Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cells ; 13(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38891095

ABSTRACT

Basal cell carcinomas (BCCs) and squamous cell carcinomas (SCCs) are high-incidence, non-melanoma skin cancers (NMSCs). The success of immune-targeted therapies in advanced NMSCs led us to anticipate that NMSCs harbored significant populations of tumor-infiltrating lymphocytes with potential anti-tumor activity. The main aim of this study was to characterize T cells infiltrating NMSCs. Flow cytometry and immunohistochemistry were used to assess, respectively, the proportions and densities of T cell subpopulations in BCCs (n = 118), SCCs (n = 33), and normal skin (NS, n = 30). CD8+ T cells, CD4+ T cell subsets, namely, Th1, Th2, Th17, Th9, and regulatory T cells (Tregs), CD8+ and CD4+ memory T cells, and γδ T cells were compared between NMSCs and NS samples. Remarkably, both BCCs and SCCs featured a significantly higher Th1/Th2 ratio (~four-fold) and an enrichment for Th17 cells. NMSCs also showed a significant enrichment for IFN-γ-producing CD8+T cells, and a depletion of γδ T cells. Using immunohistochemistry, NMSCs featured denser T cell infiltrates (CD4+, CD8+, and Tregs) than NS. Overall, these data favor a Th1-predominant response in BCCs and SCCs, providing support for immune-based treatments in NMSCs. Th17-mediated inflammation may play a role in the progression of NMSCs and thus become a potential therapeutic target in NMSCs.


Subject(s)
Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Lymphocytes, Tumor-Infiltrating , Skin Neoplasms , Th1 Cells , Th17 Cells , Humans , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Th17 Cells/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Th1 Cells/immunology , Carcinoma, Basal Cell/immunology , Carcinoma, Basal Cell/pathology , Female , Male , Aged , Cross-Sectional Studies , Middle Aged , CD8-Positive T-Lymphocytes/immunology , Aged, 80 and over , Adult
2.
Mucosal Immunol ; 16(6): 767-775, 2023 12.
Article in English | MEDLINE | ID: mdl-37783278

ABSTRACT

The early migratory phase of pulmonary helminth infections is characterized by tissue injury leading to the release of the alarmin interleukin (IL)-33 and subsequent induction of type 2 immune responses. We recently described a role for IL-17A, through suppression of interferon (IFN)-γ, as an important inducer of type 2 responses during infection with the lung-migrating rodent nematode Nippostrongylus brasiliensis. Here, we aimed to investigate the interaction between IL-17A and IL-33 during the early lung migratory stages of N. brasiliensis infection. In this brief report, we demonstrate that deficiency of IL-17A leads to impaired IL-33 expression and secretion early in infection, independent of IL-17A suppression of IFN-γ. Neutrophil-depletion experiments, which dramatically reduce lung injury, revealed that neutrophils are primarily responsible for the IL-17A-dependent release of IL-33 into the airways. Taken together, our results reveal an IL-17A-neutrophil-axis that can drive IL-33 during helminth infection, highlighting an additional pathway by which IL-17A regulates pulmonary type 2 immunity.


Subject(s)
Nematoda , Neutrophils , Animals , Mice , Interleukin-17/metabolism , Interleukin-33 , Lung , Epithelial Cells/metabolism , Mice, Inbred C57BL
4.
Semin Immunol ; 61-64: 101657, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36370671

ABSTRACT

Unconventional T cells typically group γδ T cells, invariant Natural Killer T cells (NKT) and Mucosal Associated Invariant T (MAIT) cells. With their pre-activated status and biased tropism for non-lymphoid organs, they provide a rapid (innate-like) and efficient first line of defense against pathogens at strategical barrier sites, while they can also trigger chronic inflammation, and unexpectedly contribute to steady state physiology. Thus, a tight control of their homeostasis is critical to maintain tissue integrity. In this review, we discuss the recent advances of our understanding of the factors, from neuroimmune to inflammatory regulators, shaping the size and functional properties of unconventional T cell subsets in non-lymphoid organs. We present a general overview of the mechanisms common to these populations, while also acknowledging specific aspects of their diversity. We mainly focus on their maintenance at steady state and upon inflammation, highlighting some key unresolved issues and raising upcoming technical, fundamental and translational challenges.


