factor. In this study, Xbp1 deletion in the epithelium (Xbp1ΔIEC) is shown to cause increased expression of natural killer group 2 member D (NKG2D) ligand (NKG2DL) mouse UL16-binding protein (ULBP)–like transcript 1 and its human orthologue cytomegalovirus ULBP via ER stress–related transcription factor C/EBP homology protein. Increased NKG2DL expression on mouse IECs is associated with increased numbers of intraepithelial NKG2D-expressing group 1 innate lymphoid cells (ILCs; NK cells or ILC1). Blockade of NKG2D suppresses cytolysis against ER-stressed epithelial cells in vitro and spontaneous enteritis in vivo. Pharmacological depletion of NK1.1+ cells also significantly improved enteritis, whereas enteritis was not ameliorated in Recombinase activating gene 1−/−;Xbp1ΔIEC mice. These experiments reveal innate immune sensing of ER stress in IECs as an important mechanism of intestinal inflammation.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Joep Grootjans
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Markus Tschurtschenthaler
- Department of Medicine, Division of Gastroenterology, University of Cambridge, Cambridge, England, UK
| | - Niklas Krupka
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Juan D Matute
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Magdalena B Flak
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Eduardo Martinez-Naves
- Department of Microbiology and Immunology, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Gomez Del Moral
- Department of Cell Biology, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Mizuki Ohira
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA
| | - Arthur Kaser
- Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Richard Blumberg
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
388
|
Chitadze G, Oberg HH, Wesch D, Kabelitz D. The Ambiguous Role of γδ T Lymphocytes in Antitumor Immunity. Trends Immunol 2017; 38:668-678. [PMID: 28709825 DOI: 10.1016/j.it.2017.06.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022]
Abstract
γδ T cells play a role in immune surveillance because they recognize stress-induced surface molecules and metabolic intermediates that are frequently dysregulated in transformed cells. Hence, γδ T cells have attracted much interest as effector cells in cell-based immunotherapy. Recently, however, it has been realized that γδ T cells can also promote tumorigenesis through various mechanisms including regulatory activity and IL-17 production. In this review we outline both the pathways involved in cancer cell recognition and killing by γδ T cells as well as current evidence for their protumorigenic activity in various models. Finally, we discuss strategies to improve the tumor reactivity of γδ T cells and to counteract their protumorigenic activities, which should open improved perspectives for their clinical application.
Collapse
Affiliation(s)
- Guranda Chitadze
- Institute of Immunology, University of Kiel, Arnold-Heller-Strasse 3, Building 17, 24105 Kiel, Germany
| | - Hans-Heinrich Oberg
- Institute of Immunology, University of Kiel, Arnold-Heller-Strasse 3, Building 17, 24105 Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology, University of Kiel, Arnold-Heller-Strasse 3, Building 17, 24105 Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, University of Kiel, Arnold-Heller-Strasse 3, Building 17, 24105 Kiel, Germany.
| |
Collapse
|
389
|
Anti-cancer efficacy of biotinylated chitosan nanoparticles in liver cancer. Oncotarget 2017; 8:59068-59085. [PMID: 28938619 PMCID: PMC5601715 DOI: 10.18632/oncotarget.19146] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 06/02/2017] [Indexed: 01/13/2023] Open
Abstract
The present study investigated the synthesis of biotinylated chitosan (Bio-CS) from chitosan using a nanomaterial skeleton with biotin and the successful targeting of the formulation in liver cancer cells. Bio-CS was validated by fourier transformed infrared spectroscopy and hydrogen-1 nuclear magnetic resonance spectroscopy. Bio-CS and plasmid DNA were used to construct Bio-CS/plasmid DNA nanoparticles according to the optimal molar ratio of 1:1 and the optimal pH-value of 5.5. Under these conditions, the parameters mean particle size, potential, encapsulation rate and drug loading, were 82.9 nm, +21.8 mV, 85.7% and 35.4%, respectively. Bio-CS exhibited an apparent liver cancer targeting effect in vitro and in vivo, as demonstrated by confocal laser scanning, green fluorescent protein transfection, and in vivo imaging assays. In addition, the Bio-CS/plasmid DNA nanoparticles significantly increased the survival period of the orthotropic liver cancer mouse model compared with the plasmid DNA, with no apparent side effects on the cells. Bio-CS nanomaterials stimulated an immune response in hepatoma cells via increased expression of GM-CSF, IL-21 and Rae-1 markers. The data suggest that Bio-CS increased the inhibition of liver cancer cell proliferation in vitro and the activation of the cellular immunity in vivo.
