Next Article in Journal
Investigation of Biological Factors Contributing to Individual Variation in Viral Titer after Oral Infection of Aedes aegypti Mosquitoes by Sindbis Virus
Next Article in Special Issue
Special Issue “Gamma Delta T Cells in Immune Response against Viruses”
Previous Article in Journal
A Conserved Tryptophan in the Envelope Cytoplasmic Tail Regulates HIV-1 Assembly and Spread
Previous Article in Special Issue
The Role of γδ T Cells as a Line of Defense in Viral Infections after Allogeneic Stem Cell Transplantation: Opportunities and Challenges
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

In Acute Dengue Infection, High TIM-3 Expression May Contribute to the Impairment of IFNγ Production by Circulating Vδ2 T Cells

by
Eleonora Cimini
1,
Germana Grassi
1,
Alessia Beccacece
2,
Rita Casetti
1,
Concetta Castilletti
3,
Maria Rosaria Capobianchi
4,5,6,
Emanuele Nicastri
2 and
Chiara Agrati
1,*
1
Laboratory of Cellular Immunology and Pharmacology, National Institute for Infectious Diseases “L. Spallanzani”, Via Portuense 292, 00149 Rome, Italy
2
Clinical Department, National Institute for Infectious Diseases “L. Spallanzani”, Via Portuense 292, 00149 Rome, Italy
3
Laboratory of Virology, National Institute for Infectious Diseases “L. Spallanzani”, Via Portuense 292, 00149 Rome, Italy
4
Department of Epidemiology, Pre-Clinical Research and Advanced Diagnostic, National Institute for Infectious Diseases “L. Spallanzani”, Via Portuense 292, 00149 Rome, Italy
5
Saint Camillus International University of Health Sciences, Via di Sant’Alessandro, 8, 00131 Roma, Italy
6
Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Via Don A. Sempreboni 5, 37024 Negrar di Valpolicella, Italy
*
Author to whom correspondence should be addressed.
Submission received: 23 December 2021 / Revised: 4 January 2022 / Accepted: 7 January 2022 / Published: 12 January 2022
(This article belongs to the Special Issue Gamma Delta T Cells in Immune Response against Viruses)

Abstract

:
γδ T cells are innate cells able to quickly eliminate pathogens or infected/tumoral cells by their antiviral and adjuvancy activities. The role of γδ T cells during Dengue Viral Infection (DENV) infection is not fully elucidated. Nevertheless, human primary γδ T cells have been shown to kill in vitro DENV-infected cells, thus highlighting their possible antiviral function. The aim of this work was to characterize the phenotype and function of Vδ2 T cells in DENV patients. Fifteen DENV patients were enrolled for this study and peripheral blood mononuclear cells (PBMC) were used to analyze Vδ2-T-cell frequency, differentiation profile, activation/exhaustion status, and functionality by multiparametric flow cytometry. Our data demonstrated that DENV infection was able to significantly reduce Vδ2-T-cell frequency and to increase their activation (CD38 and HLA-DR) and exhaustion markers (PD-1 and TIM-3). Furthermore, Vδ2 T cells showed a reduced capability to produce IFN-γ after phosphoantigenic stimulation that can be associated to TIM-3 expression. Several studies are needed to depict the possible clinical impact of γδ-T-cell impairment on disease severity and to define the antiviral and immunoregulatory activities of γδ T cells in the first phases of infection.

