Abstract

Due to the ability of γδ T cells to bridge adaptive and innate immunity, γδ T cells can respond to a variety of molecular cues and acquire the ability to induce a variety of cytokines such as IL-17 family, IFN-γ, IL-4, and IL-10. IL-17+ γδ T cells (γδ T17 cells) populations have recently received considerable interest as they are the major early source of IL-17A in many immune response models. However, the exact mechanism of γδ T17 cells is still poorly understood, especially in the context of cardiovascular disease (CVD). CVD is the leading cause of death in the world, and it tends to be younger. Here, we offer a review of the cardiovascular inflammatory and immune functions of γδ T17 cells in order to understand their role in CVD, which may be the key to developing new clinical applications.

INTRODUCTION

Cardiovascular disease (CVD) is the leading cause of death worldwide and poses a serious threat to human health.1–3 These diseases involved injury in the heart or blood vessels, including myocardial infarction (MI), atherosclerosis (AS), aortic aneurysm (AA), ischemia-reperfusion injury (IRI), and so on.4,5 At present, it has been recognized that γδ T cells are closely related to CVD.6

γδ T cells are a group of T lymphocytes that are distinct from αβ T cells, whose surface TCR is composed of γ chain and δ chain. γδ T cells only account for 1–5% of T lymphocytes in peripheral blood circulation and lymphatic circulation, and they were mainly distributed in mucosa and subcutaneous tissue.7–9 γδ T cells have been classified in a variety of ways, among which the most common classification is based on composition of TCR chains10 and cytokine secretion.11

γδ T cells as a bridge contact innate and adaptive immunity,12 such as they can act as APC to perform the antigen presentation function,13 and secrete a variety of cytokines, including IL-17 family, IFN-γ, IL-4, IL-10, and so on.14–17 In this group of cytokines, the IL-17 family consists of 6 structurally related cytokines,18 and the research hotspots are focused on IL-17A and IL-17F.

γδ T cells that produce IL-17 family, which are termed γδ T17 cells (or alternatively γδ17 cells, γδT-17 cells, Tγδ17 cells),19,20 and they are the primary early source of IL-17A in immune response.21 Two different types of γδ T17 cells have been found in each human and mice.22 γδ T17 cells can rapidly not only produce large amounts of IL-17A/F, but also IL-22, IL-21, and GM-CSF, which are attributed to their innate feature.23,24 In this review, we introduced the classification, differentiation, and characteristics of γδ T17 cells. Then, we sorted out the role of γδ T17 cells in CVD.

CLASSIFICATION OF γδ T CELLS

Classification based on TCR chains

γδ T cells can be divided into different subsets according to different composition of γ chains and δ chains. Human γδ T cells can be distinguished by δ chain expression, including Vδ1, Vδ2, Vδ3, and Vδ5 subtypes.10,25 However, murine γδ T cells can be distinguished by their γ chain expression.26 According to Heilig and Tonegawa nomenclature,27 there are 7 subtypes of mouse γδ T cells, Vγ1–Vγ728 (Table 1).

TABLE 1

Subsets of human and murine γδ T cells

 SubsetPaired TCR chainsTissue residentReference
Human
Murine
Vδ1 Vδ2 Vδ3 Vδ5
Vγ1 Vγ2 Vγ3 Vγ4 Vγ5 Vγ6 Vγ7
Vγ2/3/4/5/8/9 Vγ9
Vγ2/3/4 Vγ4
Vδ6.3/6.4
Vδ4 Vδ1 Vδ4 Vδ1 Vδ1 Vδ4/5/6
Skin, liver, spleen mucosal tissues, PB (peripheral blood) Skin, PB
Liver, PB PB
Skin, lung, colon, liver, PB Skin, lung, colon, liver, PB Skin
Skin, lung, colon, liver, PB, joint Skin, liver
Genital tract, tongue, lung, colon, skin, adipose tissue Intraepithelial lymphocytes
29,30,31  
32,33,34  
35,36  
37  
38,39  
40,41,42,43  
44,45  
39,46,47,48  
49,50  
51,52  
53
 SubsetPaired TCR chainsTissue residentReference
Human
Murine
Vδ1 Vδ2 Vδ3 Vδ5
Vγ1 Vγ2 Vγ3 Vγ4 Vγ5 Vγ6 Vγ7
Vγ2/3/4/5/8/9 Vγ9
Vγ2/3/4 Vγ4
Vδ6.3/6.4
Vδ4 Vδ1 Vδ4 Vδ1 Vδ1 Vδ4/5/6
Skin, liver, spleen mucosal tissues, PB (peripheral blood) Skin, PB
Liver, PB PB
Skin, lung, colon, liver, PB Skin, lung, colon, liver, PB Skin
Skin, lung, colon, liver, PB, joint Skin, liver
Genital tract, tongue, lung, colon, skin, adipose tissue Intraepithelial lymphocytes
29,30,31  
32,33,34  
35,36  
37  
38,39  
40,41,42,43  
44,45  
39,46,47,48  
49,50  
51,52  
53
TABLE 1

Subsets of human and murine γδ T cells

 SubsetPaired TCR chainsTissue residentReference
Human
Murine
Vδ1 Vδ2 Vδ3 Vδ5
Vγ1 Vγ2 Vγ3 Vγ4 Vγ5 Vγ6 Vγ7
Vγ2/3/4/5/8/9 Vγ9
Vγ2/3/4 Vγ4
Vδ6.3/6.4
Vδ4 Vδ1 Vδ4 Vδ1 Vδ1 Vδ4/5/6
Skin, liver, spleen mucosal tissues, PB (peripheral blood) Skin, PB
Liver, PB PB
Skin, lung, colon, liver, PB Skin, lung, colon, liver, PB Skin
Skin, lung, colon, liver, PB, joint Skin, liver
Genital tract, tongue, lung, colon, skin, adipose tissue Intraepithelial lymphocytes
29,30,31  
32,33,34  
35,36  
37  
38,39  
40,41,42,43  
44,45  
39,46,47,48  
49,50  
51,52  
53
 SubsetPaired TCR chainsTissue residentReference
Human
Murine
Vδ1 Vδ2 Vδ3 Vδ5
Vγ1 Vγ2 Vγ3 Vγ4 Vγ5 Vγ6 Vγ7
Vγ2/3/4/5/8/9 Vγ9
Vγ2/3/4 Vγ4
Vδ6.3/6.4
Vδ4 Vδ1 Vδ4 Vδ1 Vδ1 Vδ4/5/6
Skin, liver, spleen mucosal tissues, PB (peripheral blood) Skin, PB
Liver, PB PB
Skin, lung, colon, liver, PB Skin, lung, colon, liver, PB Skin
Skin, lung, colon, liver, PB, joint Skin, liver
Genital tract, tongue, lung, colon, skin, adipose tissue Intraepithelial lymphocytes
29,30,31  
32,33,34  
35,36  
37  
38,39  
40,41,42,43  
44,45  
39,46,47,48  
49,50  
51,52  
53

Although classification based on TCR chains has been accepted as the acknowledged method, 1 subtype of γδ T cells can secrete a variety of cytokines, making this cell subtype multifunctional. For example, the role of Vδ1 T cells is debatable. Vδ1 T cells have 2 opposite effects, both secreting IFN-γ to produce antitumor effects29,30 and secreting IL-17 to promote tumor growth.54,55 Therefore, classification based on TCR may have some limitation reflecting cell function.

Classification of cytokines secreted

According to the secreted cytokines, γδ T cells can be divided into IL-17+ γδ T cells, IFN-γ+ γδ T cells, IL-4+ γδ T cells, and so on (Table 2). This classification method can avoid the inconsistency in the description of γδ T cells due to different species. Therefore, the classification of cell function is widely used in practical applications.14,61,64

TABLE 2

Functional subsets of human and murine γδ T cells

 SubsetFunctionReference
Human
Murine
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T IL-17+ γδ T
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T KGF+ γδ T IL-17+ γδ T
Antitumor, anti-infection Anti-inflammatory Immune tolerance
Proinflammatory
Anti-infection
Anti-inflammatory, allergenic Protect liver, anti-infection
Protect the integrity of damaged epithelial surfaces Proatherogenic, promote tumor
56,57  
58  
17  
59  
60  
15,16  
61  
62  
63
 SubsetFunctionReference
Human
Murine
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T IL-17+ γδ T
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T KGF+ γδ T IL-17+ γδ T
Antitumor, anti-infection Anti-inflammatory Immune tolerance
Proinflammatory
Anti-infection
Anti-inflammatory, allergenic Protect liver, anti-infection
Protect the integrity of damaged epithelial surfaces Proatherogenic, promote tumor
56,57  
58  
17  
59  
60  
15,16  
61  
62  
63
TABLE 2

Functional subsets of human and murine γδ T cells

 SubsetFunctionReference
Human
Murine
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T IL-17+ γδ T
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T KGF+ γδ T IL-17+ γδ T
Antitumor, anti-infection Anti-inflammatory Immune tolerance
Proinflammatory
Anti-infection
Anti-inflammatory, allergenic Protect liver, anti-infection
Protect the integrity of damaged epithelial surfaces Proatherogenic, promote tumor
56,57  
58  
17  
59  
60  
15,16  
61  
62  
63
 SubsetFunctionReference
Human
Murine
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T IL-17+ γδ T
IFN-γ+ γδ T IL-4+ γδ T IL-10+ γδ T KGF+ γδ T IL-17+ γδ T
Antitumor, anti-infection Anti-inflammatory Immune tolerance
Proinflammatory
Anti-infection
Anti-inflammatory, allergenic Protect liver, anti-infection
Protect the integrity of damaged epithelial surfaces Proatherogenic, promote tumor
56,57  
58  
17  
59  
60  
15,16  
61  
62  
63

γδ T cells can secrete a variety of cytokines and chemokines, including proinflammatory cytokines, IFN-γ.57 In addition, IL-4+ γδ T cells infiltrating the spleen and heart are major early producers of IL-4, inhibiting acute cardiac inflammation in a mouse model of human viral myocarditis induced by Coxsackievirus of group B3 (CVB3) infection.15 IL-10+ γδ T cells controlled the expansion of CD8+ T cells and reduced TNF-α secretion by activated CD8+ T cells.61 Certain γδ T cells secrete specific cytokines, such as keratinocyte growth factor (KGF), that play important roles in the control of epithelial integrity, fibrin production, and wound repair.62

γδ T cells produce IL-17A, which promotes CVB3-induced viral pancreatitis.48 In addition, γδ T17 cells induced neutrophil infiltration into meninges by CXCL1 and CXCL2 and aggravated ischemic brain injury.65,66 IL-17A produced by γδ T cells in the aorta of C57BL/6 mice induced proinflammatory chemokines to recruit neutrophils and monocytes to promotes atherogenesis.63

γδ T17 CELLS

Differentiation of γδ T17 cells

The thymic programming of human γδ T cells appears to differ from that described in mice.57 In human, γδ T17 cells production occurs peripherally,57 and unlike murine γδ T cells that “naturally” produce IL-17 during thymus development, human γδ T cells acquire this ability only under inflammatory conditions, similar to Th17 cells. Therefore, γδ T17 cells are absent in the human thymus57 but can accumulate in inflamed tissues.67 Circulating Vγ9Vδ2 T cells from healthy adult donors produce little or no IL-1768, unless under inflammatory stimulation. For example, IL-17+ Vγ9Vδ2 T cells were significantly increased in patients with bacterial meningitis.67 Differentiation of human γδ T17 cells in vitro requires TCR activation69 and maximization with other cytokines, including IL-23, IL-6, TGF-β, and IL-1β.67 Human γδ T17 cells were significantly amplified by IL-7 and TCR agonist68 (Figure 1).