Collapse
|
390
|
Zumwalde NA, Sharma A, Xu X, Ma S, Schneider CL, Romero-Masters JC, Hudson AW, Gendron-Fitzpatrick A, Kenney SC, Gumperz JE. Adoptively transferred Vγ9Vδ2 T cells show potent antitumor effects in a preclinical B cell lymphomagenesis model. JCI Insight 2017; 2:93179. [PMID: 28679955 DOI: 10.1172/jci.insight.93179] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/31/2017] [Indexed: 01/09/2023] Open
Abstract
A central issue for adoptive cellular immunotherapy is overcoming immunosuppressive signals to achieve tumor clearance. While γδ T cells are known to be potent cytolytic effectors that can kill a variety of cancers, it is not clear whether they are inhibited by suppressive ligands expressed in tumor microenvironments. Here, we have used a powerful preclinical model where EBV infection drives the de novo generation of human B cell lymphomas in vivo, and autologous T lymphocytes are held in check by PD-1/CTLA-4-mediated inhibition. We show that a single dose of adoptively transferred Vδ2+ T cells has potent antitumor effects, even in the absence of checkpoint blockade or activating compounds. Vδ2+ T cell immunotherapy given within the first 5 days of EBV infection almost completely prevented the outgrowth of tumors. Vδ2+ T cell immunotherapy given more than 3 weeks after infection (after neoplastic transformation is evident) resulted in a dramatic reduction in tumor burden. The immunotherapeutic Vδ2+ T cells maintained low cell surface expression of PD-1 in vivo, and their recruitment to tumors was followed by a decrease in B cells expressing PD-L1 and PD-L2 inhibitory ligands. These results suggest that adoptively transferred PD-1lo Vδ2+ T cells circumvent the tumor checkpoint environment in vivo.
Collapse
Affiliation(s)
| | | | - Xuequn Xu
- Department of Medical Microbiology and Immunology
| | - Shidong Ma
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Christine L Schneider
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - James C Romero-Masters
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Amy W Hudson
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Annette Gendron-Fitzpatrick
- Comparative Pathology Laboratory, Research Animal Resources Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Shannon C Kenney
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | | |
Collapse
|
391
|
Vadstrup K, Galsgaard ED, Jensen H, Lanier LL, Ryan JC, Chen SY, Nolan GP, Vester-Andersen MK, Pedersen JS, Gerwien J, Jensen T, Bendtsen F. NKG2D ligand expression in Crohn's disease and NKG2D-dependent stimulation of CD8 + T cell migration. Exp Mol Pathol 2017; 103:56-70. [PMID: 28684217 DOI: 10.1016/j.yexmp.2017.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 05/24/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022]
Abstract
Interaction between the activating NKG2D receptor on lymphocytes and its ligands MICA, MICB, and ULBP1-6 modulate T and NK cell activity and may contribute to the pathogenesis of Crohn's disease (CD). NKG2D ligands are generally not expressed on the cell surface of normal, non-stressed cells, but expression of MICA and MICB in CD intestine has been reported. In this exploratory study, we further characterize the expression of NKG2D and its ligands, including the less well-described ULBP4-6, in CD, and test if NKG2D ligand interactions are involved in the migration of activated T cells into the affected mucosal compartments. Intestinal tissue from CD patients and healthy controls were analyzed by flow cytometry, mass cytometry, and immunohistochemistry for expression of NKG2D and ligands, and for cytokine release. Furthermore, NKG2D-dependent chemotaxis of activated CD8+ T cells across a monolayer of ligand-expressing human intestinal endothelial cells was examined. Activated lymphocytes down-regulated NKG2D expression upon accumulation in inflamed CD intestine. NKG2D expression on CD56+ T and γδ T cells from inflamed tissue seemed inversely correlated with CRP levels and cytokine release. B cells, monocytes, mucosal epithelium, and vascular endothelium expressed NKG2D ligands in inflamed CD intestine. The expression of NKG2D ligands was correlated with cytokine release, but was highly variable between patients. Stimulation of vascular intestinal endothelial cells in vitro induced expression of NKG2D ligands, including MICA/B and ULBP2/6. Blockade of NKG2D on CD8+ T cells inhibited the migration over ligand-expressing endothelial cells. Intestinal induction of NKG2D ligands and ligand-induced down-regulation of NKG2D in CD suggest that the NKG2D-ligand interaction may be involved in both the activation and recruitment of NKG2D+ lymphocytes into the inflamed CD intestine.
