Skip to main content
Cellular and Molecular Immunology logoLink to Cellular and Molecular Immunology
. 2012 Oct 15;10(1):50–57. doi: 10.1038/cmi.2012.43

γδ-T cells: an unpolished sword in human anti-infection immunity

Jian Zheng 1, Yinping Liu 1, Yu-Lung Lau 1, Wenwei Tu 1
PMCID: PMC4003172  PMID: 23064104

Abstract

γδ-T cells represent a small population of immune cells, but play an indispensable role in host defenses against exogenous pathogens, immune surveillance of endogenous pathogenesis and even homeostasis of the immune system. Activation and expansion of γδ-T cells are generally observed in diverse human infectious diseases and correlate with their progression and prognosis. γδ-T cells have both ‘innate' and ‘adaptive' characteristics in the immune response, and their anti-infection activities are mediated by multiple pathways that are under elaborate regulation by other immune components. In this review, we summarize the current state of the literature and the recent advancements in γδ-T cell-mediated immune responses against common human infectious pathogens. Although further investigation is needed to improve our understanding of the characteristics of different γδ-T cell subpopulations under specific conditions, γδ-T cell-based therapy has great potential for the treatment of infectious diseases.

Keywords: γδ-T cells, infection, immunity, human

Introduction

Infectious disease is one of the major threats to human health and causes substantial global morbidity and mortality. Current strategies for controlling infection principally focus on the pathogens themselves, but neglect the importance of the host factors that are involved in the process of disease.1 However, the rapid emergence of drug resistance in infectious pathogens often leads to costly therapy that is largely ineffective. Moreover, the efficiency of the adaptive immune response induced by vaccines might be significantly impaired by the rapid immune evasion of pathogens through their frequent mutations. To this extent, innate immune cells that recognize the conserved structural components of pathogens and raise rapid responses against the dangerous signals evoked by infections have great potential in anti-infection therapy.

Human γδ-T cells are critical components of the innate immune system and play critical roles in the early response to invasive pathogens. γδ-T cells represent only a minor T-cell population in peripheral blood (2%–10% of CD3+ T cells), but constitute the major subset of resident T cells in mucosa and skin.2 This preferential distribution favors their initial in situ anti-infection activities. Compared with the T-cell receptors (TCRs) of conventional αβ-T cells, the TCRs of γδ-T cells are relatively invariant and the exact ligands they recognize are still unknown.3 Nevertheless, it has been confirmed that γδ TCRs can sense the evolutionarily conserved components of exogenous pathogens as unique receptor agonists and initiate a rapid response against them.4,5

The roles of γδ-T cells are multifaceted and correlate with their distribution and differentiation.2 On the one hand, epidermal γδ-T cells play an indispensable role in limiting and eliminating invasive pathogens and recruiting inflammatory cells to infected locations,6,7 while skin γδ-T cells promote tissue repair by producing keratinocyte growth factor.8 On the other hand, some γδ-T cells, especially IL-17-producing γδ-T cells, have been confirmed to be involved in the pathogenesis of transplantation rejection,9 autoimmune diseases,10,11,12,13 inflammatory diseases14,15 and allergy16 in human and animal models. However, the scarcity of peripheral γδ-T cells and the difficulties in monitoring their fate in vivo make it difficult to achieve a comprehensive understanding of the characteristics of human γδ-T cells. Thus, the general application of γδ-T cell-based immune therapy in treating infectious diseases still needs further support from experimental investigations.

In this review, we will focus on the roles of human γδ-T cells in anti-infection immunity. With insights into the underlying mechanisms and regulation of the γδ-T cell-mediated anti-infection immune responses, this review is expected to provide perspective on the development of γδ-T cell-based immune therapy against infectious diseases in the future.

Roles of γδ-T cells in infectious diseases

Subpopulations of human γδ-T cells

Human γδ-T cells can be classified into two main populations according to their TCR expression, which is determined early in the thymus through TCR-mediated selection:17 Vδ1 and Vδ2 γδ-T cells. Vδ1 γδ-T cells are abundant in the skin, epithelia, intestine and uterus; in contrast, Vδ2 γδ-T cells are the majority of peripheral blood γδ-Τ cells.18 Consistent with their different distributions, these two γδ-Τ−cell subpopulations also exhibit distinct migratory patterns and homing capabilities.17

Although it is still controversial whether γδ-Τ cells are capable of antigen-specific memory in the same manner as αβ-T cells, the memory and activation markers CD27 and CD45RA have been found to be expressed on γδ-T cells.19,20 Similarly to αβ-T cells, γδ-T cells can also be classified into four populations based on their expression of CD27 and CD45RA: naive (CD27+CD45RA+), effector memory (CD27CD45RA), central memory (CD27+CD45RA) and terminally differentiated (CD27CD45RA+).21 More important, subpopulations of γδ-T cells identified by the expressions of CD27 and CD45RA exhibit unique functions during mycobacterial infection that correspond to the functions of their αβ-T cell analogues.21 In addition to these two markers, other surface makers are also detected to identify γδ-T cells of different characteristics. Our recent study demonstrated that human CD56+ Vδ2 γδ-T cells have a higher cytolytic capacity against influenza virus-infected cells than CD56 Vδ2 γδ-T cells, suggesting that the expression of CD56 might be a marker for subsets of γδ-T cells that protect against infection.22

Involvement of γδ-T cells in infectious diseases

The dynamic variation in the quality and quantity of human γδ-T cells affects the initiation, progression and prognosis of infectious diseases. Similarly, the nature of the pathogen affects the response of γδ-T cells. The exact roles of γδ-T-cell subpopulations during infections are dependent on their distinct functions and on the specific pathogens. In the following section, we provide an overview of the involvement of γδ-T cells during infection with different pathogens.

Viruses

Although the mechanisms underlying γδ-T cell-mediated immune responses against viruses are still incompletely understood, their protective effects have been confirmed in several acute and chronic viral infections. The activation and cytokine secretions of γδ-T cells are regarded as indicators of early viral infection.23,24

Similarly to the contribution of murine γδ-T cells during recovery after influenza-caused pneumonia,25 the beneficial effects of human Vγ9Vδ2 γδ-T cells against influenza virus infection have also recently been confirmed by our laboratory.22,26,27 Through the direct killing of virus-infected cells and the production of antiviral cytokines, Vγ9Vδ2 γδ-T cells can control infection by different strains of the influenza virus, such as human seasonal H1N1, pandemic H1N1, and the avian H5N1 and H9N2 viruses.22,26,27 Moreover, these antiviral activities can be significantly improved by phosphoantigen stimulation, which confers sufficient protection on humanized mice to prevent lethal influenza virus infection.28 In addition, another group has shown that γδ-T cells can initiate efficient adaptive immunity through processing and presenting influenza virus-derived peptides to CD4+ and CD8+ T cells.29 Moreover, γδ-T cells have been found to promote the establishment of protective adaptive immunity against West Nile virus by inducing the maturation of dendritic cells (DCs).30 However, the protective functions of γδ-T cells can be impaired by some viruses. It has been shown that herpes simplex virus31 and respiratory syncytial virus32 can directly infect local or peripheral γδ-T-cell subsets, respectively, which results in their dysfunction, although a few protective virus-reactive γδ-T cells could still be detected in patients infected with herpes simplex virus.33

The protective roles of γδ-T cells have also been confirmed in some chronic infectious diseases. Vδ2 γδ-T cells, the minor subpopulation of peripheral blood γδ-T cells, have been found to expand on a large scale during human cytomegalovirus (HCMV) infection.34 These HCMV-reactive Vδ2 γδ-T cells have a more potent ‘virus-specific' cytotoxicity than their Vδ2+ analogues and show increased elimination of pathogens.35 More importantly, this Vδ2 γδ-T cell-mediated ‘HCMV-specific immune response' can be induced in the fetal immune system during in utero infection, which offers protection in early life.36 Similarly, γδ-T cells also exhibit beneficial roles in controlling HIV infection.24 γδ-T cells in HIV-infected patients have been found to exhibit antiviral potential through their cell-lytic functions37 and cytokine secretions.38 Although the quantity and quality of γδ-T cells have been found to generally decrease with the advancement of HIV infection,39 the suppressed functions of Vγ9Vδ2 γδ-T cells can be enhanced by stimulation with phosphoantigen,39 which might become a novel target of therapeutic strategies.

γδ-T cells also help control the infection caused by Epstein–Barr virus40 and human hepatitis virus C41 in humans. However, the activation of γδ-T cells by hepatitis virus C might induce excessive inflammation and result in severe side effects.41 In addition, activated γδ-T cells can improve αβ-T cell-mediated specific immune responses against Epstein–Barr virus-induced lymphoma42 and contribute to the suppression of polyomavirus-induced tumor growth.43

Bacteria

Human γδ-T cells can recognize multiple conserved pathogen antigens and raise rapid immune responses. Although they have been found to participate in immune responses during many infections, including salmonellosis, brucellosis, legionellosis, tularemia,44 listeriolisis45 and Escherichia coli infections, the importance of human γδ-T cells in anti-bacterial activity is still controversial because the complicated immune responses initiated by the diverse components and products of bacteria make it difficult to identify the independent roles of γδ-T cells. Recently, successful control of both extracellular Gram-positive (Staphylococcus aureus) and Gram-negative (E. coli and Morganella morganii) bacterial infections in severe combined immunodeficiency mice by adoptive transfer of human Vγ9Vδ2 γδ-T cells offers solid evidence of the potent protective functions of γδ-T cells.46 More importantly, some intracellular bacterial pathogens, such as Mycobacterium tuberculosis, can specifically expand and activate Vγ9Vδ2 γδ-T cells by inducing the production of metabolites (e.g., isopentenyl pyrophosphate, IPP) in infected cells, which strongly suggests the importance of γδ-T cells in infection control.47 Consistent with this finding, the suppression of γδ-T cells by chronic tuberculosis infection can result in a disastrous outcome.48

Other infectious pathogens

In addition to their protective functions in viral and bacterial infections, γδ-T cells have been found to be activated and to help control infections caused by Leishmania49 and Toxoplasma gondii,50 although the γδ-T cell-mediated inflammation also causes some unwanted destruction of surrounding tissue.50 Similarly, the protective roles of γδ-T cells in malaria infection have been confirmed in several independent studies.51,52

HOW DO gd-T CELLS SENSE INFECTIOUS PATHOGENS?

