Skip to main content

MINI REVIEW article

Front. Immunol., 14 February 2019
Sec. T Cell Biology
This article is part of the Research Topic Understanding gamma delta T Cell Multifunctionality - Towards Immunotherapeutic Applications View all 27 articles

Human Vδ1+ T Cells in the Immune Response to Plasmodium falciparum Infection

  • 1Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
  • 2Centre for Medical Parasitology, Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
  • 3Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Ghana
  • 4Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom

Naturally acquired protective immunity to Plasmodium falciparum malaria is mainly antibody-mediated. However, other cells of the innate and adaptive immune system also play important roles. These include so-called unconventional T cells, which express a γδ T-cell receptor (TCR) rather than the αβ TCR expressed by the majority of T cells—the conventional T cells. The γδ T-cell compartment can be divided into distinct subsets. One expresses a TCR involving Vγ9 and Vδ2, while another major subset uses instead a TCR composed of Vδ1 paired with one of several types of γ chains. The former of these subsets uses a largely semi-invariant TCR repertoire and responds in an innate-like fashion to pyrophosphate antigens generated by various stressed host cells and infectious pathogens, including P. falciparum. In this short review, we focus instead on the Vδ1 subset, which appears to have a more adaptive immunobiology, but which has been much less studied in general and in malaria in particular. We discuss the evidence that Vδ1+ cells do indeed play a role in malaria and speculate on the function and specificity of this cell type, which is increasingly attracting the attention of immunologists.

Introduction

The most serious form of malaria is caused by the hemoprotozoan parasite Plasmodium falciparum. The disease is a major humanitarian and economic burden on societies affected by it, mainly in sub-Saharan Africa, and it leads to the death of about half a million children every year (1, 2). Immunity to the disease is gradually acquired after years of exposure and many disease episodes, and is mainly mediated by IgG antibodies targeting the asexual blood stages of the infection, which are responsible for all the clinical symptoms and complications (35). T cells are nevertheless also of obvious importance in acquisition of immunity, not least to enable B-cell class switching and affinity maturation.

Most circulating T cells express αβ type T-cell receptors (TCR-αβ), but a minority of T cells instead expresses the alternative γδ TCR heterodimer (TCR-γδ). The pivotal role of αβ T cells in immunity to P. falciparum malaria is well-established. The αβ T cells function both directly as cytotoxic effector cells against infected hepatocytes, and indirectly as CD4+ helper cells for a variety of innate and adaptive immune responses to all stages of the parasite life cycle in the human host. Much less is known about the function and significance of γδ T cells in this immunity.

The αβ and γδ T-cell compartments share several features. In both, the TCR constitutes the antigen recognition element of the multi-molecular TCR complex, which also includes several signal transduction components, such as CD3. TCR diversity is generated by somatic recombination events during T-cell maturation in the thymus. As for αβ T cells, the TCRs of γδ T cells are clonally distributed, such that each T-cell clone expresses a single, rearranged TCR variant, which determines the antigen specificity of the clone—at least in the case of αβ T cells.

The two compartments also exhibit important differences. Thus, αβ T cells respond predominantly to protein antigens that are processed by antigen-presenting cells (APCs) and subsequently displayed as short peptides bound to major histocompatibility complex (MHC) molecules on the APC surface. In contrast to αβ T cells, which typically express either CD4 or CD8, γδ T cells often express neither, in particular in the Vγ9+Vδ2+ subset. In keeping with this lack of MHC restriction elements, recognition of antigen by “double-negative” γδ T cells is not MHC-restricted. Furthermore, Vγ9+Vδ2+ T cells universally respond to non-peptide prenyl pyrophosphate metabolites (termed phospho-antigens, or P-Ag) (6). These antigens, which are produced by a variety of stressed cells (isopentenyl pyrophosphate, IPP, produced via the host mevalonate pathway) and by infectious pathogens, including P. falciparum [(E)-4-Hydroxy-3-methyl-but-2-enyl pyrophosphate, HMB-PP, produced via the microbial non-mevalonate pathway] are structurally related. Accordingly, the Vγ9 chains expressed by these cells are relatively invariant (7, 8) due to convergent and recurrent recombinations (9). In addition, the Vγ9+Vδ2+ TCR repertoire is already restricted from birth, and contains a high proportion of Vγ9 clonotypes that are shared by many clones in a given individual, and conserved between many individuals (i.e., “public” repertoires). Furthermore, the repertoire of these cells does not exhibit dramatic clonotypic focusing in adults relative to neonates (9, 10). The Vγ9+Vδ2+ T-cell subset, which is usually the dominant γδ T-cell subset in the peripheral blood of healthy individuals without exposure to P. falciparum, can thus be described as an “innate-like” T-cell subset.