Development of γδ T17 cells in human and mouse. γδ T17 cells do not exist in human thymus, and human γδ T17 cells only appear in peripheral inflammatory conditions, whether pathogen-associated molecular pattern (PAMP) or damage-associated molecular pattern (DAMP). In mice, γδ T17 cells are thought to be uniquely developed in the fetal/perinatal thymus. The precommitted γδ T17 cells differentiated into mature γδ T17 cells through the intrinsic developmental program and were then activated in the peripheral
FIGURE 1

Development of γδ T17 cells in human and mouse. γδ T17 cells do not exist in human thymus, and human γδ T17 cells only appear in peripheral inflammatory conditions, whether pathogen-associated molecular pattern (PAMP) or damage-associated molecular pattern (DAMP). In mice, γδ T17 cells are thought to be uniquely developed in the fetal/perinatal thymus. The precommitted γδ T17 cells differentiated into mature γδ T17 cells through the intrinsic developmental program and were then activated in the peripheral

In mice, γδ T17 cells are identified to be developed in the fetal/perinatal thymus, suggested that physiologic development of γδ T17 cells is terminated before birth and that these cells subsequently persist as a long-lived, self-renewing population.70

Murine γδ T17 cells differentiation requires IL-23, IL-1β, IL-7, and other cytokines. When γδ T17 cells in the thymus, it requires TCR signaling to develop. Subsequently, γδ T17 cells leaved thymus to periphery, in this process, the influence of TCR signaling on γδ T17 cells gradually weakens, the activity of these cells largely guided by innate signaling, such as IL-23 and IL-1β.24,71,72 But there is a research that shows that IL-23 is indispensable for the differentiation of γδ T17 cells in thymus.73,74

IL-7 is an essential cytokine for T cells development and survival, and γδ T cells is no exception.75 γδ T cells in the thymus of IL-7R–/– mice were severely reduced,76 partly due to the effect of IL-7 in V-J recombination of the TCR γ gene.77 γδ T17 cells are dependent on IL-7 for regeneration and survival, because IL-7 connects to up-regulate antiapoptotic molecules Bcl-2 and Bcl-xL.78

The role of IL-6 and IL-15Rα in γδ T17 cells development is controversial. The addition of IL-6 impair induction of γδ T17 cells,72,79 and the lack of IL-15Rα increases γδ T17 cells and their precursors.80 In the thymus, TGF-β1 plays an irreplaceable role in the acquisition of IL-17-producing capacity,73,81 but it inhibited the expansion of γδ T17 cells in vitro and negatively regulated γδ T17 cells expansion or survival.72,82

Transcription factors (TFs) and signaling pathways involved in γδ T17 cells differentiation include RORγt, SRY-related HMG-box (SOX) 4, and SOX13, B-lymphoid tyrosine kinase (Blk), PI3K/Akt, Notch/Hes-1, NF-κB, and mTOR.83 RORγt is the master TF for cytokine IL-17.84 The expression of RORγt was significantly increased in γδ T17 cells from IL-10−/– mice, indicating that RORγt is influenced by IL-10 in γδ T17 cells differentiation.85 SOX4 and SOX13 constitute the central positive regulators of γδ T17 cells differentiation.86,87 SOX4/13 are paramount in the acquisition of the γδ T17 cells effector fate by controlling Rorc (the cardinal TF for γδT17 cells) transcription, enhancing IL-7Rα signaling pathway, and inhibiting Rorc-repressing TFs.87,88 Blk, a B-cell-specific member of the Src family that encodes protein tyrosine kinases, is preferentially expressed in γδ T cells over αβ T cells. BLK affects γδ T17 cells differentiation possibly by initiating its genetic signaling pathway or regulating the γδTCR signaling threshold.89 And, RasGRP1-deficient γδ T cells are damaged and unable to produce IL-17.90

The PI3K/Akt pathway plays an important role in the transcription process of γδ T17 lineage. PI3K inhibition reduced the expression of the TFs RORγt and SOX13 in developing γδ T cells. Syk induces PI3K/Akt pathway activation under γδ TCR stimulation and significantly induces γδ T17 cells development.91,92 Notch/Hes1 signaling pathway plays a crucial part in γδ T17 cells formation. Hes1 expression was associated with IL-17 production, and hes1-deficient mice had almost no γδ T17 cells.93 The expression of IL-7R in γδ T cells can be controlled by the Notch–RBPjκ signaling pathway. Deletion of RBPJκ significantly reduced the size of γδ T17 cells banks in peripheral organs of adult mice.94 γδ T cells require RelA and RelB, members of the NF-κB family, and lymphotoxin β receptors for IL-17 production.95 mTORC1 and mTORC2 signals synergistically promote IL-17 secretion in γδ T17 cells, mTORC1 promotes IL-17 expression by mediating glucose-dependent glycolysis, and mTORC2 enhances γδ T17 differentiation by inhibiting the production of mitoROS.96

Characteristics of γδ T17 cells

Many cell surface markers have been shown to distinguish γδ T17 cells, human and murine γδ T17 cells can be defined as CD3high, IL-7Rαhigh, IL-18Rhigh, and CCR6+ cells; in addition, CD161+ is unique to human γδ T17 cells.64,97–101 γδ T17 cells mainly reside in the heart, blood vessels, lung, skin, vagina, oral barrier sites, and endotheliocyte102,103; these cells are characterized by their rapid production of IL-17A/F23, IL-2224, IL-8, TNF-α, IL-21, and GM-CSF.104 The migration of murine γδ T17 cells is regulated by chemokine receptors CCR2 (during inflammation) and CCR6 (homeostasis).105 An animal study reported that γδ T17 cells appear to directly recognize microbiome-derived lipids provided by nonclassical MHC molecule CD1d.20 CD1d is a typical lipid presenting molecule for NKT cells and can also present lipid antigens to γδ TCR and activate γδ T cells. Hepatocytes express CD1d microbiome lipid antigens rather than pathogen-associated molecular pattern (PAMP) or cytokine signaling to maintain liver-resident γδ T17 cells homeostasis. Low endogenous levels of IL-1β regulated by skin commensals to maintain γδ T17 cells homeostasis in mouse skin, and dysbiosis of the microbiome increases the secretion of IL-1β, leading to γδ T17 cells expansion, which induce dermal inflammation.106 Madecassic acid can inhibit γδ T17 cells activation through PPARγ–pTEN/Akt/GSK3β/NFAT pathway and contributes to the improvement of colitis.107

Both Th17 cells and γδ T17 cells produce IL-17,108 but the 2 cells differ in many ways (Table 3). Th17 cells and γδ T17 cells express different types of TCR, namely αβTCR or γδTCR, which are composed of different groups of somatic rearrangement TCR chains and CD3 subunits.109 The antigen recognition and differentiation requirements of γδ T17 cells are different from those of Th17 cells. Th17 cells development and function depend on αβTCR recognition of antigenic peptides presented by MHC proteins. Upon recognition of the peptide–MHC complex, αβT cells differentiate into effector Th17 cells, producing the cytokine IL-17 that activates innate immune cells or B cells, thereby preventing invasion of pathogens.110 However, TCR γδ can recognize and bind antigen molecules directly without binding MHC molecules or APC.

TABLE 3

Comparison of Th17 cells and γδT17 cells

 Th17γδT17
TCR MHC
Markers
Transcription factors Proximal regulators Effector molecules
Location Main targets
TCRαβ+T MHC
CD3+, CD4+, IL-23R+, CCR6+ and IL-1R+
RORγt, RORa, STAT3
TGF-β, IL-1β, IL-21, IL-6 and IL-23
IL-17A, IL-17F, IL-21, IL-22, IL-26, TNF-α, CCL20 and GM-CSF
Mucosal surface
Infection, autoimmune diseases, transplant rejection and tumors
TCRγδ+T
Nonclassical MHC
CD3+, IL-7Rαhigh, IL-18Rhigh CCR6+ and
CD161+ RORγt, STAT3
IL-2, IL-23, IL-1β, IL-7 and TGF-β
IL-17A, IL-17F, IL-22, IL-8, IL-21, TNF-α
and GM-CSF
Mucosa and subcutaneous tissue Infection, autoimmune diseases, tumors,
inflammation the central nervous system and psoriasis
 Th17γδT17
TCR MHC
Markers
Transcription factors Proximal regulators Effector molecules
Location Main targets
TCRαβ+T MHC
CD3+, CD4+, IL-23R+, CCR6+ and IL-1R+
RORγt, RORa, STAT3
TGF-β, IL-1β, IL-21, IL-6 and IL-23
IL-17A, IL-17F, IL-21, IL-22, IL-26, TNF-α, CCL20 and GM-CSF
Mucosal surface
Infection, autoimmune diseases, transplant rejection and tumors
TCRγδ+T
Nonclassical MHC
CD3+, IL-7Rαhigh, IL-18Rhigh CCR6+ and
CD161+ RORγt, STAT3
IL-2, IL-23, IL-1β, IL-7 and TGF-β
IL-17A, IL-17F, IL-22, IL-8, IL-21, TNF-α
and GM-CSF
Mucosa and subcutaneous tissue Infection, autoimmune diseases, tumors,
inflammation the central nervous system and psoriasis
TABLE 3

Comparison of Th17 cells and γδT17 cells

 Th17γδT17
TCR MHC
Markers
Transcription factors Proximal regulators Effector molecules
Location Main targets
TCRαβ+T MHC
CD3+, CD4+, IL-23R+, CCR6+ and IL-1R+
RORγt, RORa, STAT3
TGF-β, IL-1β, IL-21, IL-6 and IL-23
IL-17A, IL-17F, IL-21, IL-22, IL-26, TNF-α, CCL20 and GM-CSF
Mucosal surface
Infection, autoimmune diseases, transplant rejection and tumors
TCRγδ+T
Nonclassical MHC
CD3+, IL-7Rαhigh, IL-18Rhigh CCR6+ and
CD161+ RORγt, STAT3
IL-2, IL-23, IL-1β, IL-7 and TGF-β
IL-17A, IL-17F, IL-22, IL-8, IL-21, TNF-α
and GM-CSF
Mucosa and subcutaneous tissue Infection, autoimmune diseases, tumors,
inflammation the central nervous system and psoriasis
 Th17γδT17
TCR MHC
Markers
Transcription factors Proximal regulators Effector molecules
Location Main targets
TCRαβ+T MHC
CD3+, CD4+, IL-23R+, CCR6+ and IL-1R+
RORγt, RORa, STAT3
TGF-β, IL-1β, IL-21, IL-6 and IL-23
IL-17A, IL-17F, IL-21, IL-22, IL-26, TNF-α, CCL20 and GM-CSF
Mucosal surface
Infection, autoimmune diseases, transplant rejection and tumors
TCRγδ+T
Nonclassical MHC
CD3+, IL-7Rαhigh, IL-18Rhigh CCR6+ and
CD161+ RORγt, STAT3
IL-2, IL-23, IL-1β, IL-7 and TGF-β
IL-17A, IL-17F, IL-22, IL-8, IL-21, TNF-α
and GM-CSF
Mucosa and subcutaneous tissue Infection, autoimmune diseases, tumors,
inflammation the central nervous system and psoriasis

ROLE OF γδ T17 CELLS IN CVD

Myocardial infarction

Myocardial infarction (MI) is a subset of a series of acute coronary syndromes. Plaque or thrombosis results in reduced myocardial perfusion, then occurs MI.111,112 Tissue injury after MI leads to secondary inflammation; there are many immune cells involved, including γδ T cells, macrophages, and neutrophils.113–115 Beyond that, these immune cells also participate in ventricular remodeling.116 It has been reported that almost 90% of the IL-17A-secreting leukocytes were T cells, including approximately 75% γδ T cells and 10% CD4+ T cells on MI, which proved that γδ T cells were the major source of IL-17A in the infarct of the heart.117 By analyzing the clinical blood sample of patients with MI, it was found that compared with normal subjects, restrictive expression of TCR γδ repertoire and alteration expression of IL-17A gene are the important characteristics of γδ T cells in MI patients.118

In mouse, γδ T17 cells has the effects of promoting apoptosis, fibrosis, or inflammation in ventricular remodeling after MI (Figure 2). The apoptotic cardiomyocytes and damaged matrix will release “danger signal” molecules called damage-associated molecular pattern (DAMP),119 such as mitochondrial DNA, F-actin, heat-shock protein, peroxiredoxins, and so on.120–122 TLRs on the surfaces of macrophages, neutrophils, and dendritic cells respond to these DAMP.123–125 These cells were stimulated to release IL-23 and IL-1β. IL-23 is an indispensable upstream regulator of IL-17A production in γδ T cells and can recruit γδ T cells to damaged regions.126 IL-1β be produced by caspase-1 and inflammatory complex, it can work in concert with IL-23 to drive γδ T cells secrete IL-17A. This is IL-23/IL-17 axis.24,99,127,128 Some studies have shown that IL-1 receptor blockers play a protective role in post-MI heart.129,130 Importantly, production of IL-17A by γδ T cells can increase the infarct size and myocardial fibrosis after MI.131 In vitro, IL-17A produced by γδ T17 cells can promote cardiomyocyte apoptosis by regulating caspase-3 activity, pro- to antiapoptotic protein (Bax/Bcl-2) ratio, induce neutrophil migration, and directly activate the p38 MAPK-p53-Bax signaling pathway.117 By contrast, genetic IL-17A deficiency had the opposite effect.117,132

γδ T17 cells participate in the injury process of heart disease. In MI, damaged cardiomyocytes release signaling molecules, which are recognized by TLRs on macrophages and dendritic cells, stimulating the release of IL-23 and active IL-1β, act synergistically to drive IL-17A production in γδ T cells, which also act synergistically with IL-23 in direct response to TLR stimulation. Activated γδ T17 cells can produce cytokines such as IL-17A and IL-22. IL-17RA is expressed on the surface of fibroblasts and macrophages in the heart. On the one hand, it can stabilize the chemotaxis of CXCL1 and other cells in fibroblasts. The expression of the factor, CXCL1, can recruit peripheral neutrophils to the injured myocardium, and neutrophils release reactive oxygen species (ROS) and proteolytic enzymes, thereby causing damage to the cardiomyocytes. On the other hand, IL-17A enhanced M1 macrophage polarization and directly activated macrophages to express TNF-α, IL-6, and IL-1β, further aggravating myocardial injury.
FIGURE 2

γδ T17 cells participate in the injury process of heart disease. In MI, damaged cardiomyocytes release signaling molecules, which are recognized by TLRs on macrophages and dendritic cells, stimulating the release of IL-23 and active IL-1β, act synergistically to drive IL-17A production in γδ T cells, which also act synergistically with IL-23 in direct response to TLR stimulation. Activated γδ T17 cells can produce cytokines such as IL-17A and IL-22. IL-17RA is expressed on the surface of fibroblasts and macrophages in the heart. On the one hand, it can stabilize the chemotaxis of CXCL1 and other cells in fibroblasts. The expression of the factor, CXCL1, can recruit peripheral neutrophils to the injured myocardium, and neutrophils release reactive oxygen species (ROS) and proteolytic enzymes, thereby causing damage to the cardiomyocytes. On the other hand, IL-17A enhanced M1 macrophage polarization and directly activated macrophages to express TNF-α, IL-6, and IL-1β, further aggravating myocardial injury.

