101
|
Central nervous system acute lymphoblastic leukemia: role of natural killer cells. Blood 2015; 125:3420-31. [PMID: 25896649 DOI: 10.1182/blood-2014-08-595108] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 03/17/2015] [Indexed: 02/06/2023] Open
Abstract
Central nervous system acute lymphoblastic leukemia (CNS-ALL) is a major clinical problem. Prophylactic therapy is neurotoxic, and a third of the relapses involve the CNS. Increased expression of interleukin 15 (IL-15) in leukemic blasts is associated with increased risk for CNS-ALL. Using in vivo models for CNS leukemia caused by mouse T-ALL and human xenografts of ALL cells, we demonstrate that expression of IL-15 in leukemic cells is associated with the activation of natural killer (NK) cells. This activation limits the outgrowth of leukemic cells in the periphery, but less in the CNS because NK cells are excluded from the CNS. Depletion of NK cells in NOD/SCID mice enabled combined systemic and CNS leukemia of human pre-B-ALL. The killing of human leukemia lymphoblasts by NK cells depended on the expression of the NKG2D receptor. Analysis of bone marrow (BM) diagnostic samples derived from children with subsequent CNS-ALL revealed a significantly high expression of the NKG2D and NKp44 receptors. We suggest that the CNS may be an immunologic sanctuary protected from NK-cell activity. CNS prophylactic therapy may thus be needed with emerging NK cell-based therapies against hematopoietic malignancies.
Collapse
|
102
|
Yossef R, Gur C, Shemesh A, Guttman O, Hadad U, Nedvetzki S, Miletić A, Nalbandyan K, Cerwenka A, Jonjic S, Mandelboim O, Porgador A. Targeting natural killer cell reactivity by employing antibody to NKp46: implications for type 1 diabetes. PLoS One 2015; 10:e0118936. [PMID: 25719382 PMCID: PMC4342013 DOI: 10.1371/journal.pone.0118936] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 01/07/2015] [Indexed: 11/18/2022] Open
Abstract
Natural killer (NK) cells belong to the innate lymphoid cells. Their cytotoxic activity is regulated by the delicate balance between activating and inhibitory signals. NKp46 is a member of the primary activating receptors of NK cells. We previously reported that the NKp46 receptor is involved in the development of type 1 diabetes (T1D). Subsequently, we hypothesized that blocking this receptor could prevent or hinder disease development. To address this goal, we developed monoclonal antibodies for murine NKp46. One mAb, named NCR1.15, recognizes the mouse homologue protein of NKp46, named Ncr1, and was able to down-regulate the surface expression of NKp46 on primary murine NK cells following antibody injection in vivo. Additionally, NCR1.15 treatments were able to down-regulate cytotoxic activity mediated by NKp46, but not by other NK receptors. To test our primary assumption, we examined T1D development in two models, non-obese diabetic mice and low-dose streptozotocin. Our results show a significantly lower incidence of diabetic mice in the NCR1.15-treated group compared to control groups. This study directly demonstrates the involvement of NKp46 in T1D development and suggests a novel treatment strategy for early insulitis.
Collapse
Affiliation(s)
- Rami Yossef
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Chamutal Gur
- The Lautenberg Center for General and Tumor Immunology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
- Department of Medicine, Hadassah-Hebrew University Hospital, Jerusalem 91120, Israel
| | - Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ofer Guttman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Uzi Hadad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Shlomo Nedvetzki
- BioLineRx Ltd., 19 Hartum Street, P.O. Box 45158. Jerusalem 91450, Israel
| | - Antonija Miletić
- Center for Proteomics and Department for Histology and Embryology, School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | | | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center, Heidelberg 69120, Germany
| | - Stipan Jonjic
- Center for Proteomics and Department for Histology and Embryology, School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
- * E-mail:
| |
Collapse
|
103
|
Mechanisms of tumor-induced T cell immune suppression and therapeutics to counter those effects. Arch Pharm Res 2015; 38:1415-33. [PMID: 25634101 DOI: 10.1007/s12272-015-0566-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/18/2015] [Indexed: 02/07/2023]
Abstract
The theory of tumor immune surveillance states that the host immune system has means to recognize transformed cells and kills them to prevent growth and spreading of those cells. Nevertheless, cancer cells often survive and outgrow to form a tumor mass and metastasize to other tissues or organs. During the stage of immune evasion of tumor, various changes takes place both in the tumor cells and the tumor microenvironment to divert the anti-tumor immune responses by T cells and natural killer cells. Advances in the basic science in tumor immunology have led to development of many creative strategies to overcome the immune suppression imposed during tumor progression, a few of which have been approved for the treatment of cancer patients in the clinic. In the first part of this review, mechanisms of tumor-induced T cell immune suppression resulting in immune evasion of tumors will be discussed. In the second part, emerging methods to harness the immune responses against tumors will be introduced.
Collapse
|
104
|
Korrer MJ, Routes JM. Adenovirus serotype 5 E1A expressing tumor cells elicit a tumor-specific CD8+ T cell response independent of NKG2D. RESULTS IN IMMUNOLOGY 2015; 5:1-5. [PMID: 25685658 PMCID: PMC4309920 DOI: 10.1016/j.rinim.2015.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 11/22/2022]
Abstract
The expression of the Adenovirus serotype 2 or serotype 5 (Ad2/5) E1A gene in tumor cells upregulates ligands that are recognized by the NKG2D activating receptor, which is expressed on NK cells and T cells, and reduces their tumorigenicity, a process dependent on NK cells and T cells. In some model systems, the forced overexpression of NKG2D ligands on tumor cells induced antigen-specific CD8+ T cells that mediated anti-tumor immunity. We wanted to determine if the interaction of NKG2D ligands on tumor cells that express E1A with NKG2D on immune cells contributed to the ability of E1A to induce a CD8+ T cell anti-tumor response or reduce tumorigenicity. To address these questions, we used the MCA-205 tumor cell line or MCA-205 cells that expressed Ad5 E1A (MCA-205-E1A cells), a fusion protein of E1A and ovalbumin (MCA-205-E1A-OVA) or OVA (MCA-205-OVA). We found that the expression of E1A or E1A–OVA, but not OVA, upregulated the expression of the NKG2D ligand RAE-1 on the surface of MCA-205 cells. Additionally, MCA-205-E1A cells and MCA-205-E1A-OVA cells were more sensitive to NK cell lysis than MCA-205 or MCA-205-OVA cells in WT B6 mice, but not NKG2D deficient B6 mice. Next, we adoptively transferred WT or NKG2D deficient OT-1 T cells (CD8 T cells that recognize OVA residues 257–264) into WT B6 mice or B6 mice that were deficient in NKG2D respectively and measured the expansion of OT-1 cells following immunization with MCA-205-E1A-OVA or MCA-205-OVA cells. We found that the expansion of OT-1 cells following immunization of either OVA-expressing MCA-205 cell lines was not affected by the presence or absence of NKG2D in B6 mice. Finally, we found that the capacity of E1A to reduce the tumorigenicity of MCA-205 cells was not impaired in B6-NKG2D deficient mice as compared to WT B6 mice. Our results suggest that the ability of E1A to reduce the tumorigenicity of MCA-205 cells, or induce an antigen-specific CD8+ T cell response, is independent of the interaction of NKG2D ligands with the NKG2D receptor.
Collapse
Key Words
- Ad, adenovirus
- Adenovirus E1A
- B6, C57BL/6
- CD8+ T cells
- E1A, early region 1 A
- MCA, methylcholanthrene
- NK cells
- NK, natural killer
- NKG2D
- NKG2D ligands
- NKG2D, natural killer group 2 D
- OVA, ovalbumin
- RAE-1, retinoic acid early inducible
- TPD50, tumor producing dose 50
- Tumor immunology
- WT, wildtype
Collapse
Affiliation(s)
- Michael J. Korrer
- Department of Pediatrics, Division of Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Correspondence to: Medical College of Wisconsin, 9000 W. Wisconsin Avenue, Milwaukee, WI 53226-4874, USA.
| | - John M. Routes
- Department of Pediatrics, Division of Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Correspondence to: Medical College of Wisconsin, 9000 W. Wisconsin Avenue, Milwaukee, WI 53226-4874, USA.
| |
Collapse
|
105
|
Korneychuk N, Meresse B, Cerf-Bensussan N. Lessons from rodent models in celiac disease. Mucosal Immunol 2015; 8:18-28. [PMID: 25354320 DOI: 10.1038/mi.2014.102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/23/2014] [Indexed: 02/04/2023]
Abstract
Over the past 25 years, studies led in humans have considerably improved our understanding of celiac disease, a complex disease that is generally defined as an autoimmune-like enteropathy induced by dietary gluten in genetically predisposed individuals. Recently, large efforts were also invested in the development of mouse models in order to explore pathogenic hypotheses, and also with the goal to design pretherapeutic models that could be used to test innovative therapies. Yet, modeling this complex multifactorial disease has been a very challenging task. Herein, we review how approaches in rodents have provided insight into celiac disease pathophysiology and also highlight the difficulties met to fully recapitulate the human disease.
Collapse
Affiliation(s)
- N Korneychuk
- 1] INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, Paris, France [2] Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, Paris, France
| | - B Meresse
- 1] INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, Paris, France [2] Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, Paris, France
| | - N Cerf-Bensussan
- 1] INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, Paris, France [2] Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, Paris, France
| |
Collapse
|
106
|
Fujita H, Hatanaka Y, Sutoh Y, Suzuki Y, Oba K, Hatanaka KC, Mitsuhashi T, Otsuka N, Fugo K, Kasahara M, Matsuno Y. Immunohistochemical validation and expression profiling of NKG2D ligands in a wide spectrum of human epithelial neoplasms. J Histochem Cytochem 2014; 63:217-27. [PMID: 25473094 DOI: 10.1369/0022155414563800] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The MHC class I-chain-related proteins (MICs) and the UL16-binding proteins (ULBPs) are inducible stress response molecules that work as activators of a specific receptor, NKG2D, which is expressed on effector cells, such as NK cells and subsets of T cells. In this study, we sought to explore the biological significance of NKG2D ligands in human neoplasms by comprehensively examining the immunohistochemical expression profile of NKG2D ligands in a variety of human epithelial neoplasms. Following careful validation of the immunohistochemical specificity and availability of anti-human ULBP antibodies for formalin-fixed paraffin-embedded (FFPE) materials, the expression of NKG2D ligands was analyzed in FFPE tissue microarrays comprising 22 types of epithelial neoplastic tissue with their non-neoplastic counterpart from various organs. Hierarchical cluster analysis demonstrated a positive relationship among ULBP2/6, ULBP3, ULBP1, and ULBP5, whose expression patterns were similar across all of the neoplastic tissues examined. In contrast, MICA/B, as well as ULBP4, did not appear to be related to any other ligand. These expression profiles of NKG2D ligands in human neoplasms based on well-validated specific antibodies, followed by hierarchical cluster analysis, should help to clarify some functional aspects of these molecules in cancer biology, and also provide a path to the development of novel tumor-type-specific treatment strategies.
Collapse
Affiliation(s)
- Hiromi Fujita
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM),Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Yoichi Sutoh
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM),Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Yuta Suzuki
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Koji Oba
- Translational Research and Clinical Trial Center, Hokkaido University Hospital, Sapporo, Japan (KO)
| | - Kanako C Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Tomoko Mitsuhashi
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| | - Noriyuki Otsuka
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Kazunori Fugo
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Masanori Kasahara
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan (HF, YS, NO, KF, MK)
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan (HF, YH, YS, KCH, TM, YM)
| |
Collapse
|
107
|
Telcian AG, Zdrenghea MT, Caramori G, Laza-Stanca V, Message SD, Kebadze T, Kon OM, Groh V, Papi A, Johnston SL, Mallia P, Stanciu LA. Soluble major histocompatibility complex class I-related chain B molecules are increased and correlate with clinical outcomes during rhinovirus infection in healthy subjects. Chest 2014; 146:32-40. [PMID: 24556715 PMCID: PMC4077410 DOI: 10.1378/chest.13-2247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND: Surface major histocompatibility complex class I-related chain (MIC) A and B molecules are increased by IL-15 and have a role in the activation of natural killer group 2 member D-positive natural killer and CD8 T cells. MICA and MICB also exist in soluble forms (sMICA and sMICB). Rhinoviruses (RVs) are the major cause of asthma exacerbations, and IL-15 levels are decreased in the airways of subjects with asthma. The role of MIC molecules in immune responses in the lung has not been studied. Here, we determine the relationship between MICA and MICB and RV infection in vitro in respiratory epithelial cells and in vivo in healthy subjects and subjects with asthma. METHODS: Surface MICA and MICB, as well as sMICA and sMICB, in respiratory epithelial cells were measured in vitro in response to RV infection and exposure to IL-15. Levels of sMICA and sMICB in serum, sputum, and BAL were measured and correlated with blood and bronchoalveolar immune cells in healthy subjects and subjects with asthma before and during RV infection. RESULTS: RV increased MICA and MICB in vitro in epithelial cells. Exogenous IL-15 upregulated sMICB levels in RV-infected epithelial cells. Levels of sMICB molecules in serum were increased in healthy subjects compared with subjects with stable asthma. Following RV infection, airway levels of sMIC are upregulated, and there are positive correlations between sputum MICB levels and the percentage of bronchoalveolar natural killer cells in healthy subjects but not subjects with asthma. CONCLUSIONS: RV infection induces MIC molecules in respiratory epithelial cells in vitro and in vivo. Induction of MICB molecules is impaired in subjects with asthma, suggesting these molecules may have a role in the antiviral immune response to RV infections.
Collapse
Affiliation(s)
- Aurica G Telcian
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England
| | - Mihnea T Zdrenghea
- Medical Research Council, London, England; Asthma UK Centre in Allergic Mechanisms of Asthma, London, England; The Oncology Institute "Prof. Dr. Ion Chiricuţă" Cluj-Napoca and Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania.
| | - Gaetano Caramori
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF), Sezione di Malattie dell'Apparato Respiratorio, Università degli Studi di Ferrara, Ferrara, Italy
| | - Vasile Laza-Stanca
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England
| | - Simon D Message
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Imperial College Healthcare NHS Trust, London, England
| | - Tatiana Kebadze
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England
| | - Onn M Kon
- Imperial College Healthcare NHS Trust, London, England
| | | | - Alberto Papi
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF), Sezione di Malattie dell'Apparato Respiratorio, Università degli Studi di Ferrara, Ferrara, Italy
| | - Sebastian L Johnston
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Imperial College Healthcare NHS Trust, London, England
| | - Patrick Mallia
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Imperial College Healthcare NHS Trust, London, England
| | - Luminita A Stanciu
- Airways Disease Infection Section, National Heart & Lung Institute (NHLI), and Centre for Respiratory Infection, Imperial College London, London, England; Medical Research Council, London, England; Asthma UK Centre in Allergic Mechanisms of Asthma, London, England; The Oncology Institute "Prof. Dr. Ion Chiricuţă" Cluj-Napoca and Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
108
|
Klingel K, Fabritius C, Sauter M, Göldner K, Stauch D, Kandolf R, Ettischer N, Gahlen S, Schönberger T, Ebner S, Makrigiannis AP, Bélanger S, Diefenbach A, Polić B, Pratschke J, Kotsch K. The activating receptor NKG2D of natural killer cells promotes resistance against enterovirus-mediated inflammatory cardiomyopathy. J Pathol 2014; 234:164-77. [PMID: 24797160 DOI: 10.1002/path.4369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 04/08/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022]
Abstract
In enterovirus-induced cardiomyopathy, information regarding the detailed impact of natural killer (NK) cells on the outcome of the disease is limited. We therefore hypothesized that NK cells and certain NK cell receptors determine the different outcome of coxsackievirus B3 (CVB3) myocarditis. Here, we demonstrate in murine models that resistance to chronic CVB3 myocarditis in immunocompetent C57BL/6 mice is characterized by significantly more mature CD11b(high) NK cells, the presence of NKG2D on NK cells, and enhanced NKG2D-dependent cytotoxicity compared to CVB3-susceptible A.BY/SnJ mice. The highly protective role of NKG2D in myocarditis was further proven by in vivo neutralization of NKG2D as well as in NKG2D-deficient mice but was shown to be independent of CD8(+) T-cell-dependent immunity. Moreover, the adoptive transfer of immunocompetent C57BL/6 NK cells pre- (day -1) as well as post-infectionem (day +2) displayed the potential to prevent permissive A.BY/SnJ mice from a progressive outcome of CVB3 myocarditis reflected by significantly improved cardiopathology and heart function. Altogether, our results provide firm evidence for a protective role of NKG2D-activated NK cells in CVB3 myocarditis leading to an effective virus clearance, thus offering novel therapeutic options in the treatment of virus-induced myocarditis.
Collapse
Affiliation(s)
- Karin Klingel
- Department of Molecular Pathology, University Hospital Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Karimi MA, Aguilar O, Zou B, Bachmann MH, Carlyle JR, Baldwin CL, Kambayashi T. A truncated human NKG2D splice isoform negatively regulates NKG2D-mediated function. THE JOURNAL OF IMMUNOLOGY 2014; 193:2764-2771. [PMID: 25092887 DOI: 10.4049/jimmunol.1400920] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Natural killer group 2, member D (NKG2D) is a stimulatory receptor expressed by NK cells and a subset of T cells. NKG2D is crucial in diverse aspects of innate and adaptive immune functions. In this study, we characterize a novel splice variant of human NKG2D that encodes a truncated receptor lacking the ligand-binding ectodomain. This truncated NKG2D (NKG2D(TR)) isoform was detected in primary human NK and CD8(+) T cells. Overexpression of NKG2D(TR) severely attenuated cell killing and IFN-γ release mediated by full-length NKG2D (NKG2D(FL)). In contrast, specific knockdown of endogenously expressed NKG2D(TR) enhanced NKG2D-mediated cytotoxicity, suggesting that NKG2D(TR) is a negative regulator of NKG2D(FL). Biochemical studies demonstrated that NKG2D(TR) was bound to DNAX-activated protein of 10 kDa (DAP10) and interfered with the interaction of DAP10 with NKG2D(FL). In addition, NKG2D(TR) associated with NKG2D(FL), which led to forced intracellular retention, resulting in decreased surface NKG2D expression. Taken together, these data suggest that competitive interference of NKG2D/DAP10 complexes by NKG2D(TR) constitutes a novel mechanism for regulation of NKG2D-mediated function in human CD8(+) T cells and NK cells.
Collapse
Affiliation(s)
- Mobin A Karimi
- Department of Veterinary & Animal Sciences/Immunology, University of Massachusetts, Amherst, MA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Oscar Aguilar
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Baixiang Zou
- Department of Veterinary & Animal Sciences/Immunology, University of Massachusetts, Amherst, MA
| | - Michael H Bachmann
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - James R Carlyle
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Cynthia L Baldwin
- Department of Veterinary & Animal Sciences/Immunology, University of Massachusetts, Amherst, MA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
110
|
Shen Y, Wang X, Jin Y, Lu J, Qiu G, Wen X. Differentially expressed genes and interacting pathways in bladder cancer revealed by bioinformatic analysis. Mol Med Rep 2014; 10:1746-52. [PMID: 25050631 PMCID: PMC4148370 DOI: 10.3892/mmr.2014.2396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 04/16/2014] [Indexed: 01/11/2023] Open
Abstract
The goal of this study was to identify cancer-associated differentially expressed genes (DEGs), analyze their biological functions and investigate the mechanism(s) of cancer occurrence and development, which may provide a theoretical foundation for bladder cancer (BCa) therapy. We downloaded the mRNA expression profiling dataset GSE13507 from the Gene Expression Omnibus database; the dataset includes 165 BCa and 68 control samples. T-tests were used to identify DEGs. To further study the biological functions of the identified DEGs, we performed a Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Next, we built a network of potentially interacting pathways to study the synergistic relationships among DEGs. A total of 12,105 genes were identified as DEGs, of which 5,239 were upregulated and 6,866 were downregulated in BCa. The DEGs encoding activator protein 1 (AP-1), nuclear factor of activated T-cells (NFAT) proteins, nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and interleukin (IL)-10 were revealed to participate in the significantly enriched immune pathways that were downregulated in BCa. KEGG enrichment analysis revealed 7 significantly upregulated and 47 significantly downregulated pathways enriched among the DEGs. We found a crosstalk interaction among a total of 44 pathways in the network of BCa-affected pathways. In conclusion, our results show that BCa involves dysfunctions in multiple systems. Our study is expected to pave ways for immune and inflammatory research and provide molecular insights for cancer therapy.
