Submitted:
19 January 2026
Posted:
20 January 2026
You are already at the latest version
Abstract
Keywords:
Inappropriate Innate Immunity Activation
gd T Cells and the Autoinflammatory Response
gd T Cells in Monogenic Autoinflammatory Diseases
gd T Cells in Multifactorial/polygenic Autoinflammatory Disease
- Diseases of Inappropriate autoinflammatory responses to well defined endogenous triggers.
- 2.
- Diseases of Inappropriate autoinflammatory responses to poorly defined endogenous triggers.
Discussion
Funding
References
- Parentelli, AS; Lopes, AA; Rieux-Laucat, F; Hermine, O. Systemic auto-inflammatory diseases, auto-immune diseases and immune deficiencies: From independent to overlapped diseases approach. Autoimmun Rev. 2025, 24(11), 103901. [Google Scholar] [CrossRef] [PubMed]
- Kozu, KT; Nascimento, R; Aires, PP; Cordeiro, RA; Moura, TCL; Sztajnbok, FR; et al. Inflammatory turmoil within: an exploration of autoinflammatory disease genetic underpinnings, clinical presentations, and therapeutic approaches. Adv Rheumatol. 2024, 64(1), 62. [Google Scholar] [CrossRef] [PubMed]
- An, J; Marwaha, A; Laxer, RM. Autoinflammatory Diseases: A Review. J Rheumatol. 2024, 51(9), 848–61. [Google Scholar] [CrossRef]
- Klotgen, HW; Beltraminelli, H; Yawalkar, N; van Gijn, ME; Holzinger, D; Borradori, L. The expanding spectrum of clinical phenotypes associated with PSTPIP1 mutations: from PAPA to PAMI syndrome and beyond. Br J Dermatol 2018, 178(4), 982–3. [Google Scholar] [CrossRef] [PubMed]
- Bank, I; DePinho, RA; Brenner, MB; Cassimeris, J; Alt, FW; Chess, L. A functional T3 molecule associated with a novel heterodimer on the surface of immature human thymocytes. Nature 1986, 322(6075), 179–81. [Google Scholar] [CrossRef]
- Brenner, MB; McLean, J; Dialynas, DP; Strominger, JL; Smith, JA; Owen, FL; et al. Identification of a putative second T-cell receptor. Nature 1986, 322(6075), 145–9. [Google Scholar] [CrossRef]
- Hayday, AC. [gamma][delta] cells: a right time and a right place for a conserved third way of protection. Annu Rev Immunol. 2000, 18, 975–1026. [Google Scholar] [CrossRef]
- Sok, CL; Rossjohn, J; Gully, BS. The Evolving Portrait of gammadelta TCR Recognition Determinants. J Immunol. 2024, 213(5), 543–52. [Google Scholar] [CrossRef]
- Hu, Y; Hu, Q; Li, Y; Lu, L; Xiang, Z; Yin, Z; et al. gammadelta T cells: origin and fate, subsets, diseases and immunotherapy. Signal Transduct Target Ther. 2023, 8(1), 434. [Google Scholar] [CrossRef]
- Sanchez Sanchez, G; Papadopoulou, M; Azouz, A; Tafesse, Y; Mishra, A; Chan, JKY; et al. Identification of distinct functional thymic programming of fetal and pediatric human gammadelta thymocytes via single-cell analysis. Nat Commun. 2022, 13(1), 5842. [Google Scholar] [CrossRef]
- Herrmann, T; Karunakaran, MM. Butyrophilins: gammadelta T Cell Receptor Ligands, Immunomodulators and More. Front Immunol. 2022, 13, 876493. [Google Scholar] [CrossRef]
- Zhu, Y; Gao, W; Zheng, J; Bai, Y; Tian, X; Huang, T; et al. Phosphoantigen-induced inside-out stabilization of butyrophilin receptor complexes drives dimerization-dependent gammadelta TCR activation. Immunity 2025, 58(7), 1646–59 e5. [Google Scholar] [CrossRef]
- Boutin, L; Scotet, E. Towards Deciphering the Hidden Mechanisms That Contribute to the Antigenic Activation Process of Human Vgamma9Vdelta2 T Cells. Front Immunol. 2018, 9, 828. [Google Scholar] [CrossRef]
