Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
EMBO J ; 43(14): 2878-2907, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38816652

ABSTRACT

In mice, γδ-T lymphocytes that express the co-stimulatory molecule, CD27, are committed to the IFNγ-producing lineage during thymic development. In the periphery, these cells play a critical role in host defense and anti-tumor immunity. Unlike αß-T cells that rely on MHC-presented peptides to drive their terminal differentiation, it is unclear whether MHC-unrestricted γδ-T cells undergo further functional maturation after exiting the thymus. Here, we provide evidence of phenotypic and functional diversity within peripheral IFNγ-producing γδ T cells. We found that CD27+ Ly6C- cells convert into CD27+Ly6C+ cells, and these CD27+Ly6C+ cells control cancer progression in mice, while the CD27+Ly6C- cells cannot. The gene signatures of these two subsets were highly analogous to human immature and mature γδ-T cells, indicative of conservation across species. We show that IL-27 supports the cytