Next Article in Journal
PAR2 Serves an Indispensable Role in Controlling PAR4 Oncogenicity: The β-Catenin–p53 Axis
Previous Article in Journal
Prometastatic Potential of Non-Functionalized Multiwalled Carbon Nanotubes in the MDA-MB-436 Breast Cancer Cell Line Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases

1
State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China
2
Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, New Brunswick, NJ 08901-8554, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(6), 2778; https://doi.org/10.3390/ijms26062778
Submission received: 13 February 2025 / Revised: 15 March 2025 / Accepted: 17 March 2025 / Published: 19 March 2025
(This article belongs to the Section Molecular Biology)

Abstract

:
γδT cells, a distinct group of T lymphocytes, serve as a link between innate and adaptive immune responses. They are pivotal in the pathogenesis of various liver disorders, such as viral hepatitis, nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), liver fibrosis, autoimmune liver diseases, and hepatocellular carcinoma (HCC). Despite their importance, the functional diversity and regulatory mechanisms of γδT cells remain incompletely understood. Recent advances in high-throughput single-cell sequencing and spatial transcriptomics have revealed significant heterogeneity among γδT cell subsets, particularly Vδ1+ and Vδ2+, which exhibit distinct immunological roles. Vδ1+ T cells are mainly tissue-resident and contribute to tumor immunity and chronic inflammation, while Vδ2+ T cells, predominantly found in peripheral blood, play roles in systemic immune surveillance but may undergo dysfunction in chronic liver diseases. Additionally, γδT17 cells exacerbate inflammation in NAFLD and ALD, whereas IFN-γ-secreting γδT cells contribute to antiviral and antifibrotic responses. These discoveries have laid the foundation for the creation of innovative solutions. γδT cell-based immunotherapeutic approaches, such as adoptive cell transfer, immune checkpoint inhibition, and strategies targeting metabolic pathways. Future research should focus on harnessing γδT cells’ therapeutic potential through targeted interventions, offering promising prospects for precision immunotherapy in liver diseases.

1. Introduction

The liver serves not only as the body’s main organ for metabolism and detoxification but also as an essential component of the immune response, continuously surveilling and reacting to pathogens and signals associated with tissue damage [1]. With advancements in immunological research, γδT cells, a unique and multifunctional T cell subset [2], have garnered significant attention for their roles within the hepatic immune microenvironment [3,4]. These cells connect innate immunity and adaptive immunity [5] and have been proved to contribute to infection defense [6], wound healing [7], cancer immunity [8], and transplant rejection [9].
Recent developments in high-throughput single-cell sequencing and spatial transcriptomics have unveiled substantial heterogeneity among γδT cell subsets [10,11,12,13]. Studies indicate that different γδT cell subsets, particularly Vδ1+ and Vδ2+, exhibit divergent functional properties in liver pathology [14]. For instance, Vδ1+ T cells demonstrate potent anti-tumor activity in hepatocellular carcinoma (HCC) by secreting cytotoxic factors and regulating immune checkpoint molecules, thereby inhibiting tumor cell proliferation and metastasis [15]. Conversely, Vδ2+ T cells primarily function in inflammation regulation and tissue repair, contributing to immune homeostasis in conditions such as chronic hepatitis B (CHB) and alcoholic liver disease (ALD) [16]. Moreover, research conducted both in vitro and in vivo indicates that γδT cells participate in intricate interactions with macrophages, dendritic cells, and Treg cells, significantly influencing the development and resolution of liver diseases [17].

2. Background of γδT Cells

2.1. Development and Classification of γδT Cells

γδT cells, a unique subset within the T cell family, exhibit distinct developmental processes and classification standards compared to conventional αβT cells [18]. γδT cells arise from multipotent hematopoietic stem cells within the bone marrow, subsequently migrating to the thymus, where they undergo a unique differentiation and selection process to reach maturity [19]. Within the thymus, γδT cells undergo rearrangement of the γ and δ receptor chains, generating a diverse set of receptors that offer enhanced specificity and variation in immune responses [20]. In contrast to αβT cells that depend on traditional MHC molecules for antigen presentation, γδT cells have the ability to detect various non-classical antigens, such as stress-related molecules, metabolic byproducts, and lipids. This capability provides them with a distinct advantage in immune monitoring and swift immune responses [21]. While MHC-restricted γδT cells have been observed, they constitute only a minor portion of the overall γδT cell population.To provide a visual overview of their developmental pathway and distribution in peripheral tissues, these processes are depicted in Figure 1 below.
The TCR of γδT cells undergoes a somatic DNA rearrangement process involving the variable (V), diversity (D, exclusive to TRD), and joining (J) regions, generating a wide diversity of individual TCR chain combinations [22]. Furthermore, the combination of TRG and TRD chains, along with the incorporation of palindromic (P) nucleotides and non-template (N) nucleotides by terminal deoxynucleotidyl transferase (TdT) at the V(D)J junction, contributes significantly to increasing the diversity [23]. However, the clonal diversity within an individual’s γδTCR repertoire may be significantly smaller [24].

2.2. Classification of Human γδT Cells Based on δTCR Chains

In humans, γδT cells are categorized into three primary subsets—Vδ1+, Vδ2+, and Vδ3+—which are distinguished by the TCR δ chain (encoded by the TRDV gene) [25]. The Vδ2+ T cells primarily associate with the Vγ9 chain, creating the Vγ9Vδ2 TCR complex, which represents the most abundant subset of γδT cells in peripheral blood circulation [26]. In contrast to Vδ1+ T cells, which develop in the thymus months after birth, Vγ9Vδ2 T cells are generated early during fetal development. This early development indicates their function as crucial components of the body’s natural defense system, acting as the initial line of protection [27]. However, the Vγ9-negative Vδ2+ subset exhibits a broader functional diversity, sometimes showing characteristics of adaptive immunity [8].To further illustrate the diversity and functional characteristics of these human γδT cell subsets, their key features are summarized in Table 1 below.
Vδ1+ T cells are predominantly located in various tissues, including the intestinal epithelium, skin, spleen, and liver, while only a limited quantity can be detected in peripheral blood [28]. Vδ1+ TCRs pair with various Vγ chains, displaying significant flexibility, and their diversity predominantly originates from the TRD gene library rather than the TRG gene library [28]. Sequencing studies indicate that Vδ1+ T cells in adults often exhibit a distinct clonal focus, forming private Vδ1+ T cell clones [29,30]. These cells can rapidly expand in response to infections or tumors, exhibiting characteristics similar to memory T cells [28,30,31]. This contrasts with Vγ9Vδ2 T cells, which tend to undergo activation-induced cell death (AICD) after prolonged antigen stimulation [32,33]. Furthermore, non-Vγ9Vδ2 subsets, such as Vδ1+, Vγ9Vδ2+, and Vδ3+ cells, can also undergo clonal expansion in both healthy and diseased individuals, leading to the idea that they “record” the immune history of the body, reflecting antigen exposure and immune memory [12]. For example, in infections or tumors [34,35,36,37], the Vδ1+/Vδ2+ ratio is often inverted in peripheral blood and tissues [38], suggesting that Vδ1+ T cells play a crucial role through clonal expansion in chronic infections and tumor microenvironments. Approaches aimed at addressing this issue, such as the adoptive transfer of autologous Vδ2+ T cells, could potentially restore the optimal Vδ1+/Vδ2+ ratio, alleviate tumor burden, and facilitate the reorganization of the tumor microenvironment (TME) [39].
The Vδ3+ T cell subset is extremely rare in peripheral blood but is significantly enriched in the liver and intestinal epithelium [40,41,42,43,44]. Its TCR recognizes ligands similar to those recognized by Vδ1+ T cells [44] and also expresses high levels of CD16 (a low-affinity IgG Fc receptor, FcγRIII), providing antibody-dependent cellular cytotoxicity (ADCC) capabilities [45]. Vδ3+ T cells display increased cytotoxic activity and undergo clonal expansion when infected with malaria, CMV, and HCV [46], highlighting their significant involvement in immune responses to infections. Additionally, recent research demonstrates that Vδ3+ T cells infiltrate or proliferate within different tumors, implying a potential function in the immune defense against cancer [47,48,49,50]. In vitro, amplified Vδ3+ T cells have been demonstrated to stimulate B cell differentiation and enhance IgM production [51]. However, due to the rarity of Vδ3+ T cells, research evidence remains limited, and their functions in physiological and pathological states require further exploration and validation.
This review centers on Vδ1+ and Vδ2+ T cells, given their extensive representation in existing research and their significance for both experimental studies and clinical applications.

2.2.1. Vδ1+ T Cells

Vδ1+ T cells play a critical role in adaptive immunity, utilizing γδTCRs and natural killer receptors (NKRs) to facilitate the detection of tumor-associated antigens and stress signals. Although the roles of Vδ1+ and Vδ2+ T cells in anti-tumor immunity exhibit some overlap, there are notable distinctions between the two subsets. Vδ1 TCRs predominantly identify MHC-like molecules from the CD1 family (e.g., CD1c and CD1d) [52,53,54,55,56,57], as well as membrane-bound proteins such as A2 [58] and MICA/B (MHC class I chain-related proteins A and B) [54,59], which are frequently upregulated in cancerous and virus-infected cells. Studies have shown that the binding affinity of Vδ1 TCRs for CD1d is significantly higher than for MICA/B, suggesting a non-redundant role in specific immune surveillance mechanisms [25]. Similarly to Vδ2+ T cells, Vδ1+ T cells show increased expression of the stress-responsive molecule NKG2D, which binds to MICA/B present on tumor cell membranes. However, the binding regions for MICA/B vary between the Vδ1+ TCR and NKG2D, with the affinity of NKG2D for MICA/B being a thousand times greater than that of the Vδ1+ TCR-MICA/B interaction [59].
Although Vδ1+ and Vδ2+ T cells recognize antigens via different mechanisms, both subsets facilitate the induction of target cell apoptosis through perforin/granzyme B-driven cytotoxic pathways and death receptor signaling (e.g., TRAIL/TRAIL-R and Fas/FasL). These mechanisms highlight that Vδ1+ T cells are involved in detecting tumor and stress-related antigens via their specialized TCRs and also triggering cytotoxic responses through various effector pathways. As a result, Vδ1+ T cells are essential for the adaptive immune functions performed by γδT cell subsets.

2.2.2. Vδ2+ T Cells

The stimulation and antigen recognition of Vδ2+ T cells primarily depends on the existence of phosphoantigens. Ligand recognition occurs via two separate pathways: one facilitated by the γδTCR and the other by natural killer receptors (NKR) [60]. In contrast to other γδT cell subsets, the Vγ9Vδ2 TCR primarily identifies phosphoantigens, which accumulate within tumor cells as a result of disturbances in the mevalonate metabolic pathway [61,62,63,64]. However, these phosphoantigens do not directly interact with the γδTCR. Rather, they initially bind to the B30.2 domain of the BTN3A1 protein inside the cell, inducing a conformational alteration in BTN3A1 [65,66]. This, in turn, causes the hinge region of the costimulatory protein BTN2A1 to interact with the Vγ9 chain, ultimately activating Vδ2+ T cells [67,68,69,70]. This indirect recognition mechanism has not been fully elucidated, and there remains controversy over whether the Vδ2 chain directly participates in this process. Additionally, the Vγ9Vδ2 TCR can also interact with proteins that are highly expressed in cancer cells, such as F1-ATPase, apolipoprotein A1, and hMSH2, which further enhance its anti-tumor activity [71,72,73]. It is noteworthy that rodents do not possess homologous γδTCRs that can be activated by phosphoantigens, meaning traditional mouse models are unsuitable for studying the capability of Vγ9Vδ2 T cells. Recently, studies in alpacas have revealed Vγ9Vδ2 TCRs responsive to phosphoantigens [74], providing a new non-primate animal model and expanding current research capabilities.
In addition to TCR-mediated recognition, NKR signaling also plays a crucial role in the activation of Vδ2+ T cells and the lysis of tumor cells. The NKG2D receptor on the surface of Vδ2+ T cells can bind to MICA/B [75,76] and UL16 binding proteins (ULBP) [77,78] expressed by cancer cells, while DNAM-1 interacts with Nectin-like 5, collectively promoting perforin/granzyme-mediated cytotoxic responses [79]. Furthermore, Vδ2+ T cells express the CD16 molecule, enabling them to mediate antibody-dependent cellular cytotoxicity (ADCC) through interaction with tumor-specific antibodies [80,81,82]. In vitro studies have shown that this ADCC activity is primarily confined to Vδ2+ T cells, whereas Vδ1+ T cells generally lack this function [83]. However, CD16 expression has been observed on Vδ1+ T cells in patients with viral infections, suggesting functional plasticity across subsets in specific environments [45,84]. Therefore, further investigation into the phenotypic differences in antigen recognition and effector functions between Vδ2+ and Vδ1+ T cells can help clarify their specific clinical advantages in different pathological contexts, providing new insights for targeted γδT cell immunotherapies.

2.3. Effector Subsets Defined by Cytokine Release

The functional properties of γδT cells vary according to the cytokines they secrete, and they can be categorized into distinct subsets based on their effector abilities. Early studies revealed that γδT cells are a primary source of IFN-γ at the onset of immune responses, playing a critical role in anti-tumor immunity across various cancer types [85,86,87,88,89]. However, subsequent research has revealed that γδT cells can, in some contexts, promote tumor growth. For example, in IL-17 knockout mice, γδT cells lacking IL-17 inhibited tumor progression significantly, suggesting that IL-17-producing γδT cells have the potential to promote tumor growth [90,91,92,93,94,95]. Thus, it is difficult to precisely classify the immune functions of γδT cells based solely on TCR chain structures, and researchers tend to classify them into effector subsets based on the types of cytokines they secrete.
The two major effector subsets of γδT cells are γδT1 and γδT17. γδT1 cells secrete IFN-γ and play crucial roles in anti-tumor immunity and intracellular pathogen clearance, while γδT17 cells secrete IL-17, providing protection against extracellular bacterial and fungal infections but potentially contributing to inflammation and disease exacerbation in tumors and certain autoimmune diseases [2,96,97]. Additionally, γδT cells also include a γδNKT subset, which simultaneously secretes IL-4 and IFN-γ, drawing increasing attention [98,99]. The development of these γδNKT and γδT1 subsets relies on strong TCR signaling, whereas γδT17 cells can develop in the absence of TCR ligand selection, highlighting the diverse developmental mechanisms of different subsets [99,100,101,102].
The functionality of γδT cells is highly context-dependent, with their behavior being strongly influenced by the surrounding microenvironment. For instance, γδT1 cells promote anti-tumor immunity within the tumor microenvironment (TME), whereas γδT17 cells, under certain inflammatory conditions, may become dysregulated and contribute to tumor progression [103,104,105,106,107]. Additionally, research has demonstrated that γδT cells exposed to inflammatory cues in the TME can undergo reprogramming into γδTreg cells [103,104,108], which exhibit immunosuppressive functions and attenuate anti-tumor immune responses. This phenomenon is corroborated by the detection of CD73+Foxp3+Vδ1+ cells within the peripheral blood and the tumor microenvironment of breast cancer patients [109]. Similarly, in colon cancer, tumor-infiltrating γδTreg cells frequently display a CD39+Foxp3+ phenotype, reinforcing their immunosuppressive characteristics. Moreover, CD39+ and CD73+ γδTreg cells regulate immune responses by modulating extracellular adenosine metabolism, further contributing to immune suppression [110,111]. Studies have also found that under certain pathological conditions, a subset of γδT cells may initiate Th2-like responses by secreting IL-4 [112,113], indicating functional diversity and plasticity. Overall, the characteristics of these effector subsets suggest that the behavior of γδT cells in different tissue microenvironments is highly context-dependent, and their effector functions play important roles in anti-infection, anti-tumor, and immune regulatory processes [40,60,114,115].
Increasing evidence shows that the functions of Vδ1+ and Vδ2+ T cells are highly plastic and are regulated by the cytokine microenvironment. Typically, Vδ1+ T cells are considered to have pro-tumor characteristics, whereas Vδ2+ T cells exhibit anti-tumor functions. However, under certain cytokine conditions, the functions of these subsets can shift. IL-6 is a pleiotropic cytokine that is frequently overexpressed in both cancerous and non-cancerous tissues in response to infection and tissue damage [116]. When Vδ2+ T cells are exposed to a combination of IL-1β, TGF-β, IL-6, and IL-23, they are capable of differentiating into γδT17 cells that produce IL-17. When exposed to TGF-β1, IL-15, and antigenic stimuli, these cells can be prompted to upregulate FOXP3 expression, resulting in their conversion into Tregs (regulatory T cells) [117,118]. Interestingly, when vitamin C, an epigenetic modifier, is added, the degree of FOXP3 gene demethylation increases, further enhancing its immune regulatory function [118]. Moreover, IL-4 is thought to inhibit γδT cell-driven anti-tumor responses by directing the γδT cell population towards producing the immunosuppressive cytokine IL-10, thereby enhancing the expansion of the Vδ1+ subset and suppressing the IFN-γ-producing Vδ2+ subset [108]. Clinical studies have shown that in cancer patients, both IL-17-secreting Vδ2+ cells and IFN-γ-secreting Vδ1+ cells are present [119,120,121], and these two subsets exhibit distinct cytotoxic marker profiles in the tumor environment [122]. Additionally, in the liver, γδT cells are predominantly composed of polyclonal Vδ1+ T cells, whose phenotype differs from their counterparts in the blood, suggesting that Vδ1+ T cells exhibit greater functional plasticity within the tissue microenvironment [31]. Notably, research by Hayday et al. has associated Vδ1+ T cells, rather than Vδ2+ T cells, with improved outcomes in individuals diagnosed with triple-negative breast cancer (TNBC), indicating a potential protective function of Vδ1+ T cells in certain cancer types [123]. During tumor initiation and progression, γδT cells demonstrate remarkable multifunctionality, with their functional characteristics being highly influenced by tumor type, stage, and microenvironment [8,119]. Furthermore, in addition to their involvement in tumors, γδT cell subsets demonstrate considerable functional diversity and adaptability, allowing them to have either beneficial [123,124,125,126] or harmful [127,128,129] effects in the setting of infections and autoimmune conditions.