Subject(s)
Mucosal-Associated Invariant T Cells , Natural Killer T-Cells , Humans , T-Lymphocyte Subsets , Inflammation , Homeostasis
5.
EMBO Rep ; 23(1): e52234, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34821000

ABSTRACT

γδ T cells are a conserved population of lymphocytes that contributes to anti-tumor responses through its overt type 1 inflammatory and cytotoxic properties. We have previously shown that human γδ T cells acquire this profile upon stimulation with IL-2 or IL-15, in a differentiation process dependent on MAPK/ERK signaling. Here, we identify microRNA-181a as a key modulator of human γδ T cell differentiation. We observe that miR-181a is highly expressed in patients with prostate cancer and that this pattern associates with lower expression of NKG2D, a critical mediator of cancer surveillance. Interestingly, miR-181a expression negatively correlates with an activated type 1 effector profile obtained from in vitro differentiated γδ T cells and miR-181a overexpression restricts their levels of NKG2D and TNF-α. Upon in silico analysis, we identify two miR-181a candidate targets, Map3k2 and Notch2, which we validate via overexpression coupled with luciferase assays. These results reveal a novel role for miR-181a as critical regulator of human γδ T cell differentiation and highlight its potential for manipulation of γδ T cells in next-generation immunotherapies.


Subject(s)
Cell Differentiation , MicroRNAs , Receptor, Notch2 , T-Lymphocytes/cytology , Humans , Lymphocyte Activation , MAP Kinase Kinase Kinase 2/metabolism , Male , MicroRNAs/genetics , Prostatic Neoplasms , Receptor, Notch2/metabolism , Signal Transduction
6.
Cell Rep ; 36(9): 109574, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469732

ABSTRACT

Neuroinflammation in patients with Alzheimer's disease (AD) and related mouse models has been recognized for decades, but the contribution of the recently described meningeal immune population to AD pathogenesis remains to be addressed. Here, using the 3xTg-AD model, we report an accumulation of interleukin-17 (IL-17)-producing cells, mostly γδ T cells, in the brain and the meninges of female, but not male, mice, concomitant with the onset of cognitive decline. Critically, IL-17 neutralization into the ventricles is sufficient to prevent short-term memory and synaptic plasticity deficits at early stages of disease. These effects precede blood-brain barrier disruption and amyloid-beta or tau pathology, implying an early involvement of IL-17 in AD pathology. When IL-17 is neutralized at later stages of disease, the onset of short-memory deficits and amyloidosis-related splenomegaly is delayed. Altogether, our data support the idea that cognition relies on a finely regulated balance of "inflammatory" cytokines derived from the meningeal immune system.


Subject(s)
Alzheimer Disease/metabolism , Behavior, Animal , Brain/metabolism , Cognition , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Intraepithelial Lymphocytes/metabolism , Neuroinflammatory Diseases/metabolism , Synapses/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Animals , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cognition/drug effects , Disease Models, Animal , Female , Inflammation Mediators/antagonists & inhibitors , Interleukin-17/antagonists & inhibitors , Intraepithelial Lymphocytes/drug effects , Male , Memory, Short-Term , Mice, 129 Strain , Mice, Transgenic , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/prevention & control , Neuroinflammatory Diseases/psychology , Neuronal Plasticity , Synapses/drug effects , Synapses/pathology
8.
Nat Rev Immunol ; 21(4): 221-232, 2021 04.
Article in English | MEDLINE | ID: mdl-33057185

ABSTRACT

γδ T cells are a unique T cell subpopulation that are rare in secondary lymphoid organs but enriched in many peripheral tissues, such as the skin, intestines and lungs. By rapidly producing large amounts of cytokines, γδ T cells make key contributions to immune responses in these tissues. In addition to their immune surveillance activities, recent reports have unravelled exciting new roles for γδ T cells in steady-state tissue physiology, with functions ranging from the regulation of thermogenesis in adipose tissue to the control of neuronal synaptic plasticity in the central nervous system. Here, we review the roles of γδ T cells in tissue homeostasis and in surveillance of infection, aiming to illustrate their major impact on tissue integrity, tissue repair and immune protection.


Subject(s)
Immunologic Surveillance/physiology , Infections/immunology , Intraepithelial Lymphocytes/physiology , Mucous Membrane/physiology , Neuronal Plasticity/physiology , Regeneration/physiology , Thermogenesis/physiology , Adipose Tissue/metabolism , Adipose Tissue/physiology , Bone Regeneration/physiology , Butyrophilins/metabolism , Central Nervous System/physiology , Female , Genitalia, Female/physiology , Gingiva/physiology , Homeostasis , Humans , Immunologic Surveillance/immunology , Intestinal Mucosa/physiology , Intraepithelial Lymphocytes/immunology , Lung/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology
10.
Mucosal Immunol ; 14(1): 242-252, 2021 01.
Article in English | MEDLINE | ID: mdl-32733025

ABSTRACT

γδT cells represent the majority of lymphocytes in several mucosal tissues where they contribute to tissue homoeostasis, microbial defence and wound repair. Here we characterise a population of interleukin (IL) 17-producing γδ (γδ17) T cells that seed the testis of naive C57BL/6 mice, expand at puberty and persist throughout adulthood. We show that this population is foetal-derived and displays a T-cell receptor (TCR) repertoire highly biased towards Vγ6-containing rearrangements. These γδ17 cells were the major source of IL-17 in the testis, whereas αß T cells mostly provided interferon (IFN)-γ in situ. Importantly, testicular γδ17 cell homoeostasis was strongly dependent on the microbiota and Toll-like receptor (TLR4)/IL-1α/IL-23 signalling. We further found that γδ17 cells contributed to tissue surveillance in a model of experimental orchitis induced by intra-testicular inoculation of Listeria monocytogenes, as Tcrδ-/- and Il17-/- infected mice displayed higher bacterial loads than wild-type (WT) controls and died 3 days after infection. Altogether, this study identified a previously unappreciated foetal-derived γδ17 cell subset that infiltrates the testis at steady state, expands upon puberty and plays a crucial role in local tissue immune surveillance.


Subject(s)
Microbiota/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Sexual Maturation/immunology , Testis/immunology , Testis/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Cytokines , Immunohistochemistry , Immunologic Surveillance , Immunophenotyping , Interleukin-17/biosynthesis , Interleukin-23/metabolism , Male , Mice , Mice, Transgenic , Spermatogenesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
J Exp Med ; 217(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32106283

ABSTRACT

T cells are classically recognized as distinct subsets that express αß or γδ TCRs. We identify a novel population of T cells that coexpress αß and γδ TCRs in mice and humans. These hybrid αß-γδ T cells arose in the murine fetal thymus by day 16 of ontogeny, underwent αß TCR-mediated positive selection into CD4+ or CD8+ thymocytes, and constituted up to 10% of TCRδ+ cells in lymphoid organs. They expressed high levels of IL-1R1 and IL-23R and secreted IFN-γ, IL-17, and GM-CSF in response to canonically restricted peptide antigens or stimulation with IL-1ß and IL-23. Hybrid αß-γδ T cells were transcriptomically distinct from conventional γδ T cells and displayed a hyperinflammatory phenotype enriched for chemokine receptors and homing molecules that facilitate migration to sites of inflammation. These proinflammatory T cells promoted bacterial clearance after infection with Staphylococcus aureus and, by licensing encephalitogenic Th17 cells, played a key role in the development of autoimmune disease in the central nervous system.


Subject(s)
Inflammation/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Animals , Biomarkers/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Inflammation/pathology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Transcription, Genetic , Transcriptome/genetics
13.
Sci Immunol ; 4(40)2019 10 11.
Article in English | MEDLINE | ID: mdl-31604844

ABSTRACT

The notion of "immune privilege" of the brain has been revised to accommodate its infiltration, at steady state, by immune cells that participate in normal neurophysiology. However, the immune mechanisms that regulate learning and memory remain poorly understood. Here, we show that noninflammatory interleukin-17 (IL-17) derived from a previously unknown fetal-derived meningeal-resident γδ T cell subset promotes cognition. When tested in classical spatial learning paradigms, mice lacking γδ T cells or IL-17 displayed deficient short-term memory while retaining long-term memory. The plasticity of glutamatergic synapses was reduced in the absence of IL-17, resulting in impaired long-term potentiation in the hippocampus. Conversely, IL-17 enhanced glial cell production of brain-derived neurotropic factor, whose exogenous provision rescued the synaptic and behavioral phenotypes of IL-17-deficient animals. Together, our work provides previously unknown clues on the mechanisms that regulate short-term versus long-term memory and on the evolutionary and functional link between the immune and nervous systems.