Despite their active roles in diverse human infectious diseases, the pathways that are used by γδ-T cells to sense pathogens and initiate rapid responses remain largely unknown. In this section, we will explore some of the principal signals that are critical for γδ-T cell-mediated anti-infection activity.

Host cell-derived signals

It is generally accepted that most factors that sensitize γδ-T cells originate from host cells rather than from pathogens themselves.23 Although the ligands for γδ TCRs are still elusive, some molecules belonging to the phosphoantigen family have been extensively investigated for their application in activating γδ-T cells based on their specific recognition by Vγ9Vδ2 TCRs. These phosphoantigens are usually natural metabolites of isoprenoid biosynthesis in host cells, which include IPP, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate, dimethylallyl pyrophosphate (DMAPP)53,54 and bromohydrin pyrophosphate.55 These phosphoantigens can be presented by T cells, monocytes and various antigen-presenting cells (APCs), such as DCs and B cells,56,57 through expression of CD1d58,59 and as-yet unknown non-MHC molecules on host cells.60 The clinical applications of phosphoantigens in γδ-T cell-based therapy will be discussed in a later part of this review.

Similarly to αβ-T cells, γδ-T cells also need secondary signals that are provided by costimulatory molecules to achieve expansion and optimal activation.61,62 Many costimulators, such as CD2,63 CD2864,65 and CD137 (4-1BB),66 are expressed in human γδ-T cells and help to promote their activation. On the contrary, when the inhibitory molecule programmed death 1 was expressed in γδ-T cells, it was found to suppress their expansion through interaction with its ligand when the ligand was expressed on infected cells or tumor-related cells (Daniel Oliver, personal communication). In addition to their roles during activation or inhibition of γδ-T cells, costimulators can modify the differentiation of γδ-T cells. For example, the inducible costimulator–inducible costimulator ligand interaction has been found to arm γδ-T cells with some of the properties of follicular T cells and thus assist in antibody production by B cells.67 In addition to the conventional costimulators that are shared with αβ-T cells, several novel molecules can serve as the secondary signals for the activation of γδ-T cells. It has been found that the expression of junctional adhesion molecule-like protein68 and high-mobility group box 169 by γδ-T cells provides positive signals for the activation of γδ-T cells, whereas stimulation with E-cadherin downregulates their functions.70 The natural killer (NK) cell receptor family is another important group of costimulators for regulating the activity of γδ-T cells, and the balance among stimulatory and inhibitory natural killer WW (NK) cell receptor signals might determine the functions of γδ-T cells. For example, NKG2D and NKG2C promote the activation of γδ-T cells, whereas NKG2A transfers inhibitory signals to them.71,72,73

Pathogen-related antigens

Human γδ-T cells also recognize danger signals derived directly from pathogens. Toll-like receptors (TLRs) are the most important pathogen recognition receptors and are capable of recognizing a broad spectrum of pathogen-associated molecule patterns from infectious pathogens. TLRs play critical roles in the antiviral activities of γδ-T cells.74 The expression of TLRs in resting γδ-T cells is usually weak or undetectable but can be quickly upregulated in activated γδ-T cells. It has been found that activated γδ-T cells express nearly all TLRs, although the expression levels of different TLRs are distinct in different subsets.74 For example, both Vδ275 and Vδ176 γδ-T cells can be activated by TLR4 and TLR5 ligands and exhibit increased anti-bacteria responses, whereas the specific expression of TLR3 in Vδ2 γδ-T cells favors their stimulation through this pathway.74 Moreover, pan-γδ-T cells can be activated77 through stimulation of the heterodimers TLR1/2 and TLR2/6 with a mixture of TLR2 ligands, while the production of Th1 cytokines and the cytolytic activity of γδ-T cells can be enhanced by stimulation with double-stranded RNA78 and DNA79 through TLR3 and TLR9, respectively, during viral infections.

In addition to TLRs, γδ TCRs can induce the anti-infection activity of γδ-T cells by recognizing multiple pathogen-derived peptides80,81,82 and unprocessed pathogen-related proteins such as tetanus toxoid,83 herpes simplex virus glycoprotein I,84 mycobacterial purified protein derivatives,85,86 staphylococcal enterotoxin A,87 listeriolysin O88 and some heat shock proteins,85,89 which undoubtedly improves the efficiency of γδ-T cell-mediated immune responses during the very early phases of infections without the need to interact with APCs. Finally, there are still debates regarding whether γδ-T cells also possess antigen specificity similar to αβ-T cells, although rapid recall expansions of clonotypic γδ-T cells have been found in mycobacterial90 and Listeria monocytogenes53 infections. A recent investigation demonstrating receptor selection in peripheral γδ-T cells supported the existence of antigen-specific memory in human γδ-T cells.91 However, care should be taken when identifying ‘antigen-specific γδ-T cells' before clarifying whether these ‘memory-like responses' are antigen-restricted or presenting molecule-restricted.92 For example, the CD1d agonist α-GalCer can independently induce interferon (IFN)-γ production by γδ-T cells regardless of the presenting capability of CD1d,93 which does not classically belong as an antigen-specific immune response.

γδ-T cell-mediated anti-infection immunity

After being sensitized by danger signals derived from pathogens or host cells, γδ-T cells execute their anti-infection activities through multiple pathways. Additionally, the γδ-T cell-mediated immune responses are tightly controlled by multiple intrinsic and extrinsic factors, which guarantee the high efficiency of the immune responses and reduce unwanted destruction caused by excessive inflammation.

Direct anti-infection activity mediated by γδ-T cells

Direct killing of infected cells or infectious pathogens is the most prominent mechanism of the γδ-T cell-mediated anti-infection responses. γδ-T cell-mediated cytotoxicity is carried out through dozens of pathways, including Fas–Fas ligand interaction and secretion of perforin, granzyme B27 and granzyme M.94 In contrast to the cytotoxicity of NK cells, the cytotoxicity mediated by Vγ9Vδ2 γδ-T cells22 and Vδ2 γδ-T cells34 against influenza virus- and HCMV-infected cells can be initiated independent of the antibody-dependent cell-mediated cytotoxicity effect. Consistent with this mechanism, the Fc receptor CD16, a key player in antibody-dependent cell-mediated cytotoxicity, has been shown to improve the anti-viral activity of Vδ2 γδ-T cells by inducing them to produce IFN-γ during HCMV infection.95

In addition to their cytotoxic capacities, γδ-T cells are able to secrete dozens of potent soluble pro-inflammatory molecules and directly attack infectious pathogens. Similarly to its role in HCMV infection, IFN-γ secreted from Vγ9Vδ2 γδ-T cells inhibits influenza virus propagation.26 In addition, IL-17 and tumor-necrosis factor-α secreted by various γδ-T cell populations contribute to the control and elimination of several types of bacteria45,96 and Plasmodium falciparum.51 Recently, IL-22 has been suggested as a potent anti-infection cytokine in the γδ-T cell-mediated anti-infection immune response by some reports.97,98

Indirect anti-infection activity mediated by γδ-T cells

Initiating local immune responses

Local responses aroused during the early phase of infection are crucial for limiting the spread of pathogens. Both resident and circulating γδ-T cells accumulate quickly around invasive pathogens and induce efficient anti-infection responses through collaboration with neighboring immune cells.99 γδ-T cells enhance the anti-infection capabilities of resident macrophages and NK cells,100,101 promote the maturation of DCs6,102 and improve the invasion resistance of epithelial cells.103,104 Importantly, the dynamic interaction between γδ-T cells and their neighbors induces TCR selection of γδ-T cells in situ, leading to more precise responses against specific pathogens.101,103 Similarly to other sentinel cells, γδ-T cells also secrete chemokines such as CCL2, CCL3 and CCL4 to recruit pro-inflammatory neutrophils to accelerate the elimination of pathogens and the repair of damaged tissues.26

APC function

In addition to their direct anti-infection activities, human γδ-T cells act as professional APCs and take up, process and present pathogen-related antigens from both free viral particles29 and infected cells57,105 to other effector immune cells.106,107 These γδ-T APCs express approximately similar levels of HLA-DR and CD80/CD86 compared to traditional APCs29 and induce NK cell activation,108,109 antigen-specific αβ-T-cell responses29,107 and even antibody production.67 Although the physiological roles of γδ-T APCs still need to be illuminated by more in vivo studies, recent investigations of murine γδ-T cells have confirmed the general existence of γδ-T APCs.110

Modifying adaptive immunity

Although generally regarded as a component of the innate immune system based on their invariant TCRs and rapid responses to danger signals, γδ-T cells are now accepted as a crucial player in the adaptive immune system as well.111 The contribution of γδ-T cells to adaptive immunity goes beyond their APC function mentioned above. For example, γδ-T cells are capable of inducing the maturation of DCs,67 facilitating the development of αβ-T cells,13,112 killing regulatory T cells (Tregs)113,114 and migrating into the secondary lymphoid tissues and acting as follicular B helper T cells to promote antibody production.115,116 Moreover, γδ-T cells can compete with αβ-T cells for growth factors such as IL-15 and thus modify the adaptive immune response.117

Treg function

γδ-T cells have also been found to exhibit several ‘regulatory characteristics' in both humans and mice.118,119 Similarly to conventional CD4+ Tregs, a regulatory Vδ1 γδ-T cell subset identified by the expression of CD27 and CD25 in the peripheral blood of systemic lupus erythematosus patients has been found to correlate with the progression and remission of the disease.120 Moreover, IPP-induced synovial fluid Vγ9Vδ2 γδ-T-cell expansion has been found to ameliorate arthritis.121 However, the contribution of these regulatory γδ-T cells to anti-infection responses is still unclear.