Collapse
Affiliation(s)
- Yinzhou Shen
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Xuelei Wang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Yongchao Jin
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Jiasun Lu
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Guangming Qiu
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Xiaofei Wen
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
111
|
Raverdeau M, Mills KHG. Modulation of T cell and innate immune responses by retinoic Acid. THE JOURNAL OF IMMUNOLOGY 2014; 192:2953-8. [PMID: 24659788 DOI: 10.4049/jimmunol.1303245] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Retinoic acid (RA) is produced by a number of cell types, including macrophages and dendritic cells, which express retinal dehydrogenases that convert vitamin A to its main biologically active metabolite, all-trans RA. All-trans RA binds to its nuclear retinoic acid receptors that are expressed in lymphoid cells and act as transcription factors to regulate cell homing and differentiation. RA production by CD103(+) dendritic cells and alveolar macrophages functions with TGF-β to promote conversion of naive T cells into Foxp3(+) regulatory T cells and, thereby, maintain mucosal tolerance. Furthermore, RA inhibits the differentiation of naive T cells into Th17 cells. However, Th1 and Th17 responses are constrained during vitamin A deficiency and in nuclear RA receptor α-defective mice. Furthermore, RA promotes effector T cell responses during infection or autoimmune diseases. Thus, RA plays a role in immune homeostasis in the steady-state but activates pathogenic T cells in conditions of inflammation.
Collapse
Affiliation(s)
- Mathilde Raverdeau
- Immune Regulation Research Group and Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | | |
Collapse
|
112
|
Lin Z, Wang C, Xia H, Liu W, Xiao W, Qian L, Jia X, Ding Y, Ji M, Gong W. CD4(+) NKG2D(+) T cells induce NKG2D down-regulation in natural killer cells in CD86-RAE-1ε transgenic mice. Immunology 2014; 141:401-15. [PMID: 24708417 DOI: 10.1111/imm.12203] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 10/23/2013] [Accepted: 10/23/2013] [Indexed: 11/30/2022] Open
Abstract
The binding of NKG2D to its ligands strengthens the cross-talk between natural killer (NK) cells and dendritic cells, particularly at early stages, before the initiation of the adaptive immune response. We found that retinoic acid early transcript-1ε (RAE-1ε), one of the ligands of NKG2D, was persistently expressed on antigen-presenting cells in a transgenic mouse model (pCD86-RAE-1ε). By contrast, NKG2D expression on NK cells, NKG2D-dependent cytotoxicity and tumour rejection, and dextran sodium sulphate-induced colitis were all down-regulated in this mouse model. The down-regulation of NKG2D on NK cells was reversed by stimulation with poly (I:C). The ectopic expression of RAE-1ε on dendritic cells maintained NKG2D expression levels and stimulated the activity of NK cells ex vivo, but the higher frequency of CD4(+) NKG2D(+) T cells in transgenic mice led to the down-regulation of NKG2D on NK cells in vivo. Hence, high levels of RAE-1ε expression on antigen-presenting cells would be expected to induce the down-regulation of NK cell activation by a regulatory T-cell subset.
Collapse
Affiliation(s)
- Zhijie Lin
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Lee YS, Kim SW. Bioreducible polymers for therapeutic gene delivery. J Control Release 2014; 190:424-39. [PMID: 24746626 DOI: 10.1016/j.jconrel.2014.04.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 01/18/2023]
Abstract
Most currently available cationic polymers have significant acute toxicity concerns such as cellular toxicity, aggregation of erythrocytes, and entrapment in the lung capillary bed, largely due to their poor biocompatibility and non-degradability under physiological conditions. To develop more intelligent polymers, disulfide bonds are introduced in the design of biodegradable polymers. Herein, the sustained innovations of biomimetic nano-sized constructs with bioreducible poly(disulfide amine)s demonstrate a viable clinical tool for the treatment of cardiovascular disease, anemia, diabetes, and cancer.
Collapse
Affiliation(s)
- Young Sook Lee
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA.
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA; Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
114
|
Li Y, Mariuzza RA. Structural basis for recognition of cellular and viral ligands by NK cell receptors. Front Immunol 2014; 5:123. [PMID: 24723923 PMCID: PMC3972465 DOI: 10.3389/fimmu.2014.00123] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 03/10/2014] [Indexed: 11/13/2022] Open
Abstract
Natural killer (NK) cells are key components of innate immune responses to tumors and viral infections. NK cell function is regulated by NK cell receptors that recognize both cellular and viral ligands, including major histocompatibility complex (MHC), MHC-like, and non-MHC molecules. These receptors include Ly49s, killer immunoglobulin-like receptors, leukocyte immunoglobulin-like receptors, and NKG2A/CD94, which bind MHC class I (MHC-I) molecules, and NKG2D, which binds MHC-I paralogs such as the stress-induced proteins MICA and ULBP. In addition, certain viruses have evolved MHC-like immunoevasins, such as UL18 and m157 from cytomegalovirus, that act as decoy ligands for NK receptors. A growing number of NK receptor–ligand interaction pairs involving non-MHC molecules have also been identified, including NKp30–B7-H6, killer cell lectin-like receptor G1–cadherin, and NKp80–AICL. Here, we describe crystal structures determined to date of NK cell receptors bound to MHC, MHC-related, and non-MHC ligands. Collectively, these structures reveal the diverse solutions that NK receptors have developed to recognize these molecules, thereby enabling the regulation of NK cytolytic activity by both host and viral ligands.
Collapse
Affiliation(s)
- Yili Li
- W. M. Keck Laboratory for Structural Biology, Institute for Bioscience and Biotechnology Research, University of Maryland , Rockville, MD , USA ; Department of Cell Biology and Molecular Genetics, University of Maryland , College Park, MD , USA
| | - Roy A Mariuzza
- W. M. Keck Laboratory for Structural Biology, Institute for Bioscience and Biotechnology Research, University of Maryland , Rockville, MD , USA ; Department of Cell Biology and Molecular Genetics, University of Maryland , College Park, MD , USA
| |
Collapse
|
115
|
López-Soto A, Huergo-Zapico L, Acebes-Huerta A, Villa-Alvarez M, Gonzalez S. NKG2D signaling in cancer immunosurveillance. Int J Cancer 2014; 136:1741-50. [DOI: 10.1002/ijc.28775] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/23/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Alejandro López-Soto
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| | | | - Andrea Acebes-Huerta
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| | - Mónica Villa-Alvarez
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| | - Segundo Gonzalez
- Departamento de Biología Funcional; Universidad de Oviedo; IUOPA, Asturias Spain
| |
Collapse
|
116
|
Berry R, Rossjohn J, Brooks AG. The Ly49 natural killer cell receptors: a versatile tool for viral self‐discrimination. Immunol Cell Biol 2014; 92:214-20. [DOI: 10.1038/icb.2013.100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/10/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Richard Berry
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash UniversityClaytonVictoriaAustralia
| | - Jamie Rossjohn
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash UniversityClaytonVictoriaAustralia
- Institute of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUK
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
117
|
Marcus A, Gowen BG, Thompson TW, Iannello A, Ardolino M, Deng W, Wang L, Shifrin N, Raulet DH. Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol 2014; 122:91-128. [PMID: 24507156 PMCID: PMC4228931 DOI: 10.1016/b978-0-12-800267-4.00003-1] [Citation(s) in RCA: 273] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, roles of the immune system in immune surveillance of cancer have been explored using a variety of approaches. The roles of the adaptive immune system have been a major emphasis, but increasing evidence supports a role for innate immune effector cells such as natural killer (NK) cells in tumor surveillance. Here, we discuss some of the evidence for roles in tumor surveillance of innate immune cells. In particular, we focus on NK cells and other immune cells that express germline-encoded receptors, often labeled NK receptors. The impact of these receptors and the cells that express them on tumor suppression is summarized. We discuss in detail some of the pathways and events in tumor cells that induce or upregulate cell-surface expression of the ligands for these receptors, and the logic of how those pathways serve to identify malignant, or potentially malignant cells. How tumors often evade tumor suppression mediated by innate killer cells is another major subject of the review. We end with a discussion on some of the implications of the various findings with respect to possible therapeutic approaches.
Collapse
Affiliation(s)
- Assaf Marcus
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Benjamin G Gowen
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Thornton W Thompson
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Alexandre Iannello
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Michele Ardolino
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Weiwen Deng
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Lin Wang
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - Nataliya Shifrin
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA
| | - David H Raulet
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, USA.
| |
Collapse
|
118
|
Moore TC, Kumm PM, Brown DM, Petro TM. Interferon response factor 3 is crucial to poly-I:C induced NK cell activity and control of B16 melanoma growth. Cancer Lett 2013; 346:122-8. [PMID: 24368188 DOI: 10.1016/j.canlet.2013.12.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/11/2013] [Accepted: 12/14/2013] [Indexed: 01/06/2023]
Abstract
Interferon Response Factor 3 (IRF3) induces several NK-cell activating factors, is activated by poly-I:C, an experimental cancer therapeutic, but is suppressed during many viral infections. IRF3 Knockout (KO) mice exhibited enhanced B16 melanoma growth, impaired intratumoral NK cell infiltration, but not an impaired poly-I:C therapeutic effect due to direct suppression of B16 growth. IRF3 was responsible for poly-I:C decrease in TIM-3 expression by intratumoral dendritic cells, induction of NK-cell Granzyme B and IFN-γ, and induction of macrophage IL-12, IL-15, IL-6, and IRF3-dependent NK-activating molecule (INAM). Thus, IRF3 is a key factor controlling melanoma growth through NK-cell activities, especially during poly-I:C therapy.
Collapse
Affiliation(s)
- Tyler C Moore
- School of Biological Sciences, University of Nebraska-Lincoln, United States
| | - Phyllis M Kumm
- Department of Oral Biology, University of Nebraska Medical Center, United States
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, United States; Nebraska Center for Virology, University of Nebraska-Lincoln, United States
| | - Thomas M Petro
- Nebraska Center for Virology, University of Nebraska-Lincoln, United States; Department of Oral Biology, University of Nebraska Medical Center, United States.
| |
Collapse
|
119
|
Ullrich E, Koch J, Cerwenka A, Steinle A. New prospects on the NKG2D/NKG2DL system for oncology. Oncoimmunology 2013; 2:e26097. [PMID: 24353908 PMCID: PMC3862635 DOI: 10.4161/onci.26097] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 08/06/2013] [Accepted: 08/09/2013] [Indexed: 11/19/2022] Open
Abstract
The activating immunoreceptor NKG2D endows cytotoxic lymphocytes with the capacity to recognize and eliminate infected or malignant cells. The recognition of such harmful cells is enabled by binding of NKG2D to various MHC class I-related glycoproteins, which are upregulated in the course of viral infection or malignant transformation. The past years have witnessed substantial progress in our understanding of the mechanisms underlying the regulation of NKG2D ligands (NKG2DLs) by malignant cells, of tumor-associated countermeasures promoting escape from NKG2D-dependent immunosurveillance, and of therapeutic measures that may bolster the NKG2D/NKG2DL system against malignancies. Here, we summarize the current knowledge on the NKG2D/NKG2DL system and outline opportunities to exploit the tumoricidal function of NKG2D for anticancer immunotherapy.
