GET THE APP

Cytokine Intervention: A Double Edged Sword in the Nkg2d System R
Immunome Research

Immunome Research
Open Access

ISSN: 1745-7580

+44-20-4587-4809

Review Article - (2015) Volume 0, Issue 0

Cytokine Intervention: A Double Edged Sword in the Nkg2d System Regulation

Ana Montalban-Arques1, Gregor Gorkiewicz1, Victor Mulero2 and Jorge Galindo-Villegas2*
1Institute of Pathology, Medical University of Graz, Graz, Austria
2Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
*Corresponding Author: Jorge Galindo-Villegas, Department of Cell Biology and Histology, Faculty of Biology, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain, Tel: (34)-868-88-3938, Fax: (34)-868-88-3963 Email:

Abstract

The natural killer group 2 members D (NKG2D) is an activating receptor which plays a major role in immune surveillance, and the detection and elimination of malignant tumors and infected cells. NKG2D acts over both arms of the vertebrate immune response, and is expressed in some human and mouse myelopoietic, γδ T, NKT and CD4+ cells, but is present in all NK and CD8+ T cells in humans and activated mouse CD8+ T cells. In humans, eight ligands which selectively bind to the NKG2D receptor have been identified. These ligands are not systemically expressed, but are triggered in response to stress and expressed only under specific pathological states. Several research results point to the importance of cytokines for increasing expression of NKG2D to restore the functionality of NK cells as well as their ligands in the target cells. However, the NKG2D system itself in an activated state, also release pro and anti-inflammatory cytokine transcripts to establish communication with other cells or for self-regulation. Additionally, type I antiviral interferon is largely produced. Such cytokine interactions could be regarded as a double edged sword. This behavior is emphasized by a discrepancy regarding the functionality of cytokines which interact with, or on the NKG2D system. Indeed, they seem to protect the host and rather can induce ligand expression, cell proliferation or dissemination of malignant tumors, generating complicated cytokine-mediated messenger loops which are far from being fully understood. Whatever the case, cytokines related to the NKG2D system could be an attractive and useful target for immunotherapeutic approaches. Thus, here we briefly review recent findings on the main aspects involved in the regulation of this system and, particularly, attempt to clarify the role played by cytokines in the activating or inhibitory function they exert over the NKG2D system in different contexts.

<

Keywords: Cytokines; Immunotherapy; MICA/B; NKG2D system; Stress; Tumor; ULBP

Abbreviations

APC: Antigen Presenting Cell; CD: Cluster of Differentiation; CIK: Cytokine-induced Killer Cell; DC: Dendritic Cell; DAP: Disulphide Adaptor Molecule; 5-FU: Fluorouracil; γc: Gamma chain; CSF2: Granulocyte-Macrophage Colony-Stimulating Factor; Grb2: Growth Factor Receptor-Bound Protein 2; H60: Histocompatibility Antigen 60; IBD: Inflammatory Bowel Disease; IEL : Intraepithelial Lymphocyte; IFN: Interferon; IL: Interleukin; JNK: Jun N-terminal Kinase; KIR2DL1: Killer Cell Immunoglobulin-Like Receptor, Two Domains, Long Cytoplasmic Tail 1; LTA: Lymphotoxin Alpha; MHC: Major Histocompatibility Complex; MICA/B: MHC class I Chain–Related Proteins A/B; miRNA: micro RNA; MyD88: Myeloid Differentiation Primary Response 88; NK: Natural Killer; NKG2D: Natural Killer Group 2 Member D; NKRs: Natural Killer Receptors; NKT: Natural Killer T Cell; FOLFOX4: Oxaliplatin-Folinic Acid- Fluorouracil; PBMCs: Peripheral Blood Mononuclear Cells; PI3K: Phosphatidyl Inositol 3 Kinase; Src: Proto-oncogene Tyrosine-protein Kinase; RAET-1: Retinoic Acid Early Transcript; RAE-1: Ribonucleic Acid Export 1; Syk: Regulatory T Cells (Treg) Spleen Tyrosine Kinase; TGFB-1: Transforming Growth Factor Beta 1; TNF: Tumor Necrosis Factor; ULBP: UL16-Binding Proteins; YINM: Tyrosine-Isoleucine- Asparagine-Methionine Motif; ZAP70: Zeta-Chain-Associated Protein Kinase 70

Introduction

Cells communicate with one another through extracellular signalling proteins know as cytokines. Each cytokine is produced immediately in response to many different stressors and binds to the extracelular domain of either one or two matching membranebound receptors denoted as α-, β- or γ-chain [1]. Due this matching specificity, cytokines enables the rapid propagation of immune signaling and have long been regarded as the regulators of host responses to infection, immune responses, inflammation, and trauma [2]. Nevertheless, the immune system’s primary task telling friend from foe is not one that can be easily solved solely through the indiscriminate production of cytokines to pass on instructions to cells. Faced with a serious threat such as infection, wound or tumors, the system needs to mount a defense response immediately. However, it also needs to know when not to interfere with innocuous visitors. To deal with dangerous outsiders, vertebrates have developed two lines of defense. These are termed learned immunity, which is acquired by exposure to a pathogen, either from the environment or through a vaccine, and innate immunity, which is the immediate hard-wired reaction to outside invaders [3]. A wide set of immune competent cells actively interact between the two arms of the immune system to mount an effective inflammatory defense against a particular threat. However, inflammatory processes intend to mediate against invaders, if not properly controlled, could irreversibly damage host tissues [4]. At cellular level, the key ingredients in linking the innate and adaptive responses are the innate-like cells. The best example of this particular cell type is the natural killer (NK) cell, a subset of lymphocytes involved in early defenses triggered through receptors that respond to infected, transformed and/or stresed cells [5,6]. Among the different receptors expressed by NK cells, the most studied is the NKG2D, which, upon stimulation, binds to any of the eight different ligands (MICA, MICB and ULBP 1 to 6) of the NKG2D system [7]. Upon detection of some pathological alterations in autologous cells, stimulation of the receptor can lead to the enhancement of innate immune functions, mediated by NK cells and myeloid cells, and the enhancement of adaptive immunity mediated by CD8+ and γδ T cells [8]. In human NK cells, at least, crosslinking with multivalent soluble ligands of NKG2D stimulates the production of several cytokines, including interferon gamma (IFN-γ), tumor-necrosis-factor alpha (TNF-α), lymphotoxin and the colony stimulating factor 2 (granulocyte-macrophage) (CSF2), as well as chemokines such as CCL4 and CCL1 [9-11]. However, the cytokines produced by NKG2D positive cells seldom play a single role. This notion is supported by several recent investigations which have established that, upon activation [12,13] or inhibition [14,15] of the NKG2D system, the same set of cytokines might be acting in both responses. Several research groups have reported functional aspects of cytokines in which IL-2 [16,17], IL-15 [18,19] or the combination of IL- 15 and TNF-α [20], have participated as key molecules, and observed a marked increased expression of the NKG2D receptor. In other studies, the dual functional role of cytokines has been demonstrated using IL-4 [21] or IL-21[22], two well known inducers of NK cell maturation [23]. This last study suggested that both cytokines, the IL-4 and the IL-21, act as negative regulators of the NKG2D system in both NK and CD8+ T cells. Additionally, the modulation of NKG2D ligands has also been observed by IFN-γ, which downregulates the expression of MICA and ULBP2 [24,25], and IFN-α that upregulates the same [26,27]. Nevertheless, in a regular setting, the activation of the NKG2D system by any of its ligands, triggers cytokine production to activate innate or adaptive immune responses following a canonical pathway. Therefore, to define the exact role that a certain cytokine may play in the NKG2D system is far from straightforward since, as mentioned above, evidence suggests their involvement in both directions depends on the context of the study (Table 1). In this brief review, we intend to summarize and discuss recent advances made in our understanding of the double edged nature of cytokines in the regulation of the NKG2D system, highlighting the importance of the full co-operation between both components to achieve a succesful immune response.