To date, the Vγ9+Vδ2+ cells are the γδ T cells that have attracted by far the most attention in relation to malaria (11, 12). However, we focus here instead on a largely complementary subset that is characterized by a TCR composed of Vδ1 paired with a variety of γ-chains, and that appears to adopt a distinct immunobiology relative to the innate-like Vγ9+Vδ2+ subset (13). Unlike Vγ9+Vδ2+ T cells, Vδ1+ T cells typically constitute a minority (≤20%) of adult peripheral blood γδ T cells. However, the subset is enriched relative to the Vγ9+Vδ2+ T-cell subset in tissues, where they have been reported to recognize a variety of host and microbial antigens (1416). Also in marked contrast, the TCR repertoire of Vδ1+ T cells—and of Vγ9negVδ2+ T cells (17)—is highly diverse at birth, and largely non-overlapping between individuals (i.e., “private” repertoires). Furthermore, the TCR repertoire of this γδ T-cell subset becomes increasingly focused over time as a result of selective expansion of specific clonotypes, most likely following antigenic stimulation (9, 1820). The Vδ1 subset therefore appears to be much more “adaptive-like” than the Vγ9+Vδ2+ subset (21), and it bears substantial similarities to conventional αβ T cells. Nevertheless, there is certainly evidence that Vδ1+ T cells play a distinct role from αβ T cells in the immune response to several infections—including P. falciparum malaria.

Increased Proportions and Numbers of Vδ1+ T Cells in Malaria Patients and Healthy Residents From Malaria-Endemic Areas

Within a few years of the discovery of the γδ TCR, several groups reported modest but protracted expansions of γδ T cells in adult P. falciparum and P. vivax patients with little or no previous malaria parasite exposure (2224). A later study of malarious children from a highly malaria-endemic area and employing a pan-γδ TCR-specific antibody reported similar findings, and did not find significant differences in peripheral blood γδ T-cell frequencies between children with uncomplicated and severe malaria, respectively (25). The authors also reported significantly decreased absolute numbers of γδ T cells at the time of admission to hospital with malaria (regardless of severity), followed by a transient increase to numbers above normal during convalescence. This was also observed among the few adult first-time malaria patients included in the study (25). Overall, the γδ T cell-specific findings appeared similar in patients with or without prior exposure to malaria, and also resembled earlier reports regarding the αβ T-cell response to malaria, namely an inflammation-induced withdrawal of these cells from the peripheral circulation, followed by their release back into the peripheral blood after successful chemotherapy [reviewed in Hviid (26)].

Substantial γδ T-cell subset heterogeneity was also reported (2730). These early papers indicated that the γδ T-cell response to P. falciparum malaria extends beyond Vγ9+Vδ2+ cells, although that subset remained the dominant one among the non-immune patients that were studied. However, it was reported shortly after that in semi-immune African children and adults with acute P. falciparum malaria, the γδ T cells responding in vivo are completely dominated by cells expressing Vδ1, with little contribution from Vγ9+Vδ2+ T cells (31, 32). A study of children and adults from P. falciparum-endemic Lao People's Democratic Republic very recently reported similar findings (33). The expanded Vδ1+ subset had an activated phenotype, produced pro-inflammatory cytokines, used a diversity of Vγ chains, and showed spectratyping evidence of clonal focusing (3133). In fact, the Vδ1+ subset appeared to dominate even among healthy P. falciparum-exposed individuals living in areas with stable transmission of these parasites (20, 34). In the absence of acute malaria, these cells were CD45RA+, resting (CD69neg and HLA-DRneg), and about half of them were CD8+ (in contrast to the majority of Vγ9+Vδ2+ cells, which are double-negative). They were clonally restricted in most adults, but less so in children (20). They thus appear phenotypically similar to the Vδ1+ cells found in epithelia (35). While Vγ9+Vδ2+ cells from such individuals could respond when stimulated in vitro by P. falciparum pyrophosphate antigens (34)—similar to Vγ9+Vδ2+ cells from donors without previous malaria exposure [reviewed in Howard et al. (11)]—this response did not appear very prominent in vivo.

Vδ1+ T Cells in Malaria: What Do They See and What Do They Do?

Essentially nothing is known about the function or antigen specificity/specificities of the dominant Vδ1+ γδ T-cell subset in P. falciparum-exposed individuals (12). A few studies have indicated that these cells might recognize, respond to, and have a direct effector function against infected erythrocytes in a manner resembling Vγ9+Vδ2+ cells (11, 33, 36). However, already early on May Ho and colleagues speculated that the expansion of Vδ1+ T cells in P. falciparum malaria might instead involve “unidentified host factors” (29). Their prediction is supported by the findings that Vδ1+ cells from parasite-exposed individuals do not respond markedly to P. falciparum antigens in vitro (34), including the parasite-derived pyrophosphate antigens recognized by Vγ9+Vδ2+ cells (37, 38).