2.4. Molecular Mechanisms and Signaling Pathways

The activation and functional regulation of γδT cells rely on multiple key molecular interactions, including activating receptors, costimulatory molecules, inhibitory signals, metabolic adaptations, and epigenetic modifications. In contrast to traditional αβ T cells, γδT cells detect antigens independently of MHC restrictions, reacting to stress-induced molecules like MICA/B [130,131]. Among these, the activating receptor NKG2D plays a critical role in triggering cytotoxic responses and promoting the secretion of inflammatory cytokines such as IFN-γ and perforin [59,75]. Additionally, the activation of γδT cells is fine-tuned by costimulatory signals, including CD28 [132] and 4-1BB (CD137) [133]. CD28 sustains γδT cell proliferation and survival by binding to CD80/CD86 while enhancing antigen-specific responses [132]. Meanwhile, 4-1BB and its ligand 4-1BBL enhance the longevity of γδT cells, allowing them to remain functionally active in prolonged immune responses [133].
Although γδT cells possess strong effector capabilities, their activity is tightly controlled to avoid overactivation of the immune system and subsequent tissue injury. The interaction of PD-1 with its ligands, PD-L1/PD-L2, suppresses γδT cell activation, a strategy commonly employed by tumors to evade immune responses [134]. CTLA-4 competes with CD28 for CD80/CD86 binding, thereby suppressing costimulatory signaling and limiting γδT cell activation. LAG-3 interacts with MHC II to further dampen immune responses. These inhibitory signals profoundly impact γδT cells in the tumor microenvironment (TME), contributing to functional exhaustion and diminished anti-tumor immunity [135,136].
The signaling pathways governing γδT cell function involve primarily the NF-κB [137], MAPK [138,139], and PI3K/Akt [140] pathways, which regulate activation, survival, proliferation, metabolic adaptation, and inflammatory responses. However, in TME, abnormal activation of this pathway may promote immunosuppression. Additionally, hypoxic conditions within the TME influence these signaling pathways and induce metabolic reprogramming, ultimately limiting γδT cell persistence and function [141]. Beyond classical signaling pathways, epigenetic modifications significantly shape γδT cell differentiation and function [142,143,144]. Metabolites such as lactate, α-ketoglutarate (α-KG), and acetyl-CoA regulate histone modifications, impacting γδT cell transcriptional programs and determining their activation or tolerance states [145]. While certain epigenetic mechanisms in γδT cells share similarities with αβ T cells, γδT cells exhibit distinct regulatory features in transcription factor dependency and cytokine-mediated modulation. Most studies in this area rely on murine models, and further research is needed to fully elucidate epigenetic regulation in human γδT cells.
Metabolic adaptation is essential for γδT cell function, as these cells predominantly rely on glycolysis rather than oxidative phosphorylation for energy production. This metabolic preference is crucial for sustaining effector functions; however, the hypoxic conditions of the TME limit glycolytic efficiency, thereby compromising γδT cell survival and proliferation [141]. Notably, in glioblastoma models, hypoxia impairs γδT cell anti-tumor activity, while in other cancers, hypoxia enhances cytotoxicity but simultaneously restricts proliferation. Moreover, oxidative stress plays a key role in γδT cell regulation, with reactive oxygen species (ROS) in the TME acting as both inhibitory and modulatory factors [146]. While ROS can suppress γδT cell-mediated immunity, they may also indirectly enhance anti-tumor responses by inhibiting pro-tumor γδT17 subsets [147]. The precise role of ROS in γδT cell function requires further investigation to optimize immunotherapeutic strategies [148].
Exosome-mediated intercellular communication is another crucial factor in γδT cell regulation [149]. Tumor-derived exosomes (TDEs) modulate γδT cell activity via non-coding RNAs such as lncRNA SNHG16, promoting the expansion of immunosuppressive γδT cell subsets like CD73+ Vδ1+ γδTregs [109]. Furthermore, TDEs may enhance the suppressive function of myeloid-derived suppressor cells (MDSCs), further diminishing γδT cell anti-tumor activity [150]. Conversely, exosomes derived from activated Vδ2+ T cells have demonstrated anti-tumor properties and are being explored for adoptive γδT cell-based therapies [151].
Overall, γδT cells demonstrate remarkable functional plasticity, coordinating immune surveillance, tumor immunity, infection defense, and fibrosis regulation through a complex network of activating receptors, inhibitory signals, metabolic adaptations, epigenetic modifications, and intercellular communication. A more comprehensive understanding of the molecular mechanisms of γδT cells will enhance the development of targeted immunotherapies for cancer, infectious diseases, and autoimmune disorders.

3. Subset Specific Functions of γδT Cells in Various Liver Diseases

3.1. Viral Hepatitis

Viral hepatitis refers to a group of inflammatory liver diseases caused by various hepatitis viruses. Among them, the hepatitis B virus (HBV) and hepatitis C virus (HCV) are associated with high infection rates and significant global disease burden [152,153]. Chronic infections frequently lead to cirrhosis, liver failure, and hepatocellular carcinoma, presenting a significant challenge to public health [152]. With the growing understanding of its pathogenesis, it has become increasingly clear that the host immune response plays a crucial role in regulating virus replication and liver injury, particularly the involvement of γδT cells [154].
The Hepatitis B virus (HBV) represents a major global health concern and is widely distributed around the world. Currently, approximately 257 million people are affected by HBV worldwide, with 68% of cases concentrated in Africa and the Western Pacific region [155]. Among individuals with acute HBV infection, nearly 5% of adults develop chronic hepatitis B, whereas the others clear the virus spontaneously through a self-limiting process [156]. Growing research indicates that variations in HBV infection outcomes are linked to the strength of the antiviral immune response. Several studies have observed a notable rise in the quantity of γδT cells within the liver tissue of acute hepatitis B (AHB) patients, whereas their levels in peripheral blood were found to decrease [157]. Peripheral γδT cells exhibit robust activation, ultimately evolving into memory phenotypes, which enhances their cytotoxic and antiviral functions. In asymptomatic individuals infected with HBV, the proportions of peripheral Vδ1+ and Vδ2+ T cells are higher compared to healthy individuals, with a significant increase in the levels of IFN-γ+ Vδ2+ T cells [158]. In the AHB-infected mouse model, a marked expansion of γδT cells in the liver was observed as HBV markers increased. Simultaneously, the expression of activation markers, including CD69, IFN-γ, and IFN-β mRNA, was upregulated [159]. These findings imply that γδT cells may play an antiviral role in AHB patients, potentially restraining the advancement in acute infection.
Following chronic infection, HBV persists by maintaining a stable reservoir of covalently closed circular DNA (cccDNA) within host cells through intracellular genomic recycling and subsequent reinfection, making complete eradication of the virus highly challenging [160]. The role of γδT cells in chronic hepatitis B (CHB) is complex and sometimes contradictory. Several studies have reported a significant reduction in the proportion of Vδ2+ T cells in both the liver and peripheral circulation of patients with severe CHB. Additionally, IFN-γ or TNF-α-induced cytotoxicity was compromised but could be reinstated through IFN-α treatment in both in vitro and in vivo conditions [161,162]. It has been reported that, following proliferation in the periphery, human Vδ2+ T cells can suppress the release of inflammatory factors from pathogenic Th17 cells, thereby helping to alleviate liver injury [161,163]. However, some studies have indicated that the frequency of γδT cells and their subsets does not significantly differ in CHB patients, with some reports even suggesting enhanced antiviral function in Vδ2+ T cells [16]. This implies that variations in patient inclusion criteria (such as age, gender, and ethnicity) may contribute to inconsistent results [164]. In addition, some studies suggest that certain subsets of γδT cells (such as CD4 CD8 types or IL-17-producing cells) may contribute to the persistence and exacerbation of chronic HBV infection by suppressing CD8 T cell activity, promoting the activation of myeloid-derived suppressor cells (MDSCs), or directly enhancing inflammation [165,166]. Combined with the above phenomena, γδT cells often show different functional differentiation states in the process of chronic HBV infection: on the one hand, some γδT cell subsets that can produce IFN-γ or TNF-α have the potential to inhibit the virus and reduce inflammation in the early stage or under specific conditions. On the other hand, some subsets or γδT cells under certain conditions (such as Vδ2+ depletion, CD4 CD8, or IL-17 secreting subsets) may aggravate inflammation and immune imbalance, leading to liver injury or disease progression. It is worth mentioning that recent studies have compared the phenotypes of γδT cells in the liver, spleen, thymus and small intestine of mice from embryonic to adult stage, and identified a group of CXCR3+CXCR6+γδT cells with tissue resident characteristics [167]. It is retained in the liver through chemokines produced by hepatic sinusoidal endothelial cells and generates substantial amounts of IFN-γ during acute HBV infection. The adoptive transfer of these CXCR3+CXCR6+γδT cells into TCR δ/ mice results in reduced levels of HBsAg and HBeAg, suggesting their protective role in acute HBV infection [167]. It aims to amplify and activate these liver resident CXCR3+CXCR6+ γδT cells in vivo or in vitro, or it may become a specific immunotherapy for acute HBV infection.
In chronic hepatitis C (CHC), significant attention has been given to the role of γδT cells in pathogenesis. Studies have indicated that the number of γδT cells in the liver of CHC patients is typically higher compared to healthy controls, with Vδ1+ T cells predominantly localized in the liver [168,169]. However, compared to healthy controls and asymptomatic HCV carriers, the number of Vγ9Vδ2 and Vδ1+ T cells in the peripheral blood of CHC patients tends to be reduced [170]. In the mouse model, compared with the wild type, the level of γδT cells in the liver of HCV transgenic mice was significantly increased [171], suggesting that peripheral γδT cells may be recruited into the liver to participate in the pathological process of HCV infection. Some studies have shown that γδT cells in the liver of CHC patients exhibit high cytotoxic activity and the ability to secrete pro-inflammatory factors [168]. Specifically, IFN-γ+ Vδ1+ T cells are positively correlated with the extent of liver necrotizing inflammation [169], reflecting the “antiviral-inflammatory” dual effect of γδT cells to some degree. However, even after HCV clearance or antiviral therapy, some patients with CHC still have dysfunction of peripheral γδT cells [46]. It has been observed that although peripheral Vγ9Vδ2+ T cells from patients with CHC can express cytotoxic markers such as perforin, granzyme B, and CD107a, their IFN-γ secretion function is significantly impaired [172]. IFN-α intervention can upregulate cytotoxic molecules but cannot restore IFN-γ production ability [173]. Although the new direct-acting antiviral drugs (DAA) treatment can induce slight changes in γδT cell number or TCR spectrum after one year [174], Vγ9Vδ2+ T cells in some patients still find it difficult to completely restore their normal cytokine response and proliferation characteristics [46,175]. To some extent, this indicates that the γδT cell functional damage caused by CHC is persistent and can continue to affect the immune balance and liver homeostasis after the virus is cleared.
In conclusion, the function of γδT cells in viral hepatitis is complex and shows obvious heterogeneity: in human acute HBV infection, γδT cells can help inhibit disease progression through enhanced cytotoxic and antiviral activity. In chronic HBV infection, its role is due to subgroup function, inflammatory state, individual differences, and other factors that have protective or pathogenic two-way effects. In CHC, γδT cells in the liver typically exhibit strong cytotoxic activity and pro-inflammatory traits; however, the quantity or functionality of γδT cells in peripheral circulation is frequently impaired, and complete recovery remains challenging even after antiviral treatment. In combination with the above, the Vδ1+ subgroup is more likely to reside locally in the liver, participate in direct antiviral and amplify inflammation; the Vδ2+ subgroup is mainly active in peripheral blood, which is prone to dysfunction or differentiation deviation in chronic infection. With the continuous advancement in single-cell and multi-group technologies, we will have a clearer understanding of the temporal and spatial distribution, functional diversity, and regulatory network of γδT cells in viral hepatitis and provide more theoretical support for hepatitis prevention and treatment strategies based on γδT cell regulation.

3.2. Liver Bacterial and Parasitic Infections

γδT cells are the primary producers of IL-17 during hepatic Schistosoma japonicum infection, serving as the initial line of defense before the activation of the T helper 17 (Th17) response. IL-17 is crucial in inducing granulomatous inflammation and fibrosis, both of which can be alleviated by anti-IL-17 monoclonal antibodies [176]. Research has shown that γδT cells producing IL-9 play a role in immune responses in C57BL/6 mice infected with Schistosoma japonicum. In γδT cell-deficient mice infected with Listeria monocytogenes, liver injury is mainly attributed to TNF-α release from CD8+ T cells. However, this damage can be reversed by infusing Vδ4+ γδT cells that produce IL-10, which in turn modulates CD8+ T cell proliferation and decreases TNF-α secretion. However, this pathological damage can be reversed through the infusion of Vδ4+ γδT cells that secrete IL-10, which subsequently regulates CD8+ T cell proliferation and reduces TNF-α production. Therefore, γδT cells are essential for protecting hepatic tissue by modulating CD8+ T cell responses to pathogenic stimuli and ensuring the maintenance of CD8+ T cell homeostasis [177]. Research indicates that γδT cells are particularly significant during the early phase of infection, whereas αβ T cells become more active during the later stages. Moreover, Vδ4+ γδT cells’ secretion of IL-17 has been demonstrated to protect against infections. In addition to bacterial and viral pathogens, γδT cells play a critical role in defending against parasitic infections, highlighting their significant immunoregulatory and host-defense functions.

3.3. NAFLD and ALD

Recent evidence suggests that alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) share more common mechanisms than previously thought, despite their distinct etiologies. Both conditions are characterized by excessive lipid accumulation in hepatocytes, driven by dysregulated lipid metabolism [178], and exacerbated by oxidative stress from reactive oxygen species [179]. Additionally, they exhibit overlapping inflammatory pathways, including activation of innate immune responses via Kupffer cells and recruitment of pro-inflammatory cytokines (e.g., TNF-α, IL-6), as well as immune dysregulation involving γδT cells and other immune populations [180]. These shared features underscore the potential for common therapeutic targets, such as those explored in γδT cell-based strategies for liver disease. Nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) are two types of metabolic liver diseases, which are characterized by the gradual progress from steatosis to steatohepatitis and even liver fibrosis and cirrhosis [181]. Studies have found that in NAFLD models induced by a high-fat diet (HFD) and high-fat and high-cholesterol diet (hfhcd), γδT cells, especially γδT17 cells produced by IL-17A, are significantly enriched in the liver, and aggravate liver inflammation and injury by recruiting granulocytes and inducing reactive oxygen species [182]. The liver–spleen axis plays a pivotal role in NAFLD by recruiting immune cells, such as monocytes and T cells, to the liver via the portal circulation, potentially serving as a partial source of hepatic γδT cells contributing to inflammation [183]. This immune modulation, alongside cytokine release (e.g., IL-6), amplifies the chronic low-grade inflammation characteristic of NAFLD progression. Moreover, growth factors such as transforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF) contribute to NAFLD pathogenesis by promoting stellate cell activation and fibrogenesis, further exacerbating liver injury in concert with cytokines and chemokines [184]. Studies have shown that the adoptive transfer of γδT cells into hfhcd-fed mice can accelerate NAFLD, while γδT cell deficiency can protect mice from steatohepatitis, which shows that liver injury and inflammation are significantly reduced [185]. Similarly, CD161+γδT cells were enriched in the liver of NAFLD patients, suggesting that γδT cells produced by IL-17A are important regulatory factors for the progression of NAFLD [156].
Besides the IL-17 signaling pathway, γδT cells also affect the progression of NAFLD and ALD through other mechanisms. For example, chronic alcohol intake (ash), methionine choline deficiency diet (NASH), and the combination model of excessive alcohol consumption and high-fat diet showed significant infiltration of γδT cells in the liver, which was regulated by CCR2 [180] and NOD2 [186] signaling pathways. Studies have found that γδT cells are not only the main source of IL-17A but also promote liver injury by regulating the expansion of CD4+T cells and inflammatory mode [187]. In addition, γδT cells affect the presentation of lipid antigens through the CD1d-mediated metabolic immune pathway [188], which further activates NKT cells and aggravates liver injuries. It is worth noting that hepatocyte-specific CD1d deletion can improve Nash symptoms in high-fat and high-cholesterol diets [189], which suggests that the CD1d-dependent mechanism plays a key role in the development of steatohepatitis. Akkermansia muciniphila alleviates nonalcoholic steatohepatitis by regulating γδT17 cells and macrophage polarization through TLR2 activation [190].
In conclusion, whether NAFLD or ash, γδT cells produced by IL-17A and CD1d-mediated metabolic immune mechanisms play an important role in the pathogenesis of steatohepatitis. Therefore, targeting these crucial mechanisms, such as blocking IL-17A produced by γδT17 cells or inhibiting CD1d-mediated lipid antigen presentation, may provide a novel approach to treating fatty liver disease.

3.4. Liver Fibrosis and Cirrhosis

Liver fibrosis and cirrhosis are the main pathological features of chronic liver disease progression, involving the activation of hepatic stellate cells (HSC) and excessive accumulation of extracellular matrix [191]. Liver cirrhosis is an end-stage disease caused by a variety of chronic diseases. For example, in Asia, hepatitis B virus (HBV) infection is the main cause, while in developed countries, hepatitis C virus (HCV) infection and alcohol abuse are the main factors [191]. The development of liver fibrosis is accompanied by continuous necrotizing inflammatory reaction, which activates immune cells such as HSC and Kupffer cells and ultimately leads to the formation and development of fibrosis [192,193].
In the context of chronic liver injury, γδT cells are one of the important primary immune cells in the portal vein region [194]. These cells promote the activation of HSC and Kupffer cells by secreting interleukin-17 (IL-17), which aggravates the progress of fibrosis [192,195]. Studies have shown that in the Schistosoma japonicum infection model, γδT cells recruit centralized granulocytes into the liver through IL-17A, leading to severe liver fibrosis [193]. In addition, the hmgb1-tlr4-il23-il17 signal axis between macrophages and γδT cells plays a key role in the process of liver inflammation [196]. Another study also demonstrated that when IL-23 and IL-1β were present, the binding of exosomes derived from HSCs to TLR3 boosted IL-17A production in γδT cells, which subsequently activated HSCs early on and facilitated the progression of fibrosis [197]. This process highlights the complex interaction between γδT cells and HSCs.
However, γδT cells also showed the opposite effect in the process of liver fibrosis. Some studies have shown that γδT cells induce HSC apoptosis through the expression of Fas ligand (FasL), thereby inhibiting fibrosis in some cases. For example, ccr6+γδT17 cells can induce HSC apoptosis in damaged liver tissue through fas/fasl interaction, which helps to limit fibrosis and chronic inflammation [198]. In addition, γδT cells, especially the subsets secreting interferon-γ (IFN-γ), can also enhance their cytotoxic response to HSC by activating natural killer cells (NK cells). This effect depends on the involvement of cytotoxic molecules such as NKp46, trail, and FasL in NK cells, directly or indirectly killing activated HSCs, thus playing a protective role in the process of fibrosis [199].
In general, the role of γδT cells in liver fibrosis and cirrhosis is multifaceted, and its mechanism varies with different cell subsets. γδT cells secreting IL-17A aggravate fibrosis mainly by promoting HSC activation and the production of fibrosis factors, while γδT cells with cytotoxic function play an anti-fibrosis role by inducing HSC apoptosis.

3.5. Autoimmune Liver Disease

Autoimmune liver diseases include autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC). Their common characteristics are chronic inflammation, immune system imbalance, and autoimmune response to liver or bile duct tissue [200]. The typical manifestations of AIH are periportal hepatitis, elevated serum autoantibodies, and hypergammaglobulinemia [201], while PBC is mainly characterized by the destruction of small intrahepatic bile ducts and the presence of anti-mitochondrial antibodies (AMA) [202]. PSC is characterized by chronic inflammation and fibrosis of bile ducts, which is often associated with inflammatory bowel disease (IBD) [203]. Although the pathological mechanisms of these diseases are different, the potential role of γδT cells in the pathogenesis of these diseases has gradually attracted attention.
Studies have indicated a notable rise in both the proportion and total count of γδT cells in the peripheral circulation, portal venous region, and areas of bile duct proliferation in individuals with autoimmune liver disease, particularly Vδ1+T cells [204,205]. The overactivation of Vδ1+T cells is considered to play an important role in liver injury and chronic inflammation in AIH and PBC diseases. Flow cytometry analysis showed that the proportion of Vδ1+T cells in patients with active PBC was significantly higher than that in the healthy control group and UDCA (ursodeoxycholic acid) treatment full responders, while the level of Vδ1+T cells in patients with insufficient responders continued to be high. These cells showed high levels of activation markers HLA-DR, CD69, and CD38 and produced a large number of IFN-γ and TNF-α, suggesting their potential cytotoxicity [205]. In liver tissue, the high expression of Vδ1+T cells is closely related to the injury of bile duct epithelial cells and the occurrence of chronic cholangitis. However, the infiltration level of these cells around the interlobular bile duct is low, suggesting that they play a role mainly by circulating into the liver. In addition, the increased release of TNF-α and IFN-γ is considered to be associated with the progression of fibrosis [206]. Thus, the ratio of circulating Vδ1+ T cells could serve as a potential marker for monitoring PBC disease activity.
Compared with Vδ1+T cells, the content of Vδ2+T cells in AIH patients is lower [207]. In addition, γδT cell-derived IL-17A is thought to aggravate hepatocyte injury in the AIH mouse model, and neutralization of the IL-17 pathway can reduce liver inflammation and fibrosis [208]. Vγ4+γδT cells showed a protective effect in the ConA-induced liver injury model [192]. IL-17A secreted by Vγ4+γδT cells could alleviate liver injury by inhibiting the pathogenicity of NKT cells [4]. However, in the biliary atresia (BA) model, IL-17 released by γδT17 cells is considered to promote the inflammatory response of bile duct and hepatocytes [209], suggesting that γδT17 cells have dual roles in different disease environments.
The immunoregulatory effect of γδT cells also shows that it can induce Treg cell apoptosis and weaken its function in some cases, thereby enhancing the activity of effector T cells and leading to immune imbalance. In addition, γδT cells regulate their activation level by expressing NKG2D receptor [76,210], which is similar to that of NK cells and NKT cells. The study also found that γδT cells can degrade ATP to produce adenosine through extracellular enzymes such as CD39 and CD73 and play an immunosuppressive role, thus inhibiting the hepatitis induced by autoreactive T cells [211]. Therefore, γδT cells can exhibit anti-inflammatory or pro-inflammatory functions under different environmental conditions. The diversity and complexity of their roles make them an important direction in the study of immune regulation of autoimmune liver disease.
In general, γδT cells exhibit considerable functional adaptability and diversity. During the progression of alcoholic liver disease (ALD), various subsets of γδT cells may exert both protective and pathogenic effects on liver tissue. Thus, targeting specific γδT cell subsets, especially through modulation of the IL-17 signaling pathway, presents a promising new strategy for treating autoimmune liver disorders. Although existing studies provide a significant understanding of the role of γδT cells in the development of autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC), further extensive and thorough investigations are required to completely clarify the mechanisms involved.