Name Cytokine Producer cell Cell target Organ target Condition Organism Main function Ref
Interleukin 1, alpha Il1a Macrophages Tumor cell Skin Merkel Cell Carcinoma M Down-regulation of RAE-1, tumor-cell resistance to NK-mediated control of virus-induced tumors [86]
Interleukin 1, beta Il1b
Interleukin 2 IL2 T cells NK - Viral infection /tumor H Up-regulation of NKG2D-DAP10 surface expression [69], [68]
Interleukin 4 Il4 Th cells CD8+ T cells - Th2 pathology M Down-regulation of NKG2D and increasing of the activation threshold of memory CD8+ T cells [21]
Interleukin 6 IL6 dNK dFibroblast Placenta HCMV infection H Control and spreading suppression of HCMV infection by NKG2D of dNKs [72]
Interleukin 7 IL7 - NK - Viral infection /tumor H Up-regulation of NKG2D-DAP10 surface expression [69]
Interleukin 8 IL8 Myoblasts NK Muscle Inflammatory myopathies H Up-regulation of NKG2D and NK cell-mediated lysis of muscle cells [123]
Interleukin 9 IL9 CD8+ T cells in vitro - Viral infection /tumor H Decreased expression of antiinflammatory cytokines through NKG2D expression [89]
Interleukin 10 IL10 DCs DCs - HIV infection H Immune dysfunction due to NK cell-mediated elimimatiom of DCs [124]
Interleukin 12 IL12 PBMNc NK - HCMV infection H Down-regulation of NKG2D to control NK cell reactivity against normal cells expressing NKG2D ligands [125]
NK - - Infection/tumor H Up-regulate the expression of NKG2D in NK cells [9]
Interleukin 13 IL13 CD8+ T cells - - Viral infection /tumor H Decreased expression of antiinflammatory cytokines through NKG2D expression [89]
Interleukin 15 IL15 in vitro NK - H Up-regulation of NKG2D-DAP10 surface expression [69]
IELs Intestine Celiac disease H Induction of MICA surface expression [128], [66]
DCs NK - Hepatitis C H Up-regulation of MICA/B in DCs and activation of NK cells [25]
Interleukin 17 IL17 CD4+ T cells IELs Intestine Crohn's Disease H Th17 immune response [126]
Interleukin 18 IL18 Macrophages, DCs, Keratinocytes Leukemia cells - Leukemia H Up-regulation of ULBP2 [85]
in vitro NK - Tumor H Down-modulation avoidance of NKG2D by TGF- β1 (together with IL-2) [68]
Interleukin 21 IL21 Ovaric cancer cell line NK Ovary Ovarian cancer H Up-regulation of NKG2D-MICA expression and the cytokines IFN-γ and TNF-α [70]
Th cells NK/CD8+ T cells - Autoimmunity H Down-regulation of NKG2D/DAP10 in autoimmune diseases (immunotherapy) [22]
Interleukin 22 IL22 CD4+ T cells IELs Intestine Crohn's Disease H Potentiate Th17 immune response [126]
Interleukin 33 Il33 Macrophages Tumor cell Skin Merkel Cell Carcinoma M Down-regulation of RAE-1 for tumor-cell resistance to NK-mediated control of virus-induced tumors [86]
Tumor necrosis factor Tnf/TNF
in vitro ECs Endothelium Activated endothelium H Inhibition of NK cytotoxic activity by soluble MICA [90]
Interferon alpha 1 IFNA1 DCs NK - Hepatitis C H Up-regulation of MICA/B in DCs and activation of NK cells [25]
in vitro Tumor cell - Infection/tumor H Promotion of MICA expression in tumor cells, enhancing their sensitivity to NK lysis [26]
Interferon beta 1 IFNB1 PBMNc NK - HCMV infection H Down-regulation of NKG2D to control NK cell reactivity against normal cells expressing NKG2D ligands [125]
Interferon gamma ifng/IFNG CD4+ T cells IELs Intestine Crohn's Disease H Up-regulation of MICA in IELs during Crohn's disease [127]
chNKG2D T cells Tumor cells - Lymphoma/ Murin ovarian cancer M Stimulation of DCs, NK and T cells with the subsequent antitumor effect [89], [88]
in vitro ECs Endothelium Activated endothelium H Inhibition of NK cytotoxic activity by soluble MICA [90]
    Colony stimulating factor 2 (granulocyte-macrophage) Csf2/CSF2 Ovaric cancer cell line NK Ovary Ovarian cancer M Up-regulation of NKG2D-MICA expression and the cytokines IFN-γ and TNF-α [70]
chNKG2D T cells Tumor cells Lymph node/ Ovary Lymphoma/ Murin ovarian cancer M Stimulation of DCs, NK and T cells with the subsequent antitumor effect [89], [88]
NK in vitro - Infection/tumor H ULBPs induce NK cells to produce this chemokine to recruit and activate NK and other immune cells [9]
Transforming growth factor beta 1 TGFB1 in vitro NK - Viral infection /tumor H Supression of NKG2D-DAP10 surface expression and MICA [87], [67], [69]
Chemokine (C-C motif) ligand 4 CCL4 NK in vitro - Infection/tumor H ULBPs induce NK cells to produce these chemokines to recruit and activate NK and other immune cells [9]
Chemokine (C-C motif) ligand 1 CCL1 H
Chemokine (C-C motif) ligand 2 Ccl2/CCL2 Tumor cells NK Liver Carcinoma M Elimination of senescent tumors by NK cells due to p53 and NKG2D ligand expression in tumor cells [71]
dNK dFibroblast Placenta HCMV infection H Crucial role of NKG2D in dNK cells in controlling HCMV infection and spreading [72]
Chemokine (C-X-C motif) ligand 1 CXCL1 H

Table 1: Major cytokines and chemokines related to the NKG2D system Abreviations: CCL: Chemokine c-c motif Ligand; CXCL: Chemokine c-x-c motif Ligand; chNKG2D: chimeric NKG2D; DCs: Dendritic Cells; dFibroblast: Decidual Fibroblast; dNK: Decidual Natural killer; ECs: Epithelial Cells; H: Human; HCMV: Human Citomegalovirus; HIV: Human Immunodeficiency Virus; IELs: Intestinal Epithelial Cells; IFN: Interferon; IL: Interleukin; M: mouse; NK: Natural Killer; PBMCs: Peripheral Blood Mononuclear Cells; TGF: Transforming Growth Factor; Th: T helper; TNF: Tumor Necrosis Factor; (-) not specified.

Natural Killer (NK) Cells: the Link between Innate and Adaptive Immunity

Herberman [28] and Kiessling [29] described NK cells for the first time in 1975, but not until the last decade a true and unique identity of this cellular group were firmly established. NK cells are a subset of lymphocytes that lack antigen-specific cell surface receptors [30] which provide innate effector mechanisms against viruses and tumor cells through direct cytotoxic effects and the release of cytokines [5]. NK cells detect microbial insults by means of innate receptors like the Tolllike receptors, or in response to pro-inflammatory cytokines produced by dendritic cells (DCs) [31-33] macrophages and neutrophils [34]. To achieve their basic effector functions, fulfil their intrinsic development, and to survive or proliferate, all NK cells are dependent on cytokines of the common gamma chain (γc) family (IL-2, IL-7, IL- 15, and IL-21) [35-37]. Of particular interest is interleukin-2, a key cytokine which enhances NK cell proliferation both in vitro and in vivo [38] potentiates and mediates NK-cell functions, orchestrates the interaction with T-lymphocytes and DCs [39], while contributing to achieve the homeostasis of mature NK cells [36]. In a different setting, during a viral infection, type I interferons (IFN-I), especially IFN-α, and type II IFN-γ are potent cytokines that trigger NK cell activation, turning them into surveillance entities that limit viral replication by binding to and sequestering specific virus-encoded proteins [40,41]. This viral dampening is a notable feature of NK cells, although recent studies have demonstrated that, in certain conditions, activated NK cells also have the ability to produce immunosuppressive cytokines such as TGF-β1 and IL-10, which may induce tolerance in local NK cells [42,43] and contribute to virus-mediated T-cell exhaustion [44,45]. Thus, this behavior suggests that, in addition to their positive role in combating viral treats when activated by IFNs, NK cells also have a negative regulatory role, releasing anti-inflammatory cytokines during acute and a chronic virus infection [41]. In addition, a different NK feature promotes their rapid extravasation from blood vessels or specific tissue to enhance the recruitment of immune cells at infection sites or tumor foci. These is achieved through the secretion of a wide set of chemokines, which include CCL2 (MCP-1), CCL3 (MIP1-a), CCL4 (MIP1-b), CCL5 (RANTES), XCL1 (lymphotactin) and CXCL8 (IL-8) [31]. This secretion of chemokines by NKs is an additional key feature that promotes their co-localization with other hematopoietic cells in areas where inflammatory processes are taking place or in tumor immunosurveillance sites [46]. But, despite this ability of NK cells to mediate the transcription of cytokines, they need to be primed in advance by exogenous cytokines such as IL-15 [47,48], IL-12 [49] or IL-18 [50] to complete their full effector potential.