Although it is not known what drives the expansion and differentiation of the adaptive-like Vδ1+ subset in malaria, Vδ1+ T-cell expansion has been observed in several other pathological conditions (16). Examples include infections with human immunodeficiency virus (HIV) (3942), cytomegalovirus (CMV) and other herpes viruses (4345), Onchocerca volvulus parasites (46), as well as autoimmune diseases such as Takayasu arteritis (47), inflammatory bowel disease and Crohn's disease (48, 49). The possibility that the Vδ1+ T-cell response in these diseases involves recognition of host-encoded components is supported by studies of CMV. In that infection, Vδ2neg T cells display shared reactivity against both CMV-infected target cells and uninfected epithelial cells, consistent with recognition of host-encoded antigens (50). Moreover, endothelial protein C receptor (EPCR) has been identified as an antigenic target for a Vδ2neg γδ TCR expressed by a clonotype heavily expanded after infection with CMV (51), which is known to infect endothelial tissues. T-cell activation was dependent on integration of TCR/EPCR-mediated signals with a TCR-extrinsic “multi-molecular stress signature” induced upon infection of target cells that included CMV-mediated increases in ICAM-1 and LFA-1 expression. Conceivably, this may represent one route for Vδ2neg γδ T-cell recognition of “stressed self.” It may be of interest in the context of malaria that EPCR has been identified as a clinically important receptor for P. falciparum-infected erythrocytes (52, 53).

Dysregulation of the B-cell compartment might constitute another pathogen-induced change that could be sensed by “adaptive-like” γδ T cells. Of relevance, P. falciparum malaria, and indeed a number of other diseases associated with Vδ1+ T-cell expansions, is characterized by massive B-cell activation, both of B cells that are specific for the infection causing the disease and B cells that are not (54, 55). This often leads to reactivation of latent EBV (and CMV) infection, and further B-cell proliferation (5658). From this perspective, it is tempting to speculate that the selective expansion of Vδ1+ T cells observed in individuals living in areas with stable transmission of P. falciparum occurs in response to antigens expressed by activated B cells, perhaps serving as part of an auto-regulatory response to curb excessive B-cell activation and proliferation. In addition, Vδ1+ cells can recognize EBV-transformed B-cell lines (59, 60), and EBV infection can result in expansion of clonally restricted Vδ1+ cell populations after stem cell transplantation (61, 62). Conceivably, CD1c/TCT.1/Blast-1 might be an antigen recognized by these cells. Thus, CD8+ Vδ1+ cells heavily expanded in vitro have been shown to recognize this antigen (63, 64), which is expressed/upregulated on some activated and transformed B cells (65, 66). This is not least the case in the spleen, where Vδ1+ cells are also abundant (67), and further increase in numbers in response to P. falciparum malaria (68). In addition, Vδ1+ T-cell reactivity to CD1c tetramers has been demonstrated (69), although to date only involving a low percentage of the Vδ1 T-cell repertoire. It therefore remains unclear whether CD1c-specific cells overlap with in vivo expanded clonotypes (21). In summary, while other possibilities cannot be discounted, responses to “stressed self” via recognition of host antigens may contribute to Vδ1-mediated adaptive surveillance in the context of malaria, which could be linked to immune, stress-linked, or EBV/CMV-related sequelae of parasite infection. Such adaptive surveillance of stressed self has strong relevance for the proposed role of Vδ1+ T cells in cancer (16, 7072).

Concluding Remarks

There is an increasing interest in the role of γδ T cells and other similar cells, such as NK cells, in the immune response to malaria (11, 73, 74). However, the Vδ1+ subset has attracted only limited attention so far. Based on the ideas and studies highlighted in this review, we believe that there is a strong case for extending the focus of γδ T-cell studies in malaria beyond the innate-like Vγ9+Vδ2+ subset, to include adaptive-like γδ T cells. Although, we have focused here on Vδ1+ T cells, it is worth noting that clonal expansion of γδ T cells that express Vδ2 chains paired with γ-chains other than Vγ9 has been described in a variety of conditions. Those cell populations also appear to display an “adaptive-like” immunobiology, positioning them functionally much closer to Vδ1+ cells than to the innate-like Vγ9+Vδ2+ cell subset [reviewed in Davey et al. (17)]. Moreover, recent data further suggest that γδ T cells that express neither Vδ1 nor Vδ2 (e.g., Vδ3+ cells) exhibit features of such adaptive immune subsets (13, 75). In light of this emerging adaptive immunobiological human γδ T-cell paradigm, examining the contributions of γδ T-cell subsets other than Vγ9+Vδ2+ in the immune response to malaria is an underexplored and important avenue for investigation.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

CS-T is supported by a Ph.D. stipend, awarded as part of the Building Stronger Universities II collaboration between Danish Universities and University of Ghana and financed by the Danish International Development Agency (DANIDA) (grant: BSU2-UG).