3.6. Liver Cancer

The main subsets of γδT cells in the liver are Vδ1+ and Vγ9Vδ2. Among them, Vδ1+ T cells play a dominant role in the local immune response of the liver, especially in the epithelium and bile duct [212]. In contrast, Vγ9Vδ2 T cells tend to be distributed in peripheral blood and secondary lymphoid tissues [213], and their number is less in healthy liver tissues, but their proportion and functional status will change significantly in liver diseases, such as hepatocellular carcinoma (HCC) and chronic hepatitis [212].
γδT cells show strong anti-tumor ability in the liver tumor immune environment, but its effect function is significantly regulated by the tumor microenvironment (TME) [214]. In a healthy state, γδT cells recognize non-MHC restricted antigens through TCR [215], such as phosphorylated metabolites (such as isopentenyl pyrophosphate, IPP) produced in the metabolic process of tumor cells, and activate rapidly to release cytotoxic molecules such as perforin and granzyme, which can directly kill tumor cells [61,216]. At the same time, these cells enhance the activation of antigen-presenting cells and CD8+T cells in the liver by secreting cytokines such as interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), thus forming a broader anti-tumor immune response [217,218]. Studies have demonstrated that γδT cells serve as key effector cells activated following immune checkpoint blockade (ICB) in cancers with HLA class I deficiencies. Their anti-tumor activity is partially mediated through the interaction between NKG2D and its corresponding ligands [219]. However, in the TME of patients with hepatocellular carcinoma, the number of γδT cells, especially Vγ9Vδ2 T cell subsets, decreased significantly, and their effector functions were depleted to varying degrees. High-throughput single-cell analysis revealed that these depleted γδT cells expressed high levels of immune checkpoint molecules, including PD-1, LAG-3 and Tim-3 [220]. In addition, the high expression of LAG-3 is highly correlated with the abnormal glutamine metabolic pathway [220], and the elevated glutamine level is considered to be an important factor in the inhibition of γδT cell function in hepatocellular carcinoma TME [221,222]. Experiments demonstrated that blocking glutamine metabolism could partially recover the functional activity of γδT cells, offering substantial support for the advancement in combination therapies targeting metabolic pathways [220].
Although γδT cells play an important role in the immune defense of liver cancer, they may also be reprogrammed into subsets with immunosuppressive function under certain conditions [214]. In the environment of some tumors and chronic liver diseases, a part of γδT cell subsets will be transformed into regulatory T cells (γδTreg), secreting immunosuppressive cytokines such as IL-10 and TGF-β, thereby inhibiting the activity of effector T cells and NK cells and promoting tumor immune escape [223]. This phenomenon indicates that the functional state of γδT cells is regulated by the cytokine network and metabolic environment in liver TME. In addition, some studies have pointed out that the migration and tissue orientation of liver γδT cells also affect their anti-tumor function [224].CD161 has been shown to promote the migration of Vγ9Vδ2 T cells across the endothelium [225]. Some γδT cells can enhance their homing ability to the liver and tumor sites through the high expression of chemokine receptors CCR5 and CXCR3 [226], but the abnormal expression of chemokines in the immunosuppressive environment may also lead to the reduction in γδT cells’ accumulation in liver tumor tissues [220].
In the study of liver immunotherapy, γδT cells are selectively depleted due to their non-specific recognition of tumor antigen and high killing activity, and cytotoxic Vδ2+γδT cells, while Treg-like Vδ1+γδT cells are upregulated in HCC. Thus, the infusion of allogeneic Vδ2+ γδT cells may offer a promising treatment strategy for this aggressive cancer [96,227,228]. The tumor-targeting properties and efficacy of Vγ9Vδ2 T cells can be markedly enhanced through in vitro expansion [212] or genetic modification of Vδ1+ T cells to express chimeric antigen receptors (CAR) [15]. CAR-γδT cell therapy demonstrated significant anti-tumor activity in a liver cancer mouse model. By co-expressing cytokines like IL-15, the survival and persistence of γδT cells in vivo were improved [15]. Additionally, combining γδT cells with PD-1 inhibitors yielded favorable preclinical outcomes, effectively reversing the immunosuppressive environment in the tumor microenvironment (TME) and boosting the anti-tumor activity of γδT cells [229]. However, in order to achieve the wide application of γδT cell immunotherapy in clinical practice, we still need to solve the problems of homing ability, metabolic adaptability, and long-term survival in vivo.
In conclusion, γδT cell subsets play an important role in immune surveillance and anti-tumor defense in the liver. Their functional status in the tumor microenvironment is affected by many factors, such as metabolic regulation, immune checkpoint expression, and cytokine network. Further research on these key regulatory mechanisms will help to provide more accurate targeting strategies for the immunotherapy of liver diseases and liver cancer and provide a theoretical basis for optimizing γδT cell adoptive cell therapy. To visually summarize the diverse roles and interactions of γδT cells in liver cancer and other liver diseases, these mechanisms are illustrated in Figure 2 below.

4. Crosstalk of γδT Cells with Immune and Non-Immune Cells in the Liver Microenvironment

γδT cells not only play a “bridge” role between innate and adaptive immunity through their unique TCR recognition and functional differentiation in liver immune regulation [5] but also form a complex crosstalk network with other key immune cell populations. Firstly, during the polarization and activation of liver-specific macrophages (Kupffer cells), γδT cells can promote or inhibit M1/m2 polarization by secreting cytokines such as IFN-γ, IL-17, TNF-α, or directly interacting with Kupffer cell surface ligands and costimulatory molecules [230]. Some studies have shown that when γδT cells tend to express IFN-γ and TNF-α, they can stimulate Kupffer cells to transform into M1 type, amplify the inflammatory response, and mediate some liver tissue damage [231]. However, in another state, some γδT cells (such as IL-10 secreting or immunomodulatory subsets) can guide Kupffer cells to M2 polarization and then play a protective role in the repair of chronic hepatitis or fibrosis [232]. This two-way regulation of “pro-inflammatory and anti-inflammatory” is closely related to the heterogeneity and activation threshold of γδT cells and also reflects the importance of equilibrium in the liver immune microenvironment.
At the same time, γδT cells can also interact with dendritic cells (DCS) and natural killer cells (NK cells) synergistically or antagonistically. DCs are responsible for antigen uptake and processing in the early stage of liver immune response [233]. γδT cells can not only transmit pre-excitation signals to DCS, thereby enhancing the presentation efficiency of the latter for intrahepatic pathogens or tumor-related antigens but also enable antigen-like presentation function (γδTapc) under certain conditions [234]. In addition, γδT cells secreting cytokines, such as IFN-γ or GM-CSF, often promote the maturation of DCS and CD4+/CD8+T cell response [150]. On the other hand, if γδT cells differentiate into regulatory or “depletion” phenotypes, they may reduce the activation intensity of DCS and inhibit excessive inflammation [110]. The collaboration between γδT cells and NK cells primarily governs target cell identification and cytotoxic responses. γδT cells express natural killer receptors (NKRs) such as NKG2D [235], which form signaling circuits with corresponding receptors on NK cells, allowing mutual activation through soluble factors (e.g., IL-2, IL-15, IFN-γ) or costimulatory molecules, thereby enhancing cytotoxicity in a coordinated manner [236,237]. Additionally, γδT cells promote the antifibrotic capacity of conventional NK (cNK) cells and liver-resident NK (lrNK) cells by enhancing their cytotoxicity against activated hepatic stellate cells (HSCs) [199]. This crosstalk between γδT and NK cells is partially dependent on the engagement of the costimulatory receptor 4-1BB (CD137) [133,238]. To visually illustrate the intricate interactions and regulatory effects of γδT cells with Kupffer cells, DCs, and NK cells in the liver microenvironment, these relationships are depicted in Figure 3 below.
Beyond immune interactions, γδT cells engage dynamically with non-immune cells, including hepatocytes, hepatic stellate cells (HSCs), and liver sinusoidal endothelial cells (LSECs), to maintain hepatic homeostasis and influence disease progression [199]. Hepatocytes express stress-induced ligands like MHC class I-related chain A/B (MICA/B) under damage, infection, or oncogenic stress [159], which are recognized by γδT cell NKG2D receptors, triggering cytotoxic responses via perforin and granzyme, and pro-inflammatory cytokine release (e.g., IFN-γ, TNF-α). While this clears damaged cells, excessive activation may exacerbate tissue injury [3]. HSCs, key drivers of fibrogenesis, upregulate ligands such as butyrophilin-like molecules upon activation, engaging γδ TCRs and recruiting γδT cells [199]. In response, γδT cells secrete IL-17 and IFN-γ, which can promote HSC activation and fibrosis or exert immunomodulatory effects to aid resolution, depending on the context [207,230]. LSECs regulate immune trafficking and sinusoidal homeostasis via adhesion molecules (e.g., ICAM-1) and CD1d-mediated lipid antigen presentation [182], inducing γδT cell IL-17 production to amplify immunity or IL-22 to support hepatocyte repair [9]. Collectively, these interactions position γδT cells as pivotal mediators of the liver’s immunological and structural balance, with dual protective and pathogenic roles in conditions like steatohepatitis, fibrosis, and hepatocellular carcinoma [239].

5. Clinical Applications, Therapeutic Potential, and Future Directions

In the field of tumor immunotherapy, γδT cells, particularly the Vδ1+ and Vδ2+ subpopulations, have demonstrated significant therapeutic potential. Although these two subsets share certain chemokine receptors on their surface, such as CCR5 and CXCR1, they exhibit distinct chemokine response patterns and migration behaviors within the tumor microenvironment (TME). For instance, Vδ2+ T cells primarily migrate through CCR5-mediated chemotaxis, whereas Vδ1+ T cells predominantly rely on CXCR1 and CXCR3, leading to differentiated tissue migration and tumor infiltration patterns [240,241]. Understanding these distinct chemotactic behaviors across various tumor types is crucial for optimizing γδT cell-based immunotherapies. Furthermore, immunosuppressive factors in the TME may reprogram the chemotactic pathways of these cells, redirecting them toward a pro-tumoral migration pattern. Thus, developing strategies to enhance γδT cell accumulation and persistence within tumors remains a critical challenge in improving the efficacy of γδT cell-based immunotherapy.
For allogeneic Vδ1+ or Vδ2+ T cell immunotherapy, a major challenge lies in extending the persistence of these cells in vivo while maintaining their functional potency. Clinical studies have shown that Vδ2+ T cell counts decline sharply within approximately two weeks post-infusion, indicating rapid apoptosis and functional exhaustion within the TME. To sustain the anti-tumor immune response, researchers recommend periodic adoptive transfer therapy (ACT), particularly with multiple infusions before complete tumor remission [39]. Moreover, due to their potent cytotoxicity and antigen-presenting capabilities, Vδ2+ T cells may play a pivotal role in post-surgical immune reconstitution in cancer patients.
To overcome the issue of functional exhaustion in the TME, engineered γδT cells have emerged as a promising solution. Through genetic modifications and cellular engineering, researchers can enhance the anti-tumor potency of these cells and extend their persistence within the TME. Among these approaches, chimeric antigen receptor (CAR)-γδT cells enable specific tumor targeting, while genetic modifications to increase resistance to chemotherapy drugs further enhance the efficacy of γδT cells in combination therapies [242,243,244,245]. However, similar to conventional CAR T-cell therapies, CAR-γδT cell approaches face challenges such as cytokine release syndrome (CRS), a potentially severe side effect caused by the massive release of cytokines (e.g., IL-6, TNF-α) from activated T cells or responding immune cells into the bloodstream [246,247]. This systemic inflammation requires careful monitoring and management strategies to ensure the safety and efficacy of these therapies in clinical settings. Additionally, the application of bispecific antibodies can facilitate direct and efficient interactions between γδT cells and tumor cells, significantly improving their anti-tumor functionality [248,249,250,251,252,253]. These advancements not only bolster current immunotherapies but also open new avenues for treating infectious diseases and autoimmune disorders.
Significant progress has also been made in integrating allogeneic γδT cells with conventional cancer treatments [141,254,255,256]. Studies indicate that combining γδT cells with chemotherapy, immune checkpoint inhibitors (such as anti-LAG3, PD1, or PDL1 antibodies), radiotherapy, or interventional therapy can effectively mitigate TME-induced immunosuppression, thereby enhancing anti-tumor activity. Notably, anti-LAG3 monoclonal antibody (mAb) therapy combined with γδT cell-based immunotherapy has been demonstrated to substantially augment immune responses [34]. However, under TME-induced stress, γδT cells may experience impaired chemotactic ability, preventing their effective migration to tumor sites [34]. In response, the development of novel bispecific antibodies presents a promising approach to improve γδT cell migration and tumor localization, thereby further enhancing therapeutic efficacy [252]. Additionally, localized administration of anti-BTN3A mAb [257] or zoledronic acid [258] within tumors may further expand the therapeutic potential of γδT cells. However, while such strategies enhance γδT cell efficacy, integrating them with immune checkpoint inhibitors like anti-PD-1 requires careful consideration of potential risks, particularly in specific HCC patient subsets. Recent research indicates that anti-PD-1 therapy may increase tumor growth in non-alcoholic steatohepatitis (NASH)-associated hepatocellular carcinoma (HCC) rather than reducing tumor burden [259]. In a mouse model, Pfister et al. (2021) found that anti-PD-1 treatment significantly promoted tumor growth in NASH-HCC, suggesting that in patients with underlying NASH, PD-1 inhibitors may pose a risk of disease progression. This highlights the importance of cautious patient selection and monitoring in this population. While our study focuses on γδT cell therapies, future investigations should explore whether these therapies carry similar risks or offer a safer alternative for patients with NASH-associated HCC.
The study of γδT cells has not advanced as rapidly as that of αβT cells, primarily because of the absence of specialized knockout mouse models. The difficulties stem from the complexity of γδT cell development, the limited understanding of its mechanism, and the influence of multiple genetic and signaling pathways on thymus development. In addition, because γδT cells lack clear markers and key transcription factors [143,144,260], the existing gene-editing technology makes it difficult to accurately knock out specific subsets. Compared with αβT cells, the number of γδT cells in the thymus is less, which makes the targeted research more difficult. At the same time, the complex interaction between γδT cells and thymus microenvironment may affect other T cell populations, which further increases the challenge of experimental design.
Despite many difficulties, researchers are actively promoting the establishment of γδT cell targeted gene knockout mouse model [261]. At the same time, the continuous optimization of the humanized mouse model also provides new possibilities for γδT cell research. For example, the use of human TCR transgenic mice combined with the transgenic expression of human BTN molecules is expected to further reveal the biological characteristics of γδT cells and lay the foundation for its application in immunological research.To summarize the current landscape of γδT cell-based cancer therapies and ongoing clinical trials, key findings and approaches are outlined in Table 2 below.

6. Conclusions

The heterogeneity and functional roles of γδT cells within the hepatic immune microenvironment are essential for immune surveillance, tumor immunity, and the regulation of liver diseases, including hepatitis, liver fibrosis, and hepatocellular carcinoma. As research advances, a deeper understanding of γδT cell subsets, their signaling pathways, and their interactions with other immune cells has elucidated their complex immunoregulatory functions across various pathological conditions. Despite significant progress in harnessing γδT cells for immunotherapy, several challenges persist, including immune evasion within the tumor microenvironment, cellular dysfunction, and limited migratory capacity. Future research efforts will be directed toward overcoming these limitations, particularly through engineering strategies designed to enhance anti-tumor efficacy, improve persistence, and optimize tumor targeting. These advancements hold great promise for developing more precise and effective therapies for liver diseases and cancer immunotherapy.