Hallmarks of NK Cells Receptors

NK cells possess a particular set of receptors that can sense microbial and non-microbial signals emitted by target cells through a variety of lectin-type receptor families. Of particular interest are the type II transmembrane proteins, which include the natural killer receptor (NKR) family such as FcgRIIIA activator [51], or the mammalian inhibitory CD94/NKG2 receptors [6]. CD94 forms heterodimers with various NKG2 receptors but not with NKG2D, which is a homodimer. In addition to CD94/NKG2 receptors, in some rodent species the inhibitory Ly49 family of lectin-type NKRs is also present. Additionally, cytokine receptors coupled to the common (γc) IL-15R, IL-2R and IL-21R are also involved in NK cell development and their effector functions. Among these receptors, IL-15R is essential for the maturation and survival of all NK cells, while IL-1R in humans [52] and IL-18R in mice [50], which are linked to the adapter protein MyD88, have a particular role in the NK cell maturation process. Thus, based on the above evidence, it can be concluded that, despite the considerable intra-and interspecific variation in gene numbers and complexity among mammalian Ig-type and lectin-type NKRs, they are expressed on NK cells, bind to MHC I or proteins that share structural similarities with MHC I, and can inhibit or activate target cell killing and/or cytokine release through competing signaling pathways [6].

Cytokine Induced Killer Cells (CIK): Antitumor Inducers

Cytokine-induced killer cells (CIK) cells are a heterogeneous subset of ex-vivo expanded T lymphocytes with a mixed T-NK phenotype [53]. Among the several advantages of conducting research with CIKs, the most interesting are their rapid proliferation in vitro, the strong antitumor activity and the broader target tumor spectrum they possess, compared with other antitumor effector cells, have been investigated so far [54]. Additionally, CIK cells have the capacity to provide a non-crossresistant mechanism of antitumor activity that can be incorporated in surgery, radiation or chemotherapy treatments [55]. Thus, in the last years application of CIK cells in combination with chemotherapy has raised a powerful tool to treat cancer patients. In a classical approach, these cells are derived from peripheral blood mononuclear cells (PBMCs), but can also be generated from bone marrow or umbilical cord precursors [56]. The ex vivo expansion of CIK cells takes 3-4 weeks and is driven by the addition of IFN-γ, Ab-anti CD3 and IL-2 [57] to the cell culture. When the expansion is finished, the predominant subsets of CD3+ T lymphocytes are: CD3+CD56- and CD3+CD56+, formerly known as cytotoxic NK-like T cells. The antitumor activity of CIK cells relies solely on cell–cell contact [17]. Briefly, following an elegant research approach using antibodies against CD54, CD11c and NKG2D, Verneris and colleagues succeeded to attenuate the cytotoxic effect by blocking the cell-cell interaction, thus demonstrating that such effect is based on MHC-unrestricted mechanisms which rely on the interaction of the NKG2D receptor present in CIK cells and the ligands expressed in tumor cells. Therefore NKG2D-ligand interaction triggers the last step of tumor-killing, which is mediated by perforin and granzyme. These enzymes play a fundamental role in generating a pore in the cell membrane, which finally causes apoptosis of the target cell. Thus, capacity of CIKs as effector cells has been observed to be active in solid and hematological malignancies, which has been tested in vitro and confirmed in vivo with murine models of human tumor xenograft transplants. In a study of the cellular capacity of IL-2 primed CIK cells, resulted in the downregulation of the IL-18 levels which were mediated by IFN-γ [58,59]. Liu and colleagues, used oxaliplatinfolinic acid-fluorouracil (FOLFOX4) in combination with CIK cells as adjuvant to treat patients with gastric cancer [60]. The process described in their study identified the need for exogenous cytokines to prime CIKs. Briefly, the CIKs obtained from PBMCs and stimulated with INF‑γ, IL‑1α, IL‑2 and anti‑CD3 MAb for use as adjuvants resulted in increased NK cells activity and higher CD3+ and CD4+ T total cell counts, whereas CD8+ T cells decreased in number. In a parallel study, Shi and colleagues used fluorouracil (5-FU) with CIKs as adjuvant to successfully treat advanced gastric cancer [54]. The CIK cells used were activated by IFN-γ and IL-2 from the patients’ PBMCs. The mix of 5-FU and activated CIK cells prolonged the disease-free state and significantly improved overall survival in patients with intestinal-type tumors. Thus, these studies conducted by different groups demonstrate that CIK cell priming by cytokines is essential for their activation and positive activity over malignant tumors. Interestingly, to determine the clinical value of autologous immunocyte therapy as a standard treatment against cancer, patients with colorectum, lung, breast, kidney, or stomach cancer received the DC vaccination once a week for six weeks and a CIK cell injection six times within four days. A positive cell-mediated cytotoxicity response was recorded, and improvements in physical strength, appetite and sleeping status were observed. Thus, they concluded that the therapy was safe since no serious adverse sideeffects similar to those caused by chemotherapy and radiotherapy were observed [61]. Additionally, in recent publications it was suggested that CIK cells treated with IL-15 [62], or in combination with IL-2 [63] improve their cytotoxicity against leukemic cells or lung cancer cells, respectively. In both cases, the percentage of CD3+CD56+ cells was significantly increased in IL-15 stimulated CIK cells and their proliferative rate was higher. Taken together, these results realize that cytokine-activated cells may have a beneficial effect in the near future on the treatment of patients with cancer [61].

Cells Respond to Stress in Different Ways

Broadly defined, stress is the state in which cells deviate from the status quo in response to sudden environmental changes or frequent fluctuations in environmental factors [64]. Such changes can damage existing molecules, including proteins, mRNAs, DNA and lipids, and, if the damage is not dealt with, a metabolic imbalance may result in a redox alteration [65]. Several mechanisms are immediately triggered to overcome the stress responses; for example, damaged macromolecules are promptly cleared [66], molecular chaperones are induced [67,68] or growth arrest and “emergency” gene transcription kicks in [69]. When cells can no longer cope with excessive damage, straightforward cell death may occur through necroptosis. In contrast, stressors like heat shock, oxidative stress, viral infection or DNA damage may induce the expression of particular extracellular ligands [70]. Tumor cells can be stressed by multiple intrinsic or extrinsic stimuli, both of which may promote membrane expression or the release of ‘eat-me‘, ‘danger‘, or ‘killing signals’ that will facilitate immune recognition and the final eradication of stressed tumor cells [71]. Molecules from the MHC-I are ligands, which inhibit or activate receptors expressed on NK cells and T cells. The expression of MHC-I is frequently impaired in virus-infected or tumor cells, which results in lack of engagement on the inhibitory receptors and hence the activation of NKs. Hence, class I serves as a positive indicator for the integrity of cells, protecting against NK cell attack [72]. In contrast, NKG2D ligands, MICA/B and members of the ULBP/RAET1 may signal cellular distress and evoke immune responses. Although NK cells can eliminate tumor cells with the loss or aberrant expression of class I, the interaction of MICA with NKG2D may promote antitumor responses in the presence of class I, depending on the balance of multiple inhibitory and activating signals, the relative amounts of receptors and their ligands, and the state of NK cell activation. However, this balance should be achieved through the differential expression of NKG2D-ligands, the modulation of the receptor and the cytotoxic activity of NKL cells after cell-cell contact with the tumor cells [73]. Thus, the interaction of NKG2D with MICA/B or ULBP/RAET1 may enhance diverse antitumor innate NK cell and antigen-specific T-cell responses. This recognition, in which self-encoded ligands are induced in stressed cells, is known as “induced self-recognition” [74].