Collapse
Affiliation(s)
- Kasper Vadstrup
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark; Faculty of Health Sciences, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark; Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | - Helle Jensen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James C Ryan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, Veterans Affairs Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Shih-Yu Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | | | - Julie Steen Pedersen
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark
| | - Jens Gerwien
- Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark
| | - Teis Jensen
- Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark; Faculty of Health Sciences, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
392
|
Nakajima NI, Niimi A, Isono M, Oike T, Sato H, Nakano T, Shibata A. Inhibition of the HDAC/Suv39/G9a pathway restores the expression of DNA damage-dependent major histocompatibility complex class I-related chain A and B in cancer cells. Oncol Rep 2017; 38:693-702. [PMID: 28677817 PMCID: PMC5562072 DOI: 10.3892/or.2017.5773] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 05/05/2017] [Indexed: 12/29/2022] Open
Abstract
Immunotherapy is expected to be promising as a next generation cancer therapy. Immunoreceptors are often activated constitutively in cancer cells, however, such levels of ligand expression are not effectively recognized by the native immune system due to tumor microenvironmental adaptation. Studies have demonstrated that natural-killer group 2, member D (NKG2D), a major activating immunoreceptor, responds to DNA damage. The upregulation of major histocompatibility complex class I-related chain A and B (MICA/B) (members of NKG2D ligands) expression after DNA damage is associated with NK cell-mediated killing of cancer cells. However, the regulation of DNA damage-induced MICA/B expression has not been fully elucidated in the context of the types of cancer cell lines. In the present study, we found that MICA/B expression varied between cancer cell lines after DNA damage. Screening in terms of chromatin remodeling identified that inhibitors related to chromatin relaxation via post-translational modification on histone H3K9, i.e. HDAC, Suv39 or G9a inhibition, restored DNA damage-dependent MICA/B expression in insensitive cells. In addition, we revealed that the restored MICA/B expression was dependent on ATR as well as E2F1, a transcription factor. We further revealed that low‑dose treatment of an HDAC inhibitor was sufficient to restore MICA/B expression in insensitive cells. Finally, we demonstrated that HDAC inhibition restored DNA damage‑dependent cytotoxic NK activity against insensitive cells. Thus, the present study revealed that DNA damage‑dependent MICA/B expression in insensitive cancer cells can be restored by chromatin relaxation via the HDAC/Suv39/G9a pathway. Collectively, manipulation of chromatin status by therapeutic cancer drugs may potentiate the antitumor effect by enhancing immune activation following radiotherapy and DNA damage-associated chemotherapy.
Collapse
Affiliation(s)
- Nakako Izumi Nakajima
- Radiological Science Research and Development Directorate, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Atsuko Niimi
- Research Program for Heavy Ion Therapy, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Maebashi, Gunma 371-8511, Japan
| | - Mayu Isono
- Advanced Scientific Research Leaders Development Unit, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Takahiro Oike
- Department of Radiation Oncology, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Hiro Sato
- Department of Radiation Oncology, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Atsushi Shibata
- Advanced Scientific Research Leaders Development Unit, Gunma University, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
393
|
Demoulin B, Cook WJ, Murad J, Graber DJ, Sentman ML, Lonez C, Gilham DE, Sentman CL, Agaugue S. Exploiting natural killer group 2D receptors for CAR T-cell therapy. Future Oncol 2017; 13:1593-1605. [PMID: 28613086 DOI: 10.2217/fon-2017-0102] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptors (CARs) are genetically engineered proteins that combine an extracellular antigen-specific recognition domain with one or several intracellular T-cell signaling domains. When expressed in T cells, these CARs specifically trigger T-cell activation upon antigen recognition. While the clinical proof of principle of CAR T-cell therapy has been established in hematological cancers, CAR T cells are only at the early stages of being explored to tackle solid cancers. This special report discusses the concept of exploiting natural killer cell receptors as an approach that could broaden the specificity of CAR T cells and potentially enhance the efficacy of this therapy against solid tumors. New data demonstrating feasibility of this approach in humans and supporting the ongoing clinical trial are also presented.
Collapse
Affiliation(s)
- Benjamin Demoulin
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| | - W James Cook
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | | | - David J Graber
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Marie-Louise Sentman
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Caroline Lonez
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| | - David E Gilham
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| | - Charles L Sentman
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Sophie Agaugue
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| |
Collapse
|
394
|
Abstract
In contrast to conventional T lymphocytes, which carry an αβ T-cell receptor and recognize antigens as peptides presented by major histocompatibility complex class I or class II molecules, human γδ T cells recognize different metabolites such as non-peptidic pyrophosphate molecules that are secreted by microbes or overproduced by tumor cells. Hence, γδ T cells play a role in immunosurveillance of infection and cellular transformation. Until recently, it has been unknown how the γδ T-cell receptor senses such pyrophosphates in the absence of known antigen-presenting molecules. Recent studies from several groups have identified a unique role of butyrophilin (BTN) protein family members in this process, notably of BTN3A1. BTNs are a large family of transmembrane proteins with diverse functions in lipid secretion and innate and adaptive immunity. Here we discuss current models of how BTN molecules regulate γδ T-cell activation. We also address the implications of these recent findings on the design of novel immunotherapeutic strategies based on the activation of γδ T cells.
Collapse
Affiliation(s)
- Dieter Kabelitz
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3, Building 17, D-24105 Kiel, Germany
| | - Marcus Lettau
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3, Building 17, D-24105 Kiel, Germany
| | - Ottmar Janssen
- Institute of Immunology, University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3, Building 17, D-24105 Kiel, Germany
| |
Collapse
|
395
|
Fang F, Xiao W, Tian Z. NK cell-based immunotherapy for cancer. Semin Immunol 2017; 31:37-54. [DOI: 10.1016/j.smim.2017.07.009] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/24/2017] [Indexed: 12/19/2022]
|
396
|
Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nat Immunol 2017; 17:1025-36. [PMID: 27540992 DOI: 10.1038/ni.3518] [Citation(s) in RCA: 812] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022]
Abstract
Alteration in the expression of cell-surface proteins is a common consequence of malignant transformation. Natural killer (NK) cells use an array of germline-encoded activating and inhibitory receptors that scan for altered protein-expression patterns, but tumor evasion of detection by the immune system is now recognized as one of the hallmarks of cancer. NK cells display rapid and potent immunity to metastasis or hematological cancers, and major efforts are now being undertaken to fully exploit NK cell anti-tumor properties in the clinic. Diverse approaches encompass the development of large-scale NK cell-expansion protocols for adoptive transfer, the establishment of a microenvironment favorable to NK cell activity, the redirection of NK cell activity against tumor cells and the release of inhibitory signals that limit NK cell function. In this Review we detail recent advances in NK cell-based immunotherapies and discuss the advantages and limitations of these strategies.