In addition, IL-17A promote the expression of CXCL1 in cardiac fibroblasts, CXCL1 leads to infiltration of neutrophils. These neutrophils release proteolytic enzymes or reactive oxygen species to destroy surrounding muscle cells.102,131,133 And anti-IL-17A antibody can reduce the mouse cardiomyocytes death, which is cultured in low serum and hypoxia.102 IL-17A can also enhance polarization of macrophages toward M1 and directly activated the expression of TNF-α, IL-6, IL-1β, and matrix metalloproteinases (MMPs), which further aggravated myocardial injury.134–136 So, the myocardial interstitial fibrosis and increased production of MMP associated with persistent inflammation contribute to subsequent adverse remodeling.135

Besides, IL-17A increased the proliferation of mouse cardiac fibroblasts in a dose-dependent manner in vitro. One week after MI, fibroblasts selected from IL-17A-KO mice expressed lower levels of fibrosis-related genes such as collagen 1, collagen 3, periostin, and TGF-β than those of wild-type (WT) mice. The survival rate of TCR γδ-KO mice in the later stage of MI was significantly higher than that of WT mice, the lack of γδ T cells limited infarct expansion in myocardium.117,131And the number of neutrophils, macrophages, and T cells in the infarcted heart of TCR γδ-KO mice rather than IL-17A-KO mice decreased.131 The result implicates the functional significance of mediating damage by cardiac γδ T cells after MI.

In terms of γδ T cell migration, except IL-23, the sphingosine-1-phosphate receptor helps to mediate γδ T cells egress from lymph nodes,137,138 and CCL20 acts as a chemokine can recruit γδ T cells to the sites of damaged myocardium.131

Atherosclerosis

Atherosclerosis (AS) is a chronic inflammatory disease of large and medium-sized arterial walls, caused by elevated levels of low-density lipoprotein and cholesterol in the blood.139 AS is a complex disease in which blood vessels, metabolism, and immune system are involved. One of the pathologic manifestation of AS is the formation of plaque. Arterial plaques are characterized by lipid accumulation in the arterial wall and infiltration of immune cells140–143 (Figure 3).

γδ T17 cells participate in vascular disease injury process. γδ T17 cells in arteries are mainly recruited to the damaged site by CXCL16 and CCL20 released by damaged vascular endothelial cells (ECs). Second, the ligand CXCR6 of CXCL16 is also expressed on the surface of macrophages. Activated γδ T17 cells recruit macrophages and neutrophils in peripheral blood by releasing IL-17A, GM-CSF, and G-CSF and aggravate vascular damage. Third, endothelial cells stabilize recruited immune cells by releasing ICAM-1
FIGURE 3

γδ T17 cells participate in vascular disease injury process. γδ T17 cells in arteries are mainly recruited to the damaged site by CXCL16 and CCL20 released by damaged vascular endothelial cells (ECs). Second, the ligand CXCR6 of CXCL16 is also expressed on the surface of macrophages. Activated γδ T17 cells recruit macrophages and neutrophils in peripheral blood by releasing IL-17A, GM-CSF, and G-CSF and aggravate vascular damage. Third, endothelial cells stabilize recruited immune cells by releasing ICAM-1

T cells proliferation is closely related to the regulation of intracellular cholesterol level and cholesterol metabolism-related genes.144,145 Lipid rafts and raft-related proteins can activate TCR signaling pathway and enhance ERK1/2 phosphorylation, which leads to T cells activation and proliferation; this regulatory function is also applicable to γδ T cells.146,147 In apolipoprotein E-deficient (ApoE KO) mice, the number of T cells expressing IL-17A was significantly increased; γδ T17 cells are the majority of these cells. Through the mouse model of human familial hypercholesterolemia, IL-17+ cells were found in the root of the model mice aorta.148 Inhibition of IL-17 expression or signal transduction will decrease the formation of aortic lesions and leukocyte accumulation149,150; it is suggested that γδ T cells can regulate AS through the production of IL-17A.24

In the early stage of AS, γδ T17 cells were the main T cell subsets of aortic root and aortic arch lesions in ApoE KO mice and were mainly distributed in the aortic adventitia.151 ApoE/γδ T Double KO mice showed that γδ T cell deficiency reduced aortic root and arch lesion size in Western diet-fed mice. Similar to the role of IL-23/IL-17 axis in MI, IL-23R+ γδ T cells can locally promote the formation of early atherosclerotic lesions, plaque fall off, and inflammation in aortic roots.152 In fact, after γδ T17 cells are activated,153 the spread of inflammatory response depends on the effect of the IL-17 produced by γδ T17 cells on other immune cells and epithelial cells. IL-17 produced by γδ T cells induces the secretion of proinflammatory cytokines (such as IL-6, TNF-α, and G-CSF) and chemokine (CXCL1, CCL2, CXCL8, and CXCL2) through multiple pathways.18,154,155 For example, IL-17A supports the production of IL-6 and IL-8 and the chemokines CXCL1, CCL2, CCL5, CXCL12, and CXCL10 in several cell types, including endothelial, vascular smooth muscle cells (VSMCs),156,157 fibroblasts, and epithelial cells.154,158 Among them, CXCL1 can promote the recruitment of monocytes to the arterial wall during AS, resulting in a pro-∖inflammatory effect.159 And CXCR4 and its ligand CXCL12 are involved in neutrophil excretion from the bone marrow and neutrophil recruitment to atherosclerotic lesions.160

In addition, IL-17A regulates levels of G-CSF, which affects the number of neutrophils in the blood. The neutrophil infiltration was positively correlated with the degree of early aortic lesion progression in the aortic root of ApoE KO mice, and neutrophil depletion reduced lesion progression.151 And the adenovirus-mediated blockade of IL-17A resulted in decreased levels of IL-6 and G-CSF plasma and reduced the recruitment of neutrophils and macrophages in the aorta, that alleviates the damage. Therefore, γδ T cells can promote the progression of new lesions and increase neutrophils in the process of early AS.160,161

Aortic aneurysm

Aortic aneurysm (AA) is a vascular disease, which can occur anywhere in the aorta. Among them, abdominal aortic aneurysm (AAA) is more common than other aneurysms.162 AAA is characterized by hypoxia and inflammation caused by apoptosis of VSMCs.163,164 However, in addition to VSMCs, other cells are also involved in the occurrence and development of AAA, including γδ T cells165 and other immune cells.166–169 And some studies show that, CXCR5+ γδ T has more effect functions in AAs increased.170,171

The current study found that γδ T17 cells are the main source of IL-17A in AAA patients.171 Analysis of the tumor tissue of AAA patients showed that γδ T cells were involved in the inflammatory response, and IL-17A could induce the production of inflammatory cytokines, chemokines, and nitric oxide.172 Related inflammatory cytokines can further activate MMP, such as MMP-1,173 MMP-2,174 MMP-9,175 MMP-12,176 and cathepsin production.177 These factors can promote inflammation and apoptosis in the aortic wall and can further damage the smooth muscle cells leading to aneurysm expansion and rupture.

A previous study found that dilatation and rupture of AAs are closely related to hypertension, while IL-17A derived from γδ T17 cells mediates hypertension and hypertensive heart injury.178 This also indirectly provides a link between γδ T cells and AA development. In addition, γδ T cells can increase apoptosis of cells that the aortic wall by inhibiting genes involved in phosphoinositide 3-kinase/AKT signaling pathway and the proliferation phase (Sos1, Mtor, Myc), whereas apoptosis-related genes (Pten, Bcl1, Bad) were up-regulated.179 But the inflammatory cytokines including IL-1β, Mcp-1, and TNF-α were down-regulated in the aneurysm tissues of γδ T KO mice.179 And the expression of inflammation-related genes such as IL19, Nlrp3, Spp1, and IL3 down-regulates and then affects the production of related inflammatory cytokines.180–184

Ischemia–reperfusion injury

Ischemia–reperfusion injury (IRI) mostly occurs in clinical, such as organ transplantation, resection, trauma, and septic, or hemorrhagic shock and leads to significant mortality.185,186 Immune cells in the ischemia–reperfusion area release various cytokines, thereby triggering a violent inflammatory response and ultimately aggravating tissue damage. The latest research proves the γδ T17 cells play an important role in the initial stage of immune response in IRI.187

γδ T17 cells are considered to be the main source of IL-17A in myocardial ischemia–reperfusion.188 IL-17A induce cardiomyocyte apoptosis, neutrophil infiltration, and macrophage migration in myocardial injury.102,189 The apoptotic cells cause damage by releasing oxygen-free radicals, cytokines, and recruiting granulocytes in IRI.190–192 The exogenous long pentraxin-3, a member of a phylogenetically conserved group of acute-phase reactants that are involved in inflammation and innate immunity, can improve cardiomyocyte apoptosis and infiltration of neutrophils and macrophages by inhibiting the expansion of γδ T cells and reducing the expression of IL-23 and IL-17A. It plays a protective role in the IRI of cardiomyocytes.193 NKG2D, are activating or coactivating receptor on γδ T cells, stimulating γδ T cells to secrete cytokines. And the blockade of NKG2D will also reduce the damage caused by reperfusion.188 The above indirectly proves the damage effect of γδ T17 cells in myocardial ischemia–reperfusion.

Importantly, recent studies in other IRI models also show that γδ T17 cells have a vital contribution to IRI.65,66,194–196 For example, γδ T cells can be considered as mediators promoting the inflammatory response in the acute stage of intestinal IRI. Selective depletion of γδ T cells after IRI significantly reduced proinflammatory cytokine levels and neutrophil infiltration in the intestinal tract compared with the control group.196 In addition, γδ T KO delayed inflammatory reaction in early renal IRI, but with the extension of reperfusion time, the survival rate of knockout mice decreased.194,197,198

Other CVDs

In addition to the 4 types mentioned above, γδ T17 cells also promote inflammation in other types of CVD, such as stroke, hypertensive heart disease, and viral myocarditis. Stroke is an acute CVD. γδ T17 cells are rapid inflammatory effector cells involved in the delayed period of cerebral ischemia (day 1–4).199,200 IL-17 from γδ T cells acts on macrophages and brain cells directly and promotes the expression of inflammatory mediators that enhance apoptotic neuronal cell death and blood–brain barrier breakdown.201 γδ T17 cells mediate cardiac damage and fibrosis in hypertensive patients. γδ T17 cells secrete IL-17A and accelerate the differentiation of myofibroblasts by promoting cardiac fibroblasts to produce IL-6178. Early activation of γδ T17 cells has positive effect in viral myocarditis by promoting neutrophil infiltration and Th17 induction.48

DISCUSSION ABOUT THE ROLE OF γδ T17 CELLS IN OTHER DISEASES

Not limited to CVD, γδ T17 cells function applies to the whole body. γδ T17 cells have been shown to be involved in a variety of acute injuries, chronic inflammations, and tumors. γδ T17 cells mainly plays a protective role in infection202,203 and plays a pathogenic factor in inflammatory diseases, including arthritis,204,205 colitis,107 diabetes,206 and psoriasis,106,207–209 which has been reviewed and reported. In a variety of murine models, γδ T17 cells have been shown to be critical in fungal,210 bacterial,211 and viral infections,212 primarily through their ability to produce IL-17 and activate epithelial cells and recruit innate cells at the site of infection. In humans, although direct evidence is lacking, studies of loss of function mutations suggest that γδ T17 cells may be important for clearing fungal213,214 and extracellular bacterial infections.215 Psoriasis is a chronic inflammatory skin disease characterized by abnormal proliferation and differentiation of keratinocytes and an influx of inflammatory cells.216 Dysregulation of the microbiome up-regulated IL-1β, resulting in the amplification of γδ T17 cells and skin inflammation. IL-1β can directly activate γδ T17 cells in mice and also stimulate keratinocytes to secrete chemokines, thus attracting more IL-17-producing cells from the periphery, thus establishing an amplified inflammatory response, leading to skin inflammation, which is the pathogenesis of psoriasis.106