- Willcox, BE; Willcox, CR. gammadelta TCR ligands: the quest to solve a 500-million-year-old mystery. Nat Immunol. 2019, 20(2), 121–8. [Google Scholar] [CrossRef] [PubMed]
- Guo, J; Chowdhury, RR; Mallajosyula, V; Xie, J; Dubey, M; Liu, Y; et al. gammadelta T cell antigen receptor polyspecificity enables T cell responses to a broad range of immune challenges. Proc Natl Acad Sci U S A 2024, 121(4), e2315592121. [Google Scholar] [CrossRef] [PubMed]
- Hayday, AC; Vantourout, P. The Innate Biologies of Adaptive Antigen Receptors. Annu Rev Immunol. 2020, 38, 487–510. [Google Scholar] [CrossRef]
- Ribot, JC; Lopes, N; Silva-Santos, B. gammadelta T cells in tissue physiology and surveillance. Nat Rev Immunol. 2021, 21(4), 221–32. [Google Scholar] [CrossRef] [PubMed]
- Hsu, UH; Chiang, BL. gammadelta T Cells and Allergic Diseases. Clin Rev Allergy Immunol. 2023, 65(2), 172–82. [Google Scholar] [CrossRef]
- Takimoto, R; Suzawa, T; Yamada, A; Sasa, K; Miyamoto, Y; Yoshimura, K; et al. Zoledronate promotes inflammatory cytokine expression in human CD14-positive monocytes among peripheral mononuclear cells in the presence of gammadelta T cells. Immunology 2021, 162(3), 306–13. [Google Scholar] [CrossRef]
- LaMarche, NM; Kohlgruber, AC; Brenner, MB. Innate T Cells Govern Adipose Tissue Biology. J Immunol. 2018, 201(7), 1827–34. [Google Scholar] [CrossRef]
- Ribot, JC; Silva-Santos, B. Differentiation and activation of gammadelta T Lymphocytes: Focus on CD27 and CD28 costimulatory receptors. Adv Exp Med Biol. 2013, 785, 95–105. [Google Scholar]
- Papotto, PH; Ribot, JC; Silva-Santos, B. IL-17(+) gammadelta T cells as kick-starters of inflammation. Nat Immunol. 2017, 18(6), 604–11. [Google Scholar] [CrossRef]
- Gombert, D; Simeonov, J; Klein, K; Agaugue, S; Scheffold, A; Kabelitz, D; et al. Butyrophilin 3A/2A1-independent activation of human Vgamma9Vdelta2 gammadelta T cells by bacteria. PNAS Nexus 2025, 4(11), pgaf358. [Google Scholar] [CrossRef]
- Yang, X; Zhan, N; Jin, Y; Ling, H; Xiao, C; Xie, Z; et al. Tofacitinib restores the balance of gammadeltaTreg/gammadeltaT17 cells in rheumatoid arthritis by inhibiting the NLRP3 inflammasome. Theranostics 2021, 11(3), 1446–57. [Google Scholar] [CrossRef]
- Zeng, Y; Lin, J; Xiao, Z; Li, Z; Zheng, G; Zhou, Y; et al. Neutrophil extracellular traps exacerbate inflammatory arthritis by inhibiting gammadelta Treg cell differentiation via the AIM2 inflammasome. Redox Biol. 2025, 87, 103881. [Google Scholar] [CrossRef]
- Bank, I; Duvdevani, M; Livneh, A. Expansion of gammadelta T-cells in Behcet’s disease: role of disease activity and microbial flora in oral ulcers. J Lab Clin Med. 2003, 141(1), 33–40. [Google Scholar] [CrossRef]
- Al, B; Bruno, M; Roring, RJ; Moorlag, S; Suen, TK; Kluck, V; et al. Peripheral T Cell Populations are Differentially Affected in Familial Mediterranean Fever, Chronic Granulomatous Disease, and Gout. J Clin Immunol. 2023, 43(8), 2033–48. [Google Scholar] [CrossRef]
- Suen, TK; Al, B; Ulas, T; Reusch, N; Bahrar, H; Bekkering, S; et al. Human gammadelta T Cell Function Is Impaired Upon Mevalonate Pathway Inhibition. Immunology 2025, 175(3), 300–22. [Google Scholar] [CrossRef] [PubMed]
- Siedel, H; Roers, A; Rosen-Wolff, A; Luksch, H. Type I interferon-independent T cell impairment in a Tmem173 N153S/WT mouse model of STING associated vasculopathy with onset in infancy (SAVI). Clin Immunol. 2020, 216, 108466. [Google Scholar] [CrossRef] [PubMed]
- Vroman, H; Das, T; Bergen, IM; van Hulst, JAC; Ahmadi, F; van Loo, G; et al. House dust mite-driven neutrophilic airway inflammation in mice with TNFAIP3-deficient myeloid cells is IL-17-independent. Clin Exp Allergy 2018, 48(12), 1705–14. [Google Scholar] [CrossRef] [PubMed]
- Yap, JY; Moens, L; Lin, MW; Kane, A; Kelleher, A; Toong, C; et al. Intrinsic Defects in B Cell Development and Differentiation, T Cell Exhaustion and Altered Unconventional T Cell Generation Characterize Human Adenosine Deaminase Type 2 Deficiency. J Clin Immunol. 2021, 41(8), 1915–35. [Google Scholar] [CrossRef]
- Mizumaki, H; Gao, S; Wu, Z; Gutierrez-Rodrigues, F; Bissa, M; Feng, X; et al. In depth transcriptomic profiling defines a landscape of dysfunctional immune responses in patients with VEXAS syndrome. Nat Commun. 2025, 16(1), 4690. [Google Scholar] [CrossRef]
- Martinon, F; Petrilli, V; Mayor, A; Tardivel, A; Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006, 440(7081), 237–41. [Google Scholar] [CrossRef]
- So, AK; Martinon, F. Inflammation in gout: mechanisms and therapeutic targets. Nat Rev Rheumatol. 2017, 13(11), 639–47. [Google Scholar] [CrossRef]
- Liu, Y; Zhao, Q; Yin, Y; McNutt, MA; Zhang, T; Cao, Y. Serum levels of IL-17 are elevated in patients with acute gouty arthritis. Biochem Biophys Res Commun. 2018, 497(3), 897–902. [Google Scholar] [CrossRef] [PubMed]
- Yuan, Y; Gao, Z; Chen, J; Liu, Y; Zhou, J. Integrative bioinformatics analysis and experimental validation reveals key genes and regulatory mechanisms in the development of gout. Front Genet. 2025, 16, 1598835. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M; Lin, Z; Liu, W. The immune microenvironment related biomarker CCL18 for patients with gout by comprehensive analysis. Comput Biol Chem. 2025, 115, 108334. [Google Scholar] [CrossRef]
- Xiao, L; Lin, S; Zhan, F. Identification of hub genes and transcription factors in patients with primary gout complicated with atherosclerosis. Sci Rep. 2024, 14(1), 3992. [Google Scholar] [CrossRef] [PubMed]
- Duewell, P; Kono, H; Rayner, KJ; Sirois, CM; Vladimer, G; Bauernfeind, FG; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464(7293), 1357–61. [Google Scholar] [CrossRef]
- Olson, NC; Sitlani, CM; Doyle, MF; Huber, SA; Landay, AL; Tracy, RP; et al. Innate and adaptive immune cell subsets as risk factors for coronary heart disease in two population-based cohorts. Atherosclerosis 2020, 300, 47–53. [Google Scholar] [CrossRef]
- Li, Y; Jiang, S; Li, J; Yin, M; Yan, F; Chen, Y; et al. Phenotypic Changes of Peripheral gammadelta T Cell and Its Subsets in Patients With Coronary Artery Disease. Front Immunol. 2022, 13, 900334. [Google Scholar] [CrossRef]
- Zhao, E; Xie, H; Zhang, Y. Predicting Diagnostic Gene Biomarkers Associated With Immune Infiltration in Patients With Acute Myocardial Infarction. Front Cardiovasc Med. 2020, 7, 586871. [Google Scholar] [CrossRef]
- Zhao, S; Zhao, C; Wang, L; Cheng, B; Zheng, M; Wang, X. Elucidating the role of LGALS3BP in coronary atherosclerosis: integrating bioinformatics and machine learning for advanced insights. J Cardiothorac Surg. 2025, 20(1), 338. [Google Scholar] [CrossRef]
- Sinha, A; Rivera, AS; Doyle, MF; Sitlani, C; Fohner, A; Huber, SA; et al. Association of immune cell subsets with cardiac mechanics in the Multi-Ethnic Study of Atherosclerosis. JCI Insight 2021, 6(13). [Google Scholar] [CrossRef] [PubMed]