The NKG2D System

Properties of the system in infection and tumor immunity

NKG2D is a cell-activating receptor that mediates non-MHC restricted and TCR-independent lysis. Cells expressing NKG2D are modulated by cytokines. Nevertheless, the NKG2D system has effector functions in which high levels of IFN-γ and TNF-α, among other cytokines with specific cytotoxic properties are produced [75]. Due this high plasticity, NKG2D is deeply involved in tumor immunosurveillance, which plays an important role in the cytotoxic activity of NK and CD8+ T cells. NKG2D is a C-type, lectin-like, type II transmembrane glycoprotein [76,77] which functions as an activating receptor through an interaction with the adaptor signalling molecules disulphide adaptor molecule (DAP)10 and/or (DAP)12 [78,79]. When the receptor is ligated, DAP10 provides signals that recruit the p85 subunit of phosphatidylinositol 3-kinase (PI3K) and a complex of GRB2 and VAV1 to complete the activation [80], whereas DAP12 activates protein tyrosine kinase Syk and ZAP70 [81], but this activation is not through the cytoplasmic signalling motif Tyrosine-Isoleucine-Asparagine-Methionine Motif (YINM) [82]. Therefore, the engagement of NKG2D with ligands in NK cells result in the induction of degranulation and cytokine production. The NKG2D receptor, is not only expressed by all NK cells [83] but also by NKT, subsets of γδ T cells [8], CD8+ T cells [71], activated mouse macrophages and a small subset of CD4+ T cells in humans. Thus, to trigger an effective immune response mediated by NK cells, the only prerequisite is engagement of the receptor with one of its ligands by means of a stimulus. However, in mouse T cells, co-stimulation by a subset of γδ T cells resident in the skin may be needed [84,85] . This co-stimulatory function is also present in αβ CD8+ T cells, but is much more noticeable when the cells lack CD28 marker expression, which is recognized as the normal co-stimulatory receptor for T cells [86]. Interestingly, the stimulation of CD8+ T cells with IL-15 together with CD3 generates a potent activation that leads to the engagement of the ligand with NKG2D. Such NKG2D priming driven by IL-15, triggers NK and T cell cytotoxicity, which is a key negative regulator in certain T cell–mediated pathologies such as in celiac disease [87]. Therefore, NKG2D is able to generate activating signals, which in some cases may co-activate cellular killing and produce undesired cytokines by NK cells and certain subsets of T cells. In the case of TGF-β1, it has been reported to decrease DAP10 levels and, as a consequence, NKG2D protein level [88]. Thus, to avoid the-induced NKG2D downregulation by TGF-β1 in NK cells, a previous activator engagement is required by the combination of IL-2/IL-18 with the receptor [89]. However, (γc) cytokines (IL-2, IL-7, IL-15) [89,90] and IL-18 [89] promote the induction of DAP10 and, consequently, the surface expression of the receptor. In ovarian cancer mice model, IL-21 has been also reported to play an activating role for the NKG2D expression [91] as well as innate tumor rejection activity, in tumors that can elicit an NKG2D-mediated immune response [92]. Therefore, two cytokines of the gamma-chain, the IL-7 and IL-15 seems to be key mediators in the upregulation of the NKG2D-DAP10 axis expression by NK cells. For instance, while IL-15 plays a common signaling role, also primes and regulates the NKG2D expression through the phosphorylation and further upregulation of the adaptor molecule DAP10 [93].

The role of cytokines on the NKG2D system is double edged

Several examples may illustrate this cytokine double edged behaviour. In one case, the enhanced expression of IL-15 increases the impaired expression of the NKG2D ligand MICA in monocyte/ macrophages and induces the abnormal expansion of NKG2D+CD4+ T cells (NK cell-like CD4+ T cells) [75], like in a rare blood vessel disease, the granulomatosis with polyangiitis (Wegener’s) [94]. Interferons are rapidly induced when NKG2D is activated, particularly IFN-α may upregulate NKG2D expression but can also dampen the expression of inhibitory receptors like NKG2A or KIR2DL1. The IFN-γ, which plays an inhibitory role in the expression of NKG2D, promotes the expression of NKG2A [27].In the case of chemokines, CCL1, CCL2, CCL4 and CXCL1 are known for their capacity to recruit and activate NK cells to the target cells [9,95,96]. Taken together, these results point to the difficulties involved in in vivo prediction on how the NKG2D system might behave under different scenarios. Thus, from above description is clear that a selection of cytokines may be specifically required for activation or inhibition of the NKG2D receptor or their associated ligands. However, the ways in which these cytokines may behave do not always correlate with their primary associated functional role. Further studies are required to throw light on how these proteins are integrated in the signaling pathways of NK cell activation and how the engagement of different activating receptors controls their activity.

NKG2D Ligands Modulation by Cytokines

NKG2D has multiple ligands including MHC class I chain-related-A (MICA), -B (MICB), and several UL-16 binding proteins (ULBP), which are preferentially expressed after cellular stress, infection, or DNA damage [78,97]. In humans, the MICA, MICB and the ULBPs, also known as retinoic acid early transcript RAET1 proteins, have been seen to be upregulated in cancer and infected cells [8,78,81,98]. These ligands are recognized by the immune activating receptor NKG2D. Upon engagement, allows the recognition and further elimination of infected and malignant cells. In mice, there are no orthologs for the MICA and MICB genes, but a family of genes orthologous to the human ULBP/RAET1 family is present. These genes are highly polymorphic and encode proteins that fall into three subgroups of NKG2D ligands, including five different isoforms of retinoic acid early inducible-1 (RAE-1) proteins (Rae-1α, Rae-1β, Rae-1γ, Rae-1δ and Rae-1ε), one murine UL16-binding protein-like transcript 1 (MULT1) protein, and three different isoforms of H60 proteins (H60a, H60b, H60c) [99]. In fact, to make the signalling pathways even more complex, there are over 60 MICA and 20 different MICB alleles. Human RAET1 genes are also polymorphic, as are Rae-1 and the histocompatibility antigen (H60) genes in mouse [100]. The effect of such polymorphism is that all the ligands engage with the NKG2D receptor with different degrees of affinity, which may affect the threshold of NK and T cell activation [7]. Moreover, some of the NKG2D ligands may be excreted to the extracellular environment, stay attached to the cell surface, remain at the transcription level biogenesis, be stabilized on the RNA or stabilized and cleaved from the cell membrane. Lastly, the effect of cytokines is no less confusing. As example, IL-18 increases the susceptibility of target cells by inducing the surface expression of ULBP2 in leukemia cells [101]. However the pro-inflammatory cytokines IL-1α, IL-1β, IL-33, and TNF-α down regulate RAE-1 expression and susceptibility to NK cell-mediated cytotoxicity, leading to the avoidance of NK cellmediated control of virus-induced tumors in mice [102]. In a similar way, TGF- β1 inhibits the transcription of the ligand MICA in humans [103]. In turn, CSF2 upregulates NKG2D-MICA expression [91] and stimulates antigen-presenting cells (APC) to initiate an antitumor response [104,105]. This last effect is shared with IFN-γ. Furthermore, IFN-γ upregulates MICA in intestinal epithelial lymphocytes (IELs) in Crohn’s disease. However, it can also have a negative effect, leading to the inhibition of NK cytotoxicity by soluble MICA. This effect has been observed to be shared with TNF-α [106].

NKG2D Ligands Modulation by Alternative Players

Among the different cellular regulatory systems, it has recently been demonstrated that the translocation of mRNAs encoding the MICA/B in naive cells is inhibited by microRNAs (miRNA) [107]. However, upon stress, the transcription of MICA/B mRNAs is substantially upregulated and significant protein levels are detected. This observation suggests that the mRNA levels, when they exceed the amount of controlling cellular miRNAs, result in the overexpression of MICA/B proteins [70]. Until now, p53 has never been regarded as being essential for the expression of NKG2D ligands in cells suffering DNA damage. Nevertheless, co-operation between p53 inducedtumor cell senescence and the innate immune system has recently been highlighted [108]. The restoration of p53 function in established carcinomas leads to tumor regression, but only in mice with an intact immune system. Also, the inflammatory cytokines IL-15 and CSF2, and the chemokines CCL2 and CXCL1 were upregulated in tumors following p53 reactivation, correlating with the recruitment of neutrophils, macrophages and NK cells into tumors, where they are responsible in tumor shrinkage [108]. Thus, it seems that input from p53 to the NKG2D system is crucial at some stages of the signaling cascade, directly modulating the transcription of cytokines by tumor cells. Other unexpected players are the epigenetic changes, manifested by inhibitors of histone deacetylase, which can also induce the surface expression of NKG2D ligands on tumor cells [109]. From an evolutive perspective, these ligands are fairly conserved among vertebrates, and thus are not exclusively expressed in human and mice, but they are widely distributed among mammals [110]. From the information presented above, it is clear that a large variety of NKG2D ligands exist. However, so far the explanation for this variability, and the intriguingly diversity in which they are regulated, remains a matter of speculation.