However, γδ T17 cells play a dual role in tumor growth. On the one hand, the tumor-promoting function of γδ T17 cells has been demonstrated in various cancer clinical studies and animal models, such as oral cancer,217 human colorectal cancer,104 and Helicobacter felis-infected mouse models of gastric MALT lymphoma.218 Activated γδ T17 cells secrete a large number of cytokines, including IL-17, IL-8, and GM-CSF, which may mobilize and recruit polymorphonuclear myeloid suppressor cells in tumors to establish an effective immunosuppressive environment that promotes tumor progression and immune avoidance.104,219 In multiple tumor models, tumor-infiltrating γδ T17 cells promote tumor growth by supporting angiogenesis.220–223 In addition to promoting angiogenesis, γδ T17 cells also promote tumor growth by inhibiting cytotoxic T cells.224,225 On the other hand, the antitumor function of γδ T17 cells has also been reported. Aging induces γδ T cells to produce IL-17A, which plays a key role in the development of resistance to lung melanoma in elderly mice.226 Infiltration of γδ T17 cells was observed in epithelial tumors after chemotherapy. γδ T17 cells enhanced recruitment of IFN-γ-producing CD8+ T cells, which mediated antitumor function.227 In addition, γδ T17 cells in human malignant pleural effusion secrete more IL-17A than peripheral blood, thereby improved patient survival, suggesting an antitumor role of γδ T17 cells in human cancer.85 Because of the plasticity of γδ T17 cells and the complexity of the tumor microenvironment, the role of γδ T17 cells in tumors is controversial and needs further study.

CLOSING REMARKS

CVD is a general term for vascular disease and heart disease. In recent years, researchers have paid attention to the important role of γδ T cells in disease progression. We summed up that γδ T17 cells predominate in CVD, such as MI, AS, AA, and IRI. In these diseases, γδ T17 cells recruit other immune cells, such as neutrophils or macrophages, by secreting cytokines and chemokines that mediate heart or blood vessel damage.

In previous studies, immunotherapy about γδ T17 cells in CVD treatment was not found. However, there are many types of immunotherapies for CVD, such as therapeutic strategies targeting cytokines, chemokines and their receptors, and immune checkpoints. Anti-IL-1β antibody treatment is currently the only immunotherapy available to treat CVD. Anti-IL-1β therapy improved primary and secondary CVD outcomes.228 Blocking of CCR2 with MLN1202, a highly specific humanized mAb that inhibits CCL2 binding, reduced serum C-reactive protein levels in patients with risk factors for CVD in a phase 2 trial.229 The CD80/86-CD28 and CD80/86-CTLA4 immune checkpoint proteins are pivotal regulators during AS. Inhibition of B7-1 (CD80) by RhuDex® reduces LPS-mediated inflammation in human atherosclerotic lesions.230

By summarizing the pathogenesis of γδ T17 cells in CVD, it may provide some enlightenment for immunotherapy of CVD, including: (1) mAbs inhibit γδ T17 cell-associated chemokines and their receptors; (2) inhibitor blocking checkpoints in γδ T17 cells; (3) drugs modulate γδ T17 cell-related signaling pathways to ameliorate CVD; (4) miRNA inhibits the synthesis of cytokines or chemokines, such as IL-17 and CCL20.

In conclusion, this review provides a more comprehensive understanding of the biology of γδ T17 cells and their role in CVD, which may be the key to effective immunotherapy.

AUTHORSHIP

S. L. and Z. Y. wrote manuscript and designed figures. Y. L. completed the writing of the classification of γδ T cells. J. Z. and S. Z. completed the writing of the differentiation of γδ T17 cells. X. C. and Y. H. completed the writing of introduction and summary. All authors contributed to the article and approved the submitted version. S. L. and Z. Y. contributed equally to this work.

ACKNOWLEDGMENTS

This work was supported by The National Natural Science Foundation of China (No. 81873100).

DISCLOSURES

The authors declare no conflict of interest.

REFERENCES

1

Zhao
 
D
,
Liu
 
J
,
Wang
 
M
 et al.  
Epidemiology of cardiovascular disease in China: current features and implications
.
Nat Rev Cardiol
.
2019
;
16
:
203
212
.

2

Nichols
 
M
,
Townsend
 
N
,
Scarborough
 
P
 et al.  
Cardiovascular disease in Europe 2014: epidemiological update
.
Eur Heart J
.
2014
;
35
:
2950
2959
.

3

Andersson
 
C
,
Vasan
 
RS
.
Epidemiology of cardiovascular disease in young individuals
.
Nat Rev Cardiol
.
2018
;
15
:
230
240
.

4

Carnethon
 
MR
,
Pu
 
J
,
Howard
 
G
 et al.  
Cardiovascular health in African Americans: a scientific statement from the American Heart Association
.
Circulation
.
2017
;
136
:
e393
e423
.

5

Zernecke
 
A
,
Bernhagen
 
J
,
Weber
 
C
.
Macrophage migration inhibitory factor in cardiovascular disease
.
Circulation
.
2008
;
117
:
1594
1602
.

6

Passos
 
LSA
,
Lupieri
 
A
,
Becker-Greene
 
D
 et al.  
Innate and adaptive immunity in cardiovascular calcification
.
Atherosclerosis
.
2020
;
306
:
59
67
.

7

Carding
 
SR
,
Egan
 
PJ
.
Gammadelta T cells: functional plasticity and heterogeneity
.
Nat Rev Immunol
.
2002
;
2
:
336
345
.

8

Hayday
 
AC
.
Gammadelta T cells and the lymphoid stress-surveillance response
.
Immunity
.
2009
;
31
:
184
196
.

9

Cua
 
DJ
,
Tato
 
CM
.
Innate IL-17-producing cells: the sentinels of the immune system
.
Nat Rev Immunol
.
2010
;
10
:
479
489
.

10

Li
 
Y
,
Li
 
G
,
Zhang
 
J
 et al.  
The dual roles of human γδ T cells: anti-tumor or tumor-promoting
.
Front Immunol
.
2021
;
11
:619954.

11

Wu
 
YL
,
Ding
 
YP
,
Tanaka
 
Y
 et al.  
γδ T cells and their potential for immunotherapy
.
Int J Biol Sci
.
2014
;
10
:
119
135
.

12

Melandri
 
D
,
Zlatareva
 
I
,
Chaleil
 
RAG
 et al.  
The γδTCR combines innate immunity with adaptive immunity by utilizing spatially distinct regions for agonist selection and antigen responsiveness
.
Nat Immunol
.
2018
;
19
:
1352
1365
.

13

Khan
 
MW
,
Curbishley
 
SM
,
Chen
 
HC
 et al.  
Expanded human blood-derived γδ T cells display potent antigen-presentation functions
.
Front Immunol
.
2014
;
5
:
344
.

14

Muñoz-Ruiz
 
M
,
Ribot
 
JC
,
Grosso
 
AR
 et al.  
TCR signal strength controls thymic differentiation of discrete proinflammatory γδ T cell subsets
.
Nat Immunol
.
2016
;
17
:
721
727
.

15

Wan
 
F
,
Yan
 
K
,
Xu
 
D
 et al.  
Vγ1+γδT, early cardiac infiltrated innate population dominantly producing IL-4, protect mice against CVB3 myocarditis by modulating IFNγ+ T response
.
Mol Immunol
.
2017
;
81
:
16
25
.

16

Yokozeki
 
H
,
Watanabe
 
K
,
Igawa
 
K
 et al.  
Gammadelta T cells assist alphabeta T cells in the adoptive transfer of contact hypersensitivity to para-phenylenediamine
.
Clin Exp Immunol
.
2001
;
125
:
351
359
.

17

Fan
 
DX
,
Duan
 
J
,
Li
 
MQ
 et al.  
The decidual gamma-delta T cells up-regulate the biological functions of trophoblasts via IL-10 secretion in early human pregnancy
.
Clin Immunol
.
2011
;
141
:
284
292
.

18

McGeachy
 
MJ
,
Cua
 
DJ
,
Gaffen
 
SL
.
The IL-17 family of cytokines in health and disease
.
Immunity
.
2019
;
50
:
892
906
.

19

McKenzie
 
DR
,
Comerford
 
I
,
Silva-Santos
 
B
 et al.  
The emerging complexity of γδT17 cells
.
Front Immunol
.
2018
;
9
:
796
.

20

Li
 
F
,
Hao
 
X
,
Chen
 
Y
 et al.  
The microbiota maintain homeostasis of liver-resident γδT-17 cells in a lipid antigen/CD1d-dependent manner
.
Nat Commun
.
2017
;
7
:13839.

21

Fan
 
Q
,
Tao
 
R
,
Zhang
 
H
 et al.  
Dectin-1 contributes to myocardial ischemia/reperfusion injury by regulating macrophage polarization and neutrophil infiltration
.
Circulation
.
2019
;
139
:
663
678
.

22

O'Brien
 
RL
,
Born
 
WK
.
Two functionally distinct subsets of IL-17 producing γδ T cells
.
Immunol Rev
.
2020
;
298
:
10
24
.

23

Barros-Martins
 
J
,
Schmolka
 
N
,
Fontinha
 
D
 et al.  
Effector γδ T cell differentiation relies on master but not auxiliary Th cell transcription factors
.
J Immunol
.
2016
;
196
:
3642
3652
.

24

Sutton
 
CE
,
Lalor
 
SJ
,
Sweeney
 
CM
 et al.  
Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity
.
Immunity
.
2009
;
31
:
331
341
.

25

Adams
 
EJ
,
Gu
 
S
,
Luoma
 
AM
.
Human gamma delta T cells: evolution and ligand recognition
.
Cell Immunol
.
2015
;
296
:
31
40
.

26

Qi
 
C
,
Wang
 
Y
,
Li
 
P
 et al.  
Gamma delta T cells and their pathogenic role in psoriasis
.
Front Immunol
.
2021
;
12
:627139.

27

Heilig
 
JS
,
Tonegawa
 
S
.
Diversity of murine gamma genes and expression in fetal and adult T lymphocytes
.
Nature
.
1986
;
322
:
836
840
.

28

Muñoz-Ruiz
 
M
,
Sumaria
 
N
,
Pennington
 
DJ
,
Silva-Santos
 
B
.
Thymic determinants of γδ T cell differentiation
.
Trends Immunol
.
2017
;
38
:
336
344
.

29

Mikulak
 
J
,
Oriolo
 
F
,
Bruni
 
E
 et al.  
NKp46-expressing human gut-resident intraepithelial Vδ1 T cell subpopulation exhibits high antitumor activity against colorectal cancer
.
JCI Insight
.
2019
;
4
:e125884.

30

Knight
 
A
,
Mackinnon
 
S
,
Lowdell
 
MW
.
Human Vdelta1 gamma-delta T cells exert potent specific cytotoxicity against primary multiple myeloma cells
.
Cytotherapy
.
2012
;
14
:
1110
1118
.

31

Siegers
 
GM
,
Dhamko
 
H
,
Wang
 
XH
 et al.  
Human Vδ1 γδ T cells expanded from peripheral blood exhibit specific cytotoxicity against B-cell chronic lymphocytic leukemia-derived cells
.
Cytotherapy
.
2011
;
13
:
753
764
.

32

Alnaggar
 
M
,
Xu
 
Y
,
Li
 
J
 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
.

33

Meraviglia
 
S
,
Eberl
 
M
,
Vermijlen
 
D
 et al.  
In vivo manipulation of Vgamma9Vdelta2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients
.
Clin Exp Immunol
.
2010
;
161
:
290
297
.

34

Di Carlo
 
E
,
Bocca
 
P
,
Emionite
 
L
 et al.  
Mechanisms of the antitumor activity of human Vγ9Vδ2 T cells in combination with zoledronic acid in a preclinical model of neuroblastoma
.
Mol Ther
.
2013
;
21
:
1034
1043
.

35

Petrasca
 
A
,
Melo
 
AM
,
Breen
 
EP
 et al.  
Human Vδ3+ γδ T cells induce maturation and IgM secretion by B cells
.
Immunol Lett
.
2018
;
196
:
126
134
.