- Zhao, P; Yuan, K; Tang, Z; Li, Y; Yu, Y; Gao, W; et al. Investigation of hub-shared genes and regulatory mechanisms of rheumatoid arthritis and atherosclerosis. Clin Rheumatol. 2025, 44(6), 2241–56. [Google Scholar] [CrossRef] [PubMed]
- Yong, X; Kang, T; Li, M; Li, S; Yan, X; Li, J; et al. Identification of novel biomarkers for atherosclerosis using single-cell RNA sequencing and machine learning. Mamm Genome 2025, 36(1), 183–99. [Google Scholar] [CrossRef] [PubMed]
- Liu, JW; Zhang, ZH; Lv, XS; Xu, MY; Ni, B; He, B; et al. Identification of key pyroptosis-related genes and microimmune environment among peripheral arterial beds in atherosclerotic arteries. Sci Rep. 2024, 14(1), 233. [Google Scholar] [CrossRef]
- Zheng, Z; Yuan, D; Shen, C; Zhang, Z; Ye, J; Zhu, L. Identification of potential diagnostic biomarkers of atherosclerosis based on bioinformatics strategy. BMC Med Genomics 2023, 16(1), 100. [Google Scholar] [CrossRef]
- Yin, Y; Xie, Z; Chen, D; Guo, H; Han, M; Zhu, Z; et al. Integrated investigation of DNA methylation, gene expression and immune cell population revealed immune cell infiltration associated with atherosclerotic plaque formation. BMC Med Genomics 2022, 15(1), 108. [Google Scholar] [CrossRef]
- Shen, Y; Xu, LR; Tang, X; Lin, CP; Yan, D; Xue, S; et al. Identification of potential therapeutic targets for atherosclerosis by analysing the gene signature related to different immune cells and immune regulators in atheromatous plaques. BMC Med Genomics 2021, 14(1), 145. [Google Scholar] [CrossRef]
- Gil-Pulido, J; Amezaga, N; Jorgacevic, I; Manthey, HD; Rosch, M; Brand, T; et al. Interleukin-23 receptor expressing gammadelta T cells locally promote early atherosclerotic lesion formation and plaque necrosis in mice. Cardiovasc Res. 2022, 118(14), 2932–45. [Google Scholar] [CrossRef]
- Han, H; Du, R; Cheng, P; Zhang, J; Chen, Y; Li, G. Comprehensive Analysis of the Immune Infiltrates and Aberrant Pathways Activation in Atherosclerotic Plaque. Front Cardiovasc Med. 2020, 7, 602345. [Google Scholar] [CrossRef] [PubMed]
- Smith, E; Prasad, KM; Butcher, M; Dobrian, A; Kolls, JK; Ley, K; et al. Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E-deficient mice. Circulation 2010, 121(15), 1746–55. [Google Scholar] [CrossRef]
- Yu, J; Zhao, Z; Li, Y; Chen, J; Huang, N; Luo, Y. Role of NLRP3 in Parkinson’s disease: Specific activation especially in dopaminergic neurons. Heliyon 2024, 10(7), e28838. [Google Scholar] [CrossRef] [PubMed]
- Diener, C; Hart, M; Kehl, T; Becker-Dorison, A; Tanzer, T; Schub, D; et al. Time-resolved RNA signatures of CD4+ T cells in Parkinson’s disease. Cell Death Discov. 2023, 9(1), 18. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y; Liu, H; Hu, J; Han, C; Zhong, Z; Luo, W; et al. Significant Difference of Immune Cell Fractions and Their Correlations With Differential Expression Genes in Parkinson’s Disease. Front Aging Neurosci. 2021, 13, 686066. [Google Scholar] [CrossRef]
- Zhou, C; Zhou, X; He, D; Li, Z; Xie, X; Ren, Y. Reduction of Peripheral Blood iNKT and gammadeltaT Cells in Patients With Parkinson’s Disease: An Observational Study. Front Immunol. 2020, 11, 1329. [Google Scholar] [CrossRef]
- Fiszer, U; Mix, E; Fredrikson, S; Kostulas, V; Olsson, T; Link, H. gamma delta+ T cells are increased in patients with Parkinson’s disease. J Neurol Sci. 1994, 121(1), 39–45. [Google Scholar] [CrossRef]
- Ravichandran, KA; Brosseron, F; McManus, RM; Ising, C; Gorgen, S; Schmidt, S; et al. Enhancing Tyro3 signalling ameliorates IL-1beta production through STAT1 in Alzheimer’s disease models. J Leukoc Biol. 2025. [Google Scholar] [CrossRef]