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. World Health Organization. World Malaria Report 2017 (2017).

PubMed Abstract

2. Phillips MA, Burrows JN, Manyando C, Van Huijsduijnen RH, Van Voorhis WC, Wells TNC. Malaria. Nat Rev Dis Primers (2017) 3:17050. doi: 10.1038/nrdp.2017.50

CrossRef Full Text | Google Scholar

3. Hviid L. Naturally acquired immunity to Plasmodium falciparum malaria in Africa. Acta Trop. (2005) 95:270–5. doi: 10.1016/j.actatropica.2005.06.012

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol. (2008) 9:725–32. doi: 10.1038/ni.f.205

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Cowman AF, Healer J, Marapana D, Marsh K. Malaria: biology and disease. Cell (2016) 167:610–24. doi: 10.1016/j.cell.2016.07.055

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Morita CT, Jin C, Sarikonda G, Wang H. Nonpeptide antigens, presentation mechanisms, and immunological memory of human Vgamma2Vdelta2 T cells: discriminating friend from foe through the recognition of prenyl pyrophosphate antigens. Immunol Rev. (2007) 215:59–76. doi: 10.1111/j.1600-065X.2006.00479.x

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Sherwood AM, Desmarais C, Livingston RJ, Andriesen J, Haussler M, Carlson CS , et al. Deep sequencing of the human TCRγ and TCRβ repertoires suggests that TCRβ rearranges after αβ and γδ T cell commitment. Sci Transl Med. (2011) 3:90ra61. doi: 10.1126/scitranslmed.3002536

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Dimova T, Brouwer M, Gosselin F, Tassignon J, Leo O, Donner C , et al. Effector Vγ9Vδ2 cells dominate the human fetal γδT-cell repertoire. Proc Natl Acad Sci USA. (2015) 112:E556–65. doi: 10.1073/pnas.1412058112

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Davey MS, Willcox CR, Hunter S, Kasatskaya SA, Remmerswaal EBM, Salim M , et al. The human Vδ2+ T-cell compartment comprises distinct innate-like Vγ9+ and adaptive Vγ9- subsets. Nat Commun. (2018) 9:1760. doi: 10.1038/s41467-018-04076-0

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Willcox CR, Davey MS, Willcox BE. Development and selection of the human Vγ9Vδ2+ T-cell repertoire. Front Immunol. (2018) 9:1501. doi: 10.3389/fimmu.2018.01501

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Howard J, Zaidi I, Loizon S, Mercereau-Puijalon O, Dechanet-Merville J, Mamani-Matsuda M. Human Vγ9Vδ2 T lymphocytes in the immune response to P. falciparum infection. Front Immunol. (2018) 9:2760. doi: 10.3389/fimmu.2018.02760

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Deroost K, Langhorne J. Gamma/delta T cells and their role in protection against malaria. Front Immunol. (2018) 9:2973. doi: 10.3389/fimmu.2018.02973

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Davey MS, Willcox CR, Joyce SP, Ladell K, Kasatskaya SA, Mclaren JE , et al. Clonal selection in the human Vδ1 T cell repertoire indicates γδ TCR-dependent adaptive immune surveillance. Nat Commun. (2017) 8:14760. doi: 10.1038/ncomms14760

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Pang DJ, Neves JF, Sumaria N, Pennington DJ. Understanding the complexity of γδ T-cell subsets in mouse and human. Immunology (2012) 136:283–90. doi: 10.1111/j.1365-2567.2012.03582.x

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Vantourout P, Hayday A. Six-of-the-best: unique contributions of γδ T cells to immunology. Nat Rev Immunol. (2013) 13:88–100. doi: 10.1038/nri3384

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Siegers GM, Lamb LS Jr. Cytotoxic and regulatory properties of circulating Vδ1+ γδ T cells: a new player on the cell therapy field? Mol Ther. (2014) 22:1416–22. doi: 10.1038/mt.2014.104

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Davey MS, Willcox CR, Hunter S, Oo YH, Willcox BE. Vδ2+ T cells - two subsets for the price of one. Front Immunol. (2018) 9:2106. doi: 10.3389/fimmu.2018.02106

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Beldjord K, Beldjord C, Macintyre E, Even P, Sigaux F. Peripheral selection of Vdelta1+ cells with restricted T cell receptor delta gene junctional repertoire in the peripheral blood of healthy donors. J Exp Med. (1993) 178:121–7. doi: 10.1084/jem.178.1.121