Regulation of γδ-T-mediated anti-infection responses

The immune system is equipped with an elaborate regulatory network, which guarantees its efficiency while avoiding the unnecessary destruction caused by immune responses. In the battle against infectious pathogens, the activity of γδ-T cells is also regulated by other molecules that act through multiple pathways. It has been confirmed that both murine122 and human123 CD4+ Tregs can directly suppress the expansion and cytokine production of mucosal γδ-T cells, whereas effector CD4+ T cells help maintain IL-17 production in γδ-T cells in naive animals.124 These dynamic interactions exist not only between adaptive immunity and innate immunity, but also within the innate immune system itself. For example, NK cells can exhibit inhibitory effects on the activation and cytotoxicity of γδ-T cells and protect pregnant women from preeclampsia.125

In addition to cellular regulation, the development, proliferation and activation of γδ-T cells in anti-infection immune responses are also controlled by multiple soluble molecules, in particular cytokines. Similarly to NK cells, both IL-7 and IL-15 are indispensable during the development and homeostasis of γδ-T cells,126 while IL-15,127 accompanied by IL-17 and IL-22, has also been found to contribute to the activation and expansion of γδ-T cells during M. tuberculosis53 infection. A similar effect that is mediated by IL-23 has been found in Listeria monocytogenes128 infection as well. In addition, IL-7,129 IL-21130 and the caspase-1-processed IL-1 family cytokines IL-1β and IL-18131 increase the anti-infection ability of γδ-T cells by promoting expansion and cytokine production. Moreover, IFN-γ has been found to contribute to the antagonizing effects of γδ-T cells against regulation that is mediated by Tregs.132 In contrast, type I IFN has been found to constrain IL-17A production by γδ-T cells against Francisella tularensis subspecies novicida,133 while administration of either transforming growth factor-β134 or IL-10135 alone inhibits IFN-γ production by γδ-T cells in response to mycobacterial infection. The administration of transforming growth factor-β accompanied by anti-γδ TCR can even induce human and murine γδ-T cells into a regulatory status, thus dampening their anti-infection effects.119,120

Chemokines are another important group of soluble factors that determine the outcome of γδ-T cell-mediated anti-infection immune responses. It has been found that RANTES, MIP1α/β and CXCL9/10/11 secreted from infected cells guide the migration of CCR526- and CXCR3136- expressed IPP-activated Vγ9Vδ2 γδ-T cells to influenza-infected sites and facilitate the elimination of pathogens.

Potential of γδ-T cell-based therapy in infectious diseases

Obstacles to the application of γδ-T cell-based therapy

Current investigations on human γδ-T cells offer a promising future for infectious disease therapeutic strategies, although many questions that need to be answered remain before general application in the clinic will be possible. One major obstacle to γδ-T cell-based therapy is the scarcity of γδ-T cells in peripheral blood. Although phosphoantigens have been globally applied in the expansion of Vγ9Vδ2 γδ-T cells, the in vivo effects of these drugs need to be thoroughly investigated. Another impediment to studying the efficacy of phosphoantigen activation and expansion of Vγ9Vδ2 γδ-T cells is the lack of an analogue in murine systems, which can be overcome by the development of a humanized mouse model.28,137 Similarly, reliable animal models need to be established for evaluating the characteristics and clinical potential of human resident γδ-T cells in mucosa and skin.

Current strategies for activating or expanding γδ-T cells

The approach of using phosphoantigens plus IL-2 has been the most potent and widely accepted protocol for activation and expansion of Vγ9Vδ2 γδ-T cells both in vitro and in vivo.138 For adoptive transfer therapy, a single dose as large as 109 ex vivo expanded Vγ9Vδ2 γδ-T cells has been confirmed to be safe by several independent investigations.139,140,141 Encouragingly, these infused Vγ9Vδ2 γδ-T cells exhibited satisfactory clinical benefit in an anti-tumor trial.142 For in vivo applications, a single dose of (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate plus 5 days of IL-2 treatment induced an 80-fold expansion of macaque peripheral blood Vγ9Vδ2 γδ-T cells without detectable side effects.143 More excitingly, the administration of (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate and IL-2 was confirmed to significantly induce prolonged accumulation of Vγ9Vδ2 γδ-T cells in the lungs of cynomolgus monkeys and ameliorate lung lesions caused by Yersinia pestis infection.144 Presently, bisphosphonates such as Pamidronate and Zoledronate145 are ‘old' drugs that are routinely applied in treating osteoporosis or Paget's disease and have shown potential benefits in human tumor therapies146,147,148 and for treating influenza infection in humanized mice.28

In addition to phosphoantigens, stimulation that targets TLRs, natural killer (NK) cell receptors or CD3 have also been applied in the activation and expansion of pan-γδ-T cells in vitro.149 In line with these efforts, significant outcomes have been claimed for the usage of anti-γδ TCR antibody accompanied by cytokines in expanding human γδ-T cells,150 which improved the treatment of tumors in a mouse model151 and helped ameliorate autoimmune diseases.120 Nevertheless, the efficacies of these distinct protocols need to be evaluated and compared under standard conditions to achieve the optimal therapeutic effect. Moreover, the associated changes in the phenotypes of these accumulated γδ-T cells need to be clarified before their clinical application.

Prospective and potential limitations

The application of γδ-T cell-based therapy against infectious diseases has a bright future. One of its most prominent advantages comes from the recognition of danger signals rather than of pathogens themselves, which can engage in immune evasion through frequent mutation of epitopes.23 Another advantage of γδ-T cell-based therapy relies on their ability to traffic to local sites of infection.143,152 Moreover, the APC function of γδ-T cells further favors their application in inducing antigen-specific immune responses to efficiently eliminate pathogens.153 Finally, over 40 years of bisphosphonate usage in the clinic offers abundant references for their use in expanding and activating γδ-T cells in vivo, although attention must still be paid to the mild or moderate acute responses evoked by their injection.154,155

The major concern with using γδ-T cell-based anti-infection therapy comes from its pro-inflammatory characteristics. These potential risks might be induced by overdoses or bystander effects and result in γδ-T cell-triggered autoimmune diseases13 or general inflammatory diseases.156 It has been shown that one auto-aggressive γδ-T-cell subset recognizing aminoacyl-tRNA synthetases that are shared by bacteria and humans can be detected in some myositis patients and might contribute to the pathogenesis of their disease.157 Thus, a thorough understanding of the migration, differentiation and transformation of activated γδ-T cells under physiological and pathological conditions158 should be achieved before their clinical application.

Summary

In summary, γδ-T cells play distinct roles in human infectious diseases corresponding to their distribution and subpopulations. The anti-infection activities of γδ-T cells can be initiated by signals from hosts and pathogens and carried out by direct cytotoxicity against infected cells or through cytokine production or by indirect pathways involving multiple immune system components. As a bridge between innate immunity and adaptive immunity, γδ-T cells exhibit diverse advantages in anti-infection responses. Although the potential of γδ-T cells for treating human diseases has been significantly improved with the application of bisphosphonates and the advancement of our understanding of their multifaceted characteristics, the clinical usage of γδ-T cell-based therapies requires more in vivo evidence from further investigations into their roles under physiological and pathological conditions.

Acknowledgments

This work was supported in part by the Area of Excellence program on influenza, which is supported by the University Grants Committee of the Hong Kong SAR, China (Project No. AoE/M-12/06), the General Research Fund and the Research Grants Council of Hong Kong (HKU 777108M, HKU777407, HKU768108, HKU781211).