36

Mangan
 
BA
,
Dunne
 
MR
,
O'Reilly
 
VP
 et al.  
Cutting edge: cD1d restriction and Th1/Th2/Th17 cytokine secretion by human Vδ3 T cells
.
J Immunol
.
2013
;
191
:
30
34
.

37

Zhao
 
Y
,
Niu
 
C
,
Cui
 
J
.
Gamma-delta (γδ) T cells: friend or foe in cancer development
.
J Transl Med
.
2018
;
16
:
3
.

38

Pereira
 
P
,
Berthault
 
C
,
Burlen-Defranoux
 
O
 et al.  
Critical role of TCR specificity in the development of Vγ1Vδ6.3+ innate NKTγδ cells
.
J Immunol
.
2013
;
191
:
1716
1723
.

39

Huber
 
SA
,
Born
 
W
,
O'Brien
 
R
.
Dual functions of murine gammadelta cells in inflammation and autoimmunity in coxsackievirus B3-induced myocarditis: role of Vgamma1+ and Vgamma4+ cells
.
Microbes Infect
.
2005
;
7
:
537
543
.

40

Tanaka
 
Y
,
Iwasaki
 
M
,
Murata-Hirai
 
K
 et al.  
Anti-tumor activity and immunotherapeutic potential of a bisphosphonate prodrug
.
Sci Rep
.
2017
;
7
:
5987
.

41

Spidale
 
NA
,
Sylvia
 
K
,
Narayan
 
K
 et al.  
Interleukin-17-producing γδ T cells originate from SOX13+ progenitors that are independent of γδTCR signaling
.
Immunity
.
2018
;
49
:
857
872
.

42

Jee
 
MH
,
Johansen
 
JD
,
Buus
 
TB
 et al.  
Increased production of IL-17A-producing γδ T cells in the thymus of filaggrin-deficient mice
.
Front Immunol
.
2018
;
9
:
988
.

43

Zheng
 
L
,
Hu
 
Y
,
Wang
 
Y
 et al.  
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
.

44

Taveirne
 
S
,
De Colvenaer
 
V
,
Van Den Broeck
 
T
 et al.  
Langerhans cells are not required for epidermal Vgamma3 T cell homeostasis and function
.
J Leukoc Biol
.
2011
;
90
:
61
68
.

45

Aono
 
A
,
Enomoto
 
H
,
Yoshida
 
N
 et al.  
Forced expression of terminal deoxynucleotidyl transferase in fetal thymus resulted in a decrease in gammadelta T cells and random dissemination of Vgamma3Vdelta1 T cells in skin of newborn but not adult mice
.
Immunology
.
2000
;
99
:
489
497
.

46

Sun
 
LD
,
Qiao
 
S
,
Wang
 
Y
 et al.  
Vγ4+ T cells: a novel IL-17-producing γδ T subsets during the early phase of chlamydial airway infection in mice
.
Mediators Inflamm
.
2018
;
2018
:6265746.

47

Li
 
Y
,
Wu
 
J
,
Luo
 
G
 et al.  
Functions of Vγ4 T cells and dendritic epidermal t cells on skin wound healing
.
Front Immunol
.
2018
;
9
:
1099
.

48

Yan
 
K
,
Yang
 
J
,
Qian
 
Q
 et al.  
Pathogenic role of an IL-23/γδT17/neutrophil axis in coxsackievirus B3-induced pancreatitis
.
J Immunol
.
2019
;
203
:
3301
3312
.

49

Holtmeier
 
W
,
Gille
 
J
,
Zeuzem
 
S
 et al.  
Distribution and development of the postnatal murine Vδ1 T-cell receptor repertoire
.
Immunology
.
2010
;
131
:
192
201
.

50

Binz
 
C
,
Bubke
 
A
,
Sandrock
 
I
 et al.  
αβ T cells replacing dermal and epidermal γδ T cells in Tcrd−/− mice express an MHC-independent TCR repertoire
.
Eur J Immunol
.
2021
;
51
:
2618
2632
.

51

Paget
 
C
,
Chow
 
MT
,
Gherardin
 
NA
 et al.  
CD3bright signals on γδ T cells identify IL-17A-producing Vγ6Vδ1+ T cells
.
Immunol Cell Biol
.
2015
;
93
:
198
212
.

52

Sun
 
X
,
Shibata
 
K
,
Yamada
 
H
 et al.  
CD30L/CD30 is critical for maintenance of IL-17A-producing γδ T cells bearing Vγ6 in mucosa-associated tissues in mice
.
Mucosal Immunol
.
2013
;
6
:
1191
1201
.

53

Lebrero-Fernández
 
C
,
Bergström
 
JH
,
Pelaseyed
 
T
 et al.  
Murine butyrophilin-like 1 and Btnl6 form heteromeric complexes in small intestinal epithelial cells and promote proliferation of local T lymphocytes
.
Front Immunol
.
2016
;
7
:
1
.

54

Daley
 
D
,
Zambirinis
 
CP
,
Seifert
 
L
 et al.  
γδ T cells support pancreatic oncogenesis by restraining αβ T cell activation
.
Cell
.
2016
;
166
:
1485
1499
.

55

McAllister
 
F
,
Bailey
 
JM
,
Alsina
 
J
 et al.  
Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia
.
Cancer Cell
.
2014
;
25
:
621
637
.

56

Wesch
 
D
,
Glatzel
 
A
,
Kabelitz
 
D
.
Differentiation of resting human peripheral blood gamma delta T cells toward Th1- or Th2-phenotype
.
Cell Immunol
.
2001
;
212
:
110
117
.

57

Ribot
 
JC
,
Ribeiro
 
ST
,
Correia
 
DV
 et al.  
Human γδ thymocytes are functionally immature and differentiate into cytotoxic type 1 effector T cells upon IL-2/IL-15 signaling
.
J Immunol
.
2014
;
192
:
2237
2243
.

58

Li
 
X
,
Wang
 
Z
,
Chang
 
L
 et al.  
γδ T cells contribute to type 2 inflammatory profiles in eosinophilic chronic rhinosinusitis with nasal polyps
.
Clin Sci (Lond)
.
2019
;
133
:
2301
2315
.

59

Chowdhury
 
AC
,
Chaurasia
 
S
,
Mishra
 
SK
 et al.  
IL-17 and IFN-γ producing NK and γδ-T cells are preferentially expanded in synovial fluid of patients with reactive arthritis and undifferentiated spondyloarthritis
.
Clin Immunol
.
2017
;
183
:
207
212
.

60

Ribot
 
JC
,
Neres
 
R
,
Zuzarte-Luís
 
V
 et al.  
γδ-T cells promote IFN-γ-dependent Plasmodium pathogenesis upon liver-stage infection
.
Proc Natl Acad Sci USA
.
2019
;
116
:
9979
9988
.

61

Rhodes
 
KA
,
Andrew
 
EM
,
Newton
 
DJ
 et al.  
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
.

62

Smith
 
E
,
Prasad
 
KM
,
Butcher
 
M
 et al.  
Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E-deficient mice
.
Circulation
.
2010
;
121
:
1746
1755
.

63

Chen
 
Y
,
Chou
 
K
,
Fuchs
 
E
 et al.  
Protection of the intestinal mucosa by intraepithelial gamma delta T cells
.
Proc Natl Acad Sci USA
.
2002
;
99
:
14338
14343
.

64

Haas
 
JD
,
González
 
FH
,
Schmitz
 
S
 et al.  
CCR6 and NK1.1 distinguish between IL-17A and IFN-gamma-producing gammadelta effector T cells
.
Eur J Immunol
.
2009
;
39
:
3488
3497
.

65

Shichita
 
T
,
Sugiyama
 
Y
,
Ooboshi
 
H
 et al.  
Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury
.
Nat Med
.
2009
;
15
:
946
950
.

66

Benakis
 
C
,
Brea
 
D
,
Caballero
 
S
 et al.  
Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells
.
Nat Med
.
2016
;
22
:
516
523
.

67

Caccamo
 
N
,
La Mendola
 
C
,
Orlando
 
V
 et al.  
Differentiation, phenotype, and function of interleukin-17-producing human Vγ9Vδ2 T cells
.
Blood
.
2011
;
118
:
129
138
.

68

Michel
 
ML
,
Pang
 
DJ
,
Haque
 
SF
 et al.  
IL-7) selectively promotes mouse and human IL-17-producing γδ cells
.
Proc Natl Acad Sci USA
.
2012
;
7
:
17549
17554
.

69

Moens
 
E
,
Brouwer
 
M
,
Dimova
 
T
 et al.  
IL-23R and TCR signaling drives the generation of neonatal Vgamma9Vdelta2 T cells expressing high levels of cytotoxic mediators and producing IFN-gamma and IL-17
.
J Leukoc Biol
.
2011
;
89
:
743
752
.

70

Haas
 
JD
,
Ravens
 
S
,
Düber
 
S
 et al.  
Development of interleukin-17-producing γδ T cells is restricted to a functional embryonic wave
.
Immunity
.
2012
;
37
:
48
59
.

71

Wencker
 
M
,
Turchinovich
 
G
,
Di
 
Marco
,
Barros
 
R
 et al.  
Innate-like T cells straddle innate and adaptive immunity by altering antigen-receptor responsiveness
.
Nat Immunol
.
2014
;
15
:
80
87
.

72

Papotto
 
PH
,
Gonçalves-Sousa
 
N
,
Schmolka
 
N
 et al.  
IL-23 drives differentiation of peripheral γδ17 T cells from adult bone marrow-derived precursors
.
EMBO Rep
.
2017
;
18
:
1957
1967
.

73

Do
 
JS
,
Fink
 
PJ
,
Li
 
L
 et al.  
Cutting edge: spontaneous development of IL-17-producing gamma delta T cells in the thymus occurs via a TGF-beta 1-dependent mechanism
.
J Immunol
.
2010
;
184
:
1675
1679
.

74

Hasegawa
 
E
,
Sonoda
 
KH
,
Shichita
 
T
 et al.  
IL-23-independent induction of IL-17 from γδT cells and innate lymphoid cells promotes experimental intraocular neovascularization
.
J Immunol
.
2013
;
190
:
1778
1787
.

75

Shitara
 
S
,
Hara
 
T
,
Liang
 
B
 et al.  
IL-7 produced by thymic epithelial cells plays a major role in the development of thymocytes and TCRγδ+ intraepithelial lymphocytes
.
J Immunol
.
2013
;
190
:
6173
6179
.

76

Maki
 
K
,
Sunaga
 
S
,
Komagata
 
Y
 et al.  
Interleukin 7 receptor-deficient mice lack gammadelta T cells
.
Proc Natl Acad Sci USA
.
1996
;
93
:
7172
7177
.

77

Maki
 
K
,
Sunaga
 
S
,
Ikuta
 
K
.
The V-J recombination of T cell receptor-gamma genes is blocked in interleukin-7 receptor-deficient mice
.
J Exp Med
.
1996
;
184
:
2423
2427
.

78

Corpuz
 
TM
,
Stolp
 
J
,
Kim
 
HO
 et al.  
Differential responsiveness of innate-like IL-17- and IFN-γ-producing γδ T cells to homeostatic cytokines
.
J Immunol
.
2016
;
196
:
645
654
.

79

Lochner
 
M
,
Peduto
 
L
,
Cherrier
 
M
 et al.  
In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORgamma t+ T cells
.
J Exp Med
.
2008
;
205
:
1381
1393
.

80

Colpitts
 
SL
,
Puddington
 
L
,
Lefrançois
 
L
.
IL-15 receptor α signaling constrains the development of IL-17-producing γδ T cells
.
Proc Natl Acad Sci USA
.
2015
;
112
:
9692
9697
.

81

Do
 
JS
,
Visperas
 
A
,
O'Brien
 
RL
 et al.  
CD4 T cells play important roles in maintaining IL-17-producing γδ T-cell subsets in naive animals
.
Immunol Cell Biol
.
2012
;
90
:
396
403
.

82

Gupta
 
PK
,
Wagner
 
SR
,
Wu
 
Q
 et al.  
Th17 cells are not required for maintenance of IL-17A-producing γδ T cells in vivo
.
Immunol Cell Biol
.
2017
;
95
:
280
286
.

83

Kisielow
 
J
,
Kopf
 
M
.
The origin and fate of γδ T cell subsets
.
Curr Opin Immunol
.
2013
;
25
:
181
188
.

84

Kumar
 
R
,
Theiss
 
AL
,
Venuprasad
 
K
.
RORγt protein modifications and IL-17-mediated inflammation
.
Trends Immunol
.
2021
;
42
:
1037
1050
.

85

Wei
 
XS
,
Pei
 
XB
,
Liu
 
YL
 et al.  
IL-17A-producing γδ T cells inhibit the formation of malignant pleural effusions
.
Am J Respir Cell Mol Biol
.
2019
;
61
:
174
184
.