The Double Edged Activity Exerted by Cytokines on Cancer Cells

During cell transformation and tumorigenesis numerous stress pathways are activated in affected cells [81]. During malignancies, NKG2D ligands are transcriptionaly induced and highly expressed on the surface of tumor cells. The ligands expressed depend on the type of cancer, which expresses MICA/B and at least one member of the ULBP family [111]. The receptor NKG2D participates in immune surveillance and has the capacity of eliminate NKG2D ligand-positive tumor cells in an early developmental stage [7]. A major feature of the antitumor response is mediated through type 1 cytokines, like TNF-α, or the IFN-γ. Indeed, increased amounts of IFN-γ and TNF-α were measured after treatment in the serum of patients [112]. Additionally, other cytokines, LTA, IL-13, IL-10 and CSF2, are produced upon contact with susceptible tumor target cells [113]. These cytokines can exert differential effects on the regulation of NKG2D ligands [7]. For example, melanoma cells exposed to IFN-γ downregulate MICA and ULBP2, and IFN-γ also reduces the expression of mouse H60 in sarcomas [24,114]. This reduced expression of NKG2D ligands has a negative effect, diminishing the susceptibility of tumors to NK cytolysis. In the supernatant of cultured HeLa and K562 cells, soluble MICA (sMICA) was observed to be upregulated by IFN-γ, demonstrating that IFN-γ modulates MICA expression not only at the transcriptional level, but also at the post-translational level by promoting proteolytic cleavage [27]. In contrast, IFN-α increases the expression of MICA in tumor cells and thereby enhances their sensitivity to NK lysis [27]. However, chronic exposure to tumor cells expressing NKG2D ligand alters NKG2D signalling and may facilitate the evasion of tumor cells from NK cell reactions [115]. One of the mechanisms that malignant cells are known to use for this purpose is the shedding of NKG2D ligands into the sera of cancer patients [116], where they weaken the immune response by downmodulating the receptor on effector cells and producing the consequent impaired immune response [114]. TGF-β1 has also been shown to decrease the transcription of MICA, ULBP2 and ULBP4 in human glioma [117], and downregulate NKG2D receptor expression on effector cells [118], while blocking TGF-β1 can lead to increased NKG2D expression [88,119,120]. This suggests that TGF-β1 secreted by tumors is a major mechanism that tumor cells employ to evade the NKG2D-DAP10-mediated cytotoxicity. Moreover, Clayton et al. [121] reported that tumor-derived exosomes also express NKG2D ligands and directly interact with NK and CD8+ cells. This response was demonstrated to be highly dependent on exosomal TGF-β1 and to induce the reduction in surface NKG2D expression. Using a mouse model, it was demonstrated that MICA did not mediate the downregulation of the receptor NKG2D [122]. The observed downregulation was explained through the sustained stimulation with tumor cell–bound ligand that disassociates the NKG2D receptor from the intracellular calcium mobilization and the exertion of NK cell–mediated cytolysis, while it induces the continuous production of IFN-γ. These functional changes are associated with a low abundance of the NKG2D signalling adaptors DAP-10 and DAP-12 [115]. So it was concluded that the low expression of MICA and MICB on resistant tumor cells may be another mechanism that allows tumor cells to escape from CIK cell-mediated cytolysis [17]. Although cytokine stimulation of cells may overcome receptor inhibition mediated by soluble ligands. To illustrate this mechanism, Song and colleagues [89] demonstrated that the combination of IL-2 with IL-18 can protect the TGF-β1-induced NKG2D down-modulation in NK cells via the JNK pathway. In addition, NKG2D contributes to the anti-tumor responses elicited by IL-2 and IL-12 cytokine therapy. As regarded the chemokine CCL2, it has been reported to be an active player in the elimination of tumor cells through the induction of NK cell recruitment into the tumor driven by p53 [95]. This mechanism has been demonstrated to be NKG2D-dependent and mediated by the recognition of ribonucleic acid export 1 (RAE-1) proteins in mice. Moreover, by pharmacological reactivation, p53 in specific cell lines has been reported to stimulate the expression of ULBP2 [123,124]. Perhaps, the use of ectopic NKG2D– DAP10 expression triggers the tumor-promoting capacity through ligand-mediated NKG2D self-stimulation [125]. An overview of these regulation mechanisms is provided in Figure 1.

immunome-research-microenvironment-cytokines-significant

Figure 1: Within the tumor microenvironment, cytokines play a significant role in the regulation of the cytotoxic and cytolytic activity against tumor cells. Antigenpresenting cells (APC), dendritic cells (DCs) and macrophages, can up- or down-regulate the expression of NKG2D and NKG2D ligands on the surface of epithelial cells, leading to tumor elimination or tumor-cell resistance, respectively. The different output effects depend on the developmental stage of the tumor. Cytokines produced by NKG2D+ cells modulate ligand expression on the target cell and stimulate APC to exert antitumor effects. Schreiner [132] alter [133] Muntasell [134] pariente [135] allez [72]hüe [136].

A Key Player in Immunotherapy: the Cytokines

Cytokines directly stimulate immune effector and stromal cells at the tumor site and enhance recognition by cytotoxic mediators. Numerous animal model studies have demonstrated that cytokines have broad anti-tumor activity and this has been translated into a number of cytokine-based approaches for human therapy [126]. Additionally, the notable success of the targeted inhibition of several cytokines in patients with rheumatoid arthritis, psoriasis and many other diseases has fundamentally revised the treatment of inflammatory diseases. Together, these findings suggest that different conditions may share a common pathophysiology and may benefit from disruption of the cytokine network [127]. In cancer therapies, cytokines are critical for tumor immunosurveillance. Single-agent or the combination of cytokines with classical immune antibodies or TLR agonists resulted in potent CD8+ T cell-mediated antitumor effects [128,129]. The mix of CD40 antibody and IL-2 has been observed to have a synergistic antitumor effect [130,131], and a similar effect was observed when mice were treated with CpG motifs and IL-15 [129]. In both cases, the antitumor effects were dependent on the production of CD8+ T cells, IFN-γ, IL-12 and Fas ligand expression [129,130]. Additionally, it has been recognized that effector and memory CD8+ T cells express elevated levels of IL-12R and IL-18R, and secrete IFN-γ in response to stimulation with both cytokines [132]. In this setting, it is clear that the stimulation of CD8+ cells with cytokines results in antigen-nonspecific expansion, which is useful for immunotherapy in the treatment of advanced tumor models and represents a primary effector mechanism [131]. Interestingly, regulatory T (Treg) cells can inhibit NK cell cytolytic function and IFN-γ secretion, and have been shown to downregulate NKG2D in human and mouse NK cells through membrane-bound TGF-β1 [133]. However, not all the interactions between CD8+ cells and cytokines have been reproducible in NKG2D+CD4+ T cells. Even though , these cells proliferate and increase in number relative to other T cell populations, thereby causing imbalances in the lymphocyte pool and imposing an immunosuppressive cytokine milieu. In advanced cancer patients, however, cytokine mediated tumor expression and shedding, mainly of soluble MICA and B, can lead to a substantial proliferative expansions of the NKG2D+CD4+ T cells [125]. Thus, above presented evidences clearly point out that various clinical trials of immunotherapy for hematologic malignancies can induce regression to their pathophysiological state.

Conclusions

Based on the information reviewed above, it is clear that the cytokines involved on the regulation of the NKG2D system are an attractive target for therapy. However, many of the molecular processes associated in this system are not well understood and establishing their functional activity on particular scenarios remains a challenge. More knowledge is needed to understand the influences of these molecules on every single regulatory activity, ranging from target effector cells to the ligands they produce. A better molecular understanding on how cytokines regulate the effector response may provide important insights into the way in which the NKG2D system overcomes infection and combat tumors. We anticipate that answers to these questions will yield clinically useful information because the NKG2D system clearly has an essential role in removing harmful cellular components. Moreover, impairment or disabling of the system has been linked to many human pathologies including cancer, autoimmune responses like inflammatory bowel disease [134], Celiac disease [135] or several types of gastritis. With this in mind, this review will hopefully contribute to stimulate much interest into the search for new answers in this intriguing research field.