References

  1. Kaufmann SH. Robert Koch, the Nobel Prize, and the ongoing threat of tuberculosis. N Engl J Med. 2005;353:2423–2426. doi: 10.1056/NEJMp058131. [DOI] [PubMed] [Google Scholar]
  2. Born WK, Reardon CL, O'Brien RL. The function of gammadelta T cells in innate immunity. Curr Opin Immunol. 2006;18:31–38. doi: 10.1016/j.coi.2005.11.007. [DOI] [PubMed] [Google Scholar]
  3. Xi X, Guo Y, Chen H, Xu CP, Zhang HY, Hu HB, et al. Antigen specificity of gamma delta T cells primarily depends on the flanking sequences of CDR3 delta. J Biol Chem. 2009;284:27449–27455. doi: 10.1074/jbc.M109.011684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Hayday AC. γδ cells: a right time and a right place for a conserved third way of protection. Annu Rev Immunol. 2000;18:975–1026. doi: 10.1146/annurev.immunol.18.1.975. [DOI] [PubMed] [Google Scholar]
  5. Kabelitz D, Glatzel A, Wesch D. Antigen recognition by human gammadelta T lymphocytes. Int Arch Allergy Immunol. 2000;122:1–7. doi: 10.1159/000024353. [DOI] [PubMed] [Google Scholar]
  6. Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe HR, et al. IL-17 is essential for host defense against cutaneous Staphylococcus aureus infection in mice. J Clin Invest. 2010;120:1762–1773. doi: 10.1172/JCI40891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Molne L, Corthay A, Holmdahl R, Tarkowski A. Role of gamma/delta T cell receptor-expressing lymphocytes in cutaneous infection caused by Staphylococcus aureus. . Clin Exp Immunol. 2003;132:209–215. doi: 10.1046/j.1365-2249.2003.02151.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Jameson J, Ugarte K, Chen N, Yachi P, Fuchs E, Boismenu R, et al. A role for skin gammadelta T cells in wound repair. Science. 2002;296:747–749. doi: 10.1126/science.1069639. [DOI] [PubMed] [Google Scholar]
  9. Shiohara T, Moriya N, Hayakawa J, Itohara S, Ishikawa H. Resistance to cutaneous graft-vs.-host disease is not induced in T cell receptor delta gene-mutant mice. J Exp Med. 1996;183:1483–1489. doi: 10.1084/jem.183.4.1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ferri S, Longhi MS, de Molo C, Lalanne C, Muratori P, Granito A, et al. A multifaceted imbalance of T cells with regulatory function characterizes type 1 autoimmune hepatitis. Hepatology. 2010;52:999–1007. doi: 10.1002/hep.23792. [DOI] [PubMed] [Google Scholar]
  11. Ito Y, Usui T, Kobayashi S, Iguchi-Hashimoto M, Ito H, Yoshitomi H, et al. Gamma/delta T cells are the predominant source of interleukin-17 in affected joints in collagen-induced arthritis, but not in rheumatoid arthritis. Arthritis Rheum. 2009;60:2294–2303. doi: 10.1002/art.24687. [DOI] [PubMed] [Google Scholar]
  12. Lalor SJ, Dungan LS, Sutton CE, Basdeo SA, Fletcher JM, Mill KH. Caspase-1-processed cytokines IL-1β and IL-18 promote IL-17 production by γδ and CD4 T cells that mediate autoimmunity. J Immunol. 2011;186:5738–5748. doi: 10.4049/jimmunol.1003597. [DOI] [PubMed] [Google Scholar]
  13. Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH, et al. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity. 2009;31:331–341. doi: 10.1016/j.immuni.2009.08.001. [DOI] [PubMed] [Google Scholar]
  14. Fink DR, Holm D, Schlosser A, Nielsen O, Latta M, Lozano F, et al. Elevated numbers of SCART1+ gammadelta T cells in skin inflammation and inflammatory bowel disease. Mol Immunol. 2010;47:1710–1718. doi: 10.1016/j.molimm.2010.03.002. [DOI] [PubMed] [Google Scholar]
  15. Smith E, Prasad KM, Butcher M, Dobrian A, Kolls JK, Ley K, et al. Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2010;121:1746–1755. doi: 10.1161/CIRCULATIONAHA.109.924886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Murdoch JR, Lloyd CM. Resolution of allergic airway inflammation and airway hyperreactivity is mediated by IL-17 producing γδT cells. Am J Respir Crit Care Med. 2010;182:464–476. doi: 10.1164/rccm.200911-1775OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Jin Y, Xia MC, Saylor CM, Narayan K, Kang J, Wiest DL, et al. Cutting edge: intrinsic programming of thymic γδT cells for specific peripheral tissue localization. J Immunol. 2010;185:7156–7160. doi: 10.4049/jimmunol.1002781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chennupati V, Worbs T, Liu X, Malinarich FH, Schmitz S, Haas JD, et al. Intra- and intercompartmental movement of γδ T cells: intestinal intraepithelial and peripheral γδ T cells represent exclusive nonoverlapping populations with distinct migration characteristics. J Immunol. 2010;185:5160–5168. doi: 10.4049/jimmunol.1001652. [DOI] [PubMed] [Google Scholar]
  19. Eberl M, Engel R, Beck E, Jomaa H. Differentiation of human gamma-delta T cells towards distinct memory phenotypes. Cell Immunol. 2002;218:1–6. doi: 10.1016/s0008-8749(02)00519-1. [DOI] [PubMed] [Google Scholar]
  20. Ribot JC, deBarros A, Pang DJ, Neves JF, Peperzak V, Roberts SJ, et al. CD27 is a thymic determinant of the balance between interferon-gamma- and interleukin 17-producing gammadelta T cell subsets. Nat Immunol. 2009;10:427–436. doi: 10.1038/ni.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Gioia C, Agrati C, Casetti R, Cairo C, Borsellino G, Battistini L, et al. Lack of CD27-CD45RA-V gamma 9V delta 2+ T cell effectors in immunocompromised hosts and during active pulmonary tuberculosis. J Immunol. 2002;168:1484–1489. doi: 10.4049/jimmunol.168.3.1484. [DOI] [PubMed] [Google Scholar]
  22. Qin G, Liu Y, Zheng J, Xiang Z, Ng IH, Malik Peiris JS, et al. Phenotypic and functional characterization of human gammadelta T-cell subsets in response to influenza A viruses. J Infect Dis. 2012;205:1646–1653. doi: 10.1093/infdis/jis253. [DOI] [PubMed] [Google Scholar]
  23. Poccia F, Agrati C, Martini F, Capobianchi MR, Wallace M, Malkovsky M. Antiviral reactivities of gammadelta T cells. Microbes Infect. 2005;7:518–528. doi: 10.1016/j.micinf.2004.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Sciammas R, Bluestone JA. TCRgammadelta cells and viruses. Microbes Infect. 1999;1:203–212. doi: 10.1016/s1286-4579(99)80035-5. [DOI] [PubMed] [Google Scholar]
  25. Hoq MM, Suzutani T, Toyoda T, Horiike G, Yoshida I, Azuma M. Role of gamma delta TCR+ lymphocytes in the augmented resistance of trehalose 6,6′-dimycolate-treated mice to influenza virus infection. J Gen Virol. 1997;78 Pt 7:1597–1603. doi: 10.1099/0022-1317-78-7-1597. [DOI] [PubMed] [Google Scholar]
  26. Qin G, Liu Y, Zheng J, Ng IH, Xiang Z, Lam KT, et al. Type 1 responses of human Vgamma9Vdelta2 T cells to influenza A viruses. J Virol. 2011;85:10109–10116. doi: 10.1128/JVI.05341-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Qin G, Herold MJ, Kimmel B, Müller I, Rincon-Orozco B, Kunzmann V, et al. Phosphoantigen-expanded human gammadelta T cells display potent cytotoxicity against monocyte-derived macrophages infected with human and avian influenza viruses. J Infect Dis. 2009;200:858–865. doi: 10.1086/605413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Tu W, Zheng J, Liu Y, Sia SF, Liu M, Qin G, et al. The aminobisphosphonate pamidronate controls influenza pathogenesis by expanding a gammadelta T cell population in humanized mice. J Exp Med. 2011;208:1511–1522. doi: 10.1084/jem.20110226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Meuter S, Eberl M, Moser B. Prolonged antigen survival and cytosolic export in cross-presenting human γδ T cells. Proc Natl Acad Sci USA. 2010;107:8730–8735. doi: 10.1073/pnas.1002769107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fang H, Welte T, Zheng X, Chang GJ, Holbrook MR, Soong L, et al. gammadelta T cells promote the maturation of dendritic cells during West Nile virus infection. FEMS Immunol Med Microbiol. 2010;59:71–80. doi: 10.1111/j.1574-695X.2010.00663.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Puttur FK, Fernandez MA, White R, Roediger B, Cunningham AL, Weninger W, et al. Herpes simplex virus infects skin γδ T cells before Langerhans cells and impedes migration of infected langerhans cells by inducing apoptosis and blocking E-cadherin downregulation. J Immunol. 2010;185:477–487. doi: 10.4049/jimmunol.0904106. [DOI] [PubMed] [Google Scholar]