- Du, W; Yu, S; Liu, R; Kong, Q; Hao, X; Liu, Y. Precision Prediction of Alzheimer’s Disease: Integrating Mitochondrial Energy Metabolism and Immunological Insights. J Mol Neurosci. 2025, 75(1), 5. [Google Scholar] [CrossRef]
- Chen, G; Xi, E; Gu, X; Wang, H; Tang, Q. The study on cuproptosis in Alzheimer’s disease based on the cuproptosis key gene FDX1. Front Aging Neurosci. 2024, 16, 1480332. [Google Scholar] [CrossRef]
- Lin, H; Su, L; Mao, D; Yang, G; Huang, Q; Lan, Y; et al. Identification of altered immune cell types and molecular mechanisms in Alzheimer’s disease progression by single-cell RNA sequencing. Front Aging Neurosci. 2024, 16, 1477327. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y; Shen, Z; Wu, H; Yu, Z; Wu, X; Zhou, L; et al. Identification of genes related to glucose metabolism and analysis of the immune characteristics in Alzheimer’s disease. Brain Res. 2023, 1819, 148545. [Google Scholar] [CrossRef]
- Zhuang, X; Zhang, G; Bao, M; Jiang, G; Wang, H; Li, S; et al. Development of a novel immune infiltration-related diagnostic model for Alzheimer’s disease using bioinformatic strategies. Front Immunol. 2023, 14, 1147501. [Google Scholar] [CrossRef]
- Shigemizu, D; Mori, T; Akiyama, S; Higaki, S; Watanabe, H; Sakurai, T; et al. Identification of potential blood biomarkers for early diagnosis of Alzheimer’s disease through RNA sequencing analysis. Alzheimers Res Ther. 2020, 12(1), 87. [Google Scholar] [CrossRef]
- Dart, RJ; Zlatareva, I; Vantourout, P; Theodoridis, E; Amar, A; Kannambath, S; et al. Conserved gammadelta T cell selection by BTNL proteins limits progression of human inflammatory bowel disease. Science 2023, 381(6663), eadh0301. [Google Scholar] [CrossRef] [PubMed]
- Aliseychik, M; Patrikeev, A; Gusev, F; Grigorenko, A; Andreeva, T; Biragyn, A; et al. Dissection of the Human T-Cell Receptor gamma Gene Repertoire in the Brain and Peripheral Blood Identifies Age- and Alzheimer’s Disease-Associated Clonotype Profiles. Front Immunol. 2020, 11, 12. [Google Scholar] [CrossRef]
- Dressman, D; Winford, ED; Vardarajan, BN; Yidenk, M; Talcoff, R; Mazen, J; et al. Basic Science and Pathogenesis. Alzheimers Dement. 2025, 21 Suppl 1(Suppl 1), e107352. [Google Scholar] [CrossRef]
- Kloske, CM; Mahinrad, S; Barnum, CJ; Batista, AF; Bradshaw, EM; Butts, B; et al. Advancements in Immunity and Dementia Research: Highlights from the 2023 AAIC Advancements: Immunity Conference. Alzheimers Dement. 2025, 21(1), e14291. [Google Scholar] [CrossRef]
- Brigas, HC; Ribeiro, M; Coelho, JE; Gomes, R; Gomez-Murcia, V; Carvalho, K; et al. IL-17 triggers the onset of cognitive and synaptic deficits in early stages of Alzheimer’s disease. Cell Rep. 2021, 36(9), 109574. [Google Scholar] [CrossRef]
- Emmi, G; Bettiol, A; Hatemi, G; Prisco, D. Behcet’s syndrome. Lancet 2024, 403(10431), 1093–108. [Google Scholar] [CrossRef]
- Hayday, AC. gammadelta T Cell Update: Adaptate Orchestrators of Immune Surveillance. J Immunol. 2019, 203(2), 311–20. [Google Scholar] [CrossRef] [PubMed]
- Hasan, A; Fortune, F; Wilson, A; Warr, K; Shinnick, T; Mizushima, Y; et al. Role of gamma delta T cells in pathogenesis and diagnosis of Behcet’s disease. Lancet 1996, 347(9004), 789–94. [Google Scholar] [CrossRef]
- Nishida, T; Hirayama, K; Nakamura, S; Ohno, S. Proliferative response of CD8+ gamma delta+ T cells to Streptococcus sanguis in patients with Behcet’s disease. Ocul Immunol Inflamm. 1998, 6(3), 139–44. [Google Scholar] [CrossRef] [PubMed]
- DIM, A; Emery, P. Rheumatoid arthritis: a review of the key clinical features and ongoing challenges of the disease. Panminerva Med. 2024, 66(4), 427–42. [Google Scholar]
- Deniz, R; Tulunay-Virlan, A; Ture Ozdemir, F; Unal, AU; Ozen, G; Alibaz-Oner, F; et al. Th17-Inducing Conditions Lead to in vitro Activation of Both Th17 and Th1 Responses in Behcet’s Disease. Immunol Invest. 2017, 46(5), 518–25. [Google Scholar] [CrossRef]
- Freysdottir, J; Lau, S; Fortune, F. Gammadelta T cells in Behcet’s disease (BD) and recurrent aphthous stomatitis (RAS). Clin Exp Immunol. 1999, 118(3), 451–7. [Google Scholar] [CrossRef]
- Clemente Ximenis, A; Crespi Bestard, C; Cambra Conejero, A; Pallares Ferreres, L; Juan Mas, A; Olea Vallejo, JL; et al. In vitro evaluation of gammadelta T cells regulatory function in Behcet’s disease patients and healthy controls. Hum Immunol. 2016, 77(1), 20–8. [Google Scholar] [CrossRef]
- Parlakgul, G; Guney, E; Erer, B; Kilicaslan, Z; Direskeneli, H; Gul, A; et al. Expression of regulatory receptors on gammadelta T cells and their cytokine production in Behcet’s disease. Arthritis Res Ther. 2013, 15(1), R15. [Google Scholar] [CrossRef]
- Yasuoka, H; Yamaguchi, Y; Mizuki, N; Nishida, T; Kawakami, Y; Kuwana, M. Preferential activation of circulating CD8+ and gammadelta T cells in patients with active Behcet’s disease and HLA-B51. Clin Exp Rheumatol. 2008, 26((4) Suppl 50, S59–63. [Google Scholar]
- Treusch, M; Vonthein, R; Baur, M; Gunaydin, I; Koch, S; Stubiger, N; et al. Influence of human recombinant interferon-alpha2a (rhIFN-alpha2a) on altered lymphocyte subpopulations and monocytes in Behcet’s disease. Rheumatology (Oxford) 2004, 43(10), 1275–82. [Google Scholar] [CrossRef]
- Accardo-Palumbo, A; Ferrante, A; Cadelo, M; Ciccia, F; Parrinello, G; Lipari, L; et al. The level of soluble Granzyme A is elevated in the plasma and in the Vgamma9/Vdelta2 T cell culture supernatants of patients with active Behcet’s disease. Clin Exp Rheumatol. 2004, 22((4) Suppl 34, S45–9. [Google Scholar]
- Triolo, G; Accardo-Palumbo, A; Dieli, F; Ciccia, F; Ferrante, A; Giardina, E; et al. Vgamma9/Vdelta2 T lymphocytes in Italian patients with Behcet’s disease: evidence for expansion, and tumour necrosis factor receptor II and interleukin-12 receptor beta1 expression in active disease. Arthritis Res Ther. 2003, 5(5), R262–8. [Google Scholar] [CrossRef]
- Verjans, GM; van Hagen, PM; van der Kooi, A; Osterhaus, AD; Baarsma, GS. Vgamma9Vdelta2 T cells recovered from eyes of patients with Behcet’s disease recognize non-peptide prenyl pyrophosphate antigens. J Neuroimmunol. 2002, 130(1-2), 46–54. [Google Scholar] [CrossRef] [PubMed]
- Natah, SS; Hayrinen-Immonen, R; Hietanen, J; Patinen, P; Malmstrom, M; Savilahti, E; et al. Increased density of lymphocytes bearing gamma/delta T-cell receptors in recurrent aphthous ulceration (RAU). Int J Oral Maxillofac Surg. 2000, 29(5), 375–80. [Google Scholar] [CrossRef]
- Hoshino, T; Ohta, A; Nakao, M; Ota, T; Inokuchi, T; Matsueda, S; et al. TCR gamma delta + T cells in peripheral blood of patients with adult Still’s disease. J Rheumatol. 1996, 23(1), 124–9. [Google Scholar] [PubMed]
- Kessel, C; Lippitz, K; Weinhage, T; Hinze, C; Wittkowski, H; Holzinger, D; et al. Proinflammatory Cytokine Environments Can Drive Interleukin-17 Overexpression by gamma/delta T Cells in Systemic Juvenile Idiopathic Arthritis. Arthritis Rheumatol. 2017, 69(7), 1480–94. [Google Scholar] [CrossRef]