1. Introduction

γδ T cells represent unconventional T lymphocytes that are considered as a bridge between innate and adaptive immunity. Human γδ T cells recognize a wide range of self- and non-self antigens, such as soluble proteins and small peptides, prenyl-pyrophosphates, stress- or infection-induced ligands, and MHC class I chain-related protein A and B (MICA/B) [1]. γδ T cells constitute about 1–10% of total circulating lymphocytes in peripheral blood [2] and the two major γδ-T-cell populations are described, respectively, as Vδ1 and Vδ2 T cells [3]. Vδ1 T cells are abundant in tissues such as intestinal and pulmonary epithelium, in skin, and in the female reproductive tract [4]. Vδ2 T cells make up 90% of circulating γδ T cells in peripheral blood in healthy humans and they are able to quickly interact with pathogens or infected/tumoral cells to eliminate them by cytotoxic activity [5,6,7]. γδ T cells play a role in several clinical settings such as autoimmune diseases [8,9,10], allergies [11,12], and cancer [13], and are known to exert a potent antimicrobial activity against both bacterial [14] and viral infections [15,16]. Moreover, human Vδ2 T cells exert their antimicrobial activities through both cytolytic and non-cytolytic mechanisms [16,17,18,19] and can shape several others immune functions (e.g., dendritic cell activation, neutrophil recruitment/activation, Th1 polarization, soluble factor release, and NK-like cytotoxicity) by producing immune-modulating factors [20].
Among the genus Flavivirus, the dengue virus (DENV) belongs to a larger, heterogeneous group of viruses called “Arboviruses”. There are four serotypes, antigenically distinct, of DENV—DEN-1, -2, -3, and -4. Dengue infection severity may depend on several factors and in general results in a flu mild infection. Primary infection with any of the four DENV serotypes could confer long-term protection to the homologous serotype. However, a secondary infection with a different serotype represents one of the major risk factors for severe DENV infection [21]. An impairment of innate immune response has been associated with a more severe DENV infection [22], but the involvement of γδ T cells in protection/pathogenesis is still not well elucidated. In humans, primary γδ T cells can contribute to the immune response during DENV infection by providing an early source of IFN-γ, as well as by killing DENV-infected cells [23], but the modulation of their effector function is not well defined. In this work, we characterized Vδ2 T cells in DENV-infected patients, focusing on their activation and functionality.

2. Materials and Methods

2.1. Cohort of Patients

Patients returning to Italy from endemic countries were enrolled at the National Institute for Infectious Diseases (INMI), L Spallanzani of Rome, after diagnosis of DENV infection. All patients were enrolled before the COVID-19 pandemic. The local ethical committee approved this study (approval number: 9/2020), and all participants gave written informed consent. Fifteen healthy donors (HD) were enrolled as controls. Clinical data of all patients are reported in Table 1.

2.2. Serological and Virological Assays

The DENV diagnosis was performed by using both molecular and serological assays and other arboviral infections (e.g., Zika and Chikungunya viruses) were ruled out. Serum samples were tested by indirect immunofluorescence assay for both IgM and IgG antibodies against ZIKV, DENV, and Chikungunya virus (IFA Arboviral Fever Mosaic 2 IgM and IgG Euroimmun AG, Luebeck, Germany). Molecular assay were performed on serum samples by real-time PCR (RT-PCR) for ZIKV, DENV, and CHIKV RNA [24,25,26]. Virological data are reported in Table 2.

2.3. Peripheral Lymphocyte Isolation

Peripheral blood mononuclear cells (PBMC) of both DENV patients and HD were isolated by gradient centrifugation (Lympholyte, Cedarlane), counted in trypan blue and frozen in FBS (fetal bovine serum, Euroclone) with 10% DMSO (Euroclone).

2.4. Phenotypic and Intracellular Staining

PBMC from peripheral blood of patients and HD were thawed in complete medium (RPMI-1640 supplemented with 10% fetal bovine serum, 2 mmol glutamine, 50 IU/mL penicillin, and 50 µg/mL streptomycin; EuroClone, Pero, Italy) and suspended 1 × 106 cells/mL. Cell viability was analyzed by trypan blue exclusion. Phenotypic analysis was performed by flow cytometry. Specifically, Vδ2 T-cells were analyzed by using the following anti-human monoclonal antibodies: FITC/PE-conjugated anti-Vδ2, PeC5.5-conjugated anti-CD3, APC-A750-conjugated anti-CD27, APC-conjugated anti-CD38, PE-conjugated anti-perforin, PE-conjugated anti-TIM-3, and APC-conjugated anti-IFN-γ (BD Biosciences, San Jose, CA, USA) and V660-conjugated anti-CD45RA, KrO-conjugated anti-CD45, ECD-conjugated anti-HLA-DR, and Viakrome IR885 (Beckman Coulter, Brea, CA, USA). Briefly, thawed PBMC was incubated with mAbs cocktail for 20 min, washed once with buffer (PBS 1×, 0.1% NaN3, 1% BSA) and fixed with 1× para-formaldehyde (PFA, Sigma, Pero, Italy). Two functional tests were performed by flow cytometry. The cytotoxic potential was tested by analyzing the perforin content by intracellular staining of fixed PBMC. The IFN-γ production by Vδ2 T cells was analyzed after stimulation of PBMC with or without phosphoantigen (PhAg, IPH1101, 3 µM) in the presence of Brefeldin A (10 µg/mL, Serva). Intracellular staining was performed on fixed samples by staining cells with IFN-γ or perforin mAbin buffer (PBS 1×, 0.1% NaN3, 1% BSA, saponin 0.5%) for 20 min at room temperature. After incubation, cells were washed once with buffer (PBS 1×, 0.1% NaN3, 1% BSA, saponin 0.1%). Sample acquisition and data analysis were performed by Cytoflex Flow Cytometer (Beckman Coulter). Data were analyzed with Cytoflex software (BC). Results are shown as box and whiskers—the box encompasses the interquartile range of individual measurements, the horizontal bar-dividing line indicates the median value, and the whiskers represent maximum and minimum values.