CrossRef Full Text | Google Scholar

19. Giachino C, Granziero L, Modena V, Maiocco V, Lomater C, Fantini F , et al. Clonal expansions of Vδ1+ and Vδ2+ cells increase with age and limit the repertoire of human γδ T cells. Eur J Immunol. (1994) 24:1914–8. doi: 10.1002/eji.1830240830

CrossRef Full Text | Google Scholar

20. Hviid L, Akanmori BD, Loizon S, Kurtzhals JA, Ricke CH, Lim A , et al. High frequency of circulating γδ Tcells with dominance of the Vδ1 subset in a healthy population. Int Immunol. (2000) 12:797–805. doi: 10.1093/intimm/12.6.797

CrossRef Full Text | Google Scholar

21. Davey MS, Willcox CR, Baker AT, Hunter S, Willcox BE. Recasting human Vδ1 lymphocytes in an adaptive role. Trends Immunol. (2018) 39:446–59. doi: 10.1016/j.it.2018.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Ho M, Webster HK, Tongtawe P, Pattanapanyasat K, Weidanz WP. Increased γδ T cells in acute Plasmodium falciparum malaria. Immunol Lett. (1990) 25:139–42. doi: 10.1016/0165-2478(90)90105-Y

CrossRef Full Text | Google Scholar

23. Roussilhon C, Agrapart M, Ballet J-J, Bensussan A. T lymphocytes bearing the γδT cell receptor in patients with acute Plasmodium falciparum malaria. J Infect Dis. (1990) 162:283–5. doi: 10.1093/infdis/162.1.283-a

CrossRef Full Text | Google Scholar

24. Perera MK, Carter R, Goonewardene R, Mendis KN. Transient increase in circulating γ/δ T cells during Plasmodium vivax malarial paroxysms. J Exp Med. (1994) 179:311–5. doi: 10.1084/jem.179.1.311

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Hviid L, Kurtzhals JaL, Dodoo D, Rodrigues O, Ronn A, Commey JOO , et al. The γ/δ T-cell response to Plasmodium falciparum malaria in a population in which malaria is endemic. Infect Immun. (1996) 64:4359–62.

PubMed Abstract | Google Scholar

26. Hviid L. Peripheral T-cell non-responsiveness in individuals exposed to Plasmodium falciparum malaria. APMIS (1995) 103 (Suppl. 53):1–46.

Google Scholar

27. Chang W-L, Van Der Heyde H, Maki DG, Malkovsky M, Weidanz W. Subset heterogeneity among γδ T cells found in peripheral blood during Plasmodium falciparum malaria. Immunol Lett. (1992) 32:273–4.

Google Scholar

28. Schwartz E, Shapiro R, Shina S, Bank I. Delayed expansion of V δ2+ and Vδ 1+ γδ T cells after acute Plasmodium falciparum and Plasmodium vivax malaria. J Allergy Clin Immunol. (1996) 97:1387–92. doi: 10.1016/S0091-6749(96)70208-7

CrossRef Full Text | Google Scholar

29. Ho M, Tongtawe P, Kriangkum J, Wimonwattrawatee T, Pattanapanyasat K, Bryant L , et al. Polyclonal expansion of peripheral γδ T cells in human Plasmodium falciparum malaria. Infect Immun. (1994) 62:855–62.

Google Scholar

30. Roussilhon C, Agrapart M, Guglielmi P, Bensussan A, Brasseur P, Ballet JJ. Human TcR γδ+ lymphocyte response on primary exposure to Plasmodium falciparum. Clin Exp Immunol. (1994) 95:91–7.

Google Scholar

31. Hviid L, Kurtzhals JaL, Adabayeri V, Loizon S, Kemp K, Goka BQ , et al. Perturbation and proinflammatory type activation of Vδ1+ γδT cells in African children with Plasmodium falciparum malaria. Infect Immun. (2001) 69:3190–6. doi: 10.1128/IAI.69.5.3190-3196.2001

CrossRef Full Text | Google Scholar

32. Worku S, Björkman A, Troye-Blomberg M, Jemaneh L, Färnert A, Christensson B. Lymphocyte activation and subset redistribution in the peripheral blood in acute malaria illness: distinct γδ+ T cell patterns in Plasmodium falciparum and P. vivax infections. Clin Exp Immunol. (1997) 108:34–41.