Collapse
Affiliation(s)
- Evelyn Ullrich
- Children's Hospital; Department of Pediatric Hematology and Oncology; Goethe-University Frankfurt am Main; Frankfurt am Main, Germany ; Center for Cell and Gene Therapy; Goethe University Frankfurt am Main; Frankfurt am Main, Germany
| | - Joachim Koch
- Center for Cell and Gene Therapy; Goethe University Frankfurt am Main; Frankfurt am Main, Germany ; Institute for Biomedical Research: Georg-Speyer-Haus; NK Cell Biology; Frankfurt am Main, Germany
| | - Adelheid Cerwenka
- German Cancer Research Center (DKFZ); Innate Immunity Group; Heidelberg, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine; Goethe-University Frankfurt am Main; Frankfurt am Main, Germany
| |
Collapse
|
120
|
Hosomi S, Chen Z, Baker K, Chen L, Huang YH, Olszak T, Zeissig S, Wang JH, Mandelboim O, Beauchemin N, Lanier LL, Blumberg RS. CEACAM1 on activated NK cells inhibits NKG2D-mediated cytolytic function and signaling. Eur J Immunol 2013; 43:2473-83. [PMID: 23696226 PMCID: PMC3775953 DOI: 10.1002/eji.201242676] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 04/09/2013] [Accepted: 05/17/2013] [Indexed: 12/29/2022]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is expressed on activated natural killer (NK) cells wherein it inhibits lysis of CEACAM1-bearing tumor cell lines. The mechanism for this is unknown. Here, we show that interleukin-2-induced expression of CEACAM1 on both mouse and primary human NK cells impairs the ability of NK gene complex group 2 member D (NKG2D) to stimulate cytolysis of CEACAM1-bearing cells. This process requires the expression of CEACAM1 on the NK cells and on the tumor cells, which is consistent with the involvement of trans-homophilic interactions between CEACAM1. Mechanistically, co-engagement of NKG2D and CEACAM1 results in a biochemical association between these two surface receptors and the recruitment of Src homology phosphatase 1 by CEACAM1 that leads to dephosphorylation of the guanine nucleotide exchange factor Vav1 and blockade of downstream signaling that is associated with the initiation of cytolysis. Thus, CEACAM1 on activated NK cells functions as an inhibitory receptor for NKG2D-mediated cytolysis, which has important implications for understanding the means by which CEACAM1 expression adversely affects tumor immunity.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhangguo Chen
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Immunology, National Jewish Health, University of Colorado Denver, Denver, CO, USA
| | - Kristi Baker
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lanfen Chen
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Yu-Hwa Huang
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Torsten Olszak
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastian Zeissig
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing H. Wang
- Department of Immunology, National Jewish Health, University of Colorado Denver, Denver, CO, USA
| | - Ofer Mandelboim
- Lautenberg Center for General and Tumor Immunology, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Nicole Beauchemin
- Goodman Cancer Research Centre and Depts. of Biochemistry, Medicine and Oncology, McGill University, Montreal, Canada
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and the Cancer Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
121
|
Chau J, Moza D, Hossain N, Lee JK, Bienenstock J, Karimi K. Increased production of IFN-γ by natural killer cells triggered with bone marrow-derived dendritic cells cultured in the presence of retinoic acid. Eur J Pharmacol 2013; 715:321-7. [DOI: 10.1016/j.ejphar.2013.04.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 04/22/2013] [Accepted: 04/26/2013] [Indexed: 11/26/2022]
|
122
|
Wensveen FM, Lenartic M, Jelencic V, Lemmermann NAW, ten Brinke A, Jonjic S, Polic B. NKG2D induces Mcl-1 expression and mediates survival of CD8 memory T cell precursors via phosphatidylinositol 3-kinase. THE JOURNAL OF IMMUNOLOGY 2013; 191:1307-15. [PMID: 23804716 DOI: 10.4049/jimmunol.1300670] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Memory formation of activated CD8 T cells is the result of a specific combination of signals that promote long-term survival and inhibit differentiation into effector cells. Much is known about initial cues that drive memory formation, but it is poorly understood which signals are essential during the intermediate stages before terminal differentiation. NKG2D is an activating coreceptor on Ag-experienced CD8 T cells that promotes effector cell functions. Its role in memory formation is currently unknown. In this study, we show that NKG2D controls formation of CD8 memory T cells by promoting survival of precursor cells. We demonstrate that NKG2D enhances IL-15-mediated PI3K signaling of activated CD8 T cells, in a specific phase of memory cell commitment, after activation but before terminal differentiation. This signal is essential for the induction of prosurvival protein Mcl-1 and precursor cell survival. In vivo, NKG2D deficiency results in reduced memory cell formation and impaired protection against reinfection. Our findings show a new role for PI3K and the NKG2D/IL-15 axis in an underappreciated stage of effector to memory cell transition that is essential for the generation of antiviral immunity. Moreover, we provide novel insights how these receptors control both effector and memory T cell differentiation.
Collapse
Affiliation(s)
- Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | | | | | | | | | | | | |
Collapse
|
123
|
Peinado C, Kang X, Hardamon C, Arora S, Mah S, Zhang H, Ngolab J, Bui JD. The nuclear factor-κB pathway down-regulates expression of the NKG2D ligand H60a in vitro: implications for use of nuclear factor-κB inhibitors in cancer therapy. Immunology 2013; 139:265-74. [PMID: 23350962 PMCID: PMC3647192 DOI: 10.1111/imm.12080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 01/15/2013] [Accepted: 01/22/2013] [Indexed: 12/31/2022] Open
Abstract
NKG2D ligands are cell surface proteins that activate NKG2D, a receptor used by natural killer (NK) cells to detect virus-infected and transformed cells. When tumour cells express high levels of NKG2D ligands, they are rejected by the immune system. Hence, reagents that increase NKG2D ligand expression on tumour cells can be important for tumour immunotherapy. To identify genes that regulate the NKG2D ligand H60a, we performed a microarray analysis of 3'-methylcholanthrene-induced sarcoma cell lines expressing high versus low H60a levels. A20, an inhibitor of nuclear factor-κB (NF-κB) activation, was differentially expressed in H60a-hi sarcoma cells. Correspondingly, treatment of tumour cells with inhibitors of NF-κB activation, such as sulfasalazine (slz), BAY-11-7085, or a non-phosphorylatable IκB, led to increased levels of H60a protein, whereas transduction of cells with an active form of IκB kinase-β (IKKβ) led to decreased levels of H60a. The regulation probably occurred at the transcriptional level, because NF-κB pathway inhibition led to increased H60a transcripts and promoter activity. Moreover, treatment of tumour cells with slz enhanced their killing by NK cells in vitro, suggesting that NF-κB inhibition can lead to tumour cell rejection. Indeed, when we blocked the NF-κB pathway specifically in tumour cells, there was decreased tumour growth in wild-type but not immune-deficient mice. Our results suggest that reagents that can block NF-κB activity specifically in the tumour and not the host immune cells would be efficacious for tumour therapy.
Collapse
Affiliation(s)
- Carlos Peinado
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
124
|
Fratricide of natural killer cells dressed with tumor-derived NKG2D ligand. Proc Natl Acad Sci U S A 2013; 110:9421-6. [PMID: 23690625 DOI: 10.1073/pnas.1300140110] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The natural killer group 2 membrane D (NKG2D) activating receptor plays crucial roles not only in host defense against tumors and viral infections, but also in autoimmune diseases. After NKG2D-mediated activation, Natural killer (NK) cells must be regulated to avoid potentially harmful reactivity. However, the negative regulation of these activated NK cells is poorly understood. Here, we reveal that the engagement of NKG2D by its ligand elicits not only target cell lysis, but also NK cell fratricide. Conventional mouse NK cells underwent cell death when cocultured with RMA cells expressing the NKG2D ligand retinoic acid early-inducible protein 1 (Rae-1), but not with RMA cells lacking MHC class I. NK cells from mice deficient for DAP10 and DAP12 or perforin did not undergo death, highlighting the importance of the NKG2D pathway for NK cell death. However, NKG2D does not transmit direct death signals in NK cells. Rather, the interaction between NKG2D and Rae-1 allowed NK cells to acquire tumor-derived Rae-1 by a membrane transfer process known as "trogocytosis," which was associated with clathrin-dependent NKG2D endocytosis. NK cells dressed with Rae-1 were lysed by neighboring NK cells through the NKG2D-induced perforin pathway in vitro and in vivo. These results provide the unique NKG2D function in negative regulation of activated NK cells.
Collapse
|
125
|
IL-15 in tumor microenvironment causes rejection of large established tumors by T cells in a noncognate T cell receptor-dependent manner. Proc Natl Acad Sci U S A 2013; 110:8158-63. [PMID: 23637340 DOI: 10.1073/pnas.1301022110] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
A major challenge of cancer immunotherapy is the persistence and outgrowth of subpopulations that lose expression of the target antigen. IL-15 is a potent cytokine that can promote organ-specific autoimmunity when up-regulated on tissue cells. Here we report that T cells eradicated 2-wk-old solid tumors that expressed IL-15, eliminating antigen-negative cells. In contrast, control tumors that lacked IL-15 expression consistently relapsed. Interestingly, even tumors lacking expression of cognate antigen were rejected when expressing IL-15, indicating that rejection after adoptive T-cell transfer was independent of cognate antigen expression. Nevertheless, the T-cell receptor of the transferred T cells influenced the outcome, consistent with the notion that T-cell receptor activation and effector status determine whether IL-15 can confer lymphokine killer activity-like properties to T cells. The effect was limited to the microenvironment of tumors expressing IL-15; there were no noticeable effects on contralateral tumors lacking IL-15. Taken together, these results indicate that expression of IL-15 in the tumor microenvironment may prevent the escape of antigen loss variants and subsequent tumor recurrence by enabling T cells to eliminate cancer cells lacking cognate antigen expression in a locally restricted manner.