Funding

Supported by Guangxi Science and Technology Major Program (No. AA24263028) and the Scientific and Technological Innovation Major Base of Guangxi (No. 2022-36-Z05).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gao, B.; Jeong, W.-I.; Tian, Z. Liver: An Organ with Predominant Innate Immunity. Hepatology 2008, 47, 729–736. [Google Scholar] [CrossRef] [PubMed]
  2. Vantourout, P.; Hayday, A. Six-of-the-Best: Unique Contributions of Γδ T Cells to Immunology. Nat. Rev. Immunol. 2013, 13, 88–100. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, X.; Tian, Z. Γδ T Cells in Liver Diseases. Front. Med. 2018, 12, 262–268. [Google Scholar] [CrossRef]
  4. Hammerich, L.; Tacke, F. Role of Gamma-Delta T Cells in Liver Inflammation and Fibrosis. World J. Gastrointest. Pathophysiol. 2014, 5, 107–113. [Google Scholar] [CrossRef]
  5. Ferreira, L.M.R. Gammadelta T Cells: Innately Adaptive Immune Cells? Int. Rev. Immunol. 2013, 32, 223–248. [Google Scholar] [CrossRef] [PubMed]
  6. Zheng, J.; Liu, Y.; Lau, Y.-L.; Tu, W. Γδ-T Cells: An Unpolished Sword in Human Anti-Infection Immunity. Cell Mol. Immunol. 2013, 10, 50–57. [Google Scholar] [CrossRef]
  7. Munoz, L.D.; Sweeney, M.J.; Jameson, J.M. Skin Resident Γδ T Cell Function and Regulation in Wound Repair. Int. J. Mol. Sci. 2020, 21, 9286. [Google Scholar] [CrossRef]
  8. Mensurado, S.; Blanco-Domínguez, R.; Silva-Santos, B. The Emerging Roles of Γδ T Cells in Cancer Immunotherapy. Nat. Rev. Clin. Oncol. 2023, 20, 178–191. [Google Scholar] [CrossRef]
  9. Li, Y.; Huang, Z.; Yan, R.; Liu, M.; Bai, Y.; Liang, G.; Zhang, X.; Hu, X.; Chen, J.; Huang, C.; et al. Vγ4 Γδ T Cells Provide an Early Source of IL-17A and Accelerate Skin Graft Rejection. J. Investig. Dermatol. 2017, 137, 2513–2522. [Google Scholar] [CrossRef]
  10. Sanz, M.; Mann, B.T.; Ryan, P.L.; Bosque, A.; Pennington, D.J.; Hackstein, H.; Soriano-Sarabia, N. Deep Characterization of Human Γδ T Cell Subsets Defines Shared and Lineage-Specific Traits. Front. Immunol. 2023, 14, 1148988. [Google Scholar] [CrossRef]
  11. Wang, X.; Wang, H.; Lu, Z.; Liu, X.; Chai, W.; Wang, W.; Feng, J.; Yang, S.; Yang, W.; Cheng, H.; et al. Spatial and Single-Cell Analyses Reveal Heterogeneity of DNAM-1 Receptor–Ligand Interactions That Instructs Intratumoral γδT-Cell Activity. Cancer Res. 2025, 85, 277–298. [Google Scholar] [CrossRef] [PubMed]
  12. Fichtner, A.S.; Ravens, S.; Prinz, I. Human Γδ TCR Repertoires in Health and Disease. Cells 2020, 9, 800. [Google Scholar] [CrossRef]
  13. McMurray, J.L.; Von Borstel, A.; Taher, T.E.; Syrimi, E.; Taylor, G.S.; Sharif, M.; Rossjohn, J.; Remmerswaal, E.B.M.; Bemelman, F.J.; Vieira Braga, F.A.; et al. Transcriptional Profiling of Human Vδ1 T Cells Reveals a Pathogen-Driven Adaptive Differentiation Program. Cell Rep. 2022, 39, 110858. [Google Scholar] [CrossRef] [PubMed]
  14. Kabelitz, D.; Kalyan, S.; Oberg, H.-H.; Wesch, D. Human Vδ2 versus Non-Vδ2 Γδ T Cells in Antitumor Immunity. Oncoimmunology 2013, 2, e23304. [Google Scholar] [CrossRef]
  15. Makkouk, A.; Yang, X.; Barca, T.; Lucas, A.; Turkoz, M.; Wong, J.T.S.; Nishimoto, K.P.; Brodey, M.M.; Tabrizizad, M.; Gundurao, S.R.Y.; et al. Off-the-Shelf Vδ1 Gamma Delta T Cells Engineered with Glypican-3 (GPC-3)-Specific Chimeric Antigen Receptor (CAR) and Soluble IL-15 Display Robust Antitumor Efficacy against Hepatocellular Carcinoma. J. Immunother. Cancer 2021, 9, e003441. [Google Scholar] [CrossRef] [PubMed]
  16. Chang, K.-M.; Traum, D.; Park, J.-J.; Ho, S.; Ojiro, K.; Wong, D.K.; Wahed, A.S.; Terrault, N.A.; Khalili, M.; Sterling, R.K.; et al. Distinct Phenotype and Function of Circulating Vδ1+ and Vδ2+ γδT-Cells in Acute and Chronic Hepatitis B. PLoS Pathog. 2019, 15, e1007715. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, Y.; Tian, Z. Innate Lymphocytes: Pathogenesis and Therapeutic Targets of Liver Diseases and Cancer. Cell Mol. Immunol. 2021, 18, 57–72. [Google Scholar] [CrossRef]
  18. Muro, R.; Takayanagi, H.; Nitta, T. T Cell Receptor Signaling for γδT Cell Development. Inflamm. Regen. 2019, 39, 6. [Google Scholar] [CrossRef]
  19. Kisielow, J.; Kopf, M. The Origin and Fate of γδT Cell Subsets. Curr. Opin. Immunol. 2013, 25, 181–188. [Google Scholar] [CrossRef]
  20. Muñoz-Ruiz, M.; Sumaria, N.; Pennington, D.J.; Silva-Santos, B. Thymic Determinants of Γδ T Cell Differentiation. Trends Immunol. 2017, 38, 336–344. [Google Scholar] [CrossRef]
  21. Deng, J.; Yin, H. Gamma Delta (Γδ) T Cells in Cancer Immunotherapy; Where It Comes from, Where It Will Go? Eur. J. Pharmacol. 2022, 919, 174803. [Google Scholar] [CrossRef] [PubMed]
  22. Chien, Y.; Konigshofer, Y. Antigen Recognition by Gammadelta T Cells. Immunol. Rev. 2007, 215, 46–58. [Google Scholar] [CrossRef] [PubMed]
  23. Notarangelo, L.D.; Kim, M.-S.; Walter, J.E.; Lee, Y.N. Human RAG Mutations: Biochemistry and Clinical Implications. Nat. Rev. Immunol. 2016, 16, 234–246. [Google Scholar] [CrossRef] [PubMed]
  24. Hata, S.; Satyanarayana, K.; Devlin, P.; Band, H.; McLean, J.; Strominger, J.L.; Brenner, M.B.; Krangel, M.S. Extensive Junctional Diversity of Rearranged Human T Cell Receptor Delta Genes. Science 1988, 240, 1541–1544. [Google Scholar] [CrossRef]
  25. Adams, E.J.; Gu, S.; Luoma, A.M. Human Gamma Delta T Cells: Evolution and Ligand Recognition. Cell. Immunol. 2015, 296, 31–40. [Google Scholar] [CrossRef]
  26. Willcox, C.R.; Davey, M.S.; Willcox, B.E. Development and Selection of the Human Vγ9Vδ2+ T-Cell Repertoire. Front. Immunol. 2018, 9, 1501. [Google Scholar] [CrossRef]
  27. Krangel, M.S.; Yssel, H.; Brocklehurst, C.; Spits, H. A Distinct Wave of Human T Cell Receptor Gamma/Delta Lymphocytes in the Early Fetal Thymus: Evidence for Controlled Gene Rearrangement and Cytokine Production. J. Exp. Med. 1990, 172, 847–859. [Google Scholar] [CrossRef]
  28. Ravens, S.; Schultze-Florey, C.; Raha, S.; Sandrock, I.; Drenker, M.; Oberdörfer, L.; Reinhardt, A.; Ravens, I.; Beck, M.; Geffers, R.; et al. Human Γδ T Cells Are Quickly Reconstituted after Stem-Cell Transplantation and Show Adaptive Clonal Expansion in Response to Viral Infection. Nat. Immunol. 2017, 18, 393–401. [Google Scholar] [CrossRef]
  29. Di Lorenzo, B.; Ravens, S.; Silva-Santos, B. High-Throughput Analysis of the Human Thymic Vδ1+ T Cell Receptor Repertoire. Sci. Data 2019, 6, 115. [Google Scholar] [CrossRef]
  30. Davey, M.S.; Willcox, C.R.; Joyce, S.P.; Ladell, K.; Kasatskaya, S.A.; McLaren, J.E.; Hunter, S.; Salim, M.; Mohammed, F.; Price, D.A.; et al. Clonal Selection in the Human Vδ1 T Cell Repertoire Indicates Γδ TCR-Dependent Adaptive Immune Surveillance. Nat. Commun. 2017, 8, 14760. [Google Scholar] [CrossRef]
  31. Hunter, S.; Willcox, C.R.; Davey, M.S.; Kasatskaya, S.A.; Jeffery, H.C.; Chudakov, D.M.; Oo, Y.H.; Willcox, B.E. Human Liver Infiltrating Γδ T Cells Are Composed of Clonally Expanded Circulating and Tissue-Resident Populations. J. Hepatol. 2018, 69, 654–665. [Google Scholar] [CrossRef] [PubMed]
  32. Chen, D.; Guo, Y.; Jiang, J.; Wu, P.; Zhang, T.; Wei, Q.; Huang, J.; Wu, D. Γδ T Cell Exhaustion: Opportunities for Intervention. J. Leukoc. Biol. 2022, 112, 1669–1676. [Google Scholar] [CrossRef] [PubMed]
  33. Correia, D.V.; Fogli, M.; Hudspeth, K.; da Silva, M.G.; Mavilio, D.; Silva-Santos, B. Differentiation of Human Peripheral Blood Vδ1+ T Cells Expressing the Natural Cytotoxicity Receptor NKp30 for Recognition of Lymphoid Leukemia Cells. Blood 2011, 118, 992–1001. [Google Scholar] [CrossRef]
  34. Carbone, A.; Vaccher, E.; Gloghini, A. Hematologic Cancers in Individuals Infected by HIV. Blood 2022, 139, 995–1012. [Google Scholar] [CrossRef] [PubMed]
  35. Li, H.; Pauza, C.D. HIV Envelope-Mediated, CCR5/A4β7-Dependent Killing of CD4-Negative Γδ T Cells Which Are Lost during Progression to AIDS. Blood 2011, 118, 5824–5831. [Google Scholar] [CrossRef]
  36. Gioia, C.; Agrati, C.; Casetti, R.; Cairo, C.; Borsellino, G.; Battistini, L.; Mancino, G.; Goletti, D.; Colizzi, V.; Pucillo, L.P.; 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. [Google Scholar] [CrossRef]
  37. Ogongo, P.; Steyn, A.J.; Karim, F.; Dullabh, K.J.; Awala, I.; Madansein, R.; Leslie, A.; Behar, S.M. Differential Skewing of Donor-Unrestricted and Γδ T Cell Repertoires in Tuberculosis-Infected Human Lungs. J. Clin. Investig. 2020, 130, 214–230. [Google Scholar] [CrossRef]
  38. Poggi, A.; Carosio, R.; Fenoglio, D.; Brenci, S.; Murdaca, G.; Setti, M.; Indiveri, F.; Scabini, S.; Ferrero, E.; Zocchi, M.R. Migration of V Delta 1 and V Delta 2 T Cells in Response to CXCR3 and CXCR4 Ligands in Healthy Donors and HIV-1-Infected Patients: Competition by HIV-1 Tat. Blood 2004, 103, 2205–2213. [Google Scholar] [CrossRef]
  39. Hu, Y.; Hu, Q.; Li, Y.; Lu, L.; Xiang, Z.; Yin, Z.; Kabelitz, D.; Wu, Y. Γδ T Cells: Origin and Fate, Subsets, Diseases and Immunotherapy. Signal Transduct. Target. Ther. 2023, 8, 434. [Google Scholar] [CrossRef]
  40. Silva-Santos, B.; Serre, K.; Norell, H. Γδ T Cells in Cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef]
  41. Dimova, T.; Brouwer, M.; Gosselin, F.; Tassignon, J.; Leo, O.; Donner, C.; Marchant, A.; Vermijlen, D. Effector Vγ9Vδ2 T Cells Dominate the Human Fetal Γδ T-Cell Repertoire. Proc. Natl. Acad. Sci. USA 2015, 112, E556–E565. [Google Scholar] [CrossRef] [PubMed]
  42. Dunne, M.R.; Elliott, L.; Hussey, S.; Mahmud, N.; Kelly, J.; Doherty, D.G.; Feighery, C.F. Persistent Changes in Circulating and Intestinal Γδ T Cell Subsets, Invariant Natural Killer T Cells and Mucosal-Associated Invariant T Cells in Children and Adults with Coeliac Disease. PLoS ONE 2013, 8, e76008. [Google Scholar] [CrossRef]
  43. Kenna, T.; Golden-Mason, L.; Norris, S.; Hegarty, J.E.; O’Farrelly, C.; Doherty, D.G. Distinct Subpopulations of Γδ T Cells Are Present in Normal and Tumor-Bearing Human Liver. Clin. Immunol. 2004, 113, 56–63. [Google Scholar] [CrossRef]
  44. Rice, M.T.; Von Borstel, A.; Chevour, P.; Awad, W.; Howson, L.J.; Littler, D.R.; Gherardin, N.A.; Le Nours, J.; Giles, E.M.; Berry, R.; et al. Recognition of the Antigen-Presenting Molecule MR1 by a Vδ3+ Γδ T Cell Receptor. Proc. Natl. Acad. Sci. USA 2021, 118, e2110288118. [Google Scholar] [CrossRef] [PubMed]
  45. León-Lara, X.; Yang, T.; Fichtner, A.S.; Bruni, E.; Von Kaisenberg, C.; Eiz-Vesper, B.; Dodoo, D.; Adu, B.; Ravens, S. Evidence for an Adult-Like Type 1-Immunity Phenotype of Vδ1, Vδ2 and Vδ3 T Cells in Ghanaian Children With Repeated Exposure to Malaria. Front. Immunol. 2022, 13, 807765. [Google Scholar] [CrossRef]
  46. Ravens, S.; Hengst, J.; Schlapphoff, V.; Deterding, K.; Dhingra, A.; Schultze-Florey, C.; Koenecke, C.; Cornberg, M.; Wedemeyer, H.; Prinz, I. Human Γδ T Cell Receptor Repertoires in Peripheral Blood Remain Stable Despite Clearance of Persistent Hepatitis C Virus Infection by Direct-Acting Antiviral Drug Therapy. Front. Immunol. 2018, 9, 510. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, L.; Xu, M.; Wang, C.; Zhu, L.; Hu, J.; Chen, S.; Wu, X.; Li, B.; Li, Y. The Feature of Distribution and Clonality of TCR γ/δ Subfamilies T Cells in Patients with B-Cell Non-Hodgkin Lymphoma. J. Immunol. Res. 2014, 2014, 241246. [Google Scholar] [CrossRef] [PubMed]
  48. de Vries, N.L.; van de Haar, J.; Veninga, V.; Chalabi, M.; Ijsselsteijn, M.E.; van der Ploeg, M.; van den Bulk, J.; Ruano, D.; van den Berg, J.G.; Haanen, J.B.; et al. Γδ T Cells Are Effectors of Immunotherapy in Cancers with HLA Class I Defects. Nature 2023, 613, 743–750. [Google Scholar] [CrossRef] [PubMed]
  49. Melo, A.M.; Mylod, E.; Fitzgerald, V.; Donlon, N.E.; Murphy, D.M.; Foley, E.K.; Bhardwaj, A.; Reynolds, J.V.; Doherty, D.G.; Lysaght, J.; et al. Tissue Distribution of Γδ T Cell Subsets in Oesophageal Adenocarcinoma. Clin. Immunol. 2021, 229, 108797. [Google Scholar] [CrossRef]
  50. Gherardin, N.A.; Waldeck, K.; Caneborg, A.; Martelotto, L.G.; Balachander, S.; Zethoven, M.; Petrone, P.M.; Pattison, A.; Wilmott, J.S.; Quiñones-Parra, S.M.; et al. Γδ T Cells in Merkel Cell Carcinomas Have a Proinflammatory Profile Prognostic of Patient Survival. Cancer Immunol. Res. 2021, 9, 612–623. [Google Scholar] [CrossRef]
  51. Petrasca, A.; Melo, A.M.; Breen, E.P.; Doherty, D.G. Human Vδ3+ Γδ T Cells Induce Maturation and IgM Secretion by B Cells. Immunol. Lett. 2018, 196, 126–134. [Google Scholar] [CrossRef] [PubMed]
  52. Spada, F.M.; Grant, E.P.; Peters, P.J.; Sugita, M.; Melián, A.; Leslie, D.S.; Lee, H.K.; van Donselaar, E.; Hanson, D.A.; Krensky, A.M.; et al. Self-Recognition of CD1 by Gamma/Delta T Cells: Implications for Innate Immunity. J. Exp. Med. 2000, 191, 937–948. [Google Scholar] [CrossRef] [PubMed]
  53. Uldrich, A.P.; Le Nours, J.; Pellicci, D.G.; Gherardin, N.A.; McPherson, K.G.; Lim, R.T.; Patel, O.; Beddoe, T.; Gras, S.; Rossjohn, J.; et al. CD1d-Lipid Antigen Recognition by the Γδ TCR. Nat. Immunol. 2013, 14, 1137–1145. [Google Scholar] [CrossRef]
  54. Luoma, A.M.; Castro, C.D.; Mayassi, T.; Bembinster, L.A.; Bai, L.; Picard, D.; Anderson, B.; Scharf, L.; Kung, J.E.; Sibener, L.V.; et al. Crystal Structure of Vδ1 T Cell Receptor in Complex with CD1d-Sulfatide Shows MHC-like Recognition of a Self-Lipid by Human Γδ T Cells. Immunity 2013, 39, 1032–1042. [Google Scholar] [CrossRef]
  55. Luoma, A.M.; Castro, C.D.; Adams, E.J. Γδ T Cell Surveillance via CD1 Molecules. Trends Immunol. 2014, 35, 613–621. [Google Scholar] [CrossRef]
  56. Roy, S.; Ly, D.; Castro, C.D.; Li, N.-S.; Hawk, A.J.; Altman, J.D.; Meredith, S.C.; Piccirilli, J.A.; Moody, D.B.; Adams, E.J. Molecular Analysis of Lipid-Reactive Vδ1 Γδ T Cells Identified by CD1c Tetramers. J. Immunol. 2016, 196, 1933–1942. [Google Scholar] [CrossRef] [PubMed]
  57. Hayday, A.; Vantourout, P. A Long-Playing CD about the Γδ TCR Repertoire. Immunity 2013, 39, 994–996. [Google Scholar] [CrossRef]
  58. Sensing of Cell Stress by Human Γδ TCR-Dependent Recognition of Annexin A2—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/28270598/ (accessed on 22 January 2025).
  59. Xu, B.; Pizarro, J.C.; Holmes, M.A.; McBeth, C.; Groh, V.; Spies, T.; Strong, R.K. Crystal Structure of a Gammadelta T-Cell Receptor Specific for the Human MHC Class I Homolog MICA. Proc. Natl. Acad. Sci. USA 2011, 108, 2414–2419. [Google Scholar] [CrossRef]
  60. Silva-Santos, B.; Mensurado, S.; Coffelt, S.B. Γδ T Cells: Pleiotropic Immune Effectors with Therapeutic Potential in Cancer. Nat. Rev. Cancer 2019, 19, 392–404. [Google Scholar] [CrossRef]
  61. Gober, H.-J.; Kistowska, M.; Angman, L.; Jenö, P.; Mori, L.; De Libero, G. Human T Cell Receptor Gammadelta Cells Recognize Endogenous Mevalonate Metabolites in Tumor Cells. J. Exp. Med. 2003, 197, 163–168. [Google Scholar] [CrossRef]
  62. Benzaïd, I.; Mönkkönen, H.; Stresing, V.; Bonnelye, E.; Green, J.; Mönkkönen, J.; Touraine, J.-L.; Clézardin, P. High Phosphoantigen Levels in Bisphosphonate-Treated Human Breast Tumors Promote Vgamma9Vdelta2 T-Cell Chemotaxis and Cytotoxicity in Vivo. Cancer Res. 2011, 71, 4562–4572. [Google Scholar] [CrossRef] [PubMed]