  32. Aoyagi M, Shimojo N, Sekine K, Nishimuta T, Kohno Y. Respiratory syncytial virus infection suppresses IFN-gamma production of gammadelta T cells. Clin Exp Immunol. 2003;131:312–317. doi: 10.1046/j.1365-2249.2003.02062.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Maccario R, Revello MG, Comoli P, Montagna D, Locatelli F, Gerna G. HLA-unrestricted killing of HSV-1-infected mononuclear cells. Involvement of either gamma/delta+ or alpha/beta+ human cytotoxic T lymphocytes. J Immunol. 1993;150:1437–1445. [PubMed] [Google Scholar]
  34. Knight A, Madriga AJ, Grace S, Sivakumaran J, Kottaridis P, Mackinnon S, et al. The role of Vdelta2-negative gamma-delta T cells during cytomegalovirus reactivation in recipients of allogeneic stem cell transplants. Blood. 2010;116:2164–2172. doi: 10.1182/blood-2010-01-255166. [DOI] [PubMed] [Google Scholar]
  35. Devaud C, Bilhere E, Loizon S, Pitard V, Behr C, Moreau JF, et al. Antitumor activity of γδ T cells reactive against cytomegalovirus-infected cells in a mouse xenograft tumor model. Cancer Res. 2009;69:3971–3978. doi: 10.1158/0008-5472.CAN-08-3037. [DOI] [PubMed] [Google Scholar]
  36. Vermijlen D, Brouwer M, Donner C, Liesnard C, Tackoen M, van Rysselberge M, et al. Human cytomegalovirus elicits fetal γδ T cell responses in utero. . J Exp Med. 2010;207:807–821. doi: 10.1084/jem.20090348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Wallace M, Bartz SR, Chang WL, Mackenzie DA, Pauza CD, Malkovsky M. Gamma delta T lymphocyte responses to HIV. Clin Exp Immunol. 1996;103:177–184. doi: 10.1046/j.1365-2249.1996.d01-625.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Agerberth B, Charo J, Werr J, Olsson B, Idali F, Lindbom L, et al. The human antimicrobial and chemotactic peptides LL-37 and alpha-defensins are expressed by specific lymphocyte and monocyte populations. Blood. 2000;96:3086–3093. [PubMed] [Google Scholar]
  39. Poccia F, Gioia C, Martini F, Sacchi A, Piacentini P, Tempestilli M, et al. Zoledronic acid and interleukin-2 treatment improves immunocompetence in HIV-infected persons by activating Vgamma9Vdelta2 T cells. AIDS. 2009;23:555–565. doi: 10.1097/QAD.0b013e3283244619. [DOI] [PubMed] [Google Scholar]
  40. de Paoli P, Gennari D, Martelli P, Cavarzerani V, Comoretto R, Santini G. Gamma delta T cell receptor-bearing lymphocytes during Epstein–Barr virus infection. J Infect Dis. 1990;161:1013–1016. doi: 10.1093/infdis/161.5.1013. [DOI] [PubMed] [Google Scholar]
  41. Tseng CT, Miskovsky E, Houghton M, Klimpel GR. Characterization of liver T-cell receptor gammadelta T cells obtained from individuals chronically infected with hepatitis C virus (HCV): evidence for these T cells playing a role in the liver pathology associated with HCV infections. Hepatology. 2001;33:1312–1320. doi: 10.1053/jhep.2001.24269. [DOI] [PubMed] [Google Scholar]
  42. Landmeier S, Altvater B, Pscherer S, Juergens H, Varnholt L, Hansmeier A, et al. Activated human gammadelta T cells as stimulators of specific CD8+ T-cell responses to subdominant Epstein Barr virus epitopes: potential for immunotherapy of cancer. J Immunother. 2009;32:310–321. doi: 10.1097/CJI.0b013e31819b7c30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Mishra R, Chen AT, Welsh RM, Szomolanyi-Tsuda E. NK cells and gammadelta T cells mediate resistance to polyomavirus-induced tumors. PLoS Pathog. 2010;6:e1000924. doi: 10.1371/journal.ppat.1000924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Chen ZW, Letvin NL. Vgamma2Vdelta2+ T cells and anti-microbial immune responses. Microbes Infect. 2003;5:491–498. doi: 10.1016/s1286-4579(03)00074-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. O'Brien RL, Roark CL, Born WK. IL-17-producing gammadelta T cells. Eur J Immunol. 2009;39:662–666. doi: 10.1002/eji.200839120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Wang L, Kamath A, Das H, Li L, Bukowski JF. Antibacterial effect of human V gamma 2V delta 2 T cells in vivo. . J Clin Invest. 2001;108:1349–1357. doi: 10.1172/JCI13584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kabelitz D, Bender A, Schondelmaier S, Schoel B, Kaufmann SH. A large fraction of human peripheral blood gamma/delta+ T cells is activated by Mycobacterium tuberculosis but not by its 65-kD heat shock protein. J Exp Med. 1990;171:667–679. doi: 10.1084/jem.171.3.667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Chen ZW. Immunology of AIDS virus and mycobacterial co-infection. Curr HIV Res. 2004;2:351–355. doi: 10.2174/1570162043351147. [DOI] [PubMed] [Google Scholar]
  49. Xu S, Han Y, Xu X, Bao Y, Zhang M, Cao X, et al. IL-17A-producing γδT cells promote CTL responses against Listeria monocytogenes infection by enhancing dendritic cell cross-presentation. J Immunol. 2010;185:5879–5887. doi: 10.4049/jimmunol.1001763. [DOI] [PubMed] [Google Scholar]
  50. Egan CE, Dalton JE, Andrew EM, Smith JE, Gubbels MJ, Striepen B, et al. A requirement for the Vgamma1+ subset of peripheral gammadelta T cells in the control of the systemic growth of Toxoplasma gondii and infection-induced pathology. J Immunol. 2005;175:8191–8199. doi: 10.4049/jimmunol.175.12.8191. [DOI] [PubMed] [Google Scholar]
  51. Horowitz A, Newman KC, Evans JH, Korbel DS, Davis DM, Riley EM, et al. Cross-talk between T cells and NK cells generates rapid effector responses to Plasmodium falciparum-infected erythrocytes. J Immunol. 2010;184:6043–6052. doi: 10.4049/jimmunol.1000106. [DOI] [PubMed] [Google Scholar]
  52. Weidanz WP, LaFleur G, Brown A, Burns JM, Jr, Gramaglia I, van der Heyde HC. γδT cells but not NK cells are essential for cell-mediated immunity against Plasmodium chabaudi malaria. Infect Immun. 2010;78:4331–4340. doi: 10.1128/IAI.00539-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Chen ZW. Immune biology of Ag-specific gammadelta T cells in infections. Cell Mol Life Sci. 2011;68:2409–2417. doi: 10.1007/s00018-011-0703-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Morita CT, Mariuzza RA, Brenner MB. Antigen recognition by human gamma delta T cells: pattern recognition by the adaptive immune system. Springer Semin Immunopathol. 2000;22:191–217. doi: 10.1007/s002810000042. [DOI] [PubMed] [Google Scholar]
  55. Chargui J, Combaret V, Scaglione V, Iacono I, Péri V, Valteau-Couanet D, et al. Bromohydrin pyrophosphate-stimulated Vgamma9delta2 T cells expanded ex vivo from patients with poor-prognosis neuroblastoma lyse autologous primary tumor cells. J Immunother. 2010;33:591–598. doi: 10.1097/CJI.0b013e3181dda207. [DOI] [PubMed] [Google Scholar]
  56. Morita CT, Beckman EM, Bukowski JF, Tanaka Y, Band H, Bloom BR, et al. Direct presentation of nonpeptide prenyl pyrophosphate antigens to human gamma delta T cells. Immunity. 1995;3:495–507. doi: 10.1016/1074-7613(95)90178-7. [DOI] [PubMed] [Google Scholar]
  57. Wei H, Huang D, Lai X, Chen M, Zhong W, Wang R, et al. Definition of APC presentation of phosphoantigen (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate to Vgamma2Vdelta 2 TCR. J Immunol. 2008;181:4798–4806. doi: 10.4049/jimmunol.181.7.4798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Agea E, Russano A, Bistoni O, Mannucci R, Nicoletti I, Corazzi L, et al. Human CD1-restricted T cell recognition of lipids from pollens. J Exp Med. 2005;202:295–308. doi: 10.1084/jem.20050773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Dieude M, Striegl H, Tyznik AJ, Wang J, Behar SM, Piccirillo CA, et al. Cardiolipin binds to CD1d and stimulates CD1d-restricted γδ T cells in the normal murine repertoire. J Immunol. 2011;186:4771–4781. doi: 10.4049/jimmunol.1000921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Sarikonda G, Wang H, Puan KJ, Liu XH, Lee HK, Song Y, et al. Photoaffinity antigens for human γδ T Cells. J Immunol. 2008;181:7738–7750. doi: 10.4049/jimmunol.181.11.7738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Ribot JC, Debarros A, Silva-Santos B. Searching for “signal 2”: costimulation requirements of gammadelta T cells. Cell Mol Life Sci. 2011;68:2345–2355. doi: 10.1007/s00018-011-0698-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Witherden DA, Havran WL. Molecular aspects of epithelial gammadelta T cell regulation. Trends Immunol. 2011;32:265–271. doi: 10.1016/j.it.2011.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Budd RC, Russell JQ, van Houten N, Cooper SM, Yagita H, Wolfe J. CD2 expression correlates with proliferative capacity of alpha beta+ or gamma delta+ CD4−CD8− T cells in lpr mice. J Immunol. 1992;148:1055–1064. [PubMed] [Google Scholar]