Collapse
|
126
|
Molecular mechanisms of natural killer cell activation in response to cellular stress. Cell Death Differ 2013; 21:5-14. [PMID: 23579243 DOI: 10.1038/cdd.2013.26] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 01/06/2023] Open
Abstract
Protection against cellular stress from various sources, such as nutritional, physical, pathogenic, or oncogenic, results in the induction of both intrinsic and extrinsic cellular protection mechanisms that collectively limit the damage these insults inflict on the host. The major extrinsic protection mechanism against cellular stress is the immune system. Indeed, it has been well described that cells that are stressed due to association with viral infection or early malignant transformation can be directly sensed by the immune system, particularly natural killer (NK) cells. Although the ability of NK cells to directly recognize and respond to stressed cells is well appreciated, the mechanisms and the breadth of cell-intrinsic responses that are intimately linked with their activation are only beginning to be uncovered. This review will provide a brief introduction to NK cells and the relevant receptors and ligands involved in direct responses to cellular stress. This will be followed by an in-depth discussion surrounding the various intrinsic responses to stress that can naturally engage NK cells, and how therapeutic agents may induce specific activation of NK cells and other innate immune cells by activating cellular responses to stress.
Collapse
|
127
|
Thaxton JE, Nevers T, Lippe EO, Blois SM, Saito S, Sharma S. NKG2D blockade inhibits poly(I:C)-triggered fetal loss in wild type but not in IL-10-/- mice. THE JOURNAL OF IMMUNOLOGY 2013; 190:3639-47. [PMID: 23455498 DOI: 10.4049/jimmunol.1203488] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Infection and inflammation can disturb immune tolerance at the maternal-fetal interface, resulting in adverse pregnancy outcomes. However, the underlying mechanisms for detrimental immune responses remain ill defined. In this study, we provide evidence for immune programming of fetal loss in response to polyinosinic:polycytidylic acid (polyI:C), a viral mimic and an inducer of inflammatory milieu. IL-10 and uterine NK (uNK) cells expressing the activating receptor NKG2D play a critical role in poly(I:C)-induced fetal demise. In wild type (WT) mice, poly(I:C) treatment induced expansion of NKG2D(+) uNK cells and expression of Rae-1 (an NKG2D ligand) on uterine macrophages and led to fetal resorption. In IL-10(-/-) mice, NKG2D(-) T cells instead became the source of fetal resorption during the same gestation period. Interestingly, both uterine NK and T cells produced TNF-α as the key cytotoxic factor contributing to fetal loss. Treatment of WT mice with poly(I:C) resulted in excessive trophoblast migration into the decidua and increased TUNEL-positive signal. IL-10(-/-) mice supplemented with recombinant IL-10 induced fetal loss through NKG2D(+) uNK cells, similar to the response in WT mice. Blockade of NKG2D in poly(I:C)-treated WT mice led to normal pregnancy outcome. Thus, we demonstrate that pregnancy-disrupting inflammatory events mimicked by poly(I:C) are regulated by IL-10 and depend on the effector function of uterine NKG2D(+) NK cells in WT mice and NKG2D(-) T cells in IL-10 null mice.
Collapse
Affiliation(s)
- Jessica E Thaxton
- Department of Pediatrics, Women and Infants Hospital of Rhode Island-Warren Alpert Medical School of Brown University, Providence, Rhode Island 02905, USA
| | | | | | | | | | | |
Collapse
|
128
|
Duncan FJ, Silva KA, Johnson C, King B, Szatkiewicz JP, Kamdar S, Ong DE, Napoli JL, Wang J, King LE, Whiting DA, McElwee KJ, Sundberg JP, Everts HB. Endogenous retinoids in the pathogenesis of alopecia areata. J Invest Dermatol 2013; 133:334-43. [PMID: 23014334 PMCID: PMC3546144 DOI: 10.1038/jid.2012.344] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Alopecia areata (AA) is an autoimmune disease that attacks anagen hair follicles. Gene array in graft-induced C3H/HeJ mice revealed that genes involved in retinoic acid (RA) synthesis were increased, whereas RA degradation genes were decreased in AA compared with sham controls. This was confirmed by immunohistochemistry in biopsies from patients with AA and both mouse and rat AA models. RA levels were also increased in C3H/HeJ mice with AA. C3H/HeJ mice were fed a purified diet containing one of the four levels of dietary vitamin A or an unpurified diet 2 weeks before grafting and disease progression followed. High vitamin A accelerated AA, whereas mice that were not fed vitamin A had more severe disease by the end of the study. More hair follicles were in anagen in mice fed high vitamin A. Both the number and localization of granzyme B-positive cells were altered by vitamin A. IFNγ was also the lowest and IL13 highest in mice fed high vitamin A. Other cytokines were reduced and chemokines increased as the disease progressed, but no additional effects of vitamin A were seen. Combined, these results suggest that vitamin A regulates both the hair cycle and immune response to alter the progression of AA.
Collapse
Affiliation(s)
- F. Jason Duncan
- Department of Nutrition, The Ohio State University, Columbus, OH
| | | | - Charles Johnson
- Department of Nutrition, The Ohio State University, Columbus, OH
| | | | | | | | - David E. Ong
- Vanderbilt University Medical Center, Nashville, TN
| | | | | | | | | | | | - John P. Sundberg
- The Jackson Laboratory, Bar Harbor, ME
- Vanderbilt University Medical Center, Nashville, TN
| | - Helen B. Everts
- Department of Nutrition, The Ohio State University, Columbus, OH
| |
Collapse
|
129
|
Huang M, Sun R, Wei H, Tian Z. Simultaneous knockdown of multiple ligands of innate receptor NKG2D prevents natural killer cell-mediated fulminant hepatitis in mice. Hepatology 2013; 57:277-88. [PMID: 22806577 DOI: 10.1002/hep.25959] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 06/21/2012] [Indexed: 02/03/2023]
Abstract
UNLABELLED NKG2D activation plays an important role in initiating and maintaining liver inflammation, and blockade of NKG2D recognition becomes a promising approach to alleviate liver inflammation. Treatment by silencing NKG2D ligands on hepatocytes, but not NKG2D on circulating immune cells, is more liver-specific, and simultaneous knockdown of multiple NKG2D ligands on hepatocytes will be more efficient in liver disease intervention. Here, we constructed a single vector that could simultaneously express multiple short hairpin RNAs (shRNAs) against all murine NKG2D ligands including Rae1, Mult1, and H60. After hydrodynamic injection of plasmid containing the three shRNA sequences (shRae1-shMult1-shH60), also called pRNAT-shRMH, we found the expression of all three NKG2D ligands on hepatocytes was downregulated both on messenger RNA and protein levels. Moreover, natural killer (NK) cell-mediated NKG2D-dependent fulminant hepatitis of the mice was alleviated, along with inactivation of hepatic NK cells, by pRNAT-shRMH if compared with its counterpart RNA interference vectors against single or double ligands. The therapeutic efficacy of pRNAT-shRMH was equivalent to that of injecting three monoclonal antibodies against Rae1, Mult1, and H60. For better in vivo application, we constructed a recombinant adenovirus containing pRNAT-shRMH (called Ad-RMH) with efficient hepatotropic infection capacity and observed that Ad-RMH intravenous injection exerted a similar therapeutic efficiency as plasmid pRNAT-shRMH hydrodynamic injection. Noticeably, simultaneous knockdown of multiple human NKG2D ligands (MICA/B, ULBP2, and ULBP3) also significantly attenuated NK cell cytolysis against human NKG2D ligand-positive hepatocyte L-02 cells, suggesting a possible translation into human settings. CONCLUSION Simultaneous knockdown of multiple ligands of NKG2D prevents NK cell-mediated fulminant hepatitis and is a potential therapeutic approach to treat liver diseases.
Collapse
Affiliation(s)
- Mei Huang
- Department of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | | | | | | |
Collapse
|
130
|
Wang R, Natarajan K, Revilleza MJR, Boyd LF, Zhi L, Zhao H, Robinson H, Margulies DH. Structural basis of mouse cytomegalovirus m152/gp40 interaction with RAE1γ reveals a paradigm for MHC/MHC interaction in immune evasion. Proc Natl Acad Sci U S A 2012; 109:E3578-87. [PMID: 23169621 PMCID: PMC3529088 DOI: 10.1073/pnas.1214088109] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Natural killer (NK) cells are activated by engagement of the NKG2D receptor with ligands on target cells stressed by infection or tumorigenesis. Several human and rodent cytomegalovirus (CMV) immunoevasins down-regulate surface expression of NKG2D ligands. The mouse CMV MHC class I (MHC-I)-like m152/gp40 glycoprotein down-regulates retinoic acid early inducible-1 (RAE1) NKG2D ligands as well as host MHC-I. Here we describe the crystal structure of an m152/RAE1γ complex and confirm the intermolecular contacts by mutagenesis. m152 interacts in a pincer-like manner with two sites on the α1 and α2 helices of RAE1 reminiscent of the NKG2D interaction with RAE1. This structure of an MHC-I-like immunoevasin/MHC-I-like ligand complex explains the binding specificity of m152 for RAE1 and allows modeling of the interaction of m152 with classical MHC-I and of related viral immunoevasins.
Collapse
Affiliation(s)
- Rui Wang
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kannan Natarajan
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Maria Jamela R. Revilleza
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Lisa F. Boyd
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Li Zhi
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Huaying Zhao
- Dynamics of Macromolecular Assembly Section, Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomolecular Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892; and
| | - Howard Robinson
- National Synchrotron Light Source, Brookhaven National Laboratories, Upton, NY 11973
| | - David H. Margulies
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
131
|
Mitrović M, Arapović J, Traven L, Krmpotić A, Jonjić S. Innate immunity regulates adaptive immune response: lessons learned from studying the interplay between NK and CD8+ T cells during MCMV infection. Med Microbiol Immunol 2012; 201:487-95. [PMID: 22965169 DOI: 10.1007/s00430-012-0263-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 08/24/2012] [Indexed: 01/04/2023]
Abstract
Natural killer (NK) cells play a crucial role in early immune response against cytomegalovirus infection. A large and mounting body of data indicate that these cells are involved in the regulation of the adaptive immune response as well. By using mouse cytomegalovirus (MCMV) as a model, several groups provided novel insights into the role of NK cells in the development and kinetics of antiviral CD8(+) T cell response. Depending on infection conditions, virus strain and the genetic background of mice used, NK cells are either positive or negative regulators of the CD8(+) T cell response. At present, there is no unique explanation for the observed differences between various experimental systems used. In this review we discuss the mechanisms involved in the interplay between NK and CD8(+) T cells in the early control of MCMV infection.