  63. Ashihara, E.; Munaka, T.; Kimura, S.; Nakagawa, S.; Nakagawa, Y.; Kanai, M.; Hirai, H.; Abe, H.; Miida, T.; Yamato, S.; et al. Isopentenyl Pyrophosphate Secreted from Zoledronate-Stimulated Myeloma Cells, Activates the Chemotaxis of γδT Cells. Biochem. Biophys. Res. Commun. 2015, 463, 650–655. [Google Scholar] [CrossRef]
  64. Tanaka, Y.; Morita, C.T.; Tanaka, Y.; Nieves, E.; Brenner, M.B.; Bloom, B.R. Natural and Synthetic Non-Peptide Antigens Recognized by Human Gamma Delta T Cells. Nature 1995, 375, 155–158. [Google Scholar] [CrossRef] [PubMed]
  65. Harly, C.; Guillaume, Y.; Nedellec, S.; Peigné, C.-M.; Mönkkönen, H.; Mönkkönen, J.; Li, J.; Kuball, J.; Adams, E.J.; Netzer, S.; et al. Key Implication of CD277/Butyrophilin-3 (BTN3A) in Cellular Stress Sensing by a Major Human Γδ T-Cell Subset. Blood 2012, 120, 2269–2279. [Google Scholar] [CrossRef]
  66. Sandstrom, A.; Peigné, C.-M.; Léger, A.; Crooks, J.E.; Konczak, F.; Gesnel, M.-C.; Breathnach, R.; Bonneville, M.; Scotet, E.; Adams, E.J. The Intracellular B30.2 Domain of Butyrophilin 3A1 Binds Phosphoantigens to Mediate Activation of Human Vγ9Vδ2 T Cells. Immunity 2014, 40, 490–500. [Google Scholar] [CrossRef] [PubMed]
  67. Rigau, M.; Ostrouska, S.; Fulford, T.S.; Johnson, D.N.; Woods, K.; Ruan, Z.; McWilliam, H.E.G.; Hudson, C.; Tutuka, C.; Wheatley, A.K.; et al. Butyrophilin 2A1 Is Essential for Phosphoantigen Reactivity by Γδ T Cells. Science 2020, 367, eaay5516. [Google Scholar] [CrossRef]
  68. Karunakaran, M.M.; Willcox, C.R.; Salim, M.; Paletta, D.; Fichtner, A.S.; Noll, A.; Starick, L.; Nöhren, A.; Begley, C.R.; Berwick, K.A.; et al. Butyrophilin-2A1 Directly Binds Germline-Encoded Regions of the Vγ9Vδ2 TCR and Is Essential for Phosphoantigen Sensing. Immunity 2020, 52, 487–498.e6. [Google Scholar] [CrossRef]
  69. Kabelitz, D.; Serrano, R.; Kouakanou, L.; Peters, C.; Kalyan, S. Cancer Immunotherapy with Γδ T Cells: Many Paths Ahead of Us. Cell Mol. Immunol. 2020, 17, 925–939. [Google Scholar] [CrossRef]
  70. Yang, Y.; Li, L.; Yuan, L.; Zhou, X.; Duan, J.; Xiao, H.; Cai, N.; Han, S.; Ma, X.; Liu, W.; et al. A Structural Change in Butyrophilin upon Phosphoantigen Binding Underlies Phosphoantigen-Mediated Vγ9Vδ2 T Cell Activation. Immunity 2019, 50, 1043–1053.e5. [Google Scholar] [CrossRef]
  71. Scotet, E.; Martinez, L.O.; Grant, E.; Barbaras, R.; Jenö, P.; Guiraud, M.; Monsarrat, B.; Saulquin, X.; Maillet, S.; Estève, J.-P.; et al. Tumor Recognition Following Vgamma9Vdelta2 T Cell Receptor Interactions with a Surface F1-ATPase-Related Structure and Apolipoprotein A-I. Immunity 2005, 22, 71–80. [Google Scholar] [CrossRef]
  72. Chen, H.; He, X.; Wang, Z.; Wu, D.; Zhang, H.; Xu, C.; He, H.; Cui, L.; Ba, D.; He, W. Identification of Human T Cell Receptor Gammadelta-Recognized Epitopes/Proteins via CDR3delta Peptide-Based Immunobiochemical Strategy. J. Biol. Chem. 2008, 283, 12528–12537. [Google Scholar] [CrossRef] [PubMed]
  73. Dai, Y.; Chen, H.; Mo, C.; Cui, L.; He, W. Ectopically Expressed Human Tumor Biomarker MutS Homologue 2 Is a Novel Endogenous Ligand That Is Recognized by Human Γδ T Cells to Induce Innate Anti-Tumor/Virus Immunity. J. Biol. Chem. 2012, 287, 16812–16819. [Google Scholar] [CrossRef]
  74. Fichtner, A.S.; Karunakaran, M.M.; Gu, S.; Boughter, C.T.; Borowska, M.T.; Starick, L.; Nöhren, A.; Göbel, T.W.; Adams, E.J.; Herrmann, T. Alpaca (Vicugna Pacos), the First Nonprimate Species with a Phosphoantigen-Reactive Vγ9Vδ2 T Cell Subset. Proc. Natl. Acad. Sci. USA 2020, 117, 6697–6707. [Google Scholar] [CrossRef]
  75. 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. [Google Scholar] [CrossRef] [PubMed]
  76. Wrobel, P.; Shojaei, H.; Schittek, B.; Gieseler, F.; Wollenberg, B.; Kalthoff, H.; Kabelitz, D.; Wesch, D. Lysis of a Broad Range of Epithelial Tumour Cells by Human Gamma Delta T Cells: Involvement of NKG2D Ligands and T-Cell Receptor- versus NKG2D-Dependent Recognition. Scand. J. Immunol. 2007, 66, 320–328. [Google Scholar] [CrossRef]
  77. The MHC Class Ib Protein ULBP1 Is a Nonredundant Determinant of Leukemia/Lymphoma Susceptibility to Gammadelta T-Cell Cytotoxicity—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/20101024/ (accessed on 22 January 2025).
  78. Simões, A.E.; Di Lorenzo, B.; Silva-Santos, B. Molecular Determinants of Target Cell Recognition by Human Γδ T Cells. Front. Immunol. 2018, 9, 929. [Google Scholar] [CrossRef]
  79. Toutirais, O.; Cabillic, F.; Le Friec, G.; Salot, S.; Loyer, P.; Le Gallo, M.; Desille, M.; de La Pintière, C.T.; Daniel, P.; Bouet, F.; et al. DNAX Accessory Molecule-1 (CD226) Promotes Human Hepatocellular Carcinoma Cell Lysis by Vgamma9Vdelta2 T Cells. Eur. J. Immunol. 2009, 39, 1361–1368. [Google Scholar] [CrossRef] [PubMed]
  80. Tokuyama, H.; Hagi, T.; Mattarollo, S.R.; Morley, J.; Wang, Q.; So, H.-F.; Moriyasu, F.; Nieda, M.; Nicol, A.J. V Gamma 9 V Delta 2 T Cell Cytotoxicity against Tumor Cells Is Enhanced by Monoclonal Antibody Drugs--Rituximab and Trastuzumab. Int. J. Cancer 2008, 122, 2526–2534. [Google Scholar] [CrossRef]
  81. Capietto, A.-H.; Martinet, L.; Fournié, J.-J. Stimulated Γδ T Cells Increase the in Vivo Efficacy of Trastuzumab in HER-2+ Breast Cancer. J. Immunol. 2011, 187, 1031–1038. [Google Scholar] [CrossRef]
  82. Gertner-Dardenne, J.; Bonnafous, C.; Bezombes, C.; Capietto, A.-H.; Scaglione, V.; Ingoure, S.; Cendron, D.; Gross, E.; Lepage, J.-F.; Quillet-Mary, A.; et al. Bromohydrin Pyrophosphate Enhances Antibody-Dependent Cell-Mediated Cytotoxicity Induced by Therapeutic Antibodies. Blood 2009, 113, 4875–4884. [Google Scholar] [CrossRef]
  83. Fisher, J.P.H.; Yan, M.; Heuijerjans, J.; Carter, L.; Abolhassani, A.; Frosch, J.; Wallace, R.; Flutter, B.; Capsomidis, A.; Hubank, M.; et al. Neuroblastoma Killing Properties of Vδ2 and Vδ2-Negative γδT Cells Following Expansion by Artificial Antigen-Presenting Cells. Clin. Cancer Res. 2014, 20, 5720–5732. [Google Scholar] [CrossRef] [PubMed]
  84. Tuengel, J.; Ranchal, S.; Maslova, A.; Aulakh, G.; Papadopoulou, M.; Drissler, S.; Cai, B.; Mohsenzadeh-Green, C.; Soudeyns, H.; Mostafavi, S.; et al. Characterization of Adaptive-like Γδ T Cells in Ugandan Infants during Primary Cytomegalovirus Infection. Viruses 2021, 13, 1987. [Google Scholar] [CrossRef] [PubMed]
  85. Street, S.E.A.; Hayakawa, Y.; Zhan, Y.; Lew, A.M.; MacGregor, D.; Jamieson, A.M.; Diefenbach, A.; Yagita, H.; Godfrey, D.I.; Smyth, M.J. Innate Immune Surveillance of Spontaneous B Cell Lymphomas by Natural Killer Cells and Gammadelta T Cells. J. Exp. Med. 2004, 199, 879–884. [Google Scholar] [CrossRef]
  86. Liu, Z.; Eltoum, I.-E.A.; Guo, B.; Beck, B.H.; Cloud, G.A.; Lopez, R.D. Protective Immunosurveillance and Therapeutic Antitumor Activity of Gammadelta T Cells Demonstrated in a Mouse Model of Prostate Cancer. J. Immunol. 2008, 180, 6044–6053. [Google Scholar] [CrossRef]
  87. Gao, Y.; Yang, W.; Pan, M.; Scully, E.; Girardi, M.; Augenlicht, L.H.; Craft, J.; Yin, Z. Γδ T Cells Provide an Early Source of Interferon γ in Tumor Immunity. J. Exp. Med. 2003, 198, 433–442. [Google Scholar] [CrossRef]
  88. Jarry, U.; Chauvin, C.; Joalland, N.; Léger, A.; Minault, S.; Robard, M.; Bonneville, M.; Oliver, L.; Vallette, F.M.; Vié, H.; et al. Stereotaxic Administrations of Allogeneic Human Vγ9Vδ2 T Cells Efficiently Control the Development of Human Glioblastoma Brain Tumors. Oncoimmunology 2016, 5, e1168554. [Google Scholar] [CrossRef] [PubMed]
  89. Pereboeva, L.; Harkins, L.; Wong, S.; Lamb, L.S. The Safety of Allogeneic Innate Lymphocyte Therapy for Glioma Patients with Prior Cranial Irradiation. Cancer Immunol. Immunother. 2015, 64, 551–562. [Google Scholar] [CrossRef]
  90. Rei, M.; Gonçalves-Sousa, N.; Lança, T.; Thompson, R.G.; Mensurado, S.; Balkwill, F.R.; Kulbe, H.; Pennington, D.J.; Silva-Santos, B. Murine CD27(−) Vγ6(+) Γδ T Cells Producing IL-17A Promote Ovarian Cancer Growth via Mobilization of Protumor Small Peritoneal Macrophages. Proc. Natl. Acad. Sci. USA 2014, 111, E3562–E3570. [Google Scholar] [CrossRef]
  91. Ma, S.; Cheng, Q.; Cai, Y.; Gong, H.; Wu, Y.; Yu, X.; Shi, L.; Wu, D.; Dong, C.; Liu, H. IL-17A Produced by Γδ T Cells Promotes Tumor Growth in Hepatocellular Carcinoma. Cancer Res. 2014, 74, 1969–1982. [Google Scholar] [CrossRef]
  92. Patin, E.C.; Soulard, D.; Fleury, S.; Hassane, M.; Dombrowicz, D.; Faveeuw, C.; Trottein, F.; Paget, C. Type I IFN Receptor Signaling Controls IL7-Dependent Accumulation and Activity of Protumoral IL17A-Producing γδT Cells in Breast Cancer. Cancer Res. 2018, 78, 195–204. [Google Scholar] [CrossRef]
  93. Kimura, Y.; Nagai, N.; Tsunekawa, N.; Sato-Matsushita, M.; Yoshimoto, T.; Cua, D.J.; Iwakura, Y.; Yagita, H.; Okada, F.; Tahara, H.; et al. IL-17A-Producing CD30(+) Vδ1 T Cells Drive Inflammation-Induced Cancer Progression. Cancer Sci. 2016, 107, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  94. Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.-S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.J.A.C.; Jonkers, J.; et al. IL-17-Producing Γδ T Cells and Neutrophils Conspire to Promote Breast Cancer Metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef] [PubMed]
  95. Kulig, P.; Burkhard, S.; Mikita-Geoffroy, J.; Croxford, A.L.; Hövelmeyer, N.; Gyülvészi, G.; Gorzelanny, C.; Waisman, A.; Borsig, L.; Becher, B. IL17A-Mediated Endothelial Breach Promotes Metastasis Formation. Cancer Immunol. Res. 2016, 4, 26–32. [Google Scholar] [CrossRef]
  96. Xu, Y.; Xiang, Z.; Alnaggar, M.; Kouakanou, L.; Li, J.; He, J.; Yang, J.; Hu, Y.; Chen, Y.; Lin, L.; et al. Allogeneic Vγ9Vδ2 T-Cell Immunotherapy Exhibits Promising Clinical Safety and Prolongs the Survival of Patients with Late-Stage Lung or Liver Cancer. Cell Mol. Immunol. 2021, 18, 427–439. [Google Scholar] [CrossRef]
  97. Parker, M.E.; Ciofani, M. Regulation of Γδ T Cell Effector Diversification in the Thymus. Front. Immunol. 2020, 11, 42. [Google Scholar] [CrossRef] [PubMed]
  98. Pellicci, D.G.; Koay, H.-F.; Berzins, S.P. Thymic Development of Unconventional T Cells: How NKT Cells, MAIT Cells and Γδ T Cells Emerge. Nat. Rev. Immunol. 2020, 20, 756–770. [Google Scholar] [CrossRef] [PubMed]
  99. Buus, T.B.; Ødum, N.; Geisler, C.; Lauritsen, J.P.H. Three Distinct Developmental Pathways for Adaptive and Two IFN-γ-Producing Γδ T Subsets in Adult Thymus. Nat. Commun. 2017, 8, 1911. [Google Scholar] [CrossRef]
  100. Ribot, J.C.; deBarros, A.; Pang, D.J.; Neves, J.F.; Peperzak, V.; Roberts, S.J.; Girardi, M.; Borst, J.; Hayday, A.C.; Pennington, D.J.; 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. [Google Scholar] [CrossRef]
  101. Muñoz-Ruiz, M.; Ribot, J.C.; Grosso, A.R.; Gonçalves-Sousa, N.; Pamplona, A.; Pennington, D.J.; Regueiro, J.R.; Fernández-Malavé, E.; Silva-Santos, B. TCR Signal Strength Controls Thymic Differentiation of Discrete Proinflammatory Γδ T Cell Subsets. Nat. Immunol. 2016, 17, 721–727. [Google Scholar] [CrossRef]
  102. Jensen, K.D.C.; Su, X.; Shin, S.; Li, L.; Youssef, S.; Yamasaki, S.; Steinman, L.; Saito, T.; Locksley, R.M.; Davis, M.M.; et al. Thymic Selection Determines Gammadelta T Cell Effector Fate: Antigen-Naive Cells Make Interleukin-17 and Antigen-Experienced Cells Make Interferon Gamma. Immunity 2008, 29, 90–100. [Google Scholar] [CrossRef]
  103. Fleming, C.; Morrissey, S.; Cai, Y.; Yan, J. Γδ T Cells: Unexpected Regulators of Cancer Development and Progression. Trends Cancer 2017, 3, 561–570. [Google Scholar] [CrossRef] [PubMed]
  104. Lo Presti, E.; Dieli, F.; Meraviglia, S. Tumor-Infiltrating Γδ T Lymphocytes: Pathogenic Role, Clinical Significance, and Differential Programing in the Tumor Microenvironment. Front. Immunol. 2014, 5, 607. [Google Scholar] [CrossRef] [PubMed]
  105. Ye, J.; Ma, C.; Wang, F.; Hsueh, E.C.; Toth, K.; Huang, Y.; Mo, W.; Liu, S.; Han, B.; Varvares, M.A.; et al. Specific Recruitment of Γδ Regulatory T Cells in Human Breast Cancer. Cancer Res. 2013, 73, 6137–6148. [Google Scholar] [CrossRef]
  106. Xuan, L.; Wu, X.; Qiu, D.; Gao, L.; Liu, H.; Fan, Z.; Huang, F.; Jin, Z.; Sun, J.; Li, Y.; et al. Regulatory Γδ T Cells Induced by G-CSF Participate in Acute Graft-versus-Host Disease Regulation in G-CSF-Mobilized Allogeneic Peripheral Blood Stem Cell Transplantation. J. Transl. Med. 2018, 16, 144. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, X.; Zhan, N.; Jin, Y.; Ling, H.; Xiao, C.; Xie, Z.; Zhong, H.; Yu, X.; Tang, R.; Ma, J.; et al. Tofacitinib Restores the Balance of γδTreg/γδT17 Cells in Rheumatoid Arthritis by Inhibiting the NLRP3 Inflammasome. Theranostics 2021, 11, 1446–1457. [Google Scholar] [CrossRef] [PubMed]
  108. Mao, Y.; Yin, S.; Zhang, J.; Hu, Y.; Huang, B.; Cui, L.; Kang, N.; He, W. A New Effect of IL-4 on Human Γδ T Cells: Promoting Regulatory Vδ1 T Cells via IL-10 Production and Inhibiting Function of Vδ2 T Cells. Cell Mol. Immunol. 2016, 13, 217–228. [Google Scholar] [CrossRef]
  109. Ni, C.; Fang, Q.-Q.; Chen, W.-Z.; Jiang, J.-X.; Jiang, Z.; Ye, J.; Zhang, T.; Yang, L.; Meng, F.-B.; Xia, W.-J.; et al. Breast Cancer-Derived Exosomes Transmit lncRNA SNHG16 to Induce CD73+γδ1 Treg Cells. Signal Transduct. Target. Ther. 2020, 5, 41. [Google Scholar] [CrossRef] [PubMed]
  110. Hu, G.; Wu, P.; Cheng, P.; Zhang, Z.; Wang, Z.; Yu, X.; Shao, X.; Wu, D.; Ye, J.; Zhang, T.; et al. Tumor-Infiltrating CD39+γδTregs Are Novel Immunosuppressive T Cells in Human Colorectal Cancer. Oncoimmunology 2017, 6, e1277305. [Google Scholar] [CrossRef]
  111. Chabab, G.; Barjon, C.; Abdellaoui, N.; Salvador-Prince, L.; Dejou, C.; Michaud, H.-A.; Boissière-Michot, F.; Lopez-Crapez, E.; Jacot, W.; Pourquier, D.; et al. Identification of a Regulatory Vδ1 Gamma Delta T Cell Subpopulation Expressing CD73 in Human Breast Cancer. J. Leukoc. Biol. 2020, 107, 1057–1067. [Google Scholar] [CrossRef]
  112. Ferrick, D.A.; Schrenzel, M.D.; Mulvania, T.; Hsieh, B.; Ferlin, W.G.; Lepper, H. Differential Production of Interferon-Gamma and Interleukin-4 in Response to Th1- and Th2-Stimulating Pathogens by Gamma Delta T Cells in Vivo. Nature 1995, 373, 255–257. [Google Scholar] [CrossRef]
  113. Seo, N.; Tokura, Y.; Furukawa, F.; Takigawa, M. Down-Regulation of Tumoricidal NK and NK T Cell Activities by MHC Kb Molecules Expressed on Th2-Type Gammadelta T and Alphabeta T Cells Coinfiltrating in Early B16 Melanoma Lesions. J. Immunol. 1998, 161, 4138–4145. [Google Scholar] [CrossRef] [PubMed]
  114. Schmolka, N.; Serre, K.; Grosso, A.R.; Rei, M.; Pennington, D.J.; Gomes, A.Q.; Silva-Santos, B. Epigenetic and Transcriptional Signatures of Stable versus Plastic Differentiation of Proinflammatory Γδ T Cell Subsets. Nat. Immunol. 2013, 14, 1093–1100. [Google Scholar] [CrossRef]
  115. Chitadze, G.; Oberg, H.-H.; Wesch, D.; Kabelitz, D. The Ambiguous Role of Γδ T Lymphocytes in Antitumor Immunity. Trends Immunol. 2017, 38, 668–678. [Google Scholar] [CrossRef]