  64. Lafont V, Liautard J, Gross A, Liautard JP, Favero J. Tumor necrosis factor-alpha production is differently regulated in gamma delta and alpha beta human T lymphocytes. J Biol Chem. 2000;275:19282–19287. doi: 10.1074/jbc.M910487199. [DOI] [PubMed] [Google Scholar]
  65. Penninger JM, Timms E, Shahinian A, Jezo-Bremond A, Nishina H, Ionescu J, et al. Alloreactive gamma delta thymocytes utilize distinct costimulatory signals from peripheral T cells. J Immunol. 1995;155:3847–3855. [PubMed] [Google Scholar]
  66. Shao Z, Schwarz H. CD137 ligand, a member of the tumor necrosis factor family, regulates immune responses via reverse signal transduction. J Leuk Biol. 2011;89:21–29. doi: 10.1189/jlb.0510315. [DOI] [PubMed] [Google Scholar]
  67. Caccamo N, Battistini L, Bonneville M, Poccia F, Fournié JJ, Meraviglia S, et al. CXCR5 identifies a subset of Vgamma9Vdelta2 T cells which secrete IL-4 and IL-10 and help B cells for antibody production. J Immunol. 2006;177:5290–5295. doi: 10.4049/jimmunol.177.8.5290. [DOI] [PubMed] [Google Scholar]
  68. Witherden DA, Verdino P, Rieder SE, Garijo O, Mills RE, Teyton L, et al. The junctional adhesion molecule JAML is a costimulatory receptor for epithelial gammadelta T cell activation. Science. 2010;329:1205–1210. doi: 10.1126/science.1192698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ciucci A, Gabriele I, Percario ZA, Affabris E, Colizzi V, Mancino G. HMGB1 and cord blood: its role as immuno-adjuvant factor in innate immunity. PloS ONE. 2011;6:e23766. doi: 10.1371/journal.pone.0023766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Uchida Y, Kawai K, Ibusuki A, Kanekura T. Role for E-cadherin as an inhibitory receptor on epidermal γδ T cells. J Immunol. 2011;186:6945–6954. doi: 10.4049/jimmunol.1003853. [DOI] [PubMed] [Google Scholar]
  71. Das H, Groh V, Kuijl C, Sugita M, Morita CT, Spies T, et al. MICA engagement by human Vgamma2Vdelta2 T cells enhances their antigen-dependent effector function. Immunity. 2001;15:83–93. doi: 10.1016/s1074-7613(01)00168-6. [DOI] [PubMed] [Google Scholar]
  72. Rincon-Orozco B, Kunzmann V, Wrobel P, Kabelitz D, Steinle A, Herrmann T. Activation of V gamma 9V delta 2 T cells by NKG2D. J Immunol. 2005;175:2144–2151. doi: 10.4049/jimmunol.175.4.2144. [DOI] [PubMed] [Google Scholar]
  73. Angelini DF, Micucci F, Poccia F, Semenzato G, Borsellino G, Santoni A, et al. NKG2A inhibits NKG2C effector functions of γδ T cells: implications in health and disease. J Leukoc Biol. 2010;89:75–84. doi: 10.1189/jlb.0710413. [DOI] [PubMed] [Google Scholar]
  74. Wesch D, Peters C, Oberg HH, Pietschmann K, Kabelitz D. Modulation of gammadelta T cell responses by TLR ligands. Cell Mol Life Sci. 2011;68:2357–2370. doi: 10.1007/s00018-011-0699-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Devilder MC, Allain S, Dousset C, Bonneville M, Scotet E. Early triggering of exclusive IFN-gamma responses of human Vgamma9Vdelta2 T cells by TLR-activated myeloid and plasmacytoid dendritic cells. J Immunol. 2009;183:3625–3633. doi: 10.4049/jimmunol.0901571. [DOI] [PubMed] [Google Scholar]
  76. Pietschmann K, Beetz S, Welte S, Martens I, Gruen J, Oberg HH, et al. Toll-like receptor expression and function in subsets of human gammadelta T lymphocytes. Scand J Immunol. 2009;70:245–255. doi: 10.1111/j.1365-3083.2009.02290.x. [DOI] [PubMed] [Google Scholar]
  77. Oberg HH, Ly TT, Ussat S, Meyer T, Kabelitz D, Wesch D. Differential but direct abolishment of human regulatory T cell suppressive capacity by various TLR2 ligands. J Immunol. 2010;184:4733–4740. doi: 10.4049/jimmunol.0804279. [DOI] [PubMed] [Google Scholar]
  78. Wesch D, Beetz S, Oberg HH, Marget M, Krengel K, Kabelitz D. Direct costimulatory effect of TLR3 ligand poly(I:C) on human gamma delta T lymphocytes. J Immunol. 2006;176:1348–1354. doi: 10.4049/jimmunol.176.3.1348. [DOI] [PubMed] [Google Scholar]
  79. Rothenfusser S, Hornung V, Krug A, Towarowski A, Krieg AM, Endres S, et al. Distinct CpG oligonucleotide sequences activate human gamma delta T cells via interferon-alpha/-beta. Eur J Immunol. 2001;31:3525–3534. doi: 10.1002/1521-4141(200112)31:12<3525::aid-immu3525>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  80. Born WK, Zhang L, Nakayama M, Jin N, Chain JL, Huang Y, et al. Peptide antigens for gamma/delta T cells. Cell Mol Life Sci. 2011;68:2335–2343. doi: 10.1007/s00018-011-0697-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Fu YX, Cranfill R, Vollmer M, van der Zee R, O'Brien RL, Born W. In vivo response of murine gamma delta T cells to a heat shock protein-derived peptide. Proc Natl Acad Sci USA. 1993;90:322–326. doi: 10.1073/pnas.90.1.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Mohan JF, Levisetti MG, Calderon B, Herzog JW, Petzold SJ, Unanue ER, et al. Unique autoreactive T cells recognize insulin peptides generated within the islets of Langerhans in autoimmune diabetes. Nat Immunol. 2010;11:350–354. doi: 10.1038/ni.1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kozbor D, Trinchieri G, Monos DS, Isobe M, Russo G, Haney JA, et al. Human TCR-gamma+/delta+, CD8+ T lymphocytes recognize tetanus toxoid in an MHC-restricted fashion. J Exp Med. 1989;169:1847–1851. doi: 10.1084/jem.169.5.1847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Johnson RM, Lancki DW, Sperling AI, Dick RF, Spear PG, Fitch FW, et al. A murine CD4−, CD8− T cell receptor-gamma delta T lymphocyte clone specific for herpes simplex virus glycoprotein I. J Immunol . 1992;148:983–988. [PubMed] [Google Scholar]
  85. O'Brien RL, Happ MP, Dallas A, Palmer E, Kubo R, Born WK. Stimulation of a major subset of lymphocytes expressing T cell receptor gamma delta by an antigen derived from Mycobacterium tuberculosis. . Cell. 1989;57:667–674. doi: 10.1016/0092-8674(89)90135-9. [DOI] [PubMed] [Google Scholar]
  86. Happ MP, Kubo RT, Palmer E, Born WK, O'Brien RL. Limited receptor repertoire in a mycobacteria-reactive subset of gamma delta T lymphocytes. Nature. 1989;342:696–698. doi: 10.1038/342696a0. [DOI] [PubMed] [Google Scholar]
  87. Rust CJ, Verreck F, Vietor H, Koning F. Specific recognition of staphylococcal enterotoxin A by human T cells bearing receptors with the V gamma 9 region. Nature. 1990;346:572–574. doi: 10.1038/346572a0. [DOI] [PubMed] [Google Scholar]
  88. Guo Y, Ziegler HK, Safley SA, Niesel DW, Vaidya S, Klimpel GR. Human T-cell recognition of Listeria monocytogenes: recognition of listeriolysin O by TcR alpha beta+ and TcR gamma delta+ T cells. Infect Immun. 1995;63:2288–2294. doi: 10.1128/iai.63.6.2288-2294.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Holoshitz J, Koning F, Coligan JE, de Bruyn J, Strober S. Isolation of CD4− CD8− mycobacteria-reactive T lymphocyte clones from rheumatoid arthritis synovial fluid. Nature. 1989;339:226–229. doi: 10.1038/339226a0. [DOI] [PubMed] [Google Scholar]
  90. Shen Y, Zhou D, Qiu L, Lai X, Simon M, Shen L, et al. Adaptive immune response of Vgamma2Vdelta2+ T cells during mycobacterial infections. Science. 2002;295:2255–2258. doi: 10.1126/science.1068819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Bonneville M, O'Brien RL, Born WK. Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol. 2010;10:467–478. doi: 10.1038/nri2781. [DOI] [PubMed] [Google Scholar]
  92. Born WK, O'Brien RL. Antigen-restricted gammadelta T-cell receptors. Arch Immunol Ther Exp (Warsz) 2009;57:129–135. doi: 10.1007/s00005-009-0017-x. [DOI] [PubMed] [Google Scholar]
  93. Paget C, Chow MT, Duret H, Mattarollo SR, Smyth MJ. Role of gammadelta T cells in alpha-galactosylceramide-mediated immunity. J Immunology. 2012;188:3928–3939. doi: 10.4049/jimmunol.1103582. [DOI] [PubMed] [Google Scholar]
  94. de Koning PJ, Kummer JA, de Poot SA, Quadir R, Broekhuizen R, McGettrick AF, et al. The cytotoxic protease granzyme M is expressed by lymphocytes of both the innate and adaptive immune system. Mol Immunol. 2009;47:903–911. doi: 10.1016/j.molimm.2009.10.001. [DOI] [PubMed] [Google Scholar]
  95. Couzi L, Pitard V, Sicard X, Garrigue I, Hawchar O, Merville P, et al. Antibody-dependent anti-cytomegalovirus activity of human gammadelta T cells expressing CD16 (FcgammaRIIIa) Blood. 2012;119:1418–1427. doi: 10.1182/blood-2011-06-363655. [DOI] [PubMed] [Google Scholar]