Collapse
Affiliation(s)
- Maja Mitrović
- Department of Histology and Embryology, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | | | | | | | | |
Collapse
|
132
|
Hervieu A, Rébé C, Végran F, Chalmin F, Bruchard M, Vabres P, Apetoh L, Ghiringhelli F, Mignot G. Dacarbazine-mediated upregulation of NKG2D ligands on tumor cells activates NK and CD8 T cells and restrains melanoma growth. J Invest Dermatol 2012; 133:499-508. [PMID: 22951720 DOI: 10.1038/jid.2012.273] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dacarbazine (DTIC) is a cytotoxic drug widely used for melanoma treatment. However, the putative contribution of anticancer immune responses in the efficacy of DTIC has not been evaluated. By testing how DTIC affects host immune responses to cancer in a mouse model of melanoma, we unexpectedly found that both natural killer (NK) and CD8(+) T cells were indispensable for DTIC therapeutic effect. Although DTIC did not directly affect immune cells, it triggered the upregulation of NKG2D ligands on tumor cells, leading to NK cell activation and IFNγ secretion in mice and humans. NK cell-derived IFNγ subsequently favored upregulation of major histocompatibility complex class I molecules on tumor cells, rendering them sensitive to cytotoxic CD8(+) T cells. Accordingly, DTIC markedly enhanced cytotoxic T lymphocyte antigen 4 inhibition efficacy in vivo in an NK-dependent manner. These results underscore the immunogenic properties of DTIC and provide a rationale to combine DTIC with immunotherapeutic agents that relieve immunosuppression in vivo.
Collapse
|
133
|
Polymeric delivery of therapeutic RAE-1 plasmid to the pancreatic islets for the prevention of type 1 diabetes. J Control Release 2012; 162:606-11. [PMID: 22910142 DOI: 10.1016/j.jconrel.2012.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/01/2012] [Accepted: 08/06/2012] [Indexed: 12/31/2022]
Abstract
The activating receptor NKG2D plays an important role in the development of type-1 diabetes. Exploiting a natural phenomenon observed in tumors, plasmid DNA encoding for a soluble ligand to NKG2D (sRAE-1γ) was isolated and engineered into a plasmid expression system. A polymeric gene delivery system was developed to deliver the soluble RAE-1 plasmid to the pancreatic islets. The bioreducible cationic polymer poly(cystamine bisacrylamide-diamino hexane) (p(CBA-DAH)) was modified with poly(ethylene glycol) (PEG) and the targeting peptide CHVLWSTRC, known to target the EphA2 and EphA4 receptors. We observed a higher uptake of the targeting polymer Eph-PEG-p(CBA-DAH) in the pancreas of NOD mice compared to non-targeting controls. To evaluate the efficacy of preventing diabetes, the Eph-PEG-p(CBA-DAH)/RAE-1 complex (polyplex) was intravenously injected into 6-week-old female NOD mice. Within 17 weeks blood glucose levels were stabilized in animals injected with polyplex, while those treated without therapeutic plasmid developed progressive hyperglycemia. Additionally, the degree of insulitis and the infiltration of CD8⁺ T-cells in the polyplex treated group were improved over the targeting polymer only treated group. The current study suggests that the therapy of the Eph-PEG-p (CBA-DAH) delivering therapeutic sRAE-1 gene may be used to protect β-cells from autoimmune destruction and prevent type-1 diabetes.
Collapse
|
134
|
Kasahara M, Yoshida S. Immunogenetics of the NKG2D ligand gene family. Immunogenetics 2012; 64:855-67. [PMID: 22843249 DOI: 10.1007/s00251-012-0638-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 07/18/2012] [Indexed: 12/31/2022]
Abstract
NKG2D ligands (NKG2DLs) are a group of major histocompatibility complex (MHC) class I-like molecules, the expression of which is induced by cellular stresses such as infection, tumorigenesis, heat shock, tissue damage, and DNA damage. They act as a molecular danger signal alerting the immune system for infected or neoplastic cells. Mammals have two families of NKG2DL genes: the MHC-encoded MIC gene family and the ULBP gene family encoded outside the MHC region in most mammals. Rodents such as mice and rats lack the MIC family of ligands. Interestingly, some mammals have NKG2DL-like molecules named MILL that are phylogenetically related to MIC, but do not function as NKG2DLs. In this paper, we review our current knowledge of the MIC, ULBP, and MILL gene families in representative mammalian species and discuss the origin and evolution of the NKG2DL gene family.
Collapse
Affiliation(s)
- Masanori Kasahara
- Department of Pathology, Hokkaido University Graduate School of Medicine, North-15 West-7, Sapporo 060-8638, Japan.
| | | |
Collapse
|
135
|
Liu XV, Ho SSW, Tan JJ, Kamran N, Gasser S. Ras activation induces expression of Raet1 family NK receptor ligands. THE JOURNAL OF IMMUNOLOGY 2012; 189:1826-34. [PMID: 22798674 DOI: 10.4049/jimmunol.1200965] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
NK cells play a crucial role in innate immunity against tumors. In many human tumors, Ras is chronically active, and tumor cells frequently express ligands for the activating NK cell receptor NKG2D. In this study, we report that Ras activation upregulates the expression of Raet1 protein family members Rae1α and Rae1β in mouse and ULBP1-3 in human cells. In addition, Ras also induced MHC class I chain-related protein expression in some human cell lines. Overexpression of the constitutively active H-RasV12 mutant was sufficient to induce NKG2D ligand expression. H-RasV12-induced NKG2D ligand upregulation depended on Raf, MAPK/MEK, and PI3K, but not ATM or ATR, two PI3K-like kinases previously shown to induce NKG2D ligand expression. Analysis of the 5' untranslated regions of Raet1 family members suggested the presence of features known to impair translation initiation. Overexpression of the rate-limiting translation initiation factor eIF4E induced Rae1 and ULBP1 expression in a Ras- and PI3K-dependent manner. Upregulation of NKG2D ligands by H-RasV12 increased sensitivity of cells to NK cell-mediated cytotoxicity. In summary, our data suggest that chronic Ras activation is linked to innate immune responses, which may contribute to immune surveillance of H-Ras transformed cells.
Collapse
Affiliation(s)
- Xi V Liu
- Immunology Programme, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456
| | | | | | | | | |
Collapse
|
136
|
Abstract
Passive immunotherapy, including adoptive T-cell therapy and antibody therapy, has shown encouraging results in cancer treatment lately. However, active immunotherapy of solid cancers remains an elusive goal. It is now known that the human innate immune system recognizes pathogen-associated molecular patterns conserved among microbes or damage-associated molecular patterns released from tissue injuries to initiate adaptive immune responses during infection and tissue inflammation, respectively. In contrast, how the innate immune system recognizes endogenously arising cancer remains poorly understood at the molecular level, which poses a significant roadblock to the development of active cancer immunotherapy. We hereby review the current knowledge of how solid cancers directly and indirectly interact with cells of the human innate immune system, with a focus on the potential effect of such interactions to the resultant adaptive immune responses against cancer. We believe that understanding cancer and innate immune system interactions may allow us to better manipulate the adaptive immune system at the molecular level to develop effective active immunotherapy against cancer. Current and future perspectives in clinical development that exploits these molecular interactions are discussed.
Collapse
Affiliation(s)
- Yanan Liu
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1738
| | - Gang Zeng
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1738
| |
Collapse
|
137
|
Abstract
INTRODUCTION NKG2D (natural killer group 2, member D) is expressed on the surface of all mouse and human NK cells, and subpopulation of T cells. Stimulation of NK cells through NKG2D triggers cell-mediated cytotoxicity and induces the production of cytokines. NKG2D binds to family of unique ligands with structurally similar to MHC class I, however, NKG2D ligands can be up-regulated in their expression on stressed cells including tumor cells unlike conventional MHC class I molecules. Mounting evidences clearly implicate that NKG2D recognition plays an important role in tumor immune surveillance. AREAS COVERED While NKG2D detect for potentially dangerous cells, various inhibitory and/or escape mechanisms counteract immune surveillance system and thereby limit effective elimination of transformed tumor cells. In addition, tumors often generate an immunosuppressive microenvironment where inhibitory molecules or cytokines negatively effect the function of anti-tumor immune responses. NKG2D ligand expression can be up-regulated by transcriptional or posttranscriptional mechanisms, therefore, certain therapy targeting those regulatory mechanisms could regain the expression of NKG2D ligands on tumor cells to be detected by the host immune responses. EXPERT OPINION Our knowledge in the precise mechanism of anti-tumor immunity is rapidly increasing. While NKG2D is known as primary cytotoxicity receptor in NK cell activation by recognizing 'induced-self' ligands on stressed cells including tumor cells, there are increasing evidences that NKG2D recognition can result in both immune activation and immune silencing. Future combined application of conventional cancer therapy and new therapy utilizing such stress-induced recognition systems will provide a novel opportunity to control malignant tumor progression of cancer disease.
Collapse
Affiliation(s)
- Yoshihiro Hayakawa
- The University of Tokyo, Graduate School of Pharmaceutical Sciences, Laboratory of Cancer Biology and Molecular Immunology, Bunkyo-ku, Japan.
| |
Collapse
|
138
|
Vivier E, Ugolini S, Blaise D, Chabannon C, Brossay L. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol 2012; 12:239-52. [PMID: 22437937 DOI: 10.1038/nri3174] [Citation(s) in RCA: 632] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Natural killer (NK) cells and natural killer T (NKT) cells are subsets of lymphocytes that share some phenotypical and functional similarities. Both cell types can rapidly respond to the presence of tumour cells and participate in antitumour immune responses. This has prompted interest in the development of innovative cancer therapies that are based on the manipulation of NK and NKT cells. Recent studies have highlighted how the immune reactivity of NK and NKT cells is shaped by the environment in which they develop. The rational use of these cells in cancer immunotherapies awaits a better understanding of their effector functions, migratory patterns and survival properties in humans.