86

Gray
 
EE
,
Ramírez-Valle
 
F
,
Xu
 
Y
 et al.  
Deficiency in IL-17-committed Vγ4(+) γδ T cells in a spontaneous Sox13-mutant CD45.1(+) congenic mouse substrain provides protection from dermatitis
.
Nat Immunol
.
2013
;
14
:
584
592
.

87

Malhotra
 
N
,
Narayan
 
K
,
Cho
 
OH
 et al.  
A network of high-mobility group box transcription factors programs innate interleukin-17 production
.
Immunity
.
2013
;
38
:
681
693
.

88

Turchinovich
 
G
,
Hayday
 
AC
.
Skint-1 identifies a common molecular mechanism for the development of interferon-γ-secreting versus interleukin-17-secreting γδ T cells
.
Immunity
.
2011
;
35
:
59
68
.

89

Laird
 
RM
,
Laky
 
K
,
Hayes
 
SM
.
Unexpected role for the B cell-specific Src family kinase B lymphoid kinase in the development of IL-17-producing γδ T cells
.
J Immunol
.
2010
;
185
:
6518
6527
.

90

Chen
 
Y
,
Ci
 
X
,
Gorentla
 
B
 et al.  
Differential requirement of RasGRP1 for γδ T cell development and activation
.
J Immunol
.
2012
;
189
:
61
71
.

91

Muro
 
R
,
Nitta
 
T
,
Nakano
 
K
 et al.  
γδTCR recruits the Syk/PI3K axis to drive proinflammatory differentiation program
.
J Clin Invest
.
2018
;
128
:
415
426
.

92

Sumaria
 
N
,
Martin
 
S
,
Pennington
 
DJ
.
Constrained TCRγδ-associated Syk activity engages PI3K to facilitate thymic development of IL-17A-secreting γδ T cells
.
Sci Signal
.
2021
;
14
:eabc5884.

93

Shibata
 
K
,
Yamada
 
H
,
Sato
 
T
 et al.  
Notch-Hes1 pathway is required for the development of IL-17-producing γδ T cells
.
Blood
.
2011
;
118
:
586
593
.

94

Nakamura
 
M
,
Shibata
 
K
,
Hatano
 
S
 et al.  
A genome-wide analysis identifies a notch-RBP-Jκ-IL-7Rα axis that controls IL-17-producing γδ T cell homeostasis in mice
.
J Immunol
.
2015
;
194
:
243
251
.

95

Powolny-Budnicka
 
I
,
Riemann
 
M
,
Tänzer
 
S
 et al.  
RelA and RelB transcription factors in distinct thymocyte populations control lymphotoxin-dependent interleukin-17 production in γδ T cells
.
Immunity
.
2011
;
34
:
364
374
.

96

Yang
 
Q
,
Liu
 
X
,
Liu
 
Q
 et al.  
Roles of mTORC1 and mTORC2 in controlling γδ T1 and γδ T17 differentiation and function
.
Cell Death Differ
.
2020
;
27
:
2248
2262
.

97

Fiala
 
GJ
,
Gomes
 
AQ
,
Silva-Santos
 
B
.
From thymus to periphery: molecular basis of effector γδ-T cell differentiation
.
Immunol Rev
.
2020
;
298
:
47
60
.

98

Ribot
 
JC
,
deBarros
 
A
,
Pang
 
DJ
 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
.

99

Lalor
 
SJ
,
Dungan
 
LS
,
Sutton
 
CE
 et al.  
Caspase-1-processed cytokines IL-1beta and IL-18 promote IL-17 production by gammadelta and CD4 T cells that mediate autoimmunity
.
J Immunol
.
2011
;
186
:
5738
5748
.

100

Maggi
 
L
,
Santarlasci
 
V
,
Capone
 
M
 et al.  
CD161 is a marker of all human IL-17-producing T-cell subsets and is induced by RORC
.
Eur J Immunol
.
2010
;
40
:
2174
2181
.

101

Fergusson
 
JR
,
Fleming
 
VM
,
Klenerman
 
P
.
CD161-expressing human T cells
.
Front Immunol
.
2011
;
2
:
36
.

102

Liao
 
YH
,
Xia
 
N
,
Zhou
 
SF
 et al.  
Interleukin-17A contributes to myocardial ischemia/reperfusion injury by regulating cardiomyocyte apoptosis and neutrophil infiltration
.
J Am Coll Cardiol
.
2012
;
59
:
420
429
.

103

Jouan
 
Y
,
Patin
 
EC
,
Hassane
 
M
 et al.  
Thymic program directing the functional development of γδT17 cells
.
Front Immunol
.
2018
;
9
:
981
.

104

Wu
 
P
,
Wu
 
D
,
Ni
 
C
 et al.  
γδT17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer
.
Immunity
.
2014
;
40
:
785
800
.

105

McKenzie
 
DR
,
Kara
 
EE
,
Bastow
 
CR
 et al.  
IL-17-producing γδ T cells switch migratory patterns between resting and activated states
.
Nat Commun
.
2017
;
8
:15632.

106

Cai
 
Y
,
Xue
 
F
,
Quan
 
C
 et al.  
A critical role of the IL-1β-IL-1R signaling pathway in skin inflammation and psoriasis pathogenesis
.
J Invest Dermatol
.
2019
;
139
:
146
156
.

107

Yun
 
X
,
Fang
 
Y
,
Lv
 
C
 et al.  
Inhibition of the activation of γδT17 cells through PPARγ-PTEN/Akt/GSK3β/NFAT pathway contributes to the anti-colitis effect of madecassic acid
.
Cell Death Dis
.
2020
;
11
:
752
.

108

Pappu
 
R
,
Ramirez-Carrozzi
 
V
,
Ota
 
N
 et al.  
The IL-17 family cytokines in immunity and disease
.
J Clin Immunol
.
2010
;
30
:
185
195
.

109

Muro
 
R
,
Takayanagi
 
H
,
Nitta
 
T
.
T cell receptor signaling for γδT cell development
.
Inflamm Regen
.
2019
;
39
:
6
.

110

Gaud
 
G
,
Lesourne
 
R
,
Love
 
PE
.
Regulatory mechanisms in T cell receptor signalling
.
Nat Rev Immunol
.
2018
;
18
:
485
497
.

111

Thygesen
 
K
,
Alpert
 
JS
,
Jaffe
 
AS
 et al.  
Fourth universal definition of myocardial infarction
.
Circulation
.
2018
;
138
:
e618
e651
.

112

Boateng
 
S
,
Sanborn
 
T
.
Acute myocardial infarction
.
Dis Mon
.
2013
;
59
:
83
96
.

113

Sreejit
 
G
,
Abdel-Latif
 
A
,
Athmanathan
 
B
 et al.  
Neutrophil-derived S100A8/A9 amplify granulopoiesis after myocardial infarction
.
Circulation
.
2020
;
141
:
1080
1094
.

114

Alonso-Herranz
 
L
,
Sahún-Español
 
Á
,
Paredes
 
A
 et al.  
Macrophages promote endothelial-to-mesenchymal transition via MT1-MMP/TGFβ1 after myocardial infarction
.
Elife
.
2020
;
9
:e57920.

115

Ramos
 
G
,
Hofmann
 
U
,
Frantz
 
S
.
Myocardial fibrosis seen through the lenses of T-cell biology
.
J Mol Cell Cardiol
.
2016
;
92
:
41
45
.

116

Nian
 
M
,
Lee
 
P
,
Khaper
 
N
 et al.  
Inflammatory cytokines and postmyocardial infarction remodeling
.
Circ Res
.
2004
;
94
:
1543
1553
.

117

Zhou
 
SF
,
Yuan
 
J
,
Liao
 
MY
 et al.  
IL-17A promotes ventricular remodeling after myocardial infarction
.
J Mol Med (Berl)
.
2014
;
92
:
1105
1116
.

118

Chen
 
XM
,
Zhang
 
T
,
Qiu
 
D
 et al.  
Gene expression pattern of TCR repertoire and alteration expression of IL-17A gene of γδ T cells in patients with acute myocardial infarction
.
J Transl Med
.
2018
;
16
:
189
.

119

Silvis
 
MJM
,
Kaffka Genaamd Dengler
 
SE
,
Odille
 
CA
 et al.  
Damage-associated molecular patterns in myocardial infarction and heart transplantation: the road to translational success
.
Front Immunol
.
2020
;
11
:599511.

120

Shichita
 
T
,
Hasegawa
 
E
,
Kimura
 
A
 et al.  
Peroxiredoxin family proteins are key initiators of post-ischemic inflammation in the brain
.
Nat Med
.
2012
;
18
:
911
917
.

121

Oka
 
T
,
Hikoso
 
S
,
Yamaguchi
 
O
 et al.  
Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure
.
Nature
.
2012
;
485
:
251
255
.

122

Ahrens
 
S
,
Zelenay
 
S
,
Sancho
 
D
 et al.  
F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells
.
Immunity
.
2012
;
36
:
635
645
.

123

Gong
 
T
,
Liu
 
L
,
Jiang
 
W
 et al.  
DAMP-sensing receptors in sterile inflammation and inflammatory diseases
.
Nat Rev Immunol
.
2020
;
20
:
95
112
.

124

Timmers
 
L
,
Sluijter
 
JP
,
van Keulen
 
JK
 et al.  
Toll-like receptor 4 mediates maladaptive left ventricular remodeling and impairs cardiac function after myocardial infarction
.
Circ Res
.
2008
;
102
:
257
264
.

125

Li
 
M
,
Carpio
 
DF
,
Zheng
 
Y
 et al.  
An essential role of the NF-kappa B/Toll-like receptor pathway in induction of inflammatory and tissue-repair gene expression by necrotic cells
.
J Immunol
.
2001
;
166
:
7128
7135
.

126

Muschaweckh
 
A
,
Petermann
 
F
,
Korn
 
T
.
IL-1β and IL-23 promote extrathymic commitment of CD27+CD122- γδ T cells to γδT17 cells
.
J Immunol
.
2017
;
199
:
2668
2679
.

127

Cai
 
Y
,
Shen
 
X
,
Ding
 
C
 et al.  
Pivotal role of dermal IL-17-producing γδ T cells in skin inflammation
.
Immunity
.
2011
;
35
:
596
610
.

128

Mezzaroma
 
E
,
Toldo
 
S
,
Farkas
 
D
 et al.  
The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse
.
Proc Natl Acad Sci USA
.
2011
;
108
:
19725
19730
.

129

Abbate
 
A
,
Van Tassell
 
BW
,
Seropian
 
IM
 et al.  
Interleukin-1beta modulation using a genetically engineered antibody prevents adverse cardiac remodelling following acute myocardial infarction in the mouse
.
Eur J Heart Fail
.
2010
;
12
:
319
322
.

130

Abbate
 
A
,
Salloum
 
FN
,
Vecile
 
E
 et al.  
Anakinra, a recombinant human interleukin-1 receptor antagonist, inhibits apoptosis in experimental acute myocardial infarction
.
Circulation
.
2008
;
117
:
2670
2683
.

131

Yan
 
X
,
Shichita
 
T
,
Katsumata
 
Y
 et al.  
Deleterious effect of the IL-23/IL-17A axis and γδT cells on left ventricular remodeling after myocardial infarction
.
J Am Heart Assoc
.
2012
;
1
:e004408.

132

Zhu
 
H
,
Li
 
J
,
Wang
 
S
 et al.  
Hmgb1-TLR4-IL-23-IL-17A axis promote ischemia-reperfusion injury in a cardiac transplantation model
.
Transplantation
.
2013
;
95
:
1448
1454
.

133

Li
 
L
,
Huang
 
L
,
Vergis
 
AL
 et al.  
IL-17 produced by neutrophils regulates IFN-gamma-mediated neutrophil migration in mouse kidney ischemia-reperfusion injury
.
J Clin Invest
.
2010
;
120
:
331
342
.

134

Dayan
 
V
,
Yannarelli
 
G
,
Billia
 
F
 et al.  
Mesenchymal stromal cells mediate a switch to alternatively activated monocytes/macrophages after acute myocardial infarction
.
Basic Res Cardiol
.
2011
;
106
:
1299
1310
.

135

Marchant
 
DJ
,
Boyd
 
JH
,
Lin
 
DC
 et al.  
Inflammation in myocardial diseases
.
Circ Res
.
2012
;
110
:
126
144
.

136

Hu
 
Y
,
Zhang
 
H
,
Lu
 
Y
 et al.  
Class A scavenger receptor attenuates myocardial infarction-induced cardiomyocyte necrosis through suppressing M1 macrophage subset polarization
.
Basic Res Cardiol
.
2011
;
106
:
1311
1328
.

