Skip to main content

Table 2 Anti-influenza mechanisms of γδ T cells in the respiratory tract

From: Role of γδ T cells in controlling viral infections with a focus on influenza virus: implications for designing novel therapeutic approaches

Study

Host

γδ subsets

Anti-influenza mechanism

Sant et al. [61]

Human γδ T cells along with target IAV-infected THP-1 cells

Vγ9Vδ2

Vγ9Vδ2 T cell-mediated IFN-γ-dependent anti-influenza activity

Killing of target cells by Vγ9Vδ2 T cells with an equivalent efficiency to CD8+ T cells and NK cells

Schwaiger et al. [105]

Pigs, as human-like IAV infection models

pan γδ

Anti-IAV activity mediated by γδ T cells in a perforin-dependent cytotoxic pathway

An increase in the percentage of CD8 expressing γδ T cells in route of viral entry

Xi-zhi et al. [14]

A neonatal mouse model

pan γδ

IL-17A-producing γδ T cell-mediated increased lung epithelial cell-derived IL-33

IL-33-mediated lung repair through the infiltration of group 2 innate lymphoid cells and regulatory T cells to the lung

Palomino et al. [106]

C57BL/6 mice

pan γδ

Recruitment of γδ T cells to the site of infection in a CXCR3 receptor-dependent fashion

IL-17A-producing γδ T cell-mediated enhanced protective cells (neutrophils and NK cells) recruitment to the tracheal mucosa

Goldberg et al. [108]

Mx1-congenic C57BL/6 mice models

pan γδ

KD-expanded γδ T cell-mediated improved lung barrier integrity

Viral clearance during lethal influenza infection in airway epithelium by γδ T cells expressing IL-17

Carding et al. [13]

C57B1/6 J mice

Vγ4+

Vγ2+/Vγ1+

A possible role of γδ T cells in responding to HSP+ cells and resolving inflammatory process upon influenza infection

Ivanov et al. [109]

IL-22−/− and wild type mice

pan γδ

IL-22-producing γδ T cell- dependent limited lung inflammation and increased immunity to bacterial infection at early phase of IAV infection

Li et al. [9]

Human γδ T cells along with IAV-infected A549 cells

Vγ9Vδ2

NKG2D-induced Vγ9Vδ2 T cell-mediated cytotoxicity in perforin/granzyme B, TRAIL, and Fas-FasL dependent pathways

Qin et al. [11]

Human PBMC infected with IAV

pan γδ

CCR5-mediated regulation of Vγ9Vδ2 T cell trafficking to virus-infected site by binding to its ligands secreted from infected cells upon IAV infections

Dong et al. [60]

TCR-δ−/− and wild type mice

pan γδ

Unprocessed whole HA molecule-restricted activation of γδ T cells

IFN-γ-mediated antiviral immunity against IAV infection provided by HA-induced γδ T cells

γδ T cell-induced bystander activation of other immune cells by secretion of immunomodulatory cytokines

Stervbo et al. [118]

Human donors

pan γδ

Detrimental effects of age and smoking on LAIV-induced γδ T cell responses

Horvath et al. [119]

Moser et al. [110]

IL-21R KO mice, C57BL/6, and CD45.1 mice

pan γδ

Modulatory role of IL-21 and serpinb1a in the control of negative effects of overstimulation of IL-17-producing γδ T cells in the respiratory tract after IAV infection

Zhao et al. [111]

Wild type and serpinB1−/− mice

  1. IAV, influenza A virus; THP-1 cells, a human monocyte cell line; NK cells, natural killer cells; MX1, myxovirus protein 1; KD, ketogenic diet; HSP, heat shock protein; A549 cells, human lung alveolar epithelial cells; HA, hemagglutinin; LAIV, live-attenuated influenza vaccine