2.5. Statistical Analysis

Statistical significance of results was determined by Graph Pad Prism software. Statistical analysis was performed using the non-parametric Mann–Whitney assay and differences were considered significant when the p value was less than 0.05.

3. Results

To investigate the impact of DENV infection on circulating Vδ2 T cells, we characterized their frequency and differentiation/activation/exhaustion profile and their functionality by multiparametric flow cytometry. A significant reduction of Vδ2-T-cell frequency was observed in DENV patients (DENV vs. HD p < 0.05) (Figure 1A). In parallel, a significant Vδ2 T-cell activation was observed, as shown by a significant increase of CD38 (DENV vs. HD, p < 0.0001) and of HLA-DR (DENV vs. HD, p < 0.0001) expression (Figure 1B,C). In contrast, the differentiation profile of Vδ2 T cells showed no significant differences between patients and HD (data not shown), in accordance with our previous work [27].
We then investigated a possible impact of DENV infection on the expression of exhaustion markers on Vδ2 T cells. Results showed a significantly higher PD-1 (DENV vs. HD, p < 0.0002) and TIM-3 (DENV vs. HD, p < 0.0029) marker expression in DENV patients compared to HD (Figure 1D,E). The functional analysis revealed a significant enrichment of perforin-positive Vδ2 T cells (DENV vs. HD, p < 0.0001) and a reduced frequency of IFN-γ-producing Vδ2 T cells after PhAg stimulation (DENV vs. HD, p < 0.0001) when compared to controls. Finally, we evaluated a possible involvement of exhaustion marker expression in reducing the ability to produce IFN-γ after specific stimulation. A significant negative correlation was observed between the frequency of IFN-γ-producing Vδ2 T cells and TIM-3 expression (R: −0.44, p < 0.01, Figure 1H) but not with PD-1 marker expression (R: −0.38, p = 0.06).

4. Discussion

DENV infection is one of the major emerging infectious diseases. Despite a large amount of data on humoral and adaptive T-cell response against DENV, very few data are currently available about the innate immune response. During DENV infection, several innate immune pathways are activated, including type-I interferon, complement, apoptosis, and autophagy [28]. Moreover, the effective functionality of innate cells is associated with milder DENV infection, suggesting a protective role of these innate cells. In DENV-infected patients, an early activation of NK cells was also reported, which was clearly associated with mild DENV diseases [29], suggesting that an early and effective activation of innate cells may be a crucial step in controlling viral replication and disease severity. Indeed, severe infection showed a dysfunctional NK response characterized by a reduction of both IFN-γ and cytotoxic markers [30].
Few data are available about the involvement of γδ T cells in the protection/pathogenesis of DENV infection. γδ T cells killed in vitro DENV-infected cells [23] and contributed to virus clearance in an in vivo mouse model [31], suggesting the ability to mediate a protective immunity. Our results showed that γδ T cells responded to acute DENV infection increasing the expression of activation markers, thus confirming previous published data [30]. Moreover, the γδ-T-cell activation was paralleled by an increase in the expression of exhaustion markers (PD-1 and TIM-3), suggesting a possible induction of a dysfunctional state of these cells. The co-expression of both activation and exhaustion markers in immune cells is well demonstrated in other infections such as Ebola [32,33] and SARS-CoV2 [34], and suggests an over-activation-induced immune impairment. The inhibitory receptor PD-1 exerts a wide range of immunoregulatory roles in T-cell activation and tolerance [35] and represents a marker to define T-cell exhaustion [36]. Accordingly, the PD-1 expression on γδ T cells strongly reduces their effector functions [37]. A similar inhibitory function was also exerted by TIM-3 which is up-regulated both on αβ and γδ T cells after activation [38,39]. Several observations demonstrated a role of both PD-1 and TIM-3 in reducing cytokine production and cytotoxicity of γδ T cells both in viral infection and in cancer [40].
The impact of DENV on γδ-T-cell function has been studied in vitro where primary γδ T cells responded rapidly to autologous DENV-infected dendritic cells by secreting IFN-γ and upregulating CD107a [23]. In contrast, we showed that during acute DENV infection these two functions seemed to be not coordinated, with an effective production of perforin but an impaired IFN-γ production. Indeed, a significant reduction of IFN-γ-producing γδ T cells after PhAg stimulation was shown and was inversely correlated with TIM-3 expression. Accordingly, a role of TIM-3 in γδ-T-cell exhaustion was also shown in malaria patients where the high expression of TIM-3 was linked to a reduced Vδ2-T-cell pro-inflammatory cytokine production [41] and in cancer patients was associated to a reduced killing activity [42]. The direct involvement of TIM-3 in reducing Vδ2-T-cell functionality and its association with the clinical course of the diseases needs further confirmation.
A protective role of γδ T cells was also reported in Zika infection where a massive expansion of effector/activated Vδ2 T-cells occurred [24]. These cells showed, in vitro, a killing capability against ZIKV-infected cells through NKG2D/NKG2DL interaction [5], suggesting a protective role by a TCR-independent mechanism.
Together, these data showed that, on the one hand, DENV infection induced Vδ2 T-cell activation and perforin production but, on the other hand, increased TIM-3 expression, which was associated with a significant reduction of IFN-γ after TCR-mediated stimulation. Several other studies are needed to define a possible association between γδ-T-cell functionality and viral severity.