Collapse
Affiliation(s)
- Camille Guillerey
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| |
Collapse
|
397
|
Xie H, Zhang Q, Zhou H, Zhou J, Zhang J, Jiang Y, Wang J, Meng X, Zeng L, Jiang X. microRNA-889 is downregulated by histone deacetylase inhibitors and confers resistance to natural killer cytotoxicity in hepatocellular carcinoma cells. Cytotechnology 2017; 70:513-521. [PMID: 28550492 DOI: 10.1007/s10616-017-0108-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 05/11/2017] [Indexed: 12/17/2022] Open
Abstract
Major histocompatibility complex class I chain-related gene B (MICB) is expressed on tumor cells and participates in natural killer (NK) cell-mediated antitumor immune response through engagement with the NKG2D receptor. This study was undertaken to identify novel microRNA (miRNA) regulators of MICB and clarify their functions in NK cell-mediated cytotoxicity to hepatocellular carcinoma (HCC) cells. Bioinformatic analysis and luciferase reporter assay were conducted to search for MICB-targeting miRNAs. Overexpression and knockdown experiments were performed to determine the roles of candidate miRNAs in the susceptibility of HCC cells to NK lysis. miR-889 was identified as a novel MICB-targeting miRNA and overexpression of miR-889 significantly inhibited the mRNA and protein expression of MICB in HepG2 and SMMC7721 HCC cells. miR-889 expression had a negative correlation with MICB mRNA levels in HCC specimens (r = -0.392, P = 0.0146). NK cell-mediated cytotoxicity was reduced in miR-889-overexpressing HCC cells, which was reversed by restoration of MICB expression. In contrast, knockdown of miR-889 led to more pronounced NK cell-mediated lysis in HCC cells. HCC cells exposed to the histone deacetylase (HDAC) inhibitor sodium valproate showed downregulation of miR-889. Enforced expression of miR-889 prevented the upregulation of MICB and enhancement of NK cell-mediated lysis by HDAC inhibitors. In conclusion, miR-889 upregulation attenuates the susceptibility of HCC cells to NK lysis and represents a potential target for improving NK cell-based antitumor therapies.
Collapse
Affiliation(s)
- Haitao Xie
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Qiugui Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Hui Zhou
- Tumor Hospital Xiangya School of Medicine of Central South University, Changsha, China
| | - Jun Zhou
- Department of Rehabilitation, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Ji Zhang
- Laboratory of Rheumatology and Immunology, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yan Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Jinghong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Xianglin Meng
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Leping Zeng
- Department of Anatomy and Neurobiology, Biology Postdoctoral Workstation, Basic School of Medicine, Central South University, Changsha, China.
| | - Xiaoxin Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, China.
| |
Collapse
|
398
|
Godbersen C, Coupet TA, Huehls AM, Zhang T, Battles MB, Fisher JL, Ernstoff MS, Sentman CL. NKG2D Ligand-Targeted Bispecific T-Cell Engagers Lead to Robust Antitumor Activity against Diverse Human Tumors. Mol Cancer Ther 2017; 16:1335-1346. [PMID: 28500232 DOI: 10.1158/1535-7163.mct-16-0846] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/16/2017] [Accepted: 04/28/2017] [Indexed: 01/06/2023]
Abstract
Two new bispecific T-cell engaging (BiTE) molecules with specificity for NKG2D ligands were developed and functionally characterized. One, huNKG2D-OKT3, was derived from the extracellular portion of the human NKG2D receptor fused to a CD3ε binding single-chain variable fragment (scFv), known as OKT3. NKG2D has multiple ligands, including MICA, which are expressed by a variety of malignant cells. A second molecule, B2-OKT3, was created in the tandem scFv BiTE format that targets MICA on tumor cells and CD3ε on human T cells. Both BiTEs specifically activated T cells to kill human tumor cell lines. Cytotoxicity by B2-OKT3, but not huNKG2D-OKT3, is blocked by soluble rMICA. The huNKG2D-OKT3 induced greater T-cell cytokine production in comparison with B2-OKT3. No T-cell pretreatment was required for IFNγ production upon coculture of B2-OKT3 or huNKG2D-OKT3 with T cells and target cells. The effector memory T-cell compartment was the primary source of IFNγ, and culture of T cells and these BiTEs with plate-bound rMICA showed ligand density-dependent production of IFNγ from both CD4+ and CD8+ T cells. There was 2-fold more IFNγ produced per CD8+ T cell and 5-fold greater percentage of CD8+ T cells producing IFNγ compared with CD4+ T cells. In addition, both BiTEs elicited significant antitumor responses against human metastatic melanoma tumor samples using autologous or healthy donor T cells. These data demonstrate the robust antitumor activity of these NKG2D ligand-binding bispecific proteins and support their further development for clinical use. Mol Cancer Ther; 16(7); 1335-46. ©2017 AACR.