References

  1. Walsh ST (2012) Structural insights into the common gamma-chain family of cytokines and receptors from the interleukin-7 pathway. Immunol Rev 250: 303-316.
  2. Galindo-Villegas J, Garcia-Moreno D, de Oliveira S, Meseguer J, Mulero V (2012) Regulation of immunity and disease resistance by commensal microbes and chromatin modifications during zebrafish development. ProcNatlAcadSci U S A 109: E2605-2614.
  3. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S (2008) Functions of natural killer cells. Nat Immunol 9: 503-510.
  4. Yoder JA, Litman GW (2011) The phylogenetic origins of natural killer receptors and recognition: relationships, possibilities, and realities. Immunogenetics 63: 123-141.
  5. Spear P, Wu MR, Sentman ML, Sentman CL (2013) NKG2D ligands as therapeutic targets. Cancer Immun 13: 8.
  6. Raulet DH (2003) Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 3: 781-790.
  7. Sutherland CL, Chalupny NJ, Schooley K, VandenBos T, Kubin M, et al. (2002) UL16-binding proteins, novel MHC class I-related proteins, bind to NKG2D and activate multiple signaling pathways in primary NK cells. J Immunol 168: 671-679.
  8. Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, et al. (2001) ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 14: 123-133.
  9. Kubin M, Cassiano L, Chalupny J, Chin W, Cosman D, et al. (2001) ULBP1, 2, 3: novel MHC class I-related molecules that bind to human cytomegalovirus glycoprotein UL16, activate NK cells. Eur J Immunol 31: 1428-1437.
  10. Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, et al. (2004) Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nat Immunol 5: 1260-1265.
  11. Raulet DH (2004) Interplay of natural killer cells and their receptors with the adaptive immune response. Nat Immunol 5: 996-1002.
  12. Perona-Wright G, Mohrs K, Szaba FM, Kummer LW, Madan R, et al. (2009) Systemic but not local infections elicit immunosuppressive IL-10 production by natural killer cells. Cell Host Microbe 6: 503-512.
  13. Muntasell A, Magri G, Pende D, Angulo A, Lopez-Botet M (2010) Inhibition of NKG2D expression in NK cells by cytokines secreted in response to human cytomegalovirus infection. Blood 115: 5170-5179.
  14. Dhanji S, Teh HS (2003) IL-2-activated CD8+CD44high cells express both adaptive and innate immune system receptors and demonstrate specificity for syngeneic tumor cells. J Immunol 171: 3442-3450.
  15. Verneris MR, Karami M, Baker J, Jayaswal A, Negrin RS (2004) Role of NKG2D signaling in the cytotoxicity of activated and expanded CD8+ T cells. Blood 103: 3065-3072.
  16. Dann SM, Wang HC, Gambarin KJ, Actor JK, Robinson P, et al. (2005) Interleukin-15 activates human natural killer cells to clear the intestinal protozoan cryptosporidium. J Infect Dis 192: 1294-1302.
  17. Maasho K, Opoku-Anane J, Marusina AI, Coligan JE, Borrego F (2005) NKG2D is a costimulatory receptor for human naive CD8+ T cells. J Immunol 174: 4480-4484.
  18. Groh V, Bruhl A, El-Gabalawy H, Nelson JL, Spies T (2003) Stimulation of T cell autoreactivity by anomalous expression of NKG2D and its MIC ligands in rheumatoid arthritis. ProcNatlAcadSci U S A 100: 9452-9457.
  19. Ventre E, Brinza L, Schicklin S, Mafille J, Coupet CA, et al. (2012) Negative regulation of NKG2D expression by IL-4 in memory CD8 T cells. J Immunol 189: 3480-3489.
  20. Burgess SJ, Marusina AI, Pathmanathan I, Borrego F, Coligan JE (2006) IL-21 down-regulates NKG2D/DAP10 expression on human NK and CD8+ T cells. J Immunol 176: 1490-1497.
  21. Parrish-Novak J, Dillon SR, Nelson A, Hammond A, Sprecher C, et al. (2000) Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature 408: 57-63.
  22. Schwinn N, Vokhminova D, Sucker A, Textor S, Striegel S, et al. (2009) Interferon-gamma down-regulates NKG2D ligand expression and impairs the NKG2D-mediated cytolysis of MHC class I-deficient melanoma by natural killer cells. Int J Cancer 124: 1594-1604.
  23. Yadav D, Ngolab J, Lim RS, Krishnamurthy S, Bui JD (2009) Cutting edge: downregulation of MHC class I-related chain A on tumor cells by IFN-gamma-induced microRNA. J Immunol 182: 39-43.
  24. Jinushi M, Takehara T, Kanto T, Tatsumi T, Groh V, et al. (2003) Critical role of MHC class I-related chain A and B expression on IFN-alpha-stimulated dendritic cells in NK cell activation: impairment in chronic hepatitis C virus infection. J Immunol 170: 1249-1256.
  25. Zhang C, Niu J, Zhang J, Wang Y, Zhou Z, et al. (2008) Opposing effects of interferon-alpha and interferon-gamma on the expression of major histocompatibility complex class I chain-related A in tumors. Cancer Sci 99: 1279-1286.
  26. Herberman RB, Nunn ME, Lavrin DH (1975) Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int J Cancer 16: 216-229.
  27. Kiessling R, Klein E, Pross H, Wigzell H (1975) "Natural" killer cells in the mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the killer cell. Eur J Immunol 5: 117-121.
  28. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, et al. (2011) Innate or adaptive immunity? The example of natural killer cells. Science 331: 44-49.
  29. Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E (2005) Natural-killer cells and dendritic cells: "l'union fait la force". Blood 106: 2252-2258.
  30. Sivori S, Falco M, Della Chiesa M, Carlomagno S, Vitale M, et al. (2004) CpG and double-stranded RNA trigger human NK cells by Toll-like receptors: induction of cytokine release and cytotoxicity against tumors and dendritic cells. ProcNatlAcadSci U S A 101: 10116-10121.
  31. Alter G, Kavanagh D, Rihn S, Luteijn R, Brooks D, et al. (2010) IL-10 induces aberrant deletion of dendritic cells by natural killer cells in the context of HIV infection. J Clin Invest 120: 1905-1913.
  32. Moretta A, Marcenaro E, Sivori S, Della Chiesa M, Vitale M, et al. (2005) Early liaisons between cells of the innate immune system in inflamed peripheral tissues. Trends Immunol 26: 668-675.
  33. Becknell B, Caligiuri MA (2005) Interleukin-2, interleukin-15, and their roles in human natural killer cells. AdvImmunol 86: 209-239.
  34. Ma R, Xu YG, Yang XH, Hu XM, Li L, et al. (2006) Immunophenotypic features in leukemia of NK cell series. Zhongguo Shi Yan Xue Ye XueZaZhi 14: 35-38.
  35. Pillet AH, Bugault F, Theze J, Chakrabarti LA, Rose T (2009) A programmed switch from IL-15- to IL-2-dependent activation in human NK cells. J Immunol 182: 6267-6277.
  36. Kim JY, Huh K, Lee KY, Yang JM, Kim TJ (2009) Nickel induces secretion of IFN-gamma by splenic natural killer cells. ExpMol Med 41: 288-295.
  37. Caligiuri MA (1993) Low-dose recombinant interleukin-2 therapy: rationale and clinical applications. SeminOncol 20: 3-10.
  38. Lee SH, Miyagi T, Biron CA (2007) Keeping NK cells in highly regulated antiviral warfare. Trends Immunol 28: 252-259.
  39. Lang PA, Lang KS, Xu HC, Grusdat M, Parish IA, et al. (2012) Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity. ProcNatlAcadSci U S A 109: 1210-1215.
  40. Tu Z, Bozorgzadeh A, Pierce RH, Kurtis J, Crispe IN, et al. (2008) TLR-dependent cross talk between human Kupffer cells and NK cells. J Exp Med 205: 233-244.
  41. Schreiner B, Voss J, Wischhusen J, Dombrowski Y, Steinle A, et al. (2006) Expression of toll-like receptors by human muscle cells in vitro and in vivo: TLR3 is highly expressed in inflammatory and HIV myopathies, mediates IL-8 release and upregulation of NKG2D-ligands. FASEB J 20: 118-120.
  42. Lee SH, Kim KS, Fodil-Cornu N, Vidal SM, Biron CA (2009) Activating receptors promote NK cell expansion for maintenance, IL-10 production, and CD8 T cell regulation during viral infection. J Exp Med 206: 2235-2251.
  43. Brooks DG, Trifilo MJ, Edelmann KH, Teyton L, McGavern DB, et al. (2006) Interleukin-10 determines viral clearance or persistence in vivo. Nat Med 12: 1301-1309.
  44. Smyth MJ, Crowe NY, Hayakawa Y, Takeda K, Yagita H, et al. (2002) NKT cells - conductors of tumor immunity? CurrOpinImmunol 14: 165-171.
  45. Lucas M, Schachterle W, Oberle K, Aichele P, Diefenbach A (2007) Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 26: 503-517.