  116. Cao, Y. Cancer-Triggered Systemic Disease and Therapeutic Targets. Holist. Integr. Oncol. 2024, 3, 11. [Google Scholar] [CrossRef] [PubMed]
  117. Casetti, R.; Agrati, C.; Wallace, M.; Sacchi, A.; Martini, F.; Martino, A.; Rinaldi, A.; Malkovsky, M. Cutting Edge: TGF-Beta1 and IL-15 Induce FOXP3+ Gammadelta Regulatory T Cells in the Presence of Antigen Stimulation. J. Immunol. 2009, 183, 3574–3577. [Google Scholar] [CrossRef]
  118. Kouakanou, L.; Peters, C.; Sun, Q.; Floess, S.; Bhat, J.; Huehn, J.; Kabelitz, D. Vitamin C Supports Conversion of Human Γδ T Cells into FOXP3-Expressing Regulatory Cells by Epigenetic Regulation. Sci. Rep. 2020, 10, 6550. [Google Scholar] [CrossRef]
  119. Lo Presti, E.; Toia, F.; Oieni, S.; Buccheri, S.; Turdo, A.; Mangiapane, L.R.; Campisi, G.; Caputo, V.; Todaro, M.; Stassi, G.; et al. Squamous Cell Tumors Recruit Γδ T Cells Producing Either IL17 or IFNγ Depending on the Tumor Stage. Cancer Immunol. Res. 2017, 5, 397–407. [Google Scholar] [CrossRef]
  120. Sureshbabu, S.K.; Chaukar, D.; Chiplunkar, S.V. Hypoxia Regulates the Differentiation and Anti-Tumor Effector Functions of γδT Cells in Oral Cancer. Clin. Exp. Immunol. 2020, 201, 40–57. [Google Scholar] [CrossRef] [PubMed]
  121. Wu, Y.; Biswas, D.; Usaite, I.; Angelova, M.; Boeing, S.; Karasaki, T.; Veeriah, S.; Czyzewska-Khan, J.; Morton, C.; Joseph, M.; et al. A Local Human Vδ1 T Cell Population Is Associated with Survival in Nonsmall-Cell Lung Cancer. Nat. Cancer 2022, 3, 696–709. [Google Scholar] [CrossRef]
  122. Pizzolato, G.; Kaminski, H.; Tosolini, M.; Franchini, D.-M.; Pont, F.; Martins, F.; Valle, C.; Labourdette, D.; Cadot, S.; Quillet-Mary, A.; et al. Single-Cell RNA Sequencing Unveils the Shared and the Distinct Cytotoxic Hallmarks of Human TCRVδ1 and TCRVδ2 Γδ T Lymphocytes. Proc. Natl. Acad. Sci. USA 2019, 116, 11906–11915. [Google Scholar] [CrossRef]
  123. Conti, H.R.; Peterson, A.C.; Brane, L.; Huppler, A.R.; Hernández-Santos, N.; Whibley, N.; Garg, A.V.; Simpson-Abelson, M.R.; Gibson, G.A.; Mamo, A.J.; et al. Oral-Resident Natural Th17 Cells and Γδ T Cells Control Opportunistic Candida Albicans Infections. J. Exp. Med. 2014, 211, 2075–2084. [Google Scholar] [CrossRef]
  124. Peng, M.Y.; Wang, Z.H.; Yao, C.Y.; Jiang, L.N.; Jin, Q.L.; Wang, J.; Li, B.Q. Interleukin 17-Producing Gamma Delta T Cells Increased in Patients with Active Pulmonary Tuberculosis. Cell Mol. Immunol. 2008, 5, 203–208. [Google Scholar] [CrossRef] [PubMed]
  125. Murphy, A.G.; O’Keeffe, K.M.; Lalor, S.J.; Maher, B.M.; Mills, K.H.G.; McLoughlin, R.M. Staphylococcus Aureus Infection of Mice Expands a Population of Memory Γδ T Cells That Are Protective against Subsequent Infection. J. Immunol. 2014, 192, 3697–3708. [Google Scholar] [CrossRef]
  126. Ponomarev, E.D.; Novikova, M.; Yassai, M.; Szczepanik, M.; Gorski, J.; Dittel, B.N. Gamma Delta T Cell Regulation of IFN-Gamma Production by Central Nervous System-Infiltrating Encephalitogenic T Cells: Correlation with Recovery from Experimental Autoimmune Encephalomyelitis. J. Immunol. 2004, 173, 1587–1595. [Google Scholar] [CrossRef] [PubMed]
  127. Park, S.-G.; Mathur, R.; Long, M.; Hosh, N.; Hao, L.; Hayden, M.S.; Ghosh, S. T Regulatory Cells Maintain Intestinal Homeostasis by Suppressing Γδ T Cells. Immunity 2010, 33, 791–803. [Google Scholar] [CrossRef]
  128. Cui, Y.; Shao, H.; Lan, C.; Nian, H.; O’Brien, R.L.; Born, W.K.; Kaplan, H.J.; Sun, D. Major Role of Gamma Delta T Cells in the Generation of IL-17+ Uveitogenic T Cells. J. Immunol. 2009, 183, 560–567. [Google Scholar] [CrossRef]
  129. Shibata, S.; Tada, Y.; Hau, C.S.; Mitsui, A.; Kamata, M.; Asano, Y.; Sugaya, M.; Kadono, T.; Masamoto, Y.; Kurokawa, M.; et al. Adiponectin Regulates Psoriasiform Skin Inflammation by Suppressing IL-17 Production from Γδ-T Cells. Nat. Commun. 2015, 6, 7687. [Google Scholar] [CrossRef] [PubMed]
  130. Groh, V.; Steinle, A.; Bauer, S.; Spies, T. Recognition of Stress-Induced MHC Molecules by Intestinal Epithelial Gammadelta T Cells. Science 1998, 279, 1737–1740. [Google Scholar] [CrossRef]
  131. Groh, V.; Rhinehart, R.; Secrist, H.; Bauer, S.; Grabstein, K.H.; Spies, T. Broad Tumor-Associated Expression and Recognition by Tumor-Derived Gamma Delta T Cells of MICA and MICB. Proc. Natl. Acad. Sci. USA 1999, 96, 6879–6884. [Google Scholar] [CrossRef]
  132. Lai, A.Y.; Patel, A.; Brewer, F.; Evans, K.; Johannes, K.; González, L.E.; Yoo, K.J.; Fromm, G.; Wilson, K.; Schreiber, T.H.; et al. Cutting Edge: Bispecific Γδ T Cell Engager Containing Heterodimeric BTN2A1 and BTN3A1 Promotes Targeted Activation of Vγ9Vδ2+ T Cells in the Presence of Costimulation by CD28 or NKG2D. J. Immunol. 2022, 209, 1475–1480. [Google Scholar] [CrossRef]
  133. Maniar, A.; Zhang, X.; Lin, W.; Gastman, B.R.; Pauza, C.D.; Strome, S.E.; Chapoval, A.I. Human Gammadelta T Lymphocytes Induce Robust NK Cell-Mediated Antitumor Cytotoxicity through CD137 Engagement. Blood 2010, 116, 1726–1733. [Google Scholar] [CrossRef]
  134. Gao, Z.; Bai, Y.; Lin, A.; Jiang, A.; Zhou, C.; Cheng, Q.; Liu, Z.; Chen, X.; Zhang, J.; Luo, P. Gamma Delta T-Cell-Based Immune Checkpoint Therapy: Attractive Candidate for Antitumor Treatment. Mol. Cancer 2023, 22, 31. [Google Scholar] [CrossRef]
  135. Chow, A.; Perica, K.; Klebanoff, C.A.; Wolchok, J.D. Clinical Implications of T Cell Exhaustion for Cancer Immunotherapy. Nat. Rev. Clin. Oncol. 2022, 19, 775–790. [Google Scholar] [CrossRef]
  136. Demaria, O.; Cornen, S.; Daëron, M.; Morel, Y.; Medzhitov, R.; Vivier, E. Harnessing Innate Immunity in Cancer Therapy. Nature 2019, 574, 45–56. [Google Scholar] [CrossRef]
  137. Suzuki, K.; Suzuki, K.; Yabe, Y.; Iida, K.; Ishikawa, J.; Makita, S.; Kageyama, T.; Iwamoto, T.; Tanaka, S.; Yokota, M.; et al. NF-κB1 Contributes to Imiquimod-Induced Psoriasis-Like Skin Inflammation by Inducing Vγ4+Vδ4+γδT17 Cells. J. Investig. Dermatol. 2022, 142, 1639–1649.e5. [Google Scholar] [CrossRef]
  138. Bain, G.; Cravatt, C.B.; Loomans, C.; Alberola-Ila, J.; Hedrick, S.M.; Murre, C. Regulation of the Helix-Loop-Helix Proteins, E2A and Id3, by the Ras-ERK MAPK Cascade. Nat. Immunol. 2001, 2, 165–171. [Google Scholar] [CrossRef]
  139. Haks, M.C.; Lefebvre, J.M.; Lauritsen, J.P.H.; Carleton, M.; Rhodes, M.; Miyazaki, T.; Kappes, D.J.; Wiest, D.L. Attenuation of gammadeltaTCR Signaling Efficiently Diverts Thymocytes to the Alphabeta Lineage. Immunity 2005, 22, 595–606. [Google Scholar] [CrossRef]
  140. Chen, S.; Paveley, R.; Kraal, L.; Sritharan, L.; Stevens, E.; Dedi, N.; Shock, A.; Shaw, S.; Juarez, M.; Yeremenko, N.; et al. Selective Targeting of PI3Kδ Suppresses Human IL-17-Producing T Cells and Innate-like Lymphocytes and May Be Therapeutic for IL-17-Mediated Diseases. J. Autoimmun. 2020, 111, 102435. [Google Scholar] [CrossRef]
  141. Tumor Hypoxia Represses Γδ T Cell-Mediated Antitumor Immunity Against Brain Tumors—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/33574616/ (accessed on 3 February 2025).
  142. Hosokawa, H.; Rothenberg, E.V. How Transcription Factors Drive Choice of the T Cell Fate. Nat. Rev. Immunol. 2021, 21, 162–176. [Google Scholar] [CrossRef]
  143. Boehme, L.; Roels, J.; Taghon, T. Development of Γδ T Cells in the Thymus—A Human Perspective. Semin. Immunol. 2022, 61–64, 101662. [Google Scholar] [CrossRef]
  144. Roels, J.; Van Hulle, J.; Lavaert, M.; Kuchmiy, A.; Strubbe, S.; Putteman, T.; Vandekerckhove, B.; Leclercq, G.; Van Nieuwerburgh, F.; Boehme, L.; et al. Transcriptional Dynamics and Epigenetic Regulation of E and ID Protein Encoding Genes during Human T Cell Development. Front. Immunol. 2022, 13, 960918. [Google Scholar] [CrossRef]
  145. Li, Y.; Wu, Y.; Hu, Y. Metabolites in the Tumor Microenvironment Reprogram Functions of Immune Effector Cells Through Epigenetic Modifications. Front. Immunol. 2021, 12, 641883. [Google Scholar] [CrossRef]
  146. Franchina, D.G.; Dostert, C.; Brenner, D. Reactive Oxygen Species: Involvement in T Cell Signaling and Metabolism. Trends Immunol. 2018, 39, 489–502. [Google Scholar] [CrossRef] [PubMed]
  147. Mensurado, S.; Rei, M.; Lança, T.; Ioannou, M.; Gonçalves-Sousa, N.; Kubo, H.; Malissen, M.; Papayannopoulos, V.; Serre, K.; Silva-Santos, B. Tumor-Associated Neutrophils Suppress pro-Tumoral IL-17+ Γδ T Cells through Induction of Oxidative Stress. PLoS Biol. 2018, 16, e2004990. [Google Scholar] [CrossRef]
  148. The Role of ROS in Tumour Development and Progression—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/35102280/ (accessed on 3 February 2025).
  149. Microenvironmental Oxygen Pressure Orchestrates an Anti- and Pro-Tumoral Γδ T Cell Equilibrium via Tumor-Derived Exosomes—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/30546089/ (accessed on 3 February 2025).
  150. Wu, P.; Wu, D.; Ni, C.; Ye, J.; Chen, W.; Hu, G.; Wang, Z.; Wang, C.; Zhang, Z.; Xia, W.; et al. γδT17 Cells Promote the Accumulation and Expansion of Myeloid-Derived Suppressor Cells in Human Colorectal Cancer. Immunity 2014, 40, 785–800. [Google Scholar] [CrossRef]
  151. Exosomes Derived from Vδ2-T Cells Control Epstein-Barr Virus-Associated Tumors and Induce T Cell Antitumor Immunity—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/32998970/ (accessed on 3 February 2025).
  152. Updates on Chronic HBV: Current Challenges and Future Goals—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/31077059/ (accessed on 25 January 2025).
  153. Leslie, J.; Geh, D.; Elsharkawy, A.M.; Mann, D.A.; Vacca, M. Metabolic Dysfunction and Cancer in HCV: Shared Pathways and Mutual Interactions. J. Hepatol. 2022, 77, 219–236. [Google Scholar] [CrossRef]
  154. Chen, M.; Hu, P.; Peng, H.; Zeng, W.; Shi, X.; Lei, Y.; Hu, H.; Zhang, D.; Ren, H. Enhanced Peripheral γδT Cells Cytotoxicity Potential in Patients with HBV-Associated Acute-On-Chronic Liver Failure Might Contribute to the Disease Progression. J. Clin. Immunol. 2012, 32, 877–885. [Google Scholar] [CrossRef]
  155. Lin, H.; Dai, Z.; Huang, L.; Zhou, X. Hepatitis B Virus Reactivation Associated with CAR T-Cell Therapy. Holist. Integr. Oncol. 2024, 3, 16. [Google Scholar] [CrossRef]
  156. Rajoriya, N.; Fergusson, J.R.; Leithead, J.A.; Klenerman, P. Gamma Delta T-Lymphocytes in Hepatitis C and Chronic Liver Disease. Front. Immunol. 2014, 5, 400. [Google Scholar] [CrossRef]
  157. Jia, Z.-H.; Li, Y.-Y.; Wang, J.-Y.; Zhang, J.-Y.; Huang, A.; Guo, X.-D.; Zhu, Z.-Y.; Wang, F.-S.; Wu, X.-L. Activated Γδ T Cells Exhibit Cytotoxicity and the Capacity for Viral Clearance in Patients with Acute Hepatitis B. Clin. Immunol. 2019, 202, 40–48. [Google Scholar] [CrossRef]
  158. Conroy, M.J.; Mac Nicholas, R.; Taylor, M.; O’Dea, S.; Mulcahy, F.; Norris, S.; Doherty, D.G. Increased Frequencies of Circulating IFN-γ-Producing Vδ1+ and Vδ2+ Γδ T Cells in Patients with Asymptomatic Persistent Hepatitis B Virus Infection. Viral Immunol. 2015, 28, 201–208. [Google Scholar] [CrossRef] [PubMed]
  159. Hou, W.; Wu, X. Diverse Functions of Γδ T Cells in the Progression of Hepatitis B Virus and Hepatitis C Virus Infection. Front. Immunol. 2021, 11, 619872. [Google Scholar] [CrossRef] [PubMed]
  160. Jiayu, Z.; Zhang, Q. Hepatitis B Virus–Associated Diffuse Large B Cell Lymphoma: Epidemiology, Biology, Clinical Features and HBV Reactivation. Holist. Integr. Oncol. 2023, 2, 38. [Google Scholar] [CrossRef]
  161. Decreased Vδ2 Γδ T Cells Associated with Liver Damage by Regulation of Th17 Response in Patients with Chronic Hepatitis B—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/23847059/ (accessed on 27 January 2025).
  162. Chen, M.; Hu, P.; Ling, N.; Peng, H.; Lei, Y.; Hu, H.; Zhang, D.; Ren, H. Enhanced Functions of Peripheral Γδ T Cells in Chronic Hepatitis B Infection during Interferon α Treatment in Vivo and in Vitro. PLoS ONE 2015, 10, e0120086. [Google Scholar] [CrossRef]
  163. Nwabo Kamdje, A.H.; Tagne Simo, R.; Fogang Dongmo, H.P.; Bidias, A.R.; Masumbe Netongo, P. Role of Signaling Pathways in the Interaction between Microbial, Inflammation and Cancer. Holist. Integr. Oncol. 2023, 2, 42. [Google Scholar] [CrossRef]
  164. Cairo, C.; Armstrong, C.L.; Cummings, J.S.; Deetz, C.O.; Tan, M.; Lu, C.; Davis, C.E.; Pauza, C.D. Impact of Age, Gender, and Race on Circulating Γδ T Cells. Hum. Immunol. 2010, 71, 968–975. [Google Scholar] [CrossRef]
  165. TCRγδ(+)CD4(−)CD8(−) T Cells Suppress the CD8(+) T-Cell Response to Hepatitis B Virus Peptides, and Are Associated with Viral Control in Chronic Hepatitis B—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/24551107/ (accessed on 28 January 2025).
  166. Kong, X.; Sun, R.; Chen, Y.; Wei, H.; Tian, Z. γδT Cells Drive Myeloid-Derived Suppressor Cell-Mediated CD8+ T Cell Exhaustion in Hepatitis B Virus-Induced Immunotolerance. J. Immunol. 2014, 193, 1645–1653. [Google Scholar] [CrossRef]
  167. Wang, Y.; Guan, Y.; Hu, Y.; Li, Y.; Lu, N.; Zhang, C. Murine CXCR3+CXCR6+γδT Cells Reside in the Liver and Provide Protection Against HBV Infection. Front. Immunol. 2022, 12, 757379. [Google Scholar] [CrossRef]
  168. Tseng, C.T.; Miskovsky, E.; Houghton, M.; Klimpel, G.R. 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. [Google Scholar] [CrossRef]
  169. Agrati, C.; D’Offizi, G.; Narciso, P.; Abrignani, S.; Ippolito, G.; Colizzi, V.; Poccia, F. Vdelta1 T Lymphocytes Expressing a Th1 Phenotype Are the Major Gammadelta T Cell Subset Infiltrating the Liver of HCV-Infected Persons. Mol. Med. 2001, 7, 11–19. [Google Scholar] [CrossRef]
  170. Pár, G.; Rukavina, D.; Podack, E.R.; Horányi, M.; Szekeres-Barthó, J.; Hegedüs, G.; Paál, M.; Szereday, L.; Mózsik, G.; Pár, A. Decrease in CD3-Negative-CD8dim(+) and Vdelta2/Vgamma9 TcR+ Peripheral Blood Lymphocyte Counts, Low Perforin Expression and the Impairment of Natural Killer Cell Activity Is Associated with Chronic Hepatitis C Virus Infection. J. Hepatol. 2002, 37, 514–522. [Google Scholar] [CrossRef]
  171. Alonzi, T.; Agrati, C.; Costabile, B.; Cicchini, C.; Amicone, L.; Cavallari, C.; Rocca, C.D.; Folgori, A.; Fipaldini, C.; Poccia, F.; et al. Steatosis and Intrahepatic Lymphocyte Recruitment in Hepatitis C Virus Transgenic Mice. J. Gen. Virol. 2004, 85, 1509–1520. [Google Scholar] [CrossRef] [PubMed]
  172. Lee, J.-W.; Kim, W.; Kwon, E.-K.; Kim, Y.; Shin, H.M.; Kim, D.-H.; Min, C.-K.; Choi, J.-Y.; Lee, W.-W.; Choi, M.-S.; et al. Immunological Dynamics Associated with Rapid Virological Response during the Early Phase of Type I Interferon Therapy in Patients with Chronic Hepatitis C. PLoS ONE 2017, 12, e0179094. [Google Scholar] [CrossRef] [PubMed]
  173. Yin, W.; Tong, S.; Zhang, Q.; Shao, J.; Liu, Q.; Peng, H.; Hu, H.; Peng, M.; Hu, P.; Ren, H.; et al. Functional Dichotomy of Vδ2 Γδ T Cells in Chronic Hepatitis C Virus Infections: Role in Cytotoxicity but Not for IFN-γ Production. Sci. Rep. 2016, 6, 26296. [Google Scholar] [CrossRef]