Collapse
Affiliation(s)
- Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Université d'Aix-Marseille, Marseille, France.
| | | | | | | | | |
Collapse
|
139
|
Yoshida S, Mohamed RH, Kajikawa M, Koizumi J, Tanaka M, Fugo K, Otsuka N, Maenaka K, Yagita H, Chiba H, Kasahara M. Involvement of an NKG2D ligand H60c in epidermal dendritic T cell-mediated wound repair. THE JOURNAL OF IMMUNOLOGY 2012; 188:3972-9. [PMID: 22403443 DOI: 10.4049/jimmunol.1102886] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dendritic epidermal T cells (DETCs) found in mouse skin are NKG2D-positive γδ T cells involved in immune surveillance and wound repair. It is assumed that the interaction of an NKG2D receptor on DETCs and an MHC class I-like NKG2D ligand on keratinocytes activates DETCs, which then secrete cytokines promoting wound repair. However, direct evidence that DETC activation through NKG2D signaling promotes wound repair is not available. In the present study, we generated mAbs for an NKG2D ligand H60c previously suggested to be expressed specifically on skin keratinocytes. Local administration of H60c-specific mAb inhibited activation of DETCs and significantly delayed wound repair. Likewise, administration of NKG2D-specific mAb impaired wound repair to a similar extent. The delay in wound closure resulting from the blockade of the NKG2D pathway was comparable to that observed in γδ T cell-deficient mice. These results indicate that H60c/NKG2D interactions play a critical role in wound repair. Reassessment of binding affinities showed that H60c monomers bind to NKG2D with affinity (K(d) = 26 ± 3.2 nM) comparable to those of other high-affinity NKG2D ligands. H60c is transcribed not only in skin but also in tissues such as tongue and female reproductive tract known to contain epithelium-resident γδ T cells expressing invariant TCRs, suggesting a more general role for H60c in the maintenance of epithelial integrity.
Collapse
Affiliation(s)
- Shigeru Yoshida
- Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Kahraman A, Fingas CD, Syn WK, Gerken G, Canbay A. Role of stress-induced NKG2D ligands in liver diseases. Liver Int 2012; 32:370-82. [PMID: 22097967 DOI: 10.1111/j.1478-3231.2011.02608.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 06/15/2011] [Indexed: 12/24/2022]
Abstract
Cell death by apoptosis is a prominent feature in a variety of liver diseases. It is likely that apoptosis is the initial cellular response to hepatocyte and biliary injury, which then leads to the initiation of cellular and cytokine cascades culminating in hepatocyte death with subsequent fibrosis and cirrhosis. This sequence of events is of paramount clinical importance. Recently, soluble forms of the major histocompatibility complex class I-related chains A and closely related B (MIC A and B) were reported to be increased in patients with a variety of liver diseases. MIC A and B are cell surface glycoproteins that function as indicators for cellular stress and thus activate circulating cytotoxic natural killer (NK) cells. The interaction between MIC A and B with their cognate receptor natural killer group 2 member D (NKG2D) culminates in enhanced liver cell death, which is mediated in part by apoptotic mechanisms. The present overview focuses on the role of the stress-induced NKG2D ligands MIC A and B in diverse liver diseases. Critical insights into these complex relations may help to promote rationally based therapies in liver diseases. Importantly, we hope that this overview will help to stimulate further studies into mechanisms by which stress ligands mediate cell death and its sequale.
Collapse
Affiliation(s)
- Alisan Kahraman
- University Clinic Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | | | | |
Collapse
|
141
|
Blockade of NKG2D Synergized With CTLA4-Ig in Promoting Long-Term Graft Survival in Murine Models of Cardiac Transplantation [RETRACTED]. Transplantation 2012; 93:356-63. [DOI: 10.1097/tp.0b013e31823ffce7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
142
|
Zloza A, Kohlhapp FJ, Lyons GE, Schenkel JM, Moore TV, Lacek AT, O'Sullivan JA, Varanasi V, Williams JW, Jagoda MC, Bellavance EC, Marzo AL, Thomas PG, Zafirova B, Polić B, Al-Harthi L, Sperling AI, Guevara-Patiño JA. NKG2D signaling on CD8⁺ T cells represses T-bet and rescues CD4-unhelped CD8⁺ T cell memory recall but not effector responses. Nat Med 2012; 18:422-8. [PMID: 22366950 PMCID: PMC3436127 DOI: 10.1038/nm.2683] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/20/2012] [Indexed: 02/03/2023]
Abstract
CD4-unhelped CD8(+) T cells are functionally defective T cells primed in the absence of CD4(+) T cell help. Given the co-stimulatory role of natural-killer group 2, member D protein (NKG2D) on CD8(+) T cells, we investigated its ability to rescue these immunologically impotent cells. We demonstrate that augmented co-stimulation through NKG2D during priming paradoxically rescues memory, but not effector, CD8(+) T cell responses. NKG2D-mediated rescue is characterized by reversal of elevated transcription factor T-box expressed in T cells (T-bet) expression and recovery of interleukin-2 and interferon-γ production and cytolytic responses. Rescue is abrogated in CD8(+) T cells lacking NKG2D. Augmented co-stimulation through NKG2D confers a high rate of survival to mice lacking CD4(+) T cells in a CD4-dependent influenza model and rescues HIV-specific CD8(+) T cell responses from CD4-deficient HIV-positive donors. These findings demonstrate that augmented co-stimulation through NKG2D is effective in rescuing CD4-unhelped CD8(+) T cells from their pathophysiological fate and may provide therapeutic benefits.
Collapse
Affiliation(s)
- Andrew Zloza
- Department of Surgery, Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Abstract
The external surfaces of the body, such as the skin and the gastrointestinal mucosal membrane, are an important line of defence preventing the invasion of microorganisms and their products. Mucosal immune cells, especially intraepithelial lymphocytes, are involved in maintaining the integrity of these epithelial barriers. They contribute towards the tolerance to commensal organisms, which occupy these same sites, and to the immune responses against harmful organisms and their products. The composition of the microbiota is influenced by immune cells as well as external environmental factors, especially the use of antibiotics and diet. There is an increasing appreciation that the microbiota affects systemic immune responses in addition to local immunity. Failure to control the occupancy by microorganisms may result in the disruption of the delicate homeostasis between beneficial and harmful microorganisms and contribute to inflammatory pathologies. This review will discuss some of our current understanding of the impact of immune cells and diet on the microbiota.
Collapse
Affiliation(s)
- Emmanuelle Moens
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | | |
Collapse
|
144
|
Klezovich-Bénard M, Corre JP, Jusforgues-Saklani H, Fiole D, Burjek N, Tournier JN, Goossens PL. Mechanisms of NK cell-macrophage Bacillus anthracis crosstalk: a balance between stimulation by spores and differential disruption by toxins. PLoS Pathog 2012; 8:e1002481. [PMID: 22253596 PMCID: PMC3257302 DOI: 10.1371/journal.ppat.1002481] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 11/28/2011] [Indexed: 01/08/2023] Open
Abstract
NK cells are important immune effectors for preventing microbial invasion and dissemination, through natural cytotoxicity and cytokine secretion. Bacillus anthracis spores can efficiently drive IFN-γ production by NK cells. The present study provides insights into the mechanisms of cytokine and cellular signaling that underlie the process of NK-cell activation by B. anthracis and the bacterial strategies to subvert and evade this response. Infection with non-toxigenic encapsulated B. anthracis induced recruitment of NK cells and macrophages into the mouse draining lymph node. Production of edema (ET) or lethal (LT) toxin during infection impaired this cellular recruitment. NK cell depletion led to accelerated systemic bacterial dissemination. IFN-γ production by NK cells in response to B. anthracis spores was: i) contact-dependent through RAE-1-NKG2D interaction with macrophages; ii) IL-12, IL-18, and IL-15-dependent, where IL-12 played a key role and regulated both NK cell and macrophage activation; and iii) required IL-18 for only an initial short time window. B. anthracis toxins subverted both NK cell essential functions. ET and LT disrupted IFN-γ production through different mechanisms. LT acted both on macrophages and NK cells, whereas ET mainly affected macrophages and did not alter NK cell capacity of IFN-γ secretion. In contrast, ET and LT inhibited the natural cytotoxicity function of NK cells, both in vitro and in vivo. The subverting action of ET thus led to dissociation in NK cell function and blocked natural cytotoxicity without affecting IFN-γ secretion. The high efficiency of this process stresses the impact that this toxin may exert in anthrax pathogenesis, and highlights a potential usefulness for controlling excessive cytotoxic responses in immunopathological diseases. Our findings therefore exemplify the delicate balance between bacterial stimulation and evasion strategies. This highlights the potential implication of the crosstalk between host innate defences and B. anthracis in initial anthrax control mechanisms. NK cells are important immune effectors that perform a surveillance task and react to transformed, stressed, and virally infected cells. They represent a first-line defence against cancer and pathogen invasion. Different pathogens trigger distinct NK-cell activation pathways. The Bacillus anthracis spore is the highly resistant form that enters the host and provokes anthrax. This microbe kills through a combination of acute bacterial infection and devastating toxemia. In the present study, we characterise the crosstalk between NK cells and spores, as well as the strategies used by B. anthracis to evade initial control mechanisms and impact anthrax pathogenesis. Our findings exemplify the spores' property to efficiently drive a high production of IFN-γ by NK cells, as well as the complex pathways used for activation which require both cytokine and cellular signaling. B. anthracis subverts this response through its toxins by paralysing essential NK cell functions. Furthermore, edema toxin from B. anthracis blocks natural cytotoxicity without affecting IFN-γ secretion. The CyaA toxin of Bordetella pertussis possesses the same enzymatic activity and has a similar effect. The high efficiency of these toxins in blocking cytotoxicity in vivo implies possible exploitation of their subverting activity to modulate excessive cytotoxic responses in immunopathological diseases.