  46. Mortier E, Advincula R, Kim L, Chmura S, Barrera J, et al. (2009) Macrophage- and dendritic-cell-derived interleukin-15 receptor alpha supports homeostasis of distinct CD8+ T cell subsets. Immunity 31: 811-822.
  47. Guia S, Cognet C, de Beaucoudrey L, Tessmer MS, Jouanguy E, et al. (2008) A role for interleukin-12/23 in the maturation of human natural killer and CD56+ T cells in vivo. Blood 111: 5008-5016.
  48. Chaix J, Tessmer MS, Hoebe K, Fuseri N, Ryffel B, et al. (2008) Cutting edge: Priming of NK cells by IL-18. J Immunol 181: 1627-1631.
  49. Prochazka V, Papajik T, Gazdova J, Divoka M, Rozmanova S, et al. (2011) FcγRIIIA receptor genotype does not influence an outcome in patients with follicular lymphoma treated with risk-adapted immunochemotherapy. Neoplasma 58: 263-270.
  50. Hughes T, Becknell B, Freud AG, McClory S, Briercheck E, et al. (2010) Interleukin-1beta selectively expands and sustains interleukin-22+ immature human natural killer cells in secondary lymphoid tissue. Immunity 32: 803-814.
  51. Sangiolo D (2011) Cytokine induced killer cells as promising immunotherapy for solid tumors. J Cancer 2: 363-368.
  52. Shi L, Zhou Q, Wu J, Ji M, Li G, et al. (2012) Efficacy of adjuvant immunotherapy with cytokine-induced killer cells in patients with locally advanced gastric cancer. Cancer ImmunolImmunother 61: 2251-2259.
  53. Wang M, Shi SB, Qi JL, Tang XY, Tian J (2013) S-1 plus CIK as second-line treatment for advanced pancreatic cancer. Med Oncol 30: 747.
  54. Introna M, Franceschetti M, Ciocca A, Borleri G, Conti E, et al. (2006) Rapid and massive expansion of cord blood-derived cytokine-induced killer cells: an innovative proposal for the treatment of leukemia relapse after cord blood transplantation. Bone Marrow Transplant 38: 621-627.
  55. Lu PH, Negrin RS (1994) A novel population of expanded human CD3+CD56+ cells derived from T cells with potent in vivo antitumor activity in mice with severe combined immunodeficiency. J Immunol 153: 1687-1696.
  56. Huang J, Li C, Wang Y, Lv H, Guo Y, et al. (2013) Cytokine-induced killer (CIK) cells bound with anti-CD3/anti-CD133 bispecific antibodies target CD133(high) cancer stem cells in vitro and in vivo. ClinImmunol 149: 156-168.
  57. Kim JS, Park YS, Kim JY, Kim YG, Kim YJ, et al. (2012) Inhibition of human pancreatic tumor growth by cytokine-induced killer cells in nude mouse xenograft model. Immune Netw 12: 247-252.
  58. Liu H, Song J, Yang Z, Zhang X (2013) Effects of cytokine-induced killer cell treatment combined with FOLFOX4 on the recurrence and survival rates for gastric cancer following surgery. ExpTher Med 6: 953-956.
  59. Cui Y, Yang X, Zhu W, Li J, Wu X, et al. (2013) Immune response, clinical outcome and safety of dendritic cell vaccine in combination with cytokine-induced killer cell therapy in cancer patients. OncolLett 6: 537-541.
  60. Tao Q, Chen T, Tao L, Wang H, Pan Y, et al. (2013) IL-15 Improves the Cytotoxicity of Cytokine-induced Killer Cells Against Leukemia Cells by Upregulating CD3+CD56+ Cells and Downregulating Regulatory T Cells As Well As IL-35. J Immunother 36: 462-467.
  61. Wei C, Wang W, Pang W, Meng M, Jiang L, et al. (2013) The CIK cells stimulated with combination of IL-2 and IL-15 provide an improved cytotoxic capacity against human lung adenocarcinoma. Tumour Biol.
  62. Leung AK, Sharp PA (2010) MicroRNA functions in stress responses. Mol Cell 40: 205-215.
  63. Kultz D (2005) Molecular and evolutionary basis of the cellular stress response. Annu Rev Physiol 67: 225-257.
  64. Kroemer G, Marino G, Levine B (2010) Autophagy and the integrated stress response. Mol Cell 40: 280-293.
  65. Buchberger A, Bukau B, Sommer T (2010) Protein quality control in the cytosol and the endoplasmic reticulum: brothers in arms. Mol Cell 40: 238-252.
  66. Richter K, Haslbeck M, Buchner J (2010) The heat shock response: life on the verge of death. Mol Cell 40: 253-266.
  67. Spriggs KA, Bushell M, Willis AE (2010) Translational regulation of gene expression during conditions of cell stress. Mol Cell 40: 228-237.
  68. Stern-Ginossar N, Gur C, Biton M, Horwitz E, Elboim M, et al. (2008) Human microRNAs regulate stress-induced immune responses mediated by the receptor NKG2D. Nat Immunol 9: 1065-1073.
  69. Ullrich E, Bonmort M, Mignot G, Kroemer G, Zitvogel L (2008) Tumor stress, cell death and the ensuing immune response. Cell Death Differ 15: 21-28.
  70. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, et al. (1999) Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285: 727-729.
  71. Jimenez-Perez MI, Jave-Suarez LF, Ortiz-Lazareno PC, Bravo-Cuellar A, Gonzalez-Ramella O, et al. (2012) Cervical cancer cell lines expressing NKG2D-ligands are able to down-modulate the NKG2D receptor on NKL cells with functional implications. BMC Immunol 13: 7.
  72. Diefenbach A, Jensen ER, Jamieson AM, Raulet DH (2001) Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature 413: 165-171.
  73. Allez M, Tieng V, Nakazawa A, Treton X, Pacault V, et al. (2007) CD4+NKG2D+ T cells in Crohn's disease mediate inflammatory and cytotoxic responses through MICA interactions. Gastroenterology 132: 2346-2358.
  74. Hansen LK, Houchins JP, O'Leary JJ (1991) Differential regulation of HSC70, HSP70, HSP90 alpha, and HSP90 beta mRNA expression by mitogen activation and heat shock in human lymphocytes. Exp Cell Res 192: 587-596.
  75. Yabe T, McSherry C, Bach FH, Fisch P, Schall RP, et al. (1993) A multigene family on human chromosome 12 encodes natural killer-cell lectins. Immunogenetics 37: 455-460.
  76. Champsaur M, Lanier LL (2010) Effect of NKG2D ligand expression on host immune responses. Immunol Rev 235: 267-285.
  77. Wu J, Song Y, Bakker AB, Bauer S, Spies T, et al. (1999) An activating immunoreceptor complex formed by NKG2D and DAP10. Science 285: 730-732.
  78. Lanier LL (2009) DAP10- and DAP12-associated receptors in innate immunity. Immunol Rev 227: 150-160.
  79. Raulet DH, Gasser S, Gowen BG, Deng W, Jung H (2013) Regulation of ligands for the NKG2D activating receptor. Annu Rev Immunol 31: 413-441.
  80. Rhee I, Veillette A (2012) Protein tyrosine phosphatases in lymphocyte activation and autoimmunity. Nat Immunol 13: 439-447.
  81. Jamieson AM, Diefenbach A, McMahon CW, Xiong N, Carlyle JR, et al. (2002) The role of the NKG2D immunoreceptor in immune cell activation and natural killing. Immunity 17: 19-29.
  82. Whang MI, Guerra N, Raulet DH (2009) Costimulation of dendritic epidermal gammadelta T cells by a new NKG2D ligand expressed specifically in the skin. J Immunol 182: 4557-4564.
  83. Girardi M, Oppenheim DE, Steele CR, Lewis JM, Glusac E, et al. (2001) Regulation of cutaneous malignancy by gammadelta T cells. Science 294: 605-609.
  84. Groh V, Rhinehart R, Randolph-Habecker J, Topp MS, Riddell SR, et al. (2001) Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol 2: 255-260.
  85. Meresse B, Chen Z, Ciszewski C, Tretiakova M, Bhagat G, et al. (2004) Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease. Immunity 21: 357-366.
  86. Lee JC, Lee KM, Kim DW, Heo DS (2004) Elevated TGF-beta1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J Immunol 172: 7335-7340.
  87. Song H, Hur DY, Kim KE, Park H, Kim T, et al. (2006) IL-2/IL-18 prevent the down-modulation of NKG2D by TGF-beta in NK cells via the c-Jun N-terminal kinase (JNK) pathway. Cell Immunol 242: 39-45.
  88. Park YP, Choi SC, Kiesler P, Gil-Krzewska A, Borrego F, et al. (2011) Complex regulation of human NKG2D-DAP10 cell surface expression: opposing roles of the gammac cytokines and TGF-beta1. Blood 118: 3019-3027.
  89. Dou J, Wang Y, Wang J, Zhao F, Li Y, et al. (2009) Antitumor efficacy induced by human ovarian cancer cells secreting IL-21 alone or combination with GM-CSF cytokines in nude mice model. Immunobiology 214: 483-492.
  90. Takaki R, Hayakawa Y, Nelson A, Sivakumar PV, Hughes S, et al. (2005) IL-21 enhances tumor rejection through a NKG2D-dependent mechanism. J Immunol 175: 2167-2173.
  91. Horng T, Bezbradica JS, Medzhitov R (2007) NKG2D signaling is coupled to the interleukin 15 receptor signaling pathway. Nat Immunol 8: 1345-1352.