Subject(s)
Interleukin-17/immunology , Memory, Short-Term , Meninges/immunology , Neuronal Plasticity/immunology , T-Lymphocytes/immunology , Animals , Interleukin-17/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Proc Natl Acad Sci U S A ; 116(20): 9979-9988, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028144

ABSTRACT

Cerebral malaria (CM) is a major cause of death due to Plasmodium infection. Both parasite and host factors contribute to the onset of CM, but the precise cellular and molecular mechanisms that contribute to its pathogenesis remain poorly characterized. Unlike conventional αß-T cells, previous studies on murine γδ-T cells failed to identify a nonredundant role for this T cell subset in experimental cerebral malaria (ECM). Here we show that mice lacking γδ-T cells are resistant to ECM when infected with Plasmodium berghei ANKA sporozoites, the liver-infective form of the parasite and the natural route of infection, in contrast with their susceptible phenotype if challenged with P. berghei ANKA-infected red blood cells that bypass the liver stage of infection. Strikingly, the presence of γδ-T cells enhanced the expression of Plasmodium immunogenic factors and exacerbated subsequent systemic and brain-infiltrating inflammatory αß-T cell responses. These phenomena were dependent on the proinflammatory cytokine IFN-γ, which was required during liver stage for modulation of the parasite transcriptome, as well as for downstream immune-mediated pathology. Our work reveals an unanticipated critical role of γδ-T cells in the development of ECM upon Plasmodium liver-stage infection.


Subject(s)
Intraepithelial Lymphocytes/physiology , Liver/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/pathogenicity , Sporozoites/pathogenicity , Animals , Liver/parasitology , Male , Mice , Mice, Inbred C57BL , Sporozoites/growth & development
15.
EMBO Rep ; 18(11): 1957-1967, 2017 11.
Article in English | MEDLINE | ID: mdl-28855306

ABSTRACT

Pro-inflammatory interleukin (IL)-17-producing γδ (γδ17) T cells are thought to develop exclusively in the thymus during fetal/perinatal life, as adult bone marrow precursors fail to generate γδ17 T cells under homeostatic conditions. Here, we employ a model of experimental autoimmune encephalomyelitis (EAE) in which hematopoiesis is reset by bone marrow transplantation and demonstrate unequivocally that Vγ4+ γδ17 T cells can develop de novo in draining lymph nodes in response to innate stimuli. In vitro, γδ T cells from IL-17 fate-mapping reporter mice that had never activated the Il17 locus acquire IL-17 expression upon stimulation with IL-1ß and IL-23. Furthermore, IL-23R (but not IL-1R1) deficiency severely compromises the induction of γδ17 T cells in EAE, demonstrating the key role of IL-23 in the process. Finally, we show, in a composite model involving transfers of both adult bone marrow and neonatal thymocytes, that induced γδ17 T cells make up a substantial fraction of the total IL-17-producing Vγ4+ T-cell pool upon inflammation, which attests the relevance of this novel pathway of peripheral γδ17 T-cell differentiation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-23/immunology , Lymph Nodes/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Th17 Cells/immunology , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Bone Marrow Transplantation , Cell Differentiation/drug effects , Cell Lineage/immunology , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Hematopoiesis/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/pharmacology , Interleukin-23/genetics , Interleukin-23/pharmacology , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction , Th17 Cells/pathology , Thymus Gland/immunology , Thymus Gland/pathology
16.
Nat Immunol ; 18(6): 604-611, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28518154

ABSTRACT

Shortly after the discovery of interleukin 17 (IL-17)-producing CD4+ helper T cells (TH17 cells), it was found that γδ T cells can also secrete large amounts of this pro-inflammatory cytokine. A decade later, it is now known that IL-17+ γδ T cells (γδ17 T cells) are often the main providers of IL-17A in various models of inflammatory diseases, while they also contribute to protective immune responses to infectious organisms. Due to an intricate thymic program of differentiation, γδ17 T cells are able to respond faster than TH17 cells do and thus predominate in the early stages of inflammatory responses. Here we review the current knowledge of the development, activation and pathophysiological functions of γδ17 T cells, aiming to increase the awareness in the community of the therapeutic potential of this 'other side' of IL-17-mediated immune responses.


Subject(s)
Inflammation/immunology , Interleukin-17/immunology , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , Humans , Immunity, Innate/immunology , Mice , Receptors, Antigen, T-Cell, gamma-delta/immunology , Thymus Gland , V(D)J Recombination
17.
Methods Mol Biol ; 1514: 257-267, 2017.
Article in English | MEDLINE | ID: mdl-27787805

ABSTRACT

The key roles played by gamma-delta (γδ) T cells in immunity to infection and tumors critically depend on their differentiation into effectors capable of secreting cytokines (such as interferon-γ or interleukin-17), and killing infected or transformed cells. Here we detail the main methods used to investigate the differentiation of γδ T cells from murine or human origin. We describe developmental assays, such as thymic organ cultures (TOCs) and coculture of progenitors cells with OP9-DL1 stomal cells, as well as functional assays typically employed to evaluate γδ T cell cytotoxicity and cytokine production.