Collapse
Affiliation(s)
- Claire Godbersen
- Department of Microbiology & Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- The Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Tiffany A Coupet
- Department of Microbiology & Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- The Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Amelia M Huehls
- Department of Microbiology & Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- The Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Tong Zhang
- Department of Microbiology & Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- The Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Michael B Battles
- Department of Microbiology & Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- The Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | | | | | - Charles L Sentman
- Department of Microbiology & Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.
- The Center for Synthetic Immunity, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| |
Collapse
|
399
|
Idorn M, Thor Straten P. Exercise and cancer: from "healthy" to "therapeutic"? Cancer Immunol Immunother 2017; 66:667-671. [PMID: 28324125 PMCID: PMC5406418 DOI: 10.1007/s00262-017-1985-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/05/2017] [Indexed: 11/28/2022]
Abstract
Exercise improves functional capacity and patient-reported outcomes across a range of cancer diagnoses. The mechanisms behind this protection have been largely unknown, but exercise-mediated changes in body composition, sex hormone levels, systemic inflammation, and immune cell function have been suggested to play a role. We recently demonstrated that voluntary exercise leads to an influx of immune cells in tumors, and a more than 60% reduction in tumor incidence and growth across several mouse models. Given the common mechanisms of immune cell mobilization in mouse and man during exercise, we hypothesize that this link between exercise and the immune system can be exploited in cancer therapy in particular in combination with immunotherapy. Thus, we believe that exercise may not just be "healthy" but may in fact be therapeutic.
Collapse
Affiliation(s)
- Manja Idorn
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), University Hospital Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Per Thor Straten
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), University Hospital Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
400
|
Morvan MG, Champsaur M, Reizis B, Lanier LL. Chronic In Vivo Interaction of Dendritic Cells Expressing the Ligand Rae-1ε with NK Cells Impacts NKG2D Expression and Function. Immunohorizons 2017; 1:10-19. [PMID: 28815225 PMCID: PMC5555644 DOI: 10.4049/immunohorizons.1700004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
To investigate how dendritic cells (DCs) interact with NK cells in vivo, we developed a novel mouse model in which Rae-1ε, a ligand of the NKG2D receptor, is expressed in cells with high levels of CD11c. In these CD11c-Rae1 mice, expression of Rae-1 was confirmed on all subsets of DCs and a small subset of B and T cells, but not on NK cells. DC numbers and activation status were unchanged, and NK cells in these CD11c-Rae1 mice presented the same Ly49 repertoire and maturation levels as their littermate wildtype controls. Early NK cell activation after mouse CMV infection was slightly lower than in wildtype mice, but NK cell expansion and viral control were comparable. Notably, we demonstrate that chronic interaction of NK cells with NKG2D ligand-expressing DCs leads to a reversible NKG2D down-modulation, as well as impaired NKG2D-dependent NK cell functions, including tumor rejection. In addition to generating a useful mouse model, our studies reveal in vivo the functional importance of the NK cell and DC cross-talk.
Collapse
Affiliation(s)
- Maelig G. Morvan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143
| | - Marine Champsaur
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY 10016
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|