PubMed Abstract | Google Scholar

33. Taniguchi T, Mannoor KM, Nonaka D, Toma H, Li C, Narita M , et al. A unique subset of γδ T cells expands and produces IL-10 in patients with naturally acquired immunity against falciparum malaria. Front Microbiol. (2017) 8:1288. doi: 10.3389/fmicb.2017.01288

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Goodier M, Krause-Jauer M, Sanni A, Massougbodji A, Sadeler B-C, Mitchell GH , et al. γδT cells in the peripheral blood of individuals from an area of holoendemic Plasmodium falciparum transmission. Trans R Soc Trop Med Hyg. (1993) 87:692–6. doi: 10.1016/0035-9203(93)90299-6

CrossRef Full Text | Google Scholar

35. Deusch K, Luling F, Reich K, Classen M, Wagner H, Pfeffer K. A major fraction of human intraepithelial lymphocytes simultaneously expresses the g/d T cell receptor, the CD8 accessory molecule and preferentially uses the Vd1 gene segment. Eur J Immunol. (1991) 21:1053–9. doi: 10.1002/eji.1830210429

CrossRef Full Text | Google Scholar

36. Troye-Blomberg M, Worku S, Tangteerawatana P, Jamshaid R, Söderström K, Elghazali G , et al. Human γδ T cells that inhibit the in vitro growth of the asexual blood stages of the Plasmodium falciparum parasite express cytolytic and proinflammatory molecules. Scand J Immunol. (1999) 50:642–50.

Google Scholar

37. Behr C, Dubois P. Preferential expansion of Vγ9Vδ2 T cells following stimulation of peripheral blood lymphocytes with extracts of Plasmodium falciparum. Int Immunol. (1992) 4:361–6.

Google Scholar

38. Goodier M, Fey P, Eichmann K, Langhorne J. Human peripheral blood γδ T cells respond to antigens of Plasmodium falciparum. Int Immunol. (1992) 4:33–41. doi: 10.1093/intimm/4.1.33

CrossRef Full Text | Google Scholar

39. Autran B, Triebel F, Katlama C, Rozenbaum W, Hercend T, Debre P. T cell receptor γ/δ + lymphocyte subsets during HIV infection. Clin Exp Immunol. (1989) 75:206–10.

PubMed Abstract | Google Scholar

40. De Maria A, Ferrazin A, Ferrini S, Ciccone E, Terragna A, Moretta L. Selective increase of a subset of T cell receptor γδT lymphocytes in the peripheral blood of patients with human immunodeficiency virus type 1 infection. J Infect Dis. (1992) 165:917–9. doi: 10.1093/infdis/165.5.917

CrossRef Full Text | Google Scholar

41. Boullier S, Cochet M, Poccia F, Gougeon M-L. CDR3-independent γδVδ1+ T cell expansion in the peripheral blood of HIV-infected persons. J Immunol. (1995) 154:1418–31.

PubMed Abstract | Google Scholar

42. Rossol R, Dobmeyer JM, Dobmeyer TS, Klein SA, Rossol S, Wesch D , et al. Increase in Vδ1+ γδ T cells in the peripheral blood and bone marrow as a selective feature of HIV-1 but not other virus infections. Br J Haematol. (1998) 100:728–34. doi: 10.1046/j.1365-2141.1998.00630.x

CrossRef Full Text | Google Scholar

43. Déchanet J, Merville P, Bergé F, Bone-Mane G, Taupin J-L, Michel P , et al. Major expansion of γδ T lymphocytes following cytomegalovirus infection in kidney allograft recipients. J Infect Dis. (1999) 179:1–8.

PubMed Abstract | Google Scholar

44. Barcy S, De Rosa SC, Vieira J, Diem K, Ikoma M, Casper C , et al. γδ+ T cells involvement in viral immune control of chronic human herpesvirus 8 infection. J Immunol. (2008) 180:3417–25. doi: 10.4049/jimmunol.180.5.3417

CrossRef Full Text | Google Scholar

45. Knight A, Madrigal AJ, Grace S, Sivakumaran J, Kottaridis P, Mackinnon S , et al. The role of Vδ2-negative γδ T cells during cytomegalovirus reactivation in recipients of allogeneic stem cell transplantation. Blood (2010) 116:2164–72. doi: 10.1182/blood-2010-01-255166

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Munk ME, Soboslay PT, Arnoldi J, Brattig N, Schulz Key H, Kaufmann SH. Onchocerca volvulus provides ligands for the stimulation of human γ/δ T lymphocytes expressing Vδ1 chains. J Infect Dis. (1993) 168:1241–7. doi: 10.1093/infdis/168.5.1241

CrossRef Full Text | Google Scholar

47. Chauhan SK, Tripathy NK, Sinha N, Nityanand S. T-cell receptor repertoire of circulating gamma delta T-cells in Takayasu's arteritis. Clin Immunol. (2006) 118:243–9. doi: 10.1016/j.clim.2005.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Giacomelli R, Parzanese I, Frieri G, Passacantando A, Pizzuto F, Pimpo T , et al. Increase of circulating γ/δ T lymphocytes in the peripheral blood of patients affected by active inflammatory bowel disease. Clin Exp Immunol. (1994) 98:83–8. doi: 10.1111/j.1365-2249.1994.tb06611.x