137

Mandala
 
S
,
Hajdu
 
R
,
Bergstrom
 
J
 et al.  
Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists
.
Science
.
2002
;
296
:
346
349
.

138

Theilmeier
 
G
,
Schmidt
 
C
,
Herrmann
 
J
 et al.  
High-density lipoproteins and their constituent, sphingosine-1-phosphate, directly protect the heart against ischemia/reperfusion injury in vivo via the S1P3 lysophospholipid receptor
.
Circulation
.
2006
;
114
:
1403
1409
.

139

Weber
 
C
,
Noels
 
H
.
Atherosclerosis: current pathogenesis and therapeutic options
.
Nat Med
.
2011
;
17
:
1410
1422
.

140

Weber
 
C
,
Zernecke
 
A
,
Libby
 
P
.
The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models
.
Nat Rev Immunol
.
2008
;
8
:
802
815
.

141

Hansson
 
GK
,
Libby
 
P
.
The immune response in atherosclerosis: a double-edged sword
.
Nat Rev Immunol
.
2006
;
6
:
508
519
.

142

Hansson
 
GK
,
Hermansson
 
A
.
The immune system in atherosclerosis
.
Nat Immunol
.
2011
;
12
:
204
212
.

143

Frostegård
 
J
.
Immunity, atherosclerosis and cardiovascular disease
.
BMC Med
.
2013
;
11
:
117
.

144

Bensinger
 
SJ
,
Bradley
 
MN
,
Joseph
 
SB
 et al.  
LXR signaling couples sterol metabolism to proliferation in the acquired immune response
.
Cell
.
2008
;
134
:
97
111
.

145

Kabouridis
 
PS
,
Jury
 
EC
.
Lipid rafts and T-lymphocyte function: implications for autoimmunity
.
FEBS Lett
.
2008
;
582
:
3711
3718
.

146

Tough
 
DF
,
Sprent
 
J
.
Lifespan of gamma/delta T cells
.
J Exp Med
.
1998
;
187
:
357
365
.

147

Cheng
 
HY
,
Wu
 
R
,
Gebre
 
AK
 et al.  
Increased cholesterol content in gammadelta (γδ) T lymphocytes differentially regulates their activation
.
PLoS One
.
2013
;
8
:e63746.

148

Lim
 
H
,
Kim
 
YU
,
Sun
 
H
 et al.  
Proatherogenic conditions promote autoimmune T helper 17 cell responses in vivo
.
Immunity
.
2014
;
40
:
153
165
.

149

Butcher
 
MJ
,
Waseem
 
TC
,
Galkina
 
EV
.
Smooth muscle cell-derived interleukin-17C plays an atherogenic role via the recruitment of proinflammatory interleukin-17A+ T Cells to the aorta
.
Arterioscler Thromb Vasc Biol
.
2016
;
36
:
1496
1506
.

150

Akhavanpoor
 
M
,
Akhavanpoor
 
H
,
Gleissner
 
CA
 et al.  
The two faces of interleukin-17A in atherosclerosis
.
Curr Drug Targets
.
2017
;
18
:
863
873
.

151

Vu
 
DM
,
Tai
 
A
,
Tatro
 
JB
 et al.  
γδT cells are prevalent in the proximal aorta and drive nascent atherosclerotic lesion progression and neutrophilia in hypercholesterolemic mice
.
PLoS One
.
2014
;
9
:e109416.

152

Gil-Pulido
 
J
,
Amézaga
 
N
,
Jorgacevic
 
I
 et al.  
Interleukin-23 receptor expressing γδ T cells locally promote early atherosclerotic lesion formation and plaque necrosis in mice
.
Cardiovasc Res
.
2021
:cvab359.

153

Vanderlaan
 
PA
,
Reardon
 
CA
.
Thematic review series: the immune system and atherogenesis. The unusual suspects: an overview of the minor leukocyte populations in atherosclerosis
.
J Lipid Res
.
2005
;
46
:
829
838
.

154

Kolls
 
JK
,
Lindén
 
A
.
Interleukin-17 family members and inflammation
.
Immunity
.
2004
;
21
:
467
476
.

155

Nordlohne
 
J
,
von Vietinghoff
 
S
.
Interleukin 17A in atherosclerosis - Regulation and pathophysiologic effector function
.
Cytokine
.
2019
;
122
:154089.

156

Eid
 
RE
,
Rao
 
DA
,
Zhou
 
J
 et al.  
Interleukin-17 and interferon-gamma are produced concomitantly by human coronary artery-infiltrating T cells and act synergistically on vascular smooth muscle cells
.
Circulation
.
2009
;
119
:
1424
1432
.

157

Rao
 
DA
,
Eid
 
RE
,
Qin
 
L
 et al.  
Interleukin (IL)-1 promotes allogeneic T cell intimal infiltration and IL-17 production in a model of human artery rejection
.
J Exp Med
.
2008
;
205
:
3145
3158
.

158

Ye
 
P
,
Rodriguez
 
FH
,
Kanaly
 
S
 et al.  
Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense
.
J Exp Med
.
2001
;
194
:
519
527
.

159

Huo
 
Y
,
Weber
 
C
,
Forlow
 
SB
 et al.  
The chemokine KC, but not monocyte chemoattractant protein-1, triggers monocyte arrest on early atherosclerotic endothelium
.
J Clin Invest
.
2001
;
108
:
1307
1314
.

160

Zernecke
 
A
,
Bot
 
I
,
Djalali-Talab
 
Y
 et al.  
Protective role of CXC receptor 4/CXC ligand 12 unveils the importance of neutrophils in atherosclerosis
.
Circ Res
.
2008
;
102
:
209
217
.

161

Drechsler
 
M
,
Megens
 
RT
,
van Zandvoort
 
M
 et al.  
Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis
.
Circulation
.
2010
;
122
:
1837
1845
.

162

Kent
 
KC
,
Zwolak
 
RM
,
Egorova
 
NN
 et al.  
Analysis of risk factors for abdominal aortic aneurysm in a cohort of more than 3 million individuals
.
J Vasc Surg
.
2010
;
52
:
539
548
.

163

Qin
 
Y
,
Wang
 
Y
,
Liu
 
O
 et al.  
Tauroursodeoxycholic acid attenuates angiotensin ii induced abdominal aortic aneurysm formation in apolipoprotein E-deficient mice by inhibiting endoplasmic reticulum stress
.
Eur J Vasc Endovasc Surg
.
2017
;
53
:
337
345
.

164

López-Candales
 
A
,
Holmes
 
DR
,
Liao
 
S
 et al.  
Decreased vascular smooth muscle cell density in medial degeneration of human abdominal aortic aneurysms
.
Am J Pathol
.
1997
;
150
:
993
1007
.

165

Platsoucas
 
CD
,
Lu
 
S
,
Nwaneshiudu
 
I
 et al.  
Abdominal aortic aneurysm is a specific antigen-driven T cell disease
.
Ann N Y Acad Sci
.
2006
;
1085
:
224
235
.

166

Sun
 
J
,
Deng
 
H
,
Zhou
 
Z
 et al.  
Endothelium as a potential target for treatment of abdominal aortic aneurysm
.
Oxid Med Cell Longev
.
2018
;
2018
:6306542.

167

Eliason
 
JL
,
Hannawa
 
KK
,
Ailawadi
 
G
 et al.  
Neutrophil depletion inhibits experimental abdominal aortic aneurysm formation
.
Circulation
.
2005
;
112
:
232
240
.

168

Hannawa
 
KK
,
Eliason
 
JL
,
Woodrum
 
DT
 et al.  
L-selectin-mediated neutrophil recruitment in experimental rodent aneurysm formation
.
Circulation
.
2005
;
112
:
241
247
.

169

Rateri
 
DL
,
Howatt
 
DA
,
Moorleghen
 
JJ
 et al.  
Prolonged infusion of angiotensin II in apoE(-/-) mice promotes macrophage recruitment with continued expansion of abdominal aortic aneurysm
.
Am J Pathol
.
2011
;
179
:
1542
1548
.

170

Caccamo
 
N
,
Battistini
 
L
,
Bonneville
 
M
 et al.  
CXCR5 identifies a subset of Vgamma9Vdelta2 T cells which secrete IL-4 and IL-10 and help B cells for antibody production
.
J Immunol
.
2006
;
177
:
5290
5295
.

171

Seo
 
IH
,
Lee
 
SJ
,
Noh
 
TW
 et al.  
Increase of Vδ2+ T cells that robustly produce IL-17A in advanced abdominal aortic aneurysm tissues
.
Immune Netw
.
2021
;
21
:e17.

172

Korn
 
T
,
Bettelli
 
E
,
Oukka
 
M
 et al.  
IL-17 and Th17 cells
.
Annu Rev Immunol
.
2009
;
27
:
485
517
.

173

Wilson
 
WR
,
Anderton
 
M
,
Choke
 
EC
 et al.  
Elevated plasma MMP1 and MMP9 are associated with abdominal aortic aneurysm rupture
.
Eur J Vasc Endovasc Surg
.
2008
;
35
:
580
584
.

174

Longo
 
GM
,
Xiong
 
W
,
Greiner
 
TC
 et al.  
Matrix metalloproteinases 2 and 9 work in concert to produce aortic aneurysms
.
J Clin Invest
.
2002
;
110
:
625
632
.

175

Li
 
T
,
Li
 
X
,
Liu
 
X
 et al.  
The elevated expression of TLR4 and MMP9 in human abdominal aortic aneurysm tissues and its implication
.
BMC Cardiovasc Disord
.
2021
;
21
:
378
. (185).

176

Zheng
 
L
,
Xing
 
L
,
Zeng
 
C
 et al.  
Inactivation of PI3Kδ induces vascular injury and promotes aneurysm development by upregulating the AP-1/MMP-12 pathway in macrophages
.
Arterioscler Thromb Vasc Biol
.
2015
;
35
:
368
377
.

177

Fanjul-Fernández
 
M
,
Folgueras
 
AR
,
Cabrera
 
S
 et al.  
Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models
.
Biochim Biophys Acta
.
2010
;
1803
:
3
19
.

178

Li
 
Y
,
Wu
 
Y
,
Zhang
 
C
 et al.  
γδ T Cell-derived interleukin-17A via an interleukin-1β-dependent mechanism mediates cardiac injury and fibrosis in hypertension
.
Hypertension
.
2014
;
64
:
305
314
.

179

Zhang
 
S
,
Kan
 
X
,
Li
 
Y
 et al.  
Deficiency of γδ T cells protects against abdominal aortic aneurysms by regulating phosphoinositide 3-kinase/AKT signaling
.
J Vasc Surg
.
2018
;
67
:
899
908
.

180

Sutterwala
 
FS
,
Haasken
 
S
,
Cassel
 
SL
.
Mechanism of NLRP3 inflammasome activation
.
Ann NY Acad Sci
.
2014
;
1319
:
82
95
.

181

Acquaviva
 
L
,
Drogat
 
J
,
Dehé
 
PM
 et al.  
Spp1 at the crossroads of H3K4me3 regulation and meiotic recombination
.
Epigenetics
.
2013
;
8
:
355
360
.

182

Johnston
 
WF
,
Salmon
 
M
,
Su
 
G
 et al.  
Genetic and pharmacologic disruption of interleukin-1β signaling inhibits experimental aortic aneurysm formation
.
Arterioscler Thromb Vasc Biol
.
2013
;
33
:
294
304
.

183

Tieu
 
BC
,
Lee
 
C
,
Sun
 
H
 et al.  
An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice
.
J Clin Invest
.
2009
;
119
:
3637
3651
.

184

Xiong
 
W
,
MacTaggart
 
J
,
Knispel
 
R
 et al.  
Blocking TNF-alpha attenuates aneurysm formation in a murine model
.
J Immunol
.
2009
;
183
:
2741
2746
.

185

Zhou
 
J
,
Chen
 
J
,
Wei
 
Q
 et al.  
The role of ischemia/reperfusion injury in early hepatic allograft dysfunction
.
Liver Transpl
.
2020
;
26
:
1034
1048
.

186

Zhou
 
H
,
Li
 
L
,
Sun
 
H
 et al.  
Remote ischemic preconditioning attenuates hepatic ischemia/reperfusion injury after hemorrhagic shock by increasing autophagy
.
Int J Med Sci
.
2021
;
18
:
873
882
.

187

Vantourout
 
P
,
Hayday
 
A
.
Six-of-the-best: unique contributions of γδ T cells to immunology
.
Nat Rev Immunol
.
2013
;
13
:
88
100
.

188

Shen
 
B
,
Li
 
J
,
Yang
 
B
.
NKG2D blockade significantly attenuates ischemia-reperfusion injury in a cardiac transplantation model
.
Transplant Proc
.
2013
;
45
:
2513
2516
.