Collapse
MESH Headings
- Animals
- Bacillus anthracis/immunology
- Bacterial Toxins/pharmacology
- Cells, Cultured
- Female
- Homeostasis/drug effects
- Homeostasis/immunology
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Macrophage Activation/drug effects
- Macrophage Activation/immunology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Receptor Cross-Talk/drug effects
- Receptor Cross-Talk/immunology
- Spores, Bacterial/immunology
- Spores, Bacterial/physiology
Collapse
Affiliation(s)
- Maria Klezovich-Bénard
- Laboratoire Pathogénie et Toxi-Infections Bactériennes, Institut Pasteur, Paris, France
- CNRS URA 2172, Paris, France
| | - Jean-Philippe Corre
- Laboratoire Pathogénie et Toxi-Infections Bactériennes, Institut Pasteur, Paris, France
- CNRS URA 2172, Paris, France
| | | | - Daniel Fiole
- Unité Interactions Hôte-Agents Pathogènes, Département de Microbiologie, Institut de Recherche Biomédicale des Armées, La Tronche, France
- Laboratoire Interdisciplinaire de Physique, UMR 5588 CNRS/Université Joseph Fourier, St-Martin-d'Hères, France
| | - Nick Burjek
- Laboratoire Pathogénie et Toxi-Infections Bactériennes, Institut Pasteur, Paris, France
- CNRS URA 2172, Paris, France
| | - Jean-Nicolas Tournier
- Unité Interactions Hôte-Agents Pathogènes, Département de Microbiologie, Institut de Recherche Biomédicale des Armées, La Tronche, France
- École du Val-de-Grâce, Paris, France
| | - Pierre L. Goossens
- Laboratoire Pathogénie et Toxi-Infections Bactériennes, Institut Pasteur, Paris, France
- CNRS URA 2172, Paris, France
- * E-mail:
| |
Collapse
|
145
|
Markiewicz MA, Wise EL, Buchwald ZS, Pinto AK, Zafirova B, Polic B, Shaw AS. RAE1ε ligand expressed on pancreatic islets recruits NKG2D receptor-expressing cytotoxic T cells independent of T cell receptor recognition. Immunity 2012; 36:132-41. [PMID: 22244846 DOI: 10.1016/j.immuni.2011.11.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 09/01/2011] [Accepted: 11/01/2011] [Indexed: 02/07/2023]
Abstract
The mechanisms by which cytotoxic T lymphocytes (CTLs) enter and are retained in nonlymphoid tissue are not well characterized. With a transgenic mouse expressing the NKG2D ligand retinoic acid early transcript 1ε (RAE1ε) in β-islet cells of the pancreas, we found that RAE1 expression was sufficient to induce the recruitment of adoptively transferred CTLs to islets. This was dependent on NKG2D expression by the CTLs and independent of antigen recognition. Surprisingly, the recruitment of CTLs resulted in the subsequent recruitment of a large number of endogenous lymphocytes. Whereas transgenic mice did not develop diabetes, RAE1 expression was sufficient to induce insulitis in older, unmanipulated transgenic mice that was enhanced by viral infection and pancreatic inflammation. These results demonstrate that the expression of an NKG2D ligand in islets is sufficient to recruit CTLs regardless of their antigen specificity and to induce insulitis.
Collapse
Affiliation(s)
- Mary A Markiewicz
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
146
|
Slavuljica I, Krmpotić A, Jonjić S. Manipulation of NKG2D ligands by cytomegaloviruses: impact on innate and adaptive immune response. Front Immunol 2011; 2:85. [PMID: 22566874 PMCID: PMC3342069 DOI: 10.3389/fimmu.2011.00085] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 12/12/2011] [Indexed: 01/20/2023] Open
Abstract
NKG2D is a potent activating receptor expressed on NK cells, NKT cells, γδ T cells, and CD8 T cells. NKG2D recognizes cell surface molecules structurally related to MHC class I proteins induced by infection or other type of cellular stress. The engagement of NKG2D leads to NK cell cytotoxicity and cytokine secretion or to a co-stimulation of CD8 T cells. Both human and mouse cytomegalovirus (CMV) have evolved numerous mechanisms to evade NKG2D-mediated immune response. This review describes the mechanisms used by CMV to inhibit NKG2D ligand expression and the recent advances in exploiting the NKG2D recognition pathway for mounting efficient and long-lasting immune response.
Collapse
Affiliation(s)
- Irena Slavuljica
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka Rijeka, Croatia
| | | | | |
Collapse
|
147
|
Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity. Proc Natl Acad Sci U S A 2011; 109:1210-5. [PMID: 22167808 DOI: 10.1073/pnas.1118834109] [Citation(s) in RCA: 266] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Infections with HIV, hepatitis B virus, and hepatitis C virus can turn into chronic infections, which currently affect more than 500 million patients worldwide. It is generally thought that virus-mediated T-cell exhaustion limits T-cell function, thus promoting chronic disease. Here we demonstrate that natural killer (NK) cells have a negative impact on the development of T-cell immunity by using the murine lymphocytic choriomeningitis virus. NK cell-deficient (Nfil3(-/-), E4BP4(-/-)) mice exhibited a higher virus-specific T-cell response. In addition, NK cell depletion caused enhanced T-cell immunity in WT mice, which led to rapid virus control and prevented chronic infection in lymphocytic choriomeningitis virus clone 13- and reduced viral load in DOCILE-infected animals. Further experiments showed that NKG2D triggered regulatory NK cell functions, which were mediated by perforin, and limited T-cell responses. Therefore, we identified an important role of regulatory NK cells in limiting T-cell immunity during virus infection.
Collapse
|
148
|
Xia M, Guerra N, Sukhova GK, Yang K, Miller CK, Shi GP, Raulet DH, Xiong N. Immune activation resulting from NKG2D/ligand interaction promotes atherosclerosis. Circulation 2011; 124:2933-43. [PMID: 22104546 DOI: 10.1161/circulationaha.111.034850] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The interplay between the immune system and abnormal metabolic conditions sustains and propagates a vicious feedback cycle of chronic inflammation and metabolic dysfunction that is critical for atherosclerotic progression. It is well established that abnormal metabolic conditions, such as dyslipidemia and hyperglycemia, cause various cellular stress responses that induce tissue inflammation and immune cell activation, which in turn exacerbate the metabolic dysfunction. However, molecular events linking these processes are not well understood. METHODS AND RESULTS Tissues and organs of humans and mice with hyperglycemia and hyperlipidemia were examined for expression of ligands for NKG2D, a potent immune-activating receptor expressed by several types of immune cells, and the role of NKG2D in atherosclerosis and metabolic diseases was probed with the use of mice lacking NKG2D or by blocking NKG2D with monoclonal antibodies. NKG2D ligands were upregulated in multiple organs, particularly atherosclerotic aortas and inflamed livers. Ligand upregulation was induced in vitro by abnormal metabolites associated with metabolic dysfunctions. Using apolipoprotein E-deficient mouse models, we demonstrated that preventing NKG2D functions resulted in a dramatic reduction in plaque formation, suppressed systemic and organ inflammation mediated by multiple immune cell types, and alleviated abnormal metabolic conditions. CONCLUSIONS The NKG2D/ligand interaction is a critical molecular link in the vicious cycle of chronic inflammation and metabolic dysfunction that promotes atherosclerosis and might be a useful target for therapeutic intervention in the disease.
Collapse
Affiliation(s)
- Mingcan Xia
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, 115 Henning Bldg, University Park, PA 16802, USA
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Blevins KS, Jeong JH, Ou M, Brumbach JH, Kim SW. EphA2 targeting peptide tethered bioreducible poly(cystamine bisacrylamide-diamino hexane) for the delivery of therapeutic pCMV-RAE-1γ to pancreatic islets. J Control Release 2011; 158:115-22. [PMID: 22062690 DOI: 10.1016/j.jconrel.2011.10.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 10/17/2011] [Accepted: 10/20/2011] [Indexed: 02/05/2023]
Abstract
The pathogenesis of type-1 diabetes is complicated, and a clear, single mechanism has yet to be identified. Reports have indicated that the activating receptor NKG2D plays an important role in the development of disease. Exploiting a natural phenomenon observed in tumors, plasmid DNA encoding for a soluble ligand to NKG2D (sRAE-1γ) was isolated and engineered into a plasmid expression system. A polymeric gene delivery system was developed to deliver the soluble RAE-1 plasmid locally to the pancreatic islets for the prevention of type-1 diabetes. The bioreducible cationic polymer poly(cystamine bisacrylamide-diamino hexane) (p(CBA-DAH)) was modified with poly(ethylene glycol) (PEG) and the targeting peptide CHVLWSTRC, known to target the EphA2 and EphA4 receptors. The PEG serves to improve stability and tissue selectivity, while the peptide will target EphA2 and A4, overexpressed in the pancreatic microvasculature. The targeting polymer Eph-PEG-p(CBA-DAH) shows selective uptake by the target cell line, indicative of the targeting properties that will be seen in systemic administration. Using the delivery system, the therapeutic plasmid can be delivered to the pancreas, reduce interactions between the beta-cells and infiltrating NKG2D positive lymphocytes, and effectively protect beta-cells from autoimmune destruction and prevent type 1 diabetes.
Collapse
Affiliation(s)
- Katherine S Blevins
- Department of Bioengineering, University of Utah, 20 South 2030 East, Salt Lake City, UT 84112-5820, USA
| | | | | | | | | |
Collapse
|
150
|
The cooperating mutation or “second hit” determines the immunologic visibility toward MYC-induced murine lymphomas. Blood 2011; 118:4635-45. [DOI: 10.1182/blood-2010-10-313098] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractIn Eμ-myc transgenic animals lymphoma formation requires additional genetic alterations, which frequently comprise loss of p53 or overexpression of BCL-2. We describe that the nature of the “second hit” affects the ability of the immune system to contain lymphoma development. Tumors with disrupted p53 signaling killed the host more rapidly than BCL-2 overexpressing ones. Relaxing immunologic control, using Tyk2−/− mice or by Ab-mediated depletion of CD8+ T or natural killer (NK) cells accelerated formation of BCL-2–overexpressing lymphomas but not of those lacking p53. Most strikingly, enforced expression of BCL-2 prolonged disease latency in the absence of p53, whereas blocking p53 function in BCL-2–overexpressing tumors failed to accelerate disease. This shows that blocking apoptosis in p53-deficient cells by enforcing BCL-2 expression can mitigate disease progression increasing the “immunologic visibility.” In vitro cytotoxicity assays confirmed that high expression of BCL-2 protein facilitates NK and T cell–mediated killing. Moreover, we found that high BCL-2 expression is accompanied by significantly increased levels of the NKG2D ligand MULT1, which may account for the enhanced killing. Our findings provide first evidence that the nature of the second hit affects tumor immunosurveillance in c-MYC–driven lymphomas and define a potential shortcoming of antitumor therapies targeting BCL-2.
Collapse
|