  92. de Menthon M, Lambert M, Guiard E, Tognarelli S, Bienvenu B, et al. (2011) Excessive interleukin-15 transpresentation endows NKG2D+CD4+ T cells with innate-like capacity to lyse vascular endothelium in granulomatosis with polyangiitis (Wegener's). Arthritis Rheum 63: 2116-2126.
  93. Iannello A, Thompson TW, Ardolino M, Lowe SW, Raulet DH (2013) p53-dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells. J Exp Med 210: 2057-2069.
  94. Siewiera J, El Costa H, Tabiasco J, Berrebi A, Cartron G, et al. (2013) Human cytomegalovirus infection elicits new decidual natural killer cell effector functions. PLoSPathog 9: e1003257.
  95. Ho SS, Gasser S (2013) NKG2D ligands link oncogenic RAS to innate immunity. Oncoimmunology 2: e22244.
  96. Eagle RA, Trowsdale J (2007) Promiscuity and the single receptor: NKG2D. Nat Rev Immunol 7: 737-744.
  97. Stephens HA (2001) MICA and MICB genes: can the enigma of their polymorphism be resolved? Trends Immunol 22: 378-385.
  98. Romphruk AV, Romphruk A, Naruse TK, Raroengjai S, Puapairoj C, et al. (2009) Polymorphisms of NKG2D ligands: diverse RAET1/ULBP genes in northeastern Thais. Immunogenetics 61: 611-617.
  99. Song H, Kim KE, Hur D, Lim JS, Yang Y, et al. (2008) IL-18 enhances ULBP2 expression through the MAPK pathway in leukemia cells. ImmunolLett 120: 103-107.
  100. Mishra R, Polic B, Welsh RM, Szomolanyi-Tsuda E (2013) Inflammatory cytokine-mediated evasion of virus-induced tumors from NK cell control. J Immunol 191: 961-970.
  101. Friese MA, Wischhusen J, Wick W, Weiler M, Eisele G, et al. (2004) RNA interference targeting transforming growth factor-beta enhances NKG2D-mediated antiglioma immune response, inhibits glioma cell migration and invasiveness, and abrogates tumorigenicity in vivo. Cancer Res 64: 7596-7603.
  102. Zhang T, Barber A, Sentman CL (2007) Chimeric NKG2D modified T cells inhibit systemic T-cell lymphoma growth in a manner involving multiple cytokines and cytotoxic pathways. Cancer Res 67: 11029-11036.
  103. Barber A, Zhang T, Sentman CL (2008) Immunotherapy with chimeric NKG2D receptors leads to long-term tumor-free survival and development of host antitumor immunity in murine ovarian cancer. J Immunol 180: 72-78.
  104. Chauveau A, Tonnerre P, Pabois A, Gavlovsky PJ, Chatelais M, et al. (2012) Endothelial Cell Activation and Proliferation Modulate NKG2D Activity by Regulating MICA Expression and Shedding. J Innate Immun 6: 89-104.
  105. Kishikawa T, Otsuka M, Yoshikawa T, Ohno M, Takata A, et al. (2013) Regulation of the expression of the liver cancer susceptibility gene MICA by microRNAs. Sci Rep 3: 2739.
  106. Xue W, Zender L, Miething C, Dickins RA, Hernando E, et al. (2007) Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445: 656-660.
  107. Armeanu S, Bitzer M, Lauer UM, Venturelli S, Pathil A, et al. (2005) Natural killer cell-mediated lysis of hepatoma cells via specific induction of NKG2D ligands by the histone deacetylase inhibitor sodium valproate. Cancer Res 65: 6321-6329.
  108. Kasahara M, Yoshida S (2012) Immunogenetics of the NKG2D ligand gene family. Immunogenetics 64: 855-867.
  109. El-Gazzar A, Groh V, Spies T (2013) Immunobiology and conflicting roles of the human NKG2D lymphocyte receptor and its ligands in cancer. J Immunol 191: 1509-1515.
  110. Olioso P, Giancola R, Di Riti M, Contento A, Accorsi P, et al. (2009) Immunotherapy with cytokine induced killer cells in solid and hematopoietic tumours: a pilot clinical trial. HematolOncol 27: 130-139.
  111. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, et al. (2001) Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood 97: 3146-3151.
  112. Bui JD, Carayannopoulos LN, Lanier LL, Yokoyama WM, Schreiber RD (2006) IFN-dependent downregulation of the NKG2D ligand H60 on tumors. J Immunol 176: 905-913.
  113. Coudert JD, Zimmer J, Tomasello E, Cebecauer M, Colonna M, et al. (2005) Altered NKG2D function in NK cells induced by chronic exposure to NKG2D ligand-expressing tumor cells. Blood 106: 1711-1717.
  114. Salih HR, Rammensee HG, Steinle A (2002) Cutting edge: downregulation of MICA on human tumors by proteolytic shedding. J Immunol 169: 4098-4102.
  115. Eisele G, Wischhusen J, Mittelbronn M, Meyermann R, Waldhauer I, et al. (2006) TGF-beta and metalloproteinases differentially suppress NKG2D ligand surface expression on malignant glioma cells. Brain 129: 2416-2425.
  116. Dasgupta S, Bhattacharya-Chatterjee M, O'Malley BW, Jr., Chatterjee SK (2005) Inhibition of NK cell activity through TGF-beta 1 by downregulation of NKG2D in a murine model of head and neck cancer. J Immunol 175: 5541-5550.
  117. Castriconi R, Cantoni C, Della Chiesa M, Vitale M, Marcenaro E, et al. (2003) Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. ProcNatlAcadSci U S A 100: 4120-4125.
  118. Crane CA, Han SJ, Barry JJ, Ahn BJ, Lanier LL, et al. (2010) TGF-beta downregulates the activating receptor NKG2D on NK cells and CD8+ T cells in glioma patients. Neuro Oncol 12: 7-13.
  119. Clayton A, Mitchell JP, Court J, Linnane S, Mason MD, et al. (2008) Human tumor-derived exosomes down-modulate NKG2D expression. J Immunol 180: 7249-7258.
  120. Wiemann K, Mittrucker HW, Feger U, Welte SA, Yokoyama WM, et al. (2005) Systemic NKG2D downregulation impairs NK and CD8 T cell responses in vivo. J Immunol 175: 720-729.
  121. Li H, Lakshmikanth T, Garofalo C, Enge M, Spinnler C, et al. (2011) Pharmacological activation of p53 triggers anticancer innate immune response through induction of ULBP2. Cell Cycle 10: 3346-3358.
  122. Textor S, Fiegler N, Arnold A, Porgador A, Hofmann TG, et al. (2011) Human NK cells are alerted to induction of p53 in cancer cells byupregulation of the NKG2D ligands ULBP1 and ULBP2. Cancer Res 71: 5998-6009.
  123. El-Gazzar A, Cai X, Reeves RS, Dai Z, Caballero-Benitez A, et al. (2013) Effects on tumor development and metastatic dissemination by the NKG2D lymphocyte receptor expressed on cancer cells. Oncogene.
  124. Lee S, Margolin K (2011) Cytokines in cancer immunotherapy. Cancers (Basel) 3: 3856-3893.
  125. Lees JG, Duffy SS, Moalem-Taylor G (2013) Immunotherapy targeting cytokines in neuropathic pain. Front Pharmacol 4: 142.
  126. Kawarada Y, Ganss R, Garbi N, Sacher T, Arnold B, et al. (2001) NK- and CD8(+) T cell-mediated eradication of established tumors by peritumoral injection of CpG-containing oligodeoxynucleotides. J Immunol 167: 5247-5253.
  127. Berner V, Liu H, Zhou Q, Alderson KL, Sun K, et al. (2007) IFN-gamma mediates CD4+ T-cell loss and impairs secondary antitumor responses after successful initial immunotherapy. Nat Med 13: 354-360.
  128. Murphy WJ, Welniak L, Back T, Hixon J, Subleski J, et al. (2003) Synergistic anti-tumor responses after administration of agonistic antibodies to CD40 and IL-2: coordination of dendritic and CD8+ cell responses. J Immunol 170: 2727-2733.
  129. Tietze JK, Wilkins DE, Sckisel GD, Bouchlaka MN, Alderson KL, et al. (2012) Delineation of antigen-specific and antigen-nonspecific CD8(+) memory T-cell responses after cytokine-based cancer immunotherapy. Blood 119: 3073-3083.
  130. Berg RE, Crossley E, Murray S, Forman J (2003) Memory CD8+ T cells provide innate immune protection against Listeria monocytogenes in the absence of cognate antigen. J Exp Med 198: 1583-1593.
  131. Li H, Han Y, Guo Q, Zhang M, Cao X (2009) Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol 182: 240-249.
  132. Pariente B, Mocan I, Camus M, Dutertre CA, Ettersperger J, et al. (2012) Activation of the receptor NKG2D leads to production of Th17 cytokines in CD4+ T cells of patients with Crohn's disease. Gastroenterology 141: 217-226, 226 e211-212.
  133. Hue S, Mention JJ, Monteiro RC, Zhang S, Cellier C, et al. (2004) A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity 21: 367-377.
Citation: Montalban-Arques A, Gorkiewicz G, Mulero V, Galindo-Villegas J (2014) Cytokine Intervention: A Double Edged Sword in the Nkg2d System Regulation. Immunome Res S2:002.

Copyright: © 2014 Montalban-Arques A, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top