- Carter, MJ; Fish, M; Jennings, A; Doores, KJ; Wellman, P; Seow, J; et al. Peripheral immunophenotypes in children with multisystem inflammatory syndrome associated with SARS-CoV-2 infection. Nat Med. 2020, 26(11), 1701–7. [Google Scholar] [CrossRef] [PubMed]
- Mukund, K; Nayak, P; Ashokkumar, C; Rao, S; Almeda, J; Betancourt-Garcia, MM; et al. Immune Response in Severe and Non-Severe Coronavirus Disease 2019 (COVID-19) Infection: A Mechanistic Landscape. Front Immunol. 2021, 12, 738073. [Google Scholar] [CrossRef]
- Bellos, E; Santillo, D; Vantourout, P; Jackson, HR; Duret, A; Hearn, H; et al. Heterozygous BTNL8 variants in individuals with multisystem inflammatory syndrome in children (MIS-C). J Exp Med. 2024, 221(12). [Google Scholar] [CrossRef]
- Xie, Y; Shi, H; Han, B. Bioinformatic analysis of underlying mechanisms of Kawasaki disease via Weighted Gene Correlation Network Analysis (WGCNA) and the Least Absolute Shrinkage and Selection Operator method (LASSO) regression model. BMC Pediatr. 2023, 23(1), 90. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y; Yang, M; Zhang, M; Wang, H; Zheng, Y; Sun, R; et al. Single-Cell Transcriptome Reveals Potential Mechanisms for Coronary Artery Lesions in Kawasaki Disease. Arterioscler Thromb Vasc Biol. 2024, 44(4), 866–82. [Google Scholar] [CrossRef] [PubMed]
- Liu, D; Song, M; Jing, F; Liu, B; Yi, Q. Diagnostic Value of Immune-Related Genes in Kawasaki Disease. Front Genet. 2021, 12, 763496. [Google Scholar] [CrossRef]
- Kohga, K; Tatsumi, T; Tsunematsu, H; Aono, S; Shimizu, S; Kodama, T; et al. Interleukin-1beta enhances the production of soluble MICA in human hepatocellular carcinoma. Cancer Immunol Immunother. 2012, 61(9), 1425–32. [Google Scholar] [CrossRef] [PubMed]
- Kondreddy, V; Keshava, S; Esmon, CT; Pendurthi, UR; Rao, LVM. A critical role of endothelial cell protein C receptor in the intestinal homeostasis in experimental colitis. Sci Rep. 2020, 10(1), 20569. [Google Scholar] [CrossRef] [PubMed]
- Pinheiro, RGR; Silva-Santos, B. Reaching maturity: Thymic gammadelta T cell differentiation in mice and humans. Sci Immunol. 2025, 10(114), eadn2093. [Google Scholar] [CrossRef]
- Khoshbakht, S; Baskurt, D; Vural, A; Vural, S. Behcet’s Disease: A Comprehensive Review on the Role of HLA-B*51, Antigen Presentation, and Inflammatory Cascade. Int J Mol Sci. 2023, 24(22). [Google Scholar] [CrossRef]
- Brandes, M; Willimann, K; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309(5732), 264–8. [Google Scholar] [CrossRef]
- Huang, Y; Getahun, A; Heiser, RA; Detanico, TO; Aviszus, K; Kirchenbaum, GA; et al. gammadelta T Cells Shape Preimmune Peripheral B Cell Populations. J Immunol. 2016, 196(1), 217–31. [Google Scholar] [CrossRef]
- Peters, C; Kabelitz, D; Wesch, D. Regulatory functions of gammadelta T cells. Cell Mol Life Sci. 2018, 75(12), 2125–35. [Google Scholar] [CrossRef]
- Raucci, F; Iqbal, AJ; Saviano, A; Minosi, P; Piccolo, M; Irace, C; et al. IL-17A neutralizing antibody regulates monosodium urate crystal-induced gouty inflammation. Pharmacol Res. 2019, 147, 104351. [Google Scholar] [CrossRef] [PubMed]
- Talarico, R; Italiano, N; Emmi, G; Piga, M; Cantarini, L; Mattioli, I; et al. Efficacy and safety of infliximab or adalimumab in severe mucocutaneous Behcet’s syndrome refractory to traditional immunosuppressants: a 6-month, multicentre, randomised controlled, prospective, parallel group, single-blind trial. Ann Rheum Dis 2024. [Google Scholar] [CrossRef] [PubMed]
| Disease | Gene | Dominant Inflammatory Pathway | Key γδ T Cell Insight | Refs |
|---|---|---|---|---|
| Familial Mediterranean Fever (FMF) | MEFV | Pyrin inflammasome → IL-1β | Preserved numbers but reduced IFN-γ production by Vδ2⁺ cells; increased CCR8⁺ subset suggests enhanced tissue homing | [25,26] |
| Hyper-IgD Syndrome (HIDS) | MVK | Mevalonate pathway defect → IL-1β | Impaired TNF-α and IFN-γ secretion due to defective Vγ9Vδ2 activation (↓ IPP) | [27] |
| SAVI | TMEM173 | Constitutive STING → type I IFN | Relative γδ T-cell preservation amid αβ lymphopenia suggests participation in IFN-driven inflammation | [28] |
| HA20 | TNFAIP3 | NF-κB dysregulation | Reduced number, but pathogenic γδ-17 accumulation in inflamed tissues (murine models) | [29] |
| DADA2 | ADA2 | inborn error of immunity | Vd2+ cells reduced | [31] |
| VEXAS | UBA1 | Myeloid ubiquitin-stress inflammation | elevated IFNa and IFNg gene module scores | [32] |
| Disease | Key Trigger / Pathway | Dominant Autoinflammatory Mechanism | γδ T Cell Signature (Key Findings) | References |
|---|---|---|---|---|
| Gout | Monosodium urate crystals; NLRP3 | IL-1β–driven inflammasome activation with neutrophil-mediated joint inflammation | Major early source of IL-17 in blood and joints; reduced IFN-γ production by Vδ2⁺ cells; | [30,31,32,33,34] |
| Atherosclerosis | Cholesterol crystals, oxLDL; NLRP3 | Foam-cell inflammasome activation, pyroptosis, chronic plaque inflammation | Decreased γδ T cells in blood ; accumulation in plaques; associated with IL-18 signaling, macrophages, and plaque instability. | [35,36,37,38,39,40,41,42,43,44,45,46,47,48,49] |
| Parkinson’s disease | α-Synuclein; NLRP3 | Microglial inflammasome activation and neuroinflammation | Activated IL-17–CD4+γδ T cells; variable blood levels; recruitment to CNS and activation | [50,51,52,53,54] |
| Alzheimer’s disease | Amyloid-β, tau; NLRP3 | Chronic microglial inflammasome-driven neuroinflammation | Enrichment in brain tissue; skewed TCR repertoire (↓TRGV9, ↑TRGV2/4/8); IL-17–producing γδ T cells in mouse model early in disease | [55,56,57,58,59,60,61,62,63,64,65,66] |
| Behçet’s disease | Microbial, stress and heat-shock antigens | Innate-like immune activation with autoinflammatory and autoimmune features | Expansion and activation of Vγ9Vδ2⁺ and Vδ1⁺ γδ T cells; secretion of IFN-γ, TNF-α, granzyme A; prominent tissue infiltration | [67,68,69,70,71,72,73,74,75,76,77,78,79,80,81] |
| Systemic-onset JIA (Still’s disease) | IL-1β, IL-18 | Cytokine-driven innate autoinflammation | Increased circulating γδ T cells with dominant IL-17A production; phenotype partially normalizes with IL-1 blockade | [82,83] |
| MIS-C (SARS-CoV-2) | Post-infectious immune dysregulation | Systemic cytokine storm with lymphopenia | Numerical reduction but strong activation (HLA-DR⁺); BTNL8 variants impair gut Vγ4Vδ1⁺ γδ T-cell responses | [84,85,86] |
| Kawasaki disease | Unknown (likely infection-triggered inflammation) | Medium-vessel vasculitis with cytokine storm | γδ T cells enriched in coronary artery infiltrates; peripheral blood findings suggesting redistribution to inflamed tissue | [87,88,89] |
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. |
© 2026 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license.