Author Contributions

Conceptualization, C.A., E.N. and E.C.; methodology, E.C., C.C. and G.G.; software, R.C.; validation, C.A. and E.C.; formal analysis, C.A.; investigation, E.C. and A.B.; data curation, E.C.; writing—original draft preparation, E.C., C.A. and A.B.; writing—review and editing, E.C. and C.A.; visualization, all authors; supervision, E.N.; funding acquisition, E.N. and M.R.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work supported by fundings from the Italian Ministry of Health to INMI L Spallanzani-IRCCS: RC, Linea 1 and RF-2016-02364155.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board (or Ethics Committee) of The National Institute for Infectious Diseases Lazzaro Spallanzani (protocol code: number 3 of 23 December 2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are openly available at http://rawdata.inmi.it, accessed date on 6 January 2022.

Acknowledgments

We thank the patients and the medical and nursing staff of the National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS for their contribution to this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Caron, J.; Ridgley, L.A.; Bodman-Smith, M. How to Train Your Dragon: Harnessing Gamma Delta T Cells Antiviral Functions and Trained Immunity in a Pandemic Era. Front. Immunol. 2021, 12, 666983. [Google Scholar] [CrossRef]
  2. Nielsen, M.M.; Witherden, D.A.; Havran, W.L. γδ T cells in homeostasis and host defence of epithelial barrier tissues. Nat. Rev. Immunol. 2017, 17, 733–745. [Google Scholar] [CrossRef] [PubMed]
  3. Parker, C.M.; Groh, V.; Band, H.; Porcelli, S.A.; Morita, C.; Fabbi, M.; Glass, D.J.; Strominger, J.L.; Brenner, M.B. Evidence for extrathymic changes in the T cell receptor gamma/delta repertoire. J. Exp. Med. 1990, 171, 1597–1612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Khairallah, C.; Chu, T.H.; Sheridan, B.S. Tissue Adaptations of Memory and Tissue-Resident Gamma Delta T Cells. Front. Immunol. 2018, 9, 2636. [Google Scholar] [CrossRef] [PubMed]
  5. Cimini, E.; Sacchi, A.; De Minicis, S.; Bordoni, V.; Casetti, R.; Grassi, G.; Colavita, F.; Castilletti, C.; Capobianchi, M.R.; Ippolito, G.; et al. Vδ2 T-Cells Kill ZIKV-Infected Cells by NKG2D-Mediated Cytotoxicity. Microorganisms 2019, 7, 350. [Google Scholar] [CrossRef] [Green Version]
  6. Cimini, E.; Bordoni, V.; Sacchi, A.; Visco-Comandini, U.; Montalbano, M.; Taibi, C.; Casetti, R.; Lalle, E.; D’Offizi, G.; Capobianchi, M.; et al. Intrahepatic Vγ9Vδ2 T-cells from HCV-infected patients show an exhausted phenotype but can inhibit HCV replication. Virus Res. 2018, 243, 31–35. [Google Scholar] [CrossRef]
  7. Agrati, C.; Castilletti, C.; Cimini, E.; Romanelli, A.; Lapa, D.; Quartu, S.; Martini, F.; Capobianchi, M.R. Antiviral activity of human Vδ2 T-cells against WNV includes both cytolytic and non-cytolytic mechanisms. New Microbiol. 2016, 39, 139–142. [Google Scholar]
  8. Bank, I. The Role of Gamma Delta T Cells in Autoimmune Rheumatic Diseases. Cells 2020, 9, 462. [Google Scholar] [CrossRef] [Green Version]