  96. McAleer JP, Kolls JK. Mechanisms controlling Th17 cytokine expression and host defense. J Leuk Biol. 2011;90:263–270. doi: 10.1189/jlb.0211099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Li Z, Burns AR, Byeseda Miller S, Smith CW. CCL20, γδ T cells, and IL-22 in corneal epithelial healing. FASEB J. 2011;25:2659–2668. doi: 10.1096/fj.11-184804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Zenewicz LA, Flavell RA. Recent advances in IL-22 biology. Int Immunol. 2011;23:159–163. doi: 10.1093/intimm/dxr001. [DOI] [PubMed] [Google Scholar]
  99. Prinz I. Dynamics of the interaction of gammadelta T cells with their neighbors in vivo. . Cell Mol Life Sci. 2011;68:2391–2398. doi: 10.1007/s00018-011-0701-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Wands JM, Roark CL, Aydintug MK, Jin N, Hahn YS, Cook L, et al. Distribution and leukocyte contacts of gammadelta T cells in the lung. J Leuk Biol. 2005;78:1086–1096. doi: 10.1189/jlb.0505244. [DOI] [PubMed] [Google Scholar]
  101. Gazit R, Gruda R, Elboim M, Arnon TI, Katz G, Achdout H, et al. Lethal influenza infection in the absence of the natural killer cell receptor gene Ncr1. . Nat Immunol. 2006;7:517–523. doi: 10.1038/ni1322. [DOI] [PubMed] [Google Scholar]
  102. Martin B, Hirota K, Cua DJ, Stockinger B, Veldhoen M. Interleukin-17-producing gammadelta T cells selectively expand in response to pathogen products and environmental signals. Immunity. 2009;31:321–330. doi: 10.1016/j.immuni.2009.06.020. [DOI] [PubMed] [Google Scholar]
  103. Silva-Santos B, Pennington DJ, Hayday AC. Lymphotoxin-mediated regulation of gammadelta cell differentiation by alphabeta T cell progenitors. Science. 2005;307:925–928. doi: 10.1126/science.1103978. [DOI] [PubMed] [Google Scholar]
  104. Pennington DJ, Silva-Santos B, Shires J, Theodoridis E, Pollitt C, Wise EL, et al. The inter-relatedness and interdependence of mouse T cell receptor gammadelta+ and alphabeta+ cells. Nat Immunol. 2003;4:991–998. doi: 10.1038/ni979. [DOI] [PubMed] [Google Scholar]
  105. Rojas RE, Torres M, Fournie JJ, Harding CV, Boom WH. Phosphoantigen presentation by macrophages to Mycobacterium tuberculosis-reactive Vgamma9Vdelta2+ T cells: modulation by chloroquine. Infect Immun. 2002;70:4019–4027. doi: 10.1128/IAI.70.8.4019-4027.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Moser B, Eberl M. gammadelta T-APCs: a novel tool for immunotherapy. Cell Mol Life Sci. 2011;68:2443–2452. doi: 10.1007/s00018-011-0706-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Brandes M, Willimann K, Bioley G, Lévy N, Eberl M, Luo M, et al. Cross-presenting human gammadelta T cells induce robust CD8+ alphabeta T cell responses. Proc Natl Acad Sci USA. 2009;106:2307–2312. doi: 10.1073/pnas.0810059106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Maniar A, Zhang X, Lin W, Gastman BR, Pauza CD, Strome SE, et al. Human γδ T lymphocytes induce robust NK cell mediated antitumor cytotoxicity through CD137 engagement. Blood. 2010;116:1726–1733. doi: 10.1182/blood-2009-07-234211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Nussbaumer O, Gruenbacher G, Gander H, Thurnher M. DC-like cell-dependent activation of human natural killer cells by the bisphosphonate zoledronic acid is regulated by γδ T lymphocytes. Blood. 2011;118:2743–2751. doi: 10.1182/blood-2011-01-328526. [DOI] [PubMed] [Google Scholar]
  110. Cheng L, Cui Y, Shao H, Han G, Zhu L, Huang Y, et al. Mouse gammadelta T cells are capable of expressing MHC class II molecules, and of functioning as antigen-presenting cells. J Neuroimmunol. 2008;203:3–11. doi: 10.1016/j.jneuroim.2008.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Kabelitz D. gammadelta T-cells: cross-talk between innate and adaptive immunity. Cell Mol Life Sci. 2011;68:2331–2333. doi: 10.1007/s00018-011-0696-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Eberl M, Roberts GW, Meuter S, Williams JD, Topley N, Moser B. A rapid crosstalk of human gammadelta T cells and monocytes drives the acute inflammation in bacterial infections. PLoS Pathog. 2009;5:e1000308. doi: 10.1371/journal.ppat.1000308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Huber SA. gammadelta T lymphocytes kill T regulatory cells through CD1d. Immunology. 2010;131:202–209. doi: 10.1111/j.1365-2567.2010.03292.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Liu W, Huber SA. Cross-talk between CD1d-restricted NKT cells and gammadelta cells in t regulatory cell response. Virol J. 2011;8:32. doi: 10.1186/1743-422X-8-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, Moser B, et al. CXC chemokine receptor 5 expression defines follicular homing T cells with B cell helper function. J Exp Med. 2000;192:1553–1562. doi: 10.1084/jem.192.11.1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Vinuesa CG, Tangye SG, Moser B, Mackay CR. Follicular B helper T cells in antibody responses and autoimmunity. Nat Rev Immunol. 2005;5:853–865. doi: 10.1038/nri1714. [DOI] [PubMed] [Google Scholar]
  117. Do JS, Min B. IL-15 produced and trans-presented by DCs underlies homeostatic competition between CD8 and γδ T cells in vivo. . Blood. 2009;113:6361–6371. doi: 10.1182/blood-2008-12-192997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Kuhl AA, Pawlowski NN, Grollich K, Blessenohl M, Westermann J, Zeitz M, et al. Human peripheral gammadelta T cells possess regulatory potential. Immunology. 2009;128:580–588. doi: 10.1111/j.1365-2567.2009.03162.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Kang N, Tang L, Li X, Wu D, Li W, Chen X, et al. Identification and characterization of Foxp3+ gammadelta T cells in mouse and human. Immunol Lett. 2009;125:105–113. doi: 10.1016/j.imlet.2009.06.005. [DOI] [PubMed] [Google Scholar]
  120. Li X, Kang N, Zhang X, Dong X, Wei W, Cui L, et al. Generation of human regulatory γδ T cells by TCRγδ stimulation in the presence of TGF-β and their involvement in the pathogenesis of systemic lupus erythematosus. J Immunol. 2011;186:6693–6700. doi: 10.4049/jimmunol.1002776. [DOI] [PubMed] [Google Scholar]
  121. Berkun Y, Bendersky A, Gerstein M, Goldstein I, Padeh S, Bank I. γδT cells in juvenile idiopathic arthritis: higher percentages of synovial Vδ1+ and Vγ9+ T cell subsets are associated with milder disease. J Rheumatol. 2011;38:1123–1129. doi: 10.3899/jrheum.100938. [DOI] [PubMed] [Google Scholar]
  122. Yurchenko E, Levings MK, Piccirillo CA. CD4+ Foxp3+ regulatory T cells suppress gammadelta T-cell effector functions in a model of T-cell-induced mucosal inflammation. Eur J Immunol. 2011;41:3455–3466. doi: 10.1002/eji.201141814. [DOI] [PubMed] [Google Scholar]
  123. Park SG, Mathur R, Long M, Hosh N, Hao L, Hayden MS, et al. T regulatory cells maintain intestinal homeostasis by suppressing gammadelta T cells. Immunity. 2010;33:791–803. doi: 10.1016/j.immuni.2010.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Do JS, Visperas A, O'Brien RL, Min B. CD4 T cells play important roles in maintaining IL-17-producing gammadelta T-cell subsets in naive animals. Immunol Cell Biol. 2011;90:396–403. doi: 10.1038/icb.2011.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Miko E, Szereday L, Barakonyi A, Jarkovich A, Varga P, Szekeres-Bartho J, et al. Immunoactivation in preeclampsia: Vdelta2+ and regulatory T cells during the inflammatory stage of disease. J Reprod Immunol. 2009;80:100–108. doi: 10.1016/j.jri.2009.01.003. [DOI] [PubMed] [Google Scholar]
  126. Boyman O, Krieg C, Homann D, Sprent J. Homeostatic maintenance of T cells and natural killer cells. Cell Mol Life Sci. 2012;69:1597–1608. doi: 10.1007/s00018-012-0968-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Meraviglia S, Caccamo N, Salerno A, Sireci G, Dieli F. Partial and ineffective activation of V gamma 9V delta 2 T cells by Mycobacterium tuberculosis-infected dendritic cells. J Immunol. 2010;185:1770–1776. doi: 10.4049/jimmunol.1000966. [DOI] [PubMed] [Google Scholar]
  128. Meeks KD, Sieve AN, Kolls JK, Ghilardi N, Berg RE. IL-23 is required for protection against systemic infection with Listeria monocytogenes. . J Immunol. 2009;183:8026–8034. doi: 10.4049/jimmunol.0901588. [DOI] [PubMed] [Google Scholar]
  129. Laky K, Sieve AN, Kolls JK, Ghilardi N, Berg RE. Enterocyte expression of interleukin 7 induces development of gammadelta T cells and Peyer's patches. J Exp Med. 2000;191:1569–1580. doi: 10.1084/jem.191.9.1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Thedrez A, Harly C, Morice A, Salot S, Bonneville M, Scotet E. IL-21-mediated potentiation of antitumor cytolytic and proinflammatory responses of human Vγ9Vδ2 T cells for adoptive immunotherapy. J Immunol. 2009;182:3423–3431. doi: 10.4049/jimmunol.0803068. [DOI] [PubMed] [Google Scholar]