189

Kuboki
 
S
,
Sakai
 
N
,
Tschöp
 
J
 et al.  
Distinct contributions of CD4+ T cell subsets in hepatic ischemia/reperfusion injury
.
Am J Physiol Gastrointest Liver Physiol
.
2009
;
296
:
G1054
G1059
.

190

Hamacher-Brady
 
A
,
Brady
 
NR
,
Gottlieb
 
RA
.
The interplay between pro-death and pro-survival signaling pathways in myocardial ischemia/reperfusion injury: apoptosis meets autophagy
.
Cardiovasc Drugs Ther
.
2006
;
20
:
445
462
.

191

Hamacher-Brady
 
A
,
Brady
 
NR
,
Logue
 
SE
 et al.  
Response to myocardial ischemia/reperfusion injury involves Bnip3 and autophagy
.
Cell Death Differ
.
2007
;
14
:
146
157
.

192

Sharma
 
AK
,
Charles
 
EJ
,
Zhao
 
Y
 et al.  
Pannexin-1 channels on endothelial cells mediate vascular inflammation during lung ischemia-reperfusion injury
.
Am J Physiol Lung Cell Mol Physiol
.
2018
;
315
:
L301
L312
.

193

Zhu
 
H
,
Cui
 
D
,
Liu
 
K
 et al.  
Long pentraxin PTX3 attenuates ischemia reperfusion injury in a cardiac transplantation model
.
Transpl Int
.
2014
;
27
:
87
95
.

194

Hochegger
 
K
,
Schätz
 
T
,
Eller
 
P
 et al.  
Role of alpha/beta and gamma/delta T cells in renal ischemia-reperfusion injury
.
Am J Physiol Renal Physiol
.
2007
;
293
:
F741
F747
.

195

Pai
 
MH
,
Shih
 
YM
,
Shih
 
JM
 et al.  
Glutamine modulates changes in intestinal intraepithelial γδT-lymphocyte expressions in mice with ischemia/reperfusion injury
.
Shock
.
2015
;
44
:
77
82
.

196

Funken
 
D
,
Yu
 
Y
,
Feng
 
X
 et al.  
Lack of gamma delta T cells ameliorates inflammatory response after acute intestinal ischemia reperfusion in mice
.
Sci Rep
.
2021
;
11
:18628.

197

Geha
 
M
,
Tsokos
 
MG
,
Bosse
 
RE
 et al.  
IL-17A produced by innate lymphoid cells is essential for intestinal ischemia-reperfusion injury
.
J Immunol
.
2017
;
199
:
2921
2929
.

198

Lee
 
HT
,
Kim
 
M
,
Kim
 
JY
 et al.  
Critical role of interleukin-17A in murine intestinal ischemia-reperfusion injury
.
Am J Physiol Gastrointest Liver Physiol
.
2013
;
304
:
G12
G25
.

199

Zhang
 
L
,
Huang
 
Y
,
Lin
 
Y
 et al.  
Anti-inflammatory effect of cholera toxin B subunit in experimental stroke
.
J Neuroinflammation
.
2016
;
13
:
147
.

200

Shichita
 
T
,
Ago
 
T
,
Kamouchi
 
M
 et al.  
Novel therapeutic strategies targeting innate immune responses and early inflammation after stroke
.
J Neurochem
.
2012
:
29
38
. Suppl.

201

Shichita
 
T
,
Sakaguchi
 
R
,
Suzuki
 
M
 et al.  
Post-ischemic inflammation in the brain
.
Front Immunol
.
2012
May 31;
3
:
132
.

202

Hassane
 
M
,
Demon
 
D
,
Soulard
 
D
 et al.  
Neutrophilic NLRP3 inflammasome-dependent IL-1β secretion regulates the γδT17 cell response in respiratory bacterial infections
.
Mucosal Immunol
.
2017
;
10
:
1056
1068
.

203

Agerholm
 
R
,
Bekiaris
 
V
.
Evolved to protect, designed to destroy: iL-17-producing γδ T cells in infection, inflammation, and cancer
.
Eur J Immunol
.
2021
;
51
:
2164
2177
.

204

Yang
 
X
,
Zhan
 
N
,
Jin
 
Y
 et al.  
Tofacitinib restores the balance of γδTreg/γδT17 cells in rheumatoid arthritis by inhibiting the NLRP3 inflammasome
.
Theranostics
.
2021
;
11
:
1446
1457
.

205

Du
 
B
,
Zhu
 
M
,
Li
 
Y
 et al.  
The prostaglandin E2 increases the production of IL-17 and the expression of costimulatory molecules on γδ T cells in rheumatoid arthritis
.
Scand J Immunol
.
2020
;
91
:e12872.

206

Lee
 
JE
,
Lee
 
SM
,
Jung
 
J
.
Integrated omics analysis unraveled the microbiome-mediated effects of Yijin-Tang on hepatosteatosis and insulin resistance in obese mouse
.
Phytomedicine
.
2020
;
79
:153354.

207

Shibata
 
S
,
Tada
 
Y
,
Hau
 
CS
 et al.  
Adiponectin regulates psoriasiform skin inflammation by suppressing IL-17 production from γδ-T cells
.
Nat Commun
.
2015
;
6
:
7687
.

208

McGinley
 
AM
,
Sutton
 
CE
,
Edwards
 
SC
 et al.  
Interleukin-17A serves a priming role in autoimmunity by recruiting IL-1β-producing myeloid cells that promote pathogenic T cells
.
Immunity
.
2020
;
52
:
342
356
.

209

Suzuki
 
K
,
Suzuki
 
K
,
Yabe
 
Y
 et al.  
NF-κB1 contributes to imiquimod-induced psoriasis-like skin inflammation by inducing Vγ4+Vδ4+γδT17 cells
.
J Invest Dermatol
.
2021
. S0022-202X(21)02483-0.

210

St Leger
 
AJ
,
Desai
 
JV
,
Drummond
 
RA
 et al.  
An ocular commensal protects against corneal infection by driving an interleukin-17 response from mucosal γδ T cells
.
Immunity
.
2017
;
47
:
148
158
.

211

Paudel
 
S
,
Ghimire
 
L
,
Jin
 
L
 et al.  
NLRC4 suppresses IL-17A-mediated neutrophil-dependent host defense through upregulation of IL-18 and induction of necroptosis during Gram-positive pneumonia
.
Mucosal Immunol
.
2019
;
12
:
247
257
.

212

Kong
 
X
,
Sun
 
R
,
Chen
 
Y
 et al.  
γδ T cells drive myeloid-derived suppressor cell-mediated CD8+ T cell exhaustion in hepatitis B virus-induced immunotolerance
.
J Immunol
.
2014
;
193
:
1645
1653
.

213

Fenoglio
 
D
,
Poggi
 
A
,
Catellani
 
S
 et al.  
Vdelta1 T lymphocytes producing IFN-gamma and IL-17 are expanded in HIV-1-infected patients and respond to Candida albicans
.
Blood
.
2009
;
113
:
6611
6618
.

214

Maher
 
CO
,
Dunne
 
K
,
Comerford
 
R
 et al.  
Candida albicans stimulates IL-23 release by human dendritic cells and downstream IL-17 secretion by Vδ1 T cells
.
J Immunol
.
2015
;
194
:
5953
5960
.

215

Okada
 
S
,
Markle
 
JG
,
Deenick
 
EK
 et al.  
Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations
.
Science
.
2015
;
349
:
606
613
.

216

Ha
 
HL
,
Wang
 
H
,
Pisitkun
 
P
 et al.  
IL-17 drives psoriatic inflammation via distinct, target cell-specific mechanisms
.
Proc Natl Acad Sci USA
.
2014
;
111
:
E3422
E3431
.

217

Sureshbabu
 
SK
,
Chaukar
 
D
,
Chiplunkar
 
SV
.
Hypoxia regulates the differentiation and anti-tumor effector functions of γδT cells in oral cancer
.
Clin Exp Immunol
.
2020
;
201
:
40
57
.

218

Zhao
 
Y
,
Lu
 
F
,
Ye
 
J
 et al.  
Myeloid-derived suppressor cells and γδT17 cells contribute to the development of gastric MALT lymphoma in H. felis-infected mice
.
Front Immunol
.
2020
;
10
:
3104
.

219

Yan
 
J
,
Huang
 
J
.
Innate γδT17 cells convert cancer-elicited inflammation into immunosuppression through myeloid-derived suppressor cells
.
Oncoimmunology
.
2014
;
3
:e953423.

220

Wakita
 
D
,
Sumida
 
K
,
Iwakura
 
Y
 et al.  
Tumor-infiltrating IL-17-producing gammadelta T cells support the progression of tumor by promoting angiogenesis
.
Eur J Immunol
.
2010
;
40
:
1927
1937
.

221

Rei
 
M
,
Gonçalves-Sousa
 
N
,
Lança
 
T
 et al.  
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
.

222

Van Hede
 
D
,
Polese
 
B
,
Humblet
 
C
 et al.  
Human papillomavirus oncoproteins induce a reorganization of epithelial-associated γδ T cells promoting tumor formation
.
Proc Natl Acad Sci USA
.
2017
;
114
:
E9056
E9065
.

223

Patil
 
RS
,
Shah
 
SU
,
Shrikhande
 
SV
 et al.  
IL17 producing γδT cells induce angiogenesis and are associated with poor survival in gallbladder cancer patients
.
Int J Cancer
.
2016
;
139
:
869
881
.

224

Ma
 
S
,
Cheng
 
Q
,
Cai
 
Y
 et al.  
IL-17A produced by γδ T cells promotes tumor growth in hepatocellular carcinoma
.
Cancer Res
.
2014
;
74
:
1969
1982
.

225

Coffelt
 
SB
,
Kersten
 
K
,
Doornebal
 
CW
 et al.  
IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis
.
Nature
.
2015
;
522
:
345
348
.

226

Cheng
 
M
,
Chen
 
Y
,
Huang
 
D
 et al.  
Intrinsically altered lung-resident γδ T cells control lung melanoma by producing interleukin-17A in the elderly
.
Aging Cell
.
2020
;
19
:e13099.

227

Ma
 
Y
,
Aymeric
 
L
,
Locher
 
C
 et al.  
Contribution of IL-17-producing gamma delta T cells to the efficacy of anticancer chemotherapy
.
J Exp Med
.
2011
;
208
:
491
503
.

228

Ridker
 
PM
,
Everett
 
BM
,
Thuren
 
T
 et al.  
Antiinflammatory therapy with canakinumab for atherosclerotic disease
.
N Engl J Med
.
2017
;
377
:
1119
1131
.

229

Gilbert
 
J
,
Lekstrom-Himes
 
J
,
Donaldson
 
D
 et al.  
Effect of CC chemokine receptor 2 CCR2 blockade on serum C-reactive protein in individuals at atherosclerotic risk and with a single nucleotide polymorphism of the monocyte chemoattractant protein-1 promoter region
.
Am J Cardiol
.
2011
;
107
:
906
911
.

230

Doesch
 
AO
,
Zhao
 
L
,
Gleissner
 
CA
 et al.  
Inhibition of B7-1 (CD80) by RhuDex® reduces lipopolysaccharide-mediated inflammation in human atherosclerotic lesions
.
Drug Des Devel Ther
.
2014
;
8
:
447
457
.

Abbreviations

     
  • CVD

    ardiovascular disease

  •  
  • MI

    myocardial infarction

  •  
  • AS

    atherosclerosis

  •  
  • AA

    aortic aneurysm

  •  
  • IRI

    ischemia-reperfusion injury

  •  
  • TCR

    T cells receptors

  •  
  • CVB3

    Coxsackievirus of group B3

  •  
  • KGF

    keratinocyte growth factor

  •  
  • TFs

    Transcription factors

  •  
  • BLK

    B-lymphoid tyrosine kinase

  •  
  • LTβR

    lymphotoxin β receptors

  •  
  • MHC

    major histocompatibility complex

  •  
  • NKT

    natural killer T

  •  
  • PAMP

    pathogen-associated molecular pattern

  •  
  • MA

    Madecassic acid

  •  
  • pMHC

    peptide-MHC

  •  
  • DAMP

    damage-associated molecular pattern

  •  
  • HSP

    heat-shock protein

  •  
  • TLRs

    Toll-like receptors

  •  
  • DCs

    dendritic cells

  •  
  • ROS

    reactive oxygen species

  •  
  • MMPs

    matrix metalloproteinases

  •  
  • WT

    wild-type

  •  
  • LDL

    low density lipoprotein

  •  
  • VSMCs

    vascular smooth muscle cells

  •  
  • AAA

    abdominal aortic aneurysm

  •  
  • MALT

    mucosa-associated lymphoid tissue

  •  
  • PMN-MDSC

    polymorphonuclear myeloid suppressor cells

  •  
  • MPE

    malignant pleural effusion

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)