  9. Zarobkiewicz, M.K.; Kowalska, W.; Roliński, J.; Bojarska-Junak, A.A. γδ T lymphocytes in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2019, 330, 67–73. [Google Scholar] [CrossRef] [PubMed]
  10. Shiromizu, C.M.; Jancic, C.C. γδ T Lymphocytes: An Effector Cell in Autoimmunity and Infection. Front. Immunol. 2018, 9, 2389. [Google Scholar] [CrossRef]
  11. Castillo-González, R.; Cibrian, D.; Sánchez-Madrid, F. Dissecting the complexity of γδ T-cell subsets in skin homeostasis, inflammation, and malignancy. J. Allergy Clin. Immunol. 2021, 147, 2030–2042. [Google Scholar] [CrossRef] [PubMed]
  12. Zarobkiewicz, M.K.; Wawryk-Gawda, E.; Kowalska, W.; Janiszewska, M.; Bojarska-Junak, A. γδ T Lymphocytes in Asthma: A Complicated Picture. Arch. Immunol. Ther. Exp. 2021, 69, 4. [Google Scholar] [CrossRef]
  13. Miyashita, M.; Shimizu, T.; Ashihara, E.; Ukimura, O. Strategies to Improve the Antitumor Effect of γδ T Cell Immunotherapy for Clinical Application. Int. J. Mol. Sci. 2021, 22, 8910. [Google Scholar] [CrossRef] [PubMed]
  14. Gao, Y.; Williams, A.P. Role of Innate T Cells in Anti-Bacterial Immunity. Front. Immunol. 2015, 6, 302. [Google Scholar] [CrossRef] [Green Version]
  15. Sabbaghi, A.; Miri, S.M.; Keshavarz, M.; Mahooti, M.; Zebardast, A.; Ghaemi, A. Role of gammadelta T cells in controlling viral infections with a focus on influenza virus: Implications for designing novel therapeutic approaches. Virol. J. 2020, 17, 174. [Google Scholar] [CrossRef]
  16. Poccia, F.; Agrati, C.; Martini, F.; Mejia, G.; Wallace, M.; Malkovsky, M. Vγ9Vδ2 T cell-mediated non-cytolytic antiviral mechanisms and their potential for cell-based therapy. Immunol. Lett. 2005, 100, 14–20. [Google Scholar] [CrossRef] [PubMed]
  17. Agrati, C.; Castilletti, C.; De Santis, R.; Cimini, E.; Bordi, L.; Malkovsky, M.; Poccia, F.; Capobianchi, M.R. Interferon-γ–Mediated Antiviral Immunity against Orthopoxvirus Infection Is Provided by γδ T Cells. J. Infect. Dis. 2006, 193, 1606–1607. [Google Scholar] [CrossRef] [Green Version]
  18. Agrati, C.; Alonzi, T.; De Santis, R.; Castilletti, C.; Abbate, I.; Capobianchi, M.R.; D’Offizi, G.; Siepi, F.; Fimia, G.M.; Tripodi, M.; et al. Activation of Vγ9Vδ2 T cells by non-peptidic antigens induces the inhibition of subgenomic HCV replication. Int. Immunol. 2005, 18, 11–18. [Google Scholar] [CrossRef]
  19. Poccia, F.; Agrati, C.; Castilletti, C.; Bordi, L.; Gioia, C.; Horejsh, D.; Ippolito, G.; Chan, P.; Hui, D.; Sung, J.J.Y.; et al. Anti–Severe Acute Respiratory Syndrome Coronavirus Immune Responses: The Role Played by Vγ9Vδ2 T Cells. J. Infect. Dis. 2006, 193, 1244–1249. [Google Scholar] [CrossRef]
  20. Agrati, C.; D’Offizi, G.; Gougeon, M.-L.; Malkovsky, M.; Sacchi, A.; Casetti, R.; Bordoni, V.; Cimini, E.; Martini, F. Innate gamma/delta T-cells during HIV infection: Terra relatively Incognita in novel vaccination strategies? AIDS Rev. 2011, 13, 3–12. [Google Scholar] [PubMed]
  21. Jayachandran, B.; Chanda, K.; Balamurali, M.M. Overview of Pathogenesis, Diagnostics, and Therapeutics of Infectious Diseases: Dengue and Zika. ACS Omega 2021, 6, 22487–22496. [Google Scholar] [CrossRef]
  22. Guzman, M.; Gubler, D.; Izquierdo, A.; Martinez, E.; Halstead, S.B. Dengue infection. Nat. Rev. Dis. Primers 2016, 2, 16055. [Google Scholar] [CrossRef]
  23. Tsai, C.-Y.; Liong, K.H.; Gunalan, M.G.; Liang, L.D.S.; Lim, D.S.L.; Fisher, D.A.; Macary, P.A.; Leo, Y.S.; Wong, S.-C.; Puan, K.J.; et al. Type I IFNs and IL-18 Regulate the Antiviral Response of Primary Human γδ T Cells against Dendritic Cells Infected with Dengue Virus. J. Immunol. 2015, 194, 3890–3900. [Google Scholar] [CrossRef]
  24. CDC DENV-1-4 Real-Time RT-PCR Assay. Available online: https://www.cdc.gov/dengue/healthcare-providers/testing/molecular-tests/realtime.html (accessed on 7 April 2016).
  25. Faye, O.; Faye, O.; Diallo, D.; Diallo, M.; Weidmann, M.; Sall, A.A. Quantitative real-time PCR detection of Zika virus and evaluation with field-caught Mosquitoes. Virol. J. 2013, 10, 311. [Google Scholar] [CrossRef] [Green Version]
  26. Pastorino, B.; Bessaud, M.; Grandadam, M.; Murri, S.; Tolou, H.J.; Peyrefitte, C.N. Development of a TaqMan® RT-PCR assay without RNA extraction step for the detection and quantification of African Chikungunya viruses. J. Virol. Methods 2005, 124, 65–71. [Google Scholar] [CrossRef]
  27. Cimini, E.; Castilletti, C.; Sacchi, A.; Casetti, R.; Bordoni, V.; Romanelli, A.; Turchi, F.; Martini, F.; Tumino, N.; Nicastri, E.; et al. Human Zika infection induces a reduction of IFN-γ producing CD4 T-cells and a parallel expansion of effector Vδ2 T-cells. Sci. Rep. 2017, 7, 6313. [Google Scholar] [CrossRef]
  28. Uno, N.; Ross, T.M. Dengue virus and the host innate immune response. Emerg. Microbes Infect. 2018, 7, 167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Epetitdemange, C.; Ewauquier, N.; Erey, J.; Ehervier, B.; Leroy, E.; Evieillard, V. Control of Acute Dengue Virus Infection by Natural Killer Cells. Front. Immunol. 2014, 5, 209. [Google Scholar] [CrossRef] [Green Version]
  30. Azeredo, E.L.; De Oliveira-Pinto, L.M.; Zagne, S.M.; Cerqueira, D.I.S.; Nogueira, R.M.R.; Kubelka, C.F. NK cells, displaying early activation, cytotoxicity and adhesion molecules, are associated with mild dengue disease. Clin. Exp. Immunol. 2006, 143, 345–356. [Google Scholar] [CrossRef] [PubMed]
  31. Mantri, C.K.; John, A.L.S. Immune synapses between mast cells and γδ T cells limit viral infection. J. Clin. Investig. 2019, 129, 1094–1108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Agrati, C.; Castilletti, C.; Casetti, R.; Sacchi, A.; Falasca, L.; Turchi, F.; Tumino, N.; Bordoni, V.; Cimini, E.; Viola, D.; et al. Longitudinal characterization of dysfunctional T cell-activation during human acute Ebola infection. Cell Death Dis. 2016, 7, e2164. [Google Scholar] [CrossRef] [Green Version]
  33. Cimini, E.; Viola, D.; Cabeza-Cabrerizo, M.; Romanelli, A.; Tumino, N.; Sacchi, A.; Bordoni, V.; Casetti, R.; Turchi, F.; Martini, F.; et al. Different features of Vδ2 T and NK cells in fatal and non-fatal human Ebola infections. PLoS Negl. Trop. Dis. 2017, 11, e0005645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. De Biasi, S.; Meschiari, M.; Gibellini, L.; Bellinazzi, C.; Borella, R.; Fidanza, L.; Gozzi, L.; Iannone, A.; Tartaro, D.L.; Mattioli, M.; et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat. Commun. 2020, 11, 3434. [Google Scholar] [CrossRef] [PubMed]
  35. Jin, H.-T.; Ahmed, R.; Okazaki, T. Role of PD-1 in Regulating T-Cell Immunity. Curr. Top. Microbiol. Immunol. 2010, 350, 17–37. [Google Scholar] [CrossRef]
  36. He, X.; Xu, C. PD-1: A Driver or Passenger of T Cell Exhaustion? Mol. Cell 2020, 77, 930–931. [Google Scholar] [CrossRef] [PubMed]
  37. Iwasaki, M.; Tanaka, Y.; Kobayashi, H.; Murata-Hirai, K.; Miyabe, H.; Sugie, T.; Toi, M.; Minato, N. Expression and function of PD-1 in human γδ T cells that recognize phosphoantigens. Eur. J. Immunol. 2010, 41, 345–355. [Google Scholar] [CrossRef] [Green Version]
  38. Avery, L.; Filderman, J.; Szymczak-Workman, A.L.; Kane, L.P. Tim-3 co-stimulation promotes short-lived effector T cells, restricts memory precursors, and is dispensable for T cell exhaustion. Proc. Natl. Acad. Sci. USA 2018, 115, 2455–2460. [Google Scholar] [CrossRef] [Green Version]
  39. Han, G.; Chen, G.; Shen, B.; Li, Y. Tim-3: An Activation Marker and Activation Limiter of Innate Immune Cells. Front. Immunol. 2013, 4, 449. [Google Scholar] [CrossRef] [Green Version]
  40. Catafal-Tardos, E.; Baglioni, M.V.; Bekiaris, V. Inhibiting the Unconventionals: Importance of Immune Checkpoint Receptors in γδ T, MAIT, and NKT Cells. Cancers 2021, 13, 4647. [Google Scholar] [CrossRef]
  41. Schofield, L.; Ioannidis, L.J.; Karl, S.; Robinson, L.J.; Tan, Q.Y.; Poole, D.P.; Betuela, I.; Hill, D.L.; Siba, P.M.; Hansen, D.S.; et al. Synergistic effect of IL-12 and IL-18 induces TIM3 regulation of γδ T cell function and decreases the risk of clinical malaria in children living in Papua New Guinea. BMC Med. 2017, 15, 114. [Google Scholar] [CrossRef]
  42. Li, X.; Lu, H.; Gu, Y.; Zhang, X.; Zhang, G.; Shi, T.; Chen, W. Tim-3 suppresses the killing effect of Vγ9Vδ2 T cells on colon cancer cells by reducing perforin and granzyme B expression. Exp. Cell Res. 2020, 386, 111719. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Phenotypic and functionality characteristics of γδ T cells. Frequency (A) activation markers (B,C: CD38, HLA-DR), exhaustion markers (D,E: PD-1, TIM-3), and functionality (F,G: perforin, IFN-γ) were performed on PBMC of DENV patients and HD by multiparametric flow cytometry. Correlations (H,I: IFN-γ/PD-1; IFN-γ/TIM-3) were performed by Spearman test for non-parametric data. *: p < 0.05; **: p < 0.01; ***: p < 0.001; ****: p < 0.0001.
Figure 1. Phenotypic and functionality characteristics of γδ T cells. Frequency (A) activation markers (B,C: CD38, HLA-DR), exhaustion markers (D,E: PD-1, TIM-3), and functionality (F,G: perforin, IFN-γ) were performed on PBMC of DENV patients and HD by multiparametric flow cytometry. Correlations (H,I: IFN-γ/PD-1; IFN-γ/TIM-3) were performed by Spearman test for non-parametric data. *: p < 0.05; **: p < 0.01; ***: p < 0.001; ****: p < 0.0001.
Viruses 14 00130 g001
Table 1. Characteristics of 15 DENV patients included in the study.
Table 1. Characteristics of 15 DENV patients included in the study.
Age
(Mean ± SD)
(Years)
Gender (M/F)Travel History (pts)Symptom
Onset
(Range, Days)
SymptomsDENV
RT-PCR
DENV Serotype
42 ± 116/9India (2 pts)
Thailand (2 pts)
Philippines (2 pts)
Singapore (1 pts)
Maldives (3 pts)
Brazil (1 pts)
Indonesia (2 pts)
Jamaica (1 pts)
Cuba (1 pts)
2–5Cutaneous rash: 8/15
Fever: 15/15
Headache: 11/15
Arthralgia: 11/15
Positive: 15
Negative: 0
Ser 1: 4
Ser 2: 3
Ser 3: 5
Ser 4: 0
ND: 3
M, male; F, female; pts, patients; DENV, dengue virus; ND, not determined.
Table 2. Virological data of 15 DENV patients included in the study.
Table 2. Virological data of 15 DENV patients included in the study.
Patient ID
(Gender/DENV Serotype)
Flavivirus PCRZIKV RT-PCRDENV RT-PCRCHIKV RT-PCRAnti-ZIKV IgGAnti-ZIKV IgMAnti-DENV IgGAnti-DENV IgMAnti-CHIKV IgGAnti-CHIKV IgM
PT1
(F/DENV 1)
PositiveNDPositiveNDNDND<1:20<1:20NDND
PT2
(F/DENV ND)
PositiveNDPositiveNDNDND1:201:40NDND
PT3
(F/DENV 3)
PositiveNegativePositiveNegative<1:20<1:201:801:80<1:20<1:20
PT4 (F/DENV 3)PositiveNegativePositiveNegative<1:20<1:201:801:160<1:20<1:20
PT5
(M/DENV 1)
PositiveNegativePositiveNegative<1:20<1:201:201:80<1:20<1:20
PT6
(M/DENV 3)
PositiveNegativePositiveNegative<1:20<1:20<1:201:20<1:20<1:20
PT7
(M/DENV ND)
PositiveNegativePositiveNegative<1:20<1:201:401:80<1:20<1:20
PT8
(F/DENV ND)
PositiveNegativePositiveNegative<1:20<1:201:160<1:20<1:20<1:20
PT9
(F/DENV 1)
PositiveNegativePositiveNegative<1:20<1:20<1:20<1:20<1:20<1:20
PT10
(F/DENV 3)
PositiveNegativePositiveNegative<1:20<1:20<1:80<1:40<1:20<1:20
PT11
(M/DENV 1)
PositiveNDPositiveNegativeNDND1:320<1:20<1:20<1:20
PT12
(M/DENV 2)
PositiveNegativePositiveNegative<1:20<1:20<1:20<1:40<1:20<1:20
PT13
(F/DENV 3)
PositiveNDPositiveNegative<1:20<1:20<1:20<1:20<1:20<1:20
PT14
(M/DENV 2)
PositiveNegativePositiveNegative<1:20<1:20<1:20<1:20<1:20<1:20
PT15
(F/DENV 2)
PositiveNegativePositiveNegative<1:20<1:20<1:20<1:20<1:20<1:20
M, male; F, female; DENV, dengue virus; ZIKV, Zika virus; CHIKV, Chikungunya virus; ND, not done. RT-PCR, Real Time PCR.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cimini, E.; Grassi, G.; Beccacece, A.; Casetti, R.; Castilletti, C.; Capobianchi, M.R.; Nicastri, E.; Agrati, C. In Acute Dengue Infection, High TIM-3 Expression May Contribute to the Impairment of IFNγ Production by Circulating Vδ2 T Cells. Viruses 2022, 14, 130. https://0-doi-org.brum.beds.ac.uk/10.3390/v14010130

AMA Style

Cimini E, Grassi G, Beccacece A, Casetti R, Castilletti C, Capobianchi MR, Nicastri E, Agrati C. In Acute Dengue Infection, High TIM-3 Expression May Contribute to the Impairment of IFNγ Production by Circulating Vδ2 T Cells. Viruses. 2022; 14(1):130. https://0-doi-org.brum.beds.ac.uk/10.3390/v14010130

Chicago/Turabian Style

Cimini, Eleonora, Germana Grassi, Alessia Beccacece, Rita Casetti, Concetta Castilletti, Maria Rosaria Capobianchi, Emanuele Nicastri, and Chiara Agrati. 2022. "In Acute Dengue Infection, High TIM-3 Expression May Contribute to the Impairment of IFNγ Production by Circulating Vδ2 T Cells" Viruses 14, no. 1: 130. https://0-doi-org.brum.beds.ac.uk/10.3390/v14010130

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

Article Metrics

Back to TopTop