Subject(s)
Cell Differentiation/genetics , Intraepithelial Lymphocytes/cytology , Organ Culture Techniques/methods , T-Lymphocyte Subsets/cytology , Animals , Cell Differentiation/immunology , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Intraepithelial Lymphocytes/immunology , Mice , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology
18.
Nat Immunol ; 17(6): 721-727, 2016 06.
Article in English | MEDLINE | ID: mdl-27043412

ABSTRACT

The mouse thymus produces discrete γδ T cell subsets that make either interferon-γ (IFN-γ) or interleukin 17 (IL-17), but the role of the T cell antigen receptor (TCR) in this developmental process remains controversial. Here we show that Cd3g(+/-) Cd3d(+/-) (CD3 double-haploinsufficient (CD3DH)) mice have reduced TCR expression and signaling strength on γδ T cells. CD3DH mice had normal numbers and phenotypes of αß thymocyte subsets, but impaired differentiation of fetal Vγ6(+) (but not Vγ4(+)) IL-17-producing γδ T cells and a marked depletion of IFN-γ-producing CD122(+) NK1.1(+) γδ T cells throughout ontogeny. Adult CD3DH mice showed reduced peripheral IFN-γ(+) γδ T cells and were resistant to experimental cerebral malaria. Thus, TCR signal strength within specific thymic developmental windows is a major determinant of the generation of proinflammatory γδ T cell subsets and their impact on pathophysiology.


Subject(s)
Cell Differentiation , Inflammation/immunology , Malaria, Cerebral/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/physiology , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Antigens, Ly/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction
19.
Front Immunol ; 6: 15, 2015.
Article in English | MEDLINE | ID: mdl-25674089

ABSTRACT

The contributions of γδ T-cells to immunity to infection or tumors critically depend on their activation and differentiation into effectors capable of secreting cytokines and killing infected or transformed cells. These processes are molecularly controlled by surface receptors that capture key extracellular cues and convey downstream intracellular signals that regulate γδ T-cell physiology. The understanding of how environmental signals are integrated by γδ T-cells is critical for their manipulation in clinical settings. Here, we discuss how different classes of surface receptors impact on human and murine γδ T-cell differentiation, activation, and expansion. In particular, we review the role of five receptor types: the T-cell receptor (TCR), costimulatory receptors, cytokine receptors, NK receptors, and inhibitory receptors. Some of the key players are the costimulatory receptors CD27 and CD28, which differentially impact on pro-inflammatory subsets of γδ T-cells; the cytokine receptors IL-2R, IL-7R, and IL-15R, which drive functional differentiation and expansion of γδ T-cells; the NK receptor NKG2D and its contribution to γδ T-cell cytotoxicity; and the inhibitory receptors PD-1 and BTLA that control γδ T-cell homeostasis. We discuss these and other receptors in the context of a five-step model of receptor signaling in γδ T-cell differentiation and activation, and discuss its implications for the manipulation of γδ T-cells in immunotherapy.

20.
J Immunol ; 192(5): 2237-43, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489097

ABSTRACT

Cytotoxicity and IFN-γ production by human γδ T cells underlie their potent antitumor functions. However, it remains unclear where and how human γδ T cells acquire these key effector properties. Given the recent disclosure of a major contribution of the thymus to murine γδ T cell functional differentiation, in this study we have analyzed a series of human pediatric thymuses. We found that ex vivo-isolated γδ thymocytes produced negligible IFN-γ and lacked cytolytic activity against leukemia cells. However, these properties were selectively acquired upon stimulation with IL-2 or IL-15, but not IL-4 or IL-7. Unexpectedly, TCR activation was dispensable for these stages of functional differentiation. The effects of IL-2/IL-15 depended on MAPK/ERK signaling and induced de novo expression of the transcription factors T-bet and eomesodermin, as well as the cytolytic enzyme perforin, required for the cytotoxic type 1 program. These findings have implications for the manipulation of γδ T cells in cancer immunotherapy.


Subject(s)
Cell Differentiation/physiology , Interleukin-15/immunology , Interleukin-2/immunology , MAP Kinase Signaling System/physiology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Child , Child, Preschool , Female , Humans , Immunotherapy , Infant , Infant, Newborn , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Male , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...