  174. Bono, V.; Tincati, C.; Van Den Bogaart, L.; Cannizzo, E.S.; Rovito, R.; Augello, M.; De Bona, A.; D’Arminio Monforte, A.; Milazzo, L.; Marchetti, G. Gamma-Delta T-Cell Phenotype and Function in DAA-Treated HIV-HCV Co-Infected and HCV-Mono-Infected Subjects. Viruses 2022, 14, 1594. [Google Scholar] [CrossRef]
  175. Ghosh, A.; Mondal, R.K.; Romani, S.; Bagchi, S.; Cairo, C.; Pauza, C.D.; Kottilil, S.; Poonia, B. Persistent Gamma Delta T-Cell Dysfunction in Chronic HCV Infection despite Direct-Acting Antiviral Therapy Induced Cure. J. Viral Hepat. 2019, 26, 1105–1116. [Google Scholar] [CrossRef]
  176. Chen, D.; Luo, X.; Xie, H.; Gao, Z.; Fang, H.; Huang, J. Characteristics of IL-17 Induction by Schistosoma Japonicum Infection in C57BL/6 Mouse Liver. Immunology 2013, 139, 523–532. [Google Scholar] [CrossRef] [PubMed]
  177. Rhodes, K.A.; Andrew, E.M.; Newton, D.J.; Tramonti, D.; Carding, S.R. A Subset of IL-10-Producing Gammadelta T Cells Protect the Liver from Listeria-Elicited, CD8(+) T Cell-Mediated Injury. Eur. J. Immunol. 2008, 38, 2274–2283. [Google Scholar] [CrossRef]
  178. Paul, B.; Lewinska, M.; Andersen, J.B. Lipid Alterations in Chronic Liver Disease and Liver Cancer. JHEP Rep. 2022, 4, 100479. [Google Scholar] [CrossRef]
  179. Cichoż-Lach, H.; Michalak, A. Oxidative Stress as a Crucial Factor in Liver Diseases. World J. Gastroenterol. 2014, 20, 8082–8091. [Google Scholar] [CrossRef]
  180. Hammerich, L.; Tacke, F. Hepatic Inflammatory Responses in Liver Fibrosis. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 633–646. [Google Scholar] [CrossRef]
  181. Israelsen, M.; Francque, S.; Tsochatzis, E.A.; Krag, A. Steatotic Liver Disease. Lancet 2024, 404, 1761–1778. [Google Scholar] [CrossRef]
  182. Li, F.; Hao, X.; Chen, Y.; Bai, L.; Gao, X.; Lian, Z.; Wei, H.; Sun, R.; Tian, Z. The Microbiota Maintain Homeostasis of Liver-Resident γδT-17 Cells in a Lipid Antigen/CD1d-Dependent Manner. Nat. Commun. 2017, 8, 13839. [Google Scholar] [CrossRef]
  183. Tarantino, G.; Citro, V.; Balsano, C. Liver-Spleen Axis in Nonalcoholic Fatty Liver Disease. Expert. Rev. Gastroenterol. Hepatol. 2021, 15, 759–769. [Google Scholar] [CrossRef]
  184. Wallace, S.J.; Tacke, F.; Schwabe, R.F.; Henderson, N.C. Understanding the Cellular Interactome of Non-Alcoholic Fatty Liver Disease. JHEP Rep. 2022, 4, 100524. [Google Scholar] [CrossRef]
  185. Xu, R.; Tao, A.; Zhang, S.; Zhang, M. Neutralization of Interleukin-17 Attenuates High Fat Diet-Induced Non-Alcoholic Fatty Liver Disease in Mice. Acta Biochim. Biophys. Sin. 2013, 45, 726–733. [Google Scholar] [CrossRef]
  186. Jin, M.; Lai, Y.; Zhao, P.; Shen, Q.; Su, W.; Yin, Y.; Zhang, W. Effects of Peptidoglycan on the Development of Steatohepatitis. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158595. [Google Scholar] [CrossRef]
  187. Gao, C.; Wang, S.; Xie, X.; Ramadori, P.; Li, X.; Liu, X.; Ding, X.; Liang, J.; Xu, B.; Feng, Y.; et al. Single-Cell Profiling of Intrahepatic Immune Cells Reveals an Expansion of Tissue-Resident Cytotoxic CD4+ T Lymphocyte Subset Associated With Pathogenesis of Alcoholic-Associated Liver Diseases. Cell Mol. Gastroenterol. Hepatol. 2025, 19, 101411. [Google Scholar] [CrossRef]
  188. Torres-Hernandez, A.; Wang, W.; Nikiforov, Y.; Tejada, K.; Torres, L.; Kalabin, A.; Adam, S.; Wu, J.; Lu, L.; Chen, R.; et al. Γδ T Cells Promote Steatohepatitis by Orchestrating Innate and Adaptive Immune Programming. Hepatology 2020, 71, 477–494. [Google Scholar] [CrossRef]
  189. Lei, Z.; Yu, J.; Wu, Y.; Shen, J.; Lin, S.; Xue, W.; Mao, C.; Tang, R.; Sun, H.; Qi, X.; et al. CD1d Protects against Hepatocyte Apoptosis in Non-Alcoholic Steatohepatitis. J. Hepatol. 2024, 80, 194–208. [Google Scholar] [CrossRef]
  190. Han, Y.; Ling, Q.; Wu, L.; Wang, X.; Wang, Z.; Chen, J.; Zheng, Z.; Zhou, Z.; Jia, L.; Li, L.; et al. Akkermansia Muciniphila Inhibits Nonalcoholic Steatohepatitis by Orchestrating TLR2-Activated γδT17 Cell and Macrophage Polarization. Gut Microbes 2023, 15, 2221485. [Google Scholar] [CrossRef]
  191. Tsochatzis, E.A.; Bosch, J.; Burroughs, A.K. Liver Cirrhosis. Lancet 2014, 383, 1749–1761. [Google Scholar] [CrossRef] [PubMed]
  192. Meng, F.; Wang, K.; Aoyama, T.; Grivennikov, S.I.; Paik, Y.; Scholten, D.; Cong, M.; Iwaisako, K.; Liu, X.; Zhang, M.; et al. Interleukin-17 Signaling in Inflammatory, Kupffer Cells, and Hepatic Stellate Cells Exacerbates Liver Fibrosis in Mice. Gastroenterology 2012, 143, 765–776.e3. [Google Scholar] [CrossRef]
  193. Zheng, L.; Hu, Y.; Wang, Y.; Huang, X.; Xu, Y.; Shen, Y.; Cao, J. Recruitment of Neutrophils Mediated by Vγ2 Γδ T Cells Deteriorates Liver Fibrosis Induced by Schistosoma Japonicum Infection in C57BL/6 Mice. Infect. Immun. 2017, 85, e01020-16. [Google Scholar] [CrossRef] [PubMed]
  194. Kanayama, K.; Morise, K.; Nagura, H. Immunohistochemical Study of T Cell Receptor Gamma Delta Cells in Chronic Liver Disease. Am. J. Gastroenterol. 1992, 87, 1018–1022. [Google Scholar] [PubMed]
  195. Tan, Z.; Qian, X.; Jiang, R.; Liu, Q.; Wang, Y.; Chen, C.; Wang, X.; Ryffel, B.; Sun, B. IL-17A Plays a Critical Role in the Pathogenesis of Liver Fibrosis through Hepatic Stellate Cell Activation. J. Immunol. 2013, 191, 1835–1844. [Google Scholar] [CrossRef]
  196. Wang, X.; Sun, R.; Wei, H.; Tian, Z. High-Mobility Group Box 1 (HMGB1)-Toll-like Receptor (TLR)4-Interleukin (IL)-23-IL-17A Axis in Drug-Induced Damage-Associated Lethal Hepatitis: Interaction of Γδ T Cells with Macrophages. Hepatology 2013, 57, 373–384. [Google Scholar] [CrossRef]
  197. Seo, W.; Eun, H.S.; Kim, S.Y.; Yi, H.; Lee, Y.; Park, S.; Jang, M.; Jo, E.; Kim, S.C.; Han, Y.; et al. Exosome-mediated Activation of Toll-like Receptor 3 in Stellate Cells Stimulates Interleukin-17 Production by Γδ T Cells in Liver Fibrosis. Hepatology 2016, 64, 616–631. [Google Scholar] [CrossRef]
  198. Hammerich, L.; Bangen, J.M.; Govaere, O.; Zimmermann, H.W.; Gassler, N.; Huss, S.; Liedtke, C.; Prinz, I.; Lira, S.A.; Luedde, T.; et al. Chemokine Receptor CCR6-Dependent Accumulation of Γδ T Cells in Injured Liver Restricts Hepatic Inflammation and Fibrosis: Hepatology, Vol. 00, No. 0, 2013 Hammerich et Al. Hepatology 2014, 59, 630–642. [Google Scholar] [CrossRef]
  199. Liu, M.; Hu, Y.; Yuan, Y.; Tian, Z.; Zhang, C. γδT Cells Suppress Liver Fibrosis via Strong Cytolysis and Enhanced NK Cell-Mediated Cytotoxicity Against Hepatic Stellate Cells. Front. Immunol. 2019, 10, 477. [Google Scholar] [CrossRef]
  200. Mieli-Vergani, G.; Vergani, D. Autoimmune Hepatitis. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 320–329. [Google Scholar] [CrossRef] [PubMed]
  201. Aizawa, Y.; Hokari, A. Autoimmune Hepatitis: Current Challenges and Future Prospects. Clin. Exp. Gastroenterol. 2017, 10, 9–18. [Google Scholar] [CrossRef]
  202. Carey, E.J.; Ali, A.H.; Lindor, K.D. Primary Biliary Cirrhosis. Lancet 2015, 386, 1565–1575. [Google Scholar] [CrossRef]
  203. Singh, S.; Talwalkar, J.A. Primary Sclerosing Cholangitis: Diagnosis, Prognosis, and Management. Clin. Gastroenterol. Hepatol. 2013, 11, 898–907. [Google Scholar] [CrossRef] [PubMed]
  204. Martins, E.B.; Graham, A.K.; Chapman, R.W.; Fleming, K.A. Elevation of Γδ T Lymphocytes in Peripheral Blood and Livers of Patients with Primary Sclerosing Cholangitis and Other Autoimmune Liver Diseases. Hepatology 1996, 23, 988–993. [Google Scholar] [CrossRef] [PubMed]
  205. Hua, F.; Wang, L.; Rong, X.; Hu, Y.; Zhang, J.M.; He, W.; Zhang, F.C. Elevation of Vδ1 T Cells in Peripheral Blood and Livers of Patients with Primary Biliary Cholangitis. Clin. Exp. Immunol. 2016, 186, 347–355. [Google Scholar] [CrossRef] [PubMed]
  206. Haas, W.; Pereira, P.; Tonegawa, S. Gamma/Delta Cells. Annu. Rev. Immunol. 1993, 11, 637–685. [Google Scholar] [CrossRef]
  207. Zhou, Q.-H.; Wu, F.-T.; Pang, L.-T.; Zhang, T.-B.; Chen, Z. Role of γδT Cells in Liver Diseases and Its Relationship with Intestinal Microbiota. World J. Gastroenterol. 2020, 26, 2559–2569. [Google Scholar] [CrossRef]
  208. He, Q.; Lu, Y.; Tian, W.; Jiang, R.; Yu, W.; Liu, Y.; Sun, M.; Wang, F.; Zhang, H.; Wu, N.; et al. TOX Deficiency Facilitates the Differentiation of IL-17A-Producing Γδ T Cells to Drive Autoimmune Hepatitis. Cell Mol. Immunol. 2022, 19, 1102–1116. [Google Scholar] [CrossRef]
  209. Klemann, C.; Schröder, A.; Dreier, A.; Möhn, N.; Dippel, S.; Winterberg, T.; Wilde, A.; Yu, Y.; Thorenz, A.; Gueler, F.; et al. Interleukin 17, Produced by Γδ T Cells, Contributes to Hepatic Inflammation in a Mouse Model of Biliary Atresia and Is Increased in Livers of Patients. Gastroenterology 2016, 150, 229–241.e5. [Google Scholar] [CrossRef]
  210. Cui, T.X.; Brady, A.E.; Zhang, Y.-J.; Anderson, C.; Popova, A.P. IL-17a-Producing γδT Cells and NKG2D Signaling Mediate Bacterial Endotoxin-Induced Neonatal Lung Injury: Implications for Bronchopulmonary Dysplasia. Front. Immunol. 2023, 14, 1156842. [Google Scholar] [CrossRef] [PubMed]
  211. Ujiie, H.; Shevach, E.M. Γδ T Cells Protect the Liver and Lungs of Mice from Autoimmunity Induced by Scurfy Lymphocytes. J. Immunol. 2016, 196, 1517–1528. [Google Scholar] [CrossRef] [PubMed]
  212. Zakeri, N.; Hall, A.; Swadling, L.; Pallett, L.J.; Schmidt, N.M.; Diniz, M.O.; Kucykowicz, S.; Amin, O.E.; Gander, A.; Pinzani, M.; et al. Characterisation and Induction of Tissue-Resident Gamma Delta T-Cells to Target Hepatocellular Carcinoma. Nat. Commun. 2022, 13, 1372. [Google Scholar] [CrossRef] [PubMed]
  213. Ontogeny of Innate T Lymphocytes—Some Innate Lymphocytes Are More Innate than Others—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/25346734/ (accessed on 1 February 2025).
  214. Xia, J.; Wang, C.; Li, B. Hepatocellular Carcinoma Cells Induce Γδ T Cells through Metabolic Reprogramming into Tumor-Progressive Subpopulation. Front. Oncol. 2024, 14, 1451650. [Google Scholar] [CrossRef]
  215. Serrano, R.; Wesch, D.; Kabelitz, D. Activation of Human Γδ T Cells: Modulation by Toll-Like Receptor 8 Ligands and Role of Monocytes. Cells 2020, 9, 713. [Google Scholar] [CrossRef]
  216. Bonneville, M.; Fournié, J.-J. Sensing Cell Stress and Transformation through Vgamma9Vdelta2 T Cell-Mediated Recognition of the Isoprenoid Pathway Metabolites. Microbes Infect. 2005, 7, 503–509. [Google Scholar] [CrossRef]
  217. Deniger, D.C.; Maiti, S.N.; Mi, T.; Switzer, K.C.; Ramachandran, V.; Hurton, L.V.; Ang, S.; Olivares, S.; Rabinovich, B.A.; Huls, M.H.; et al. Activating and Propagating Polyclonal Gamma Delta T Cells with Broad Specificity for Malignancies. Clin. Cancer Res. 2014, 20, 5708–5719. [Google Scholar] [CrossRef]
  218. Pauza, C.D.; Liou, M.-L.; Lahusen, T.; Xiao, L.; Lapidus, R.G.; Cairo, C.; Li, H. Gamma Delta T Cell Therapy for Cancer: It Is Good to Be Local. Front. Immunol. 2018, 9, 1305. [Google Scholar] [CrossRef]
  219. Fan, X. Recent Highlights of Cancer Immunotherapy. Holist. Integr. Oncol. 2023, 2, 37. [Google Scholar] [CrossRef]
  220. He, W.; Hu, Y.; Chen, D.; Li, Y.; Ye, D.; Zhao, Q.; Lin, L.; Shi, X.; Lu, L.; Yin, Z.; et al. Hepatocellular Carcinoma-infiltrating Γδ T Cells Are Functionally Defected and Allogenic Vδ2 + Γδ T Cell Can Be a Promising Complement. Clin. Transl. Med. 2022, 12, e800. [Google Scholar] [CrossRef]
  221. Leone, R.D.; Powell, J.D. Metabolism of Immune Cells in Cancer. Nat. Rev. Cancer 2020, 20, 516–531. [Google Scholar] [CrossRef]
  222. Tajan, M.; Hock, A.K.; Blagih, J.; Robertson, N.A.; Labuschagne, C.F.; Kruiswijk, F.; Humpton, T.J.; Adams, P.D.; Vousden, K.H. A Role for P53 in the Adaptation to Glutamine Starvation through the Expression of SLC1A3. Cell Metab. 2018, 28, 721–736.e6. [Google Scholar] [CrossRef] [PubMed]
  223. Yi, Y.; He, H.-W.; Wang, J.-X.; Cai, X.-Y.; Li, Y.-W.; Zhou, J.; Cheng, Y.-F.; Jin, J.-J.; Fan, J.; Qiu, S.-J. The Functional Impairment of HCC-Infiltrating Γδ T Cells, Partially Mediated by Regulatory T Cells in a TGFβ- and IL-10-Dependent Manner. J. Hepatol. 2013, 58, 977–983. [Google Scholar] [CrossRef] [PubMed]
  224. Wu, D.; Wu, P.; Qiu, F.; Wei, Q.; Huang, J. Human γδT-Cell Subsets and Their Involvement in Tumor Immunity. Cell Mol. Immunol. 2017, 14, 245–253. [Google Scholar] [CrossRef]
  225. Poggi, A.; Zocchi, M.R.; Costa, P.; Ferrero, E.; Borsellino, G.; Placido, R.; Galgani, S.; Salvetti, M.; Gasperini, C.; Ristori, G.; et al. IL-12-Mediated NKRP1A up-Regulation and Consequent Enhancement of Endothelial Transmigration of V Delta 2+ TCR Gamma Delta+ T Lymphocytes from Healthy Donors and Multiple Sclerosis Patients. J. Immunol. 1999, 162, 4349–4354. [Google Scholar] [CrossRef] [PubMed]
  226. Bouet-Toussaint, F.; Cabillic, F.; Toutirais, O.; Le Gallo, M.; Thomas De La Pintière, C.; Daniel, P.; Genetet, N.; Meunier, B.; Dupont-Bierre, E.; Boudjema, K.; et al. Vγ9Vδ2 T Cell-Mediated Recognition of Human Solid Tumors. Potential for Immunotherapy of Hepatocellular and Colorectal Carcinomas. Cancer Immunol. Immunother. 2008, 57, 531–539. [Google Scholar] [CrossRef]
  227. Hu, Y.; Chen, D.; Hong, M.; Liu, J.; Li, Y.; Hao, J.; Lu, L.; Yin, Z.; Wu, Y. Apoptosis, Pyroptosis, and Ferroptosis Conspiringly Induce Immunosuppressive Hepatocellular Carcinoma Microenvironment and Γδ T-Cell Imbalance. Front. Immunol. 2022, 13, 845974. [Google Scholar] [CrossRef]
  228. Alnaggar, M.; Xu, Y.; Li, J.; He, J.; Chen, J.; Li, M.; Wu, Q.; Lin, L.; Liang, Y.; Wang, X.; et al. Allogenic Vγ9Vδ2 T Cell as New Potential Immunotherapy Drug for Solid Tumor: A Case Study for Cholangiocarcinoma. J. Immunother. Cancer 2019, 7, 36. [Google Scholar] [CrossRef]
  229. Jiang, H.; Yang, Z.; Song, Z.; Green, M.; Song, H.; Shao, Q. Γδ T Cells in Hepatocellular Carcinoma Patients Present Cytotoxic Activity but Are Reduced in Potency Due to IL-2 and IL-21 Pathways. Int. Immunopharmacol. 2019, 70, 167–173. [Google Scholar] [CrossRef]
  230. Rao, R.; Graffeo, C.S.; Gulati, R.; Jamal, M.; Narayan, S.; Zambirinis, C.P.; Barilla, R.; Deutsch, M.; Greco, S.H.; Ochi, A.; et al. Interleukin 17–Producing γδT Cells Promote Hepatic Regeneration in Mice. Gastroenterology 2014, 147, 473–484.e2. [Google Scholar] [CrossRef]
  231. Wu, L.; Deng, H.; Feng, X.; Xie, D.; Li, Z.; Chen, J.; Mo, Z.; Zhao, Q.; Hu, Z.; Yi, S.; et al. Interferon-Γ+ Th1 Activates Intrahepatic Resident Memory T Cells to Promote HBsAg Loss by Inducing M1 Macrophage Polarization. J. Med. Virol. 2024, 96, e29627. [Google Scholar] [CrossRef] [PubMed]
  232. Chang, L.; Gao, J.; Yu, Y.; Liao, B.; Zhou, Y.; Zhang, J.; Ma, X.; Hou, W.; Zhou, T.; Xu, Q. MMP10 Alleviates Non-Alcoholic Steatohepatitis by Regulating Macrophage M2 Polarization. Int. Immunopharmacol. 2023, 124, 111045. [Google Scholar] [CrossRef] [PubMed]
  233. Wang, Y.; Zhang, C. The Roles of Liver-Resident Lymphocytes in Liver Diseases. Front. Immunol. 2019, 10, 1582. [Google Scholar] [CrossRef] [PubMed]
  234. Khan, M.W.A.; Curbishley, S.M.; Chen, H.-C.; Thomas, A.D.; Pircher, H.; Mavilio, D.; Steven, N.M.; Eberl, M.; Moser, B. Expanded Human Blood-Derived γδT Cells Display Potent Antigen-Presentation Functions. Front. Immunol. 2014, 5, 344. [Google Scholar] [CrossRef]