  131. Dungan LS, Mills KH. Caspase-1-processed IL-1 family cytokines play a vital role in driving innate IL-17. Cytokine. 2011;15:126–132. doi: 10.1016/j.cyto.2011.07.007. [DOI] [PubMed] [Google Scholar]
  132. Gong G, Shao L, Wang Y, Chen CY, Huang D, Yao S, et al. Phosphoantigen-activated V gamma 2V delta 2 T cells antagonize IL-2-induced CD4+CD25+Foxp3+ T regulatory cells in mycobacterial infection. Blood. 2009;113:837–845. doi: 10.1182/blood-2008-06-162792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Henry T, Kirimanjeswara GS, Ruby T, Jones JW, Peng K, Perret M, et al. Type I IFN signaling constrains IL-17A/F secretion by γδ T cells during bacterial infections. J Immunol. 2010;184:3755–3767. doi: 10.4049/jimmunol.0902065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Rojas RE, Balaji KN, Subramanian A, Boom WH. Regulation of human CD4+ alphabeta T-cell-receptor-positive (TCR+) and gammadelta TCR+ T-cell responses to Mycobacterium tuberculosis by interleukin-10 and transforming growth factor beta. Infect Immun. 1999;67:6461–6472. doi: 10.1128/iai.67.12.6461-6472.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Pechhold K, Wesch D, Schondelmaier S, Kabelitz D. Primary activation of V gamma 9-expressing gamma delta T cells by Mycobacterium tuberculosis. Requirement for Th1-type CD4 T cell help and inhibition by IL-10. J Immunol. 1994;152:4984–4992. [PubMed] [Google Scholar]
  136. Dieli F, Poccia F, Lipp M, Sireci G, Caccamo N, Di Sano C, et al. Differentiation of effector/memory Vdelta2 T cells and migratory routes in lymph nodes or inflammatory sites. J Exp Med. 2003;198:391–397. doi: 10.1084/jem.20030235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Shultz LD, Saito Y, Najima Y, Tanaka S, Ochi T, Tomizawa M, et al. Generation of functional human T-cell subsets with HLA-restricted immune responses in HLA class I expressing NOD/SCID/IL2rγnull humanized mice. Proc Natl Acad Sci USA. 2010;107:13022–13027. doi: 10.1073/pnas.1000475107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Sato K, Kondo M, Sakuta K, Hosoi A, Noji S, Sugiura M, et al. Impact of culture medium on the expansion of T cells for immunotherapy. Cytotherapy. 2009;11:936–946. doi: 10.3109/14653240903219114. [DOI] [PubMed] [Google Scholar]
  139. Bennouna J, Bompas E, Neidhardt EM, Rolland F, Philip I, Galéa C, et al. Phase-I study of Innacell gammadelta, an autologous cell-therapy product highly enriched in gamma9delta2 T lymphocytes, in combination with IL-2, in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother. 2008;57:1599–1609. doi: 10.1007/s00262-008-0491-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Kobayashi H, Tanaka Y, Yagi J, Osaka Y, Nakazawa H, Uchiyama T, et al. Safety profile and anti-tumor effects of adoptive immunotherapy using gamma-delta T cells against advanced renal cell carcinoma: a pilot study. Cancer Immunol Immunother. 2007;56:469–476. doi: 10.1007/s00262-006-0199-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Nakajima J, Murakawa T, Fukami T, Goto S, Kaneko T, Yoshida Y, et al. A phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous gammadelta T cells. Eur J Cardiothorac Surg. 2010;37:1191–1197. doi: 10.1016/j.ejcts.2009.11.051. [DOI] [PubMed] [Google Scholar]
  142. Kobayashi H, Tanaka Y, Shimmura H, Minato N, Tanabe K. Complete remission of lung metastasis following adoptive immunotherapy using activated autologous γδ T-cells in a patient with renal cell carcinoma. Anticancer Res. 2010;30:575–579. [PubMed] [Google Scholar]
  143. Ali Z, Shao L, Halliday L, Reichenberg A, Hintz M, Jomaa H, et al. Prolonged (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate-driven antimicrobial and cytotoxic responses of pulmonary and systemic Vgamma2Vdelta2 T cells in macaques. J Immunol. 2007;179:8287–8296. doi: 10.4049/jimmunol.179.12.8287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Huang D, Chen CY, Ali Z, Shao L, Shen L, Lockman HA, et al. Antigen-specific Vgamma2Vdelta2 T effector cells confer homeostatic protection against pneumonic plaque lesions. Proc Natl Acad Sci USA. 2009;106:7553–7558. doi: 10.1073/pnas.0811250106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Russell RG. Bisphosphonates: the first 40 years. Bone. 2011;49:2–19. doi: 10.1016/j.bone.2011.04.022. [DOI] [PubMed] [Google Scholar]
  146. Castella B, Riganti C, Fiore F, Pantaleoni F, Canepari ME, Peola S, et al. Immune modulation by zoledronic acid in human myeloma: an advantageous cross talk between Vγ9Vδ2 T cells, γδ CD8+ T cells, regulatory T cells, and dendritic cells. J Immunol. 2011;187:1578–1590. doi: 10.4049/jimmunol.1002514. [DOI] [PubMed] [Google Scholar]
  147. Kabelitz D, Wesch D, He W. Perspectives of gammadelta T cells in tumor immunology. Cancer Res. 2007;67:5–8. doi: 10.1158/0008-5472.CAN-06-3069. [DOI] [PubMed] [Google Scholar]
  148. Laggner U, Lopez JS, Perera G, Warbey VS, Sita-Lumsden A, O'Doherty MJ, et al. Regression of melanoma metastases following treatment with the n-bisphosphonate zoledronate and localised radiotherapy. Clin Immunol. 2009;131:367–373. doi: 10.1016/j.clim.2009.01.008. [DOI] [PubMed] [Google Scholar]
  149. Mehrle S, Watzl C, von Lilienfeld-Toal M, Amoroso A, Schmidt J, Märten A. Comparison of phenotype of gammadelta T cells generated using various cultivation methods. Immunol Lett. 2009;125:53–58. doi: 10.1016/j.imlet.2009.05.009. [DOI] [PubMed] [Google Scholar]
  150. Salot S, Bercegeay S, Dreno B, Saïagh S, Scaglione V, Bonnafous C, et al. Large scale expansion of Vgamma9Vdelta 2 T lymphocytes from human peripheral blood mononuclear cells after a positive selection using MACS “TCR Gamma/Delta+ T Cell Isolation Kit”. J Immunol Methods. 2009;347:12–18. doi: 10.1016/j.jim.2009.05.006. [DOI] [PubMed] [Google Scholar]
  151. Zhou J, Kang N, Cui L, Ba D, He W. Anti-gammadelta TCR antibody-expanded gammadelta T cells: a better choice for the adoptive immunotherapy of lymphoid malignancies. Cell Mol Immunol. 2011;9:34–44. doi: 10.1038/cmi.2011.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Huang D, Shen Y, Qiu L, Chen C, Shen L, Estep J, et al. Immune distribution and localization of phosphoantigen-specific Vgamma2Vdelta2 T cells in lymphoid and nonlymphoid tissues in Mycobacterium tuberculosis infection. Infect Immun. 2008;76:426–436. doi: 10.1128/IAI.01008-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Altvater B, Pscherer S, Landmeier S, Kailayangiri S, Savoldo B, Juergens H, et al. Activated human gammadelta T cells induce peptide-specific CD8+ T-cell responses to tumor-associated self-antigens. Cancer Immunol Immunother. 2011;61:385–396. doi: 10.1007/s00262-011-1111-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Dieli F, Gebbia N, Poccia F, Caccamo N, Montesano C, Fulfaro F, et al. Induction of gammadelta T-lymphocyte effector functions by bisphosphonate zoledronic acid in cancer patients in vivo. . Blood. 2003;102:2310–2311. doi: 10.1182/blood-2003-05-1655. [DOI] [PubMed] [Google Scholar]
  155. Kunzmann V, Bauer E, Wilhelm M. Gamma/delta T-cell stimulation by pamidronate. N Engl J Med. 1999;340:737–738. doi: 10.1056/NEJM199903043400914. [DOI] [PubMed] [Google Scholar]
  156. Lukens JR, Barr MJ, Chaplin DD, Chi H, Kanneganti TD. Inflammasome-derived IL-1beta regulates the production of GM-CSF by CD4+ T cells and gammadelta T cells. J Immunol. 2012;188:3107–3115. doi: 10.4049/jimmunol.1103308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Bruder J, Siewert K, Obermeier B, Malotka J, Scheinert P, Kellermann J, et al. Target specificity of an autoreactive human gammadelta-T cell receptor in myositis. J Biol Chem. 2012;287:20986–20995. doi: 10.1074/jbc.M112.356709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Placido R, Auricchio G, Gabriele I, Galli E, Brunetti E, Colizzi V, et al. Characterization of the immune response of human cord-blood derived gamma/delta T cells to stimulation with aminobisphosphonate compounds. Int J Immunopathol Pharmacol. 2011;24:101–110. doi: 10.1177/039463201102400112. [DOI] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Immunology are provided here courtesy of Nature Publishing Group

RESOURCES