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Kadivar M, Petersson J, Svensson L, Marsal J. CD8 αβ+ γδ T cells: a novel T cell subset with a potential role in inflammatory bowel disease. J Immunol. (2016) 197:4584–92. doi: 10.4049/jimmunol.1601146

CrossRef Full Text | Google Scholar

50. Halary F, Pitard V, Dlubek D, Krzysiek R, De La Salle H, Merville P , et al. Shared reactivity of Vδ2neg γδ T cells against cytomegalovirus-infected cells and tumor intestinal epithelial cells. J Exp Med. (2005) 201:1567–78. doi: 10.1084/jem.20041851

CrossRef Full Text | Google Scholar

51. Willcox CR, Pitard V, Netzer S, Couzi L, Salim M, Silberzahn T , et al. Cytomegalovirus and tumor stress surveillance by binding of a human γδ T cell antigen receptor to endothelial protein C receptor. Nat Immunol. (2012) 13:872–9. doi: 10.1038/ni.2394

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Turner L, Lavstsen T, Berger SS, Wang CW, Petersen JE, Avril M , et al. Severe malaria is associated with parasite binding to endothelial protein C receptor. Nature (2013) 498:502–5. doi: 10.1038/nature12216

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Lennartz F, Adams Y, Bengtsson A, Olsen RW, Turner L, Ndam NT , et al. Structure-guided identification of a family of dual receptor-binding PfEMP1 that is associated with cerebral malaria. Cell Host Microbe (2017) 21:403–14. doi: 10.1016/j.chom.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Mcgregor IA, Gilles HM, Walters JH, Davies AH, Pearson FA. Effects of heavy and repeated malarial infections on Gambian infants and children. Effects of erythrocytic parasitization. Br Med J. (1956) 32:686–92. doi: 10.1136/bmj.2.4994.686

CrossRef Full Text | Google Scholar

55. Donati D, Zhang LP, Chene A, Cheng Q, Flick K, Nystrom M , et al. Identification of a polyclonal B-cell activator in Plasmodium falciparum. Infect Immun. (2004) 72:5412–8. doi: 10.1128/IAI.72.9.5412-5418.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Whittle HC, Brown J, Marsh K, Blackman M, Jobe O, Shenton F. The effects of Plasmodium falciparum malaria on immune control of B lymphocytes in Gambian children. Clin Exp Immunol. (1990) 80:213–8. doi: 10.1111/j.1365-2249.1990.tb05236.x

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Lam KMC, Syed N, Whittle H, Crawford DH. Circulating Epstein-Barr virus-carrying B cells in acute malaria. Lancet (1991) 337:876–8. doi: 10.1016/0140-6736(91)90203-2

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Moormann AM, Chelimo K, Sumba OP, Lutzke ML, Ploutz-Snyder R, Newton D , et al. Exposure to holoendemic malaria results in elevated Epstein-Barr virus loads in children. J Infect Dis. (2005) 191:1233–8. doi: 10.1086/428910

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Hacker G, Kromer S, Falk M, Heeg K, Wagner H, Pfeffer K. Vδ1+ subset of human γδ T cells responds to ligands expressed by EBV-infected Burkitt lymphoma cells and transformed B lymphocytes. J Immunol. (1992) 149:3984–9.

Google Scholar

60. Siegers GM, Dhamko H, Wang XH, Mathieson AM, Kosaka Y, Felizardo TC , et al. Human Vδ1 γδ T cells expanded from peripheral blood exhibit specific cytotoxicity against B-cell chronic lymphocytic leukemia-derived cells. Cytotherapy (2011) 13:753–64. doi: 10.3109/14653249.2011.553595

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Fujishima N, Hirokawa M, Fujishima M, Yamashita J, Saitoh H, Ichikawa Y , et al. Skewed T cell receptor repertoire of Vδ1+ γδ T lymphocytes after human allogeneic haematopoietic stem cell transplantation and the potential role for Epstein-Barr virus-infected B cells in clonal restriction. Clin Exp Immunol. (2007) 149:70–9. doi: 10.1111/j.1365-2249.2007.03388.x

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Farnault L, Gertner-Dardenne J, Gondois-Rey F, Michel G, Chambost H, Hirsch I , et al. Clinical evidence implicating gamma-delta T cells in EBV control following cord blood transplantation. Bone Marrow Transplant. (2013) 48:1478–9. doi: 10.1038/bmt.2013.75

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Spada FM, Grant EP, Peters PJ, Sugita M, Melián A, Leslie DS , et al. Self-recognition of CD1 by γ/δ T cells: implications for innate immunity. J Exp Med. (2000) 191:937–48. doi: 10.1084/jem.191.6.937

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Del Porto P, Mami-Chouaib F, Bruneau JM, Jitsukawa S, Dumas J, Harnois M , et al. TCT.1, a target molecule for γ/δ T cells, is encoded by an immunoglobulin superfamily gene (Blast-1) located in the CD1 region of human chromosome 1. J Exp Med. (1991) 173:1339–44.

PubMed Abstract | Google Scholar

65. Delia D, Cattoretti G, Polli N, Fontanella E, Aiello A, Giardini R , et al. CD1c but neither CD1a nor CD1b molecules are expressed on normal, activated, and malignant human B cells: identification of a new B-cell subset. Blood (1988) 72:241–7.

PubMed Abstract | Google Scholar

66. Allan LL, Stax AM, Zheng DJ, Chung BK, Kozak FK, Tan R , et al. CD1d and CD1c expression in human B cells is regulated by activation and retinoic acid receptor signaling. J Immunol. (2011) 186:5261–72. doi: 10.4049/jimmunol.1003615

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Falini B, Flenghi L, Pileri S, Pelicci P, Fagioli M, Martelli MF , et al. Distribution of T cells bearing different forms of the T cell receptor γ/δ in normal and pathological human tissues. J Immunol. (1989) 143:2480–8.

PubMed Abstract | Google Scholar

68. Bordessoule D, Gaulard P, Mason DY. Preferential localisation of human lymphocytes bearing γδ T cell receptors to the red pulp of the spleen. J Clin Pathol. (1990) 43:461–4. doi: 10.1136/jcp.43.6.461

CrossRef Full Text | Google Scholar

69. Roy S, Ly D, Castro CD, Li NS, Hawk AJ, Altman JD , et al. Molecular analysis of lipid-reactive Vδ1 γδ T cells identified by CD1c tetramers. J Immunol. (2016) 196:1933–42. doi: 10.4049/jimmunol.1502202

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Halary F, Fournie JJ, Bonneville M. Activation and control of self-reactive γδ T cells. Microb Infect. (1999) 1:247–53. doi: 10.1016/s1286-4579(99)80041-0

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Born W, Cady C, Jones-Carson J, Mukasa A, Lahn M, O'brien R. Immunoregulatory functions of γδT cells. Adv Immunol. (1999) 71:77–144.

Google Scholar

72. Wesch D, Peters C, Siegers GM. Human gamma delta T regulatory cells in cancer: fact or fiction? Front Immunol. (2014) 5:598. doi: 10.3389/fimmu.2014.00598

CrossRef Full Text | Google Scholar

73. Arora G, Hart GT, Manzella-Lapeira J, Doritchamou JY, Narum DL, Thomas LM , et al. NK cells inhibit Plasmodium falciparum growth in red blood cells via antibody-dependent cellular cytotoxicity. Elife (2018) 7:e36806. doi: 10.7554/eLife.36806

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Wolf AS, Sherratt S, Riley EM. NK cells: uncertain allies against malaria. Front Immunol. (2017) 8:212. doi: 10.3389/fimmu.2017.00212

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Hunter S, Willcox CR, Davey MS, Kasatskaya SA, Jeffery HC, Chudakov DM , et al. Human liver infiltrating γδT cells are composed of clonally expanded circulating and tissue-resident populations. J Hepatol. (2018) 69:654–65. doi: 10.1016/j.jhep.2018.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: gamma-delta (γ/δ) T lymphocytes, Vdelta1 gamma delta T cells, malaria, Plasmodium falciparum, innate immunity, acquired immunity, immune regulation

Citation: Hviid L, Smith-Togobo C and Willcox BE (2019) Human Vδ1+ T Cells in the Immune Response to Plasmodium falciparum Infection. Front. Immunol. 10:259. doi: 10.3389/fimmu.2019.00259

Received: 09 November 2018; Accepted: 29 January 2019;
Published: 14 February 2019.

Edited by:

Francesco Dieli, Università degli Studi di Palermo, Italy

Reviewed by:

Marita Troye Blomberg, Stockholm University, Sweden
Serena Meraviglia, Università degli Studi di Palermo, Italy

Copyright © 2019 Hviid, Smith-Togobo and Willcox. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Lars Hviid, lhviid@sund.ku.dk
orcid.org/0000-0002-1698-4927

Cecilia Smith-Togobo orcid.org/0000-0002-5583-3304
Benjamin E. Willcox orcid.org/0000-0002-6113-2109

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.