  235. Dakhel, S.; Galbiati, A.; Migliorini, F.; Comacchio, C.; Oehler, S.; Prati, L.; Scheuermann, J.; Cazzamalli, S.; Neri, D.; Bassi, G.; et al. Isolation of a Natural Killer Group 2D Small-Molecule Ligand from DNA-Encoded Chemical Libraries. ChemMedChem 2022, 17, e202200350. [Google Scholar] [CrossRef]
  236. Yamaguchi, T.; Suzuki, Y.; Katakura, R.; Ebina, T.; Yokoyama, J.; Fujimiya, Y. Interleukin-15 Effectively Potentiates the in Vitro Tumor-Specific Activity and Proliferation of Peripheral Blood gammadeltaT Cells Isolated from Glioblastoma Patients. Cancer Immunol. Immunother. 1998, 47, 97–103. [Google Scholar] [CrossRef]
  237. Vacca, P.; Pietra, G.; Tumino, N.; Munari, E.; Mingari, M.C.; Moretta, L. Exploiting Human NK Cells in Tumor Therapy. Front. Immunol. 2019, 10, 3013. [Google Scholar] [CrossRef]
  238. Cairo, C.; Surendran, N.; Harris, K.M.; Mazan-Mamczarz, K.; Sakoda, Y.; Diaz-Mendez, F.; Tamada, K.; Gartenhaus, R.B.; Mann, D.L.; Pauza, C.D. Vγ2Vδ2 T Cell Costimulation Increases NK Cell Killing of Monocyte-Derived Dendritic Cells. Immunology 2014, 144, 422–430. [Google Scholar] [CrossRef]
  239. Park, J.H.; Lee, H.K. Function of Γδ T Cells in Tumor Immunology and Their Application to Cancer Therapy. Exp. Mol. Med. 2021, 53, 318–327. [Google Scholar] [CrossRef]
  240. Glatzel, A.; Wesch, D.; Schiemann, F.; Brandt, E.; Janssen, O.; Kabelitz, D. Patterns of Chemokine Receptor Expression on Peripheral Blood Gamma Delta T Lymphocytes: Strong Expression of CCR5 Is a Selective Feature of V Delta 2/V Gamma 9 Gamma Delta T Cells. J. Immunol. 2002, 168, 4920–4929. [Google Scholar] [CrossRef]
  241. Kabelitz, D.; Wesch, D. Features and Functions of Gamma Delta T Lymphocytes: Focus on Chemokines and Their Receptors. Crit. Rev. Immunol. 2003, 23, 339–370. [Google Scholar] [CrossRef]
  242. Wawrzyniecka, P.A.; Ibrahim, L.; Gritti, G.; Pule, M.A.; Maciocia, P.M. Chimeric Antigen Receptor T Cells for Gamma–Delta T Cell Malignancies. Leukemia 2022, 36, 577–579. [Google Scholar] [CrossRef] [PubMed]
  243. Mirzaei, H.R.; Mirzaei, H.; Lee, S.Y.; Hadjati, J.; Till, B.G. Prospects for Chimeric Antigen Receptor (CAR) Γδ T Cells: A Potential Game Changer for Adoptive T Cell Cancer Immunotherapy. Cancer Lett. 2016, 380, 413–423. [Google Scholar] [CrossRef] [PubMed]
  244. Ganapathy, T.; Radhakrishnan, R.; Sakshi, S.; Martin, S. CAR Γδ T Cells for Cancer Immunotherapy. Is the Field More Yellow than Green? Cancer Immunol. Immunother. 2023, 72, 277–286. [Google Scholar] [CrossRef]
  245. Wang, Y.; Wang, L.; Seo, N.; Okumura, S.; Hayashi, T.; Akahori, Y.; Fujiwara, H.; Amaishi, Y.; Okamoto, S.; Mineno, J.; et al. CAR-Modified Vγ9Vδ2 T Cells Propagated Using a Novel Bisphosphonate Prodrug for Allogeneic Adoptive Immunotherapy. Int. J. Mol. Sci. 2023, 24, 10873. [Google Scholar] [CrossRef] [PubMed]
  246. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T Cells Expressing CD19 Chimeric Antigen Receptors for Acute Lymphoblastic Leukaemia in Children and Young Adults: A Phase 1 Dose-Escalation Trial. Lancet 2015, 385, 517–528. [Google Scholar] [CrossRef]
  247. Brudno, J.N.; Kochenderfer, J.N. Toxicities of Chimeric Antigen Receptor T Cells: Recognition and Management. Blood 2016, 127, 3321–3330. [Google Scholar] [CrossRef]
  248. Oberg, H.-H.; Peipp, M.; Kellner, C.; Sebens, S.; Krause, S.; Petrick, D.; Adam-Klages, S.; Röcken, C.; Becker, T.; Vogel, I.; et al. Novel Bispecific Antibodies Increase Γδ T-Cell Cytotoxicity against Pancreatic Cancer Cells. Cancer Res. 2014, 74, 1349–1360. [Google Scholar] [CrossRef]
  249. Ganesan, R.; Chennupati, V.; Ramachandran, B.; Hansen, M.R.; Singh, S.; Grewal, I.S. Selective Recruitment of Γδ T Cells by a Bispecific Antibody for the Treatment of Acute Myeloid Leukemia. Leukemia 2021, 35, 2274–2284. [Google Scholar] [CrossRef]
  250. de Weerdt, I.; Lameris, R.; Scheffer, G.L.; Vree, J.; de Boer, R.; Stam, A.G.; van de Ven, R.; Levin, M.-D.; Pals, S.T.; Roovers, R.C.; et al. A Bispecific Antibody Antagonizes Prosurvival CD40 Signaling and Promotes Vγ9Vδ2 T Cell-Mediated Antitumor Responses in Human B-Cell Malignancies. Cancer Immunol. Res. 2021, 9, 50–61. [Google Scholar] [CrossRef]
  251. Lameris, R.; Ruben, J.M.; Iglesias-Guimarais, V.; de Jong, M.; Veth, M.; van de Bovenkamp, F.S.; de Weerdt, I.; Kater, A.P.; Zweegman, S.; Horbach, S.; et al. A Bispecific T Cell Engager Recruits Both Type 1 NKT and Vγ9Vδ2-T Cells for the Treatment of CD1d-Expressing Hematological Malignancies. Cell Rep. Med. 2023, 4, 100961. [Google Scholar] [CrossRef] [PubMed]
  252. Saura-Esteller, J.; de Jong, M.; King, L.A.; Ensing, E.; Winograd, B.; de Gruijl, T.D.; Parren, P.W.H.I.; van der Vliet, H.J. Gamma Delta T-Cell Based Cancer Immunotherapy: Past-Present-Future. Front. Immunol. 2022, 13, 915837. [Google Scholar] [CrossRef] [PubMed]
  253. Guo, Q.; Zhao, P.; Zhang, Z.; Zhang, J.; Zhang, Z.; Hua, Y.; Han, B.; Li, N.; Zhao, X.; Hou, L. TIM-3 Blockade Combined with Bispecific Antibody MT110 Enhances the Anti-Tumor Effect of Γδ T Cells. Cancer Immunol. Immunother. 2020, 69, 2571–2587. [Google Scholar] [CrossRef] [PubMed]
  254. Mu, X.; Xiang, Z.; Xu, Y.; He, J.; Lu, J.; Chen, Y.; Wang, X.; Tu, C.R.; Zhang, Y.; Zhang, W.; et al. Glucose Metabolism Controls Human Γδ T-Cell-Mediated Tumor Immunosurveillance in Diabetes. Cell Mol. Immunol. 2022, 19, 944–956. [Google Scholar] [CrossRef]
  255. Mattarollo, S.R.; Kenna, T.; Nieda, M.; Nicol, A.J. Chemotherapy and Zoledronate Sensitize Solid Tumour Cells to Vgamma9Vdelta2 T Cell Cytotoxicity. Cancer Immunol. Immunother. 2007, 56, 1285–1297. [Google Scholar] [CrossRef]
  256. Todaro, M.; Meraviglia, S.; Caccamo, N.; Stassi, G.; Dieli, F. Combining Conventional Chemotherapy and Γδ T Cell-Based Immunotherapy to Target Cancer-Initiating Cells. Oncoimmunology 2013, 2, e25821. [Google Scholar] [CrossRef]
  257. Benyamine, A.; Loncle, C.; Foucher, E.; Blazquez, J.-L.; Castanier, C.; Chrétien, A.-S.; Modesti, M.; Secq, V.; Chouaib, S.; Gironella, M.; et al. BTN3A Is a Prognosis Marker and a Promising Target for Vγ9Vδ2 T Cells Based-Immunotherapy in Pancreatic Ductal Adenocarcinoma (PDAC). Oncoimmunology 2017, 7, e1372080. [Google Scholar] [CrossRef]
  258. Bold, A.; Gross, H.; Holzmann, E.; Knop, S.; Hoeres, T.; Wilhelm, M. An Optimized Cultivation Method for Future in Vivo Application of Γδ T Cells. Front. Immunol. 2023, 14, 1185564. [Google Scholar] [CrossRef]
  259. Pfister, D.; Núñez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A.; et al. NASH Limits Anti-Tumour Surveillance in Immunotherapy-Treated HCC. Nature 2021, 592, 450–456. [Google Scholar] [CrossRef]
  260. Rodríguez-Caparrós, A.; García, V.; Casal, Á.; López-Ros, J.; García-Mariscal, A.; Tani-Ichi, S.; Ikuta, K.; Hernández-Munain, C. Notch Signaling Controls Transcription via the Recruitment of RUNX1 and MYB to Enhancers during T Cell Development. J. Immunol. 2019, 202, 2460–2472. [Google Scholar] [CrossRef]
  261. Zhu, X.; Sakamoto, S.; Ishii, C.; Smith, M.D.; Ito, K.; Obayashi, M.; Unger, L.; Hasegawa, Y.; Kurokawa, S.; Kishimoto, T.; et al. Dectin-1 Signaling on Colonic Γδ T Cells Promotes Psychosocial Stress Responses. Nat. Immunol. 2023, 24, 625–636. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Development and Peripheral Distribution of γδT Cells.
Figure 1. Development and Peripheral Distribution of γδT Cells.
Ijms 26 02778 g001
Figure 2. This figure illustrates the dual roles of γδT cells in liver diseases, including viral hepatitis, autoimmune liver disease, NAFLD, ALD, fibrosis, bacterial parasitic infections, and liver cancer. γδTcells exert protective effects by producing IFN-γ, TNF-a, perforin, and granzymes, enhancing immune defense, and regulating immune homeostasis via IL-10 and NKT activation. Conversely, they contribute to fibrosis and tumor progression through IL-17, TNF-a, and FASL, activating hepatic stellate cells (HSCs) and suppressing Tregs. Cytotoxicity is mediated via TRAIL, FASL, and CD16l, while CD137 signaling modulates interactions with NK cells. These insights highlight γδT cells as potential therapeutic targets in liver diseases.
Figure 2. This figure illustrates the dual roles of γδT cells in liver diseases, including viral hepatitis, autoimmune liver disease, NAFLD, ALD, fibrosis, bacterial parasitic infections, and liver cancer. γδTcells exert protective effects by producing IFN-γ, TNF-a, perforin, and granzymes, enhancing immune defense, and regulating immune homeostasis via IL-10 and NKT activation. Conversely, they contribute to fibrosis and tumor progression through IL-17, TNF-a, and FASL, activating hepatic stellate cells (HSCs) and suppressing Tregs. Cytotoxicity is mediated via TRAIL, FASL, and CD16l, while CD137 signaling modulates interactions with NK cells. These insights highlight γδT cells as potential therapeutic targets in liver diseases.
Ijms 26 02778 g002
Figure 3. Crosstalk between γδT cells and other immune cells in the liver microenvironment.
Figure 3. Crosstalk between γδT cells and other immune cells in the liver microenvironment.
Ijms 26 02778 g003
Table 1. Subsets of Human γδT Cells and Their Functional Characteristics.
Table 1. Subsets of Human γδT Cells and Their Functional Characteristics.
Vδ ChainsPaired Vγ Gene
Usage
DistributionDevelopment in ThymusRecognition/AntigenNotes
Vδ1Vγ2/3/4/5/8/9Peripheral blood, skin, gut, spleen, liverMid-gestation onwardsMICA/B, ULBPs, B7-H6, CD1c, CD1d, SEBPaired with diverse Vγ chains.
Vδ2Vγ9Peripheral bloodDetectable 5 to 6 weeks in fetal liverPhosphoantigens, F1-ATPase, BTN3A1, hMSH2, MICA/B, SEs, TSST-1, Nectin-like 5Vδ2/Vγ9 exclusively pairs.
Vδ3Vγ2/3Peripheral blood, liverPredominant in late-fetal and neonatal bloodCD1dRepresent approximately 0.2% of the total circulating T cells and react to CD1d.
Vδ5Vγ4Peripheral blood-Endothelial protein C receptor (EPCR)Identifying transformed cells by interacting with the endothelial protein C receptor.
Vδ6/Peripheral blood of lymphoma patients---
Abbreviations: MICA/B, MHC Class I Chain-Related Proteins A and B; ULBP, UL16 Binding Protein; CD1c, Cluster of Differentiation 1c; CD1d, Cluster of Differentiation 1d; SEB, Staphylococcal Enterotoxin B; F1-ATPase, Mitochondrial ATP Synthase F1 Subunit; BTN3A1, Butyrophilin Subfamily 3 Member A1; hMSH2, Human MutS Homolog 2; TSST-1, Toxic Shock Syndrome Toxin-1.
Table 2. γδT Cell-Based Cancer Therapies and Clinical Trials.
Table 2. γδT Cell-Based Cancer Therapies and Clinical Trials.
Therapeutic
Approach
Specific StrategyTarget Indications
(Cancer Types)
NCT NumberLocation
ACT (Adoptive Cell Transfer)Donor/Allogeneic γδT Cell InfusionRelapsed/Refractory Leukemia (e.g., AML)NCT04439721Suzhou, China
Hepatocellular Carcinoma (HCC)NCT04518774Beijing, China
Relapsed/Refractory NHL or PTCLNCT04696705Tianjin, China
Relapsed Hematologic MalignanciesNCT05755854Hefei, China
Acute Myeloid Leukemia (AML)NCT03790072Praha, Czechia
Autologous γδT Cell InfusionAdvanced HBV-Related Hepatocellular Carcinoma (HCC)NCT04032392Beijing, China
Relapsed/Refractory B-NHL, CLL, PTCLNCT04028440Tianjin, China
γδT Cells + Tumor Reduction Surgery (Cryoablation/IRE)Hepatocellular Carcinoma (HCC)NCT02425735,Guangzhou, China
Liver cancer
NCT03183219
Breast Cancer (Her-, Er-, Pr-)NCT02418481, NCT03183206Guangzhou, China
Breast Cancer
Non-Small Cell Lung Cancer (No EGFR Mutation)NCT02425748,Guangzhou, China
Lung cancer
NCT03183232
Locally Advanced Pancreatic CancerNCT03180437Guangzhou, China
γδT Cells + CIKGastric CancerNCT02585908Beijing, China
CAR-γδT Cell TherapyCD19-CAR-γδT CellsB-Cell Lymphoma, ALL, CLLNCT02656147Beijing, China
Relapsed/Refractory B-Cell NHLNCT05554939, NCT06838832, NCT06503211Beijing, China
Nanjing, China
Relapsed/Refractory B-Cell ALLNCT06696833, NCT06056752Suzhou, China
Hefei, China
Relapsed/Refractory B-Cell Hematologic MalignanciesNCT06092047Suzhou, China
B-Cell Malignancies (Follicular, Mantle Cell, Marginal Zone, Mediastinal)NCT04735471Stanford, CA, USA
B7H3-CAR-γδT CellsAdvanced Solid TumorsNCT06825455Beijing, China
Relapsed/Refractory B7H3+ Malignant GliomaNCT06018363Suzhou, China
B7H3+ Solid Tumor Leptomeningeal MetastasesNCT06592092Beijing, China
GPC3/Mesothelin-CAR-γδT CellsSolid Cancers (Expressing GPC3 or Mesothelin, e.g., HCC)NCT06196294Guangzhou, China
Universal CAR-γδT CellsPost-Transplant Relapsed AMLNCT04796441,Yanda, China
Refractory/Relapsed AML
NCT05388305Wuhan, China
CD7-CAR-γδT CellsRelapsed/Refractory CD7+ T-Cell MalignanciesNCT04702841Hefei, China
UTAA17 (CAR-γδT Cells)Relapsed/Refractory Multiple Myeloma (MM)NCT06279026Suzhou, China
NKG2DL-CAR-γδT CellsRelapsed/Refractory Solid Tumors (Colorectal, TNBC, Sarcoma, Nasopharyngeal)NCT04107142Johor, Malaysia
Advanced Solid or Hematologic MalignanciesNCT05302037Singapore
CD70-CAR-γδT CellsRelapsed/Refractory Clear Cell Renal Cell Carcinoma (ccRCC)NCT06480565Nashville, TN, USA
Drug-Resistant γδT Cells (DRI)Newly Diagnosed Glioblastoma (GBM)NCT04165941Birmingham, AL, USA
Combination TherapyγδT Cells + Immunomodulators (Interferon/PD-1)Stage III–IV Resectable MelanomaNCT06212388Xi’an, China
TKC (NK + γδT Cells) + ChemotherapyAdvanced Non-Small Cell Lung Cancer (NSCLC)NCT04990063Xi’an, China
Note: All data in this table are sourced from https://clinicaltrials.gov/. Abbreviations: ACT (Adoptive Cell Transfer), CAR-γδT (Chimeric Antigen Receptor Gamma Delta T), NCT (National Clinical Trial), HCC (Hepatocellular Carcinoma), AML (Acute Myeloid Leukemia), NHL (Non-Hodgkin Lymphoma), PTCL (Peripheral T-Cell Lymphoma), B-NHL (B-Cell Non-Hodgkin Lymphoma), CLL (Chronic Lymphocytic Leukemia), ALL (Acute Lymphocytic Leukemia), MM (Multiple Myeloma), GBM (Glioblastoma Multiforme), TNBC (Triple-Negative Breast Cancer), ccRCC (Clear Cell Renal Cell Carcinoma), NSCLC (Non-Small Cell Lung Cancer), CIK (Cytokine-Induced Killer), IRE (Irreversible Electroporation).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhan, C.; Peng, C.; Wei, H.; Wei, K.; Ou, Y.; Zhang, Z. Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases. Int. J. Mol. Sci. 2025, 26, 2778. https://doi.org/10.3390/ijms26062778

AMA Style

Zhan C, Peng C, Wei H, Wei K, Ou Y, Zhang Z. Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases. International Journal of Molecular Sciences. 2025; 26(6):2778. https://doi.org/10.3390/ijms26062778

Chicago/Turabian Style

Zhan, Chenjie, Chunxiu Peng, Huaxiu Wei, Ke Wei, Yangzhi Ou, and Zhiyong Zhang. 2025. "Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases" International Journal of Molecular Sciences 26, no. 6: 2778. https://doi.org/10.3390/ijms26062778

APA Style

Zhan, C., Peng, C., Wei, H., Wei, K., Ou, Y., & Zhang, Z. (2025). Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases. International Journal of Molecular Sciences, 26(6), 2778. https://doi.org/10.3390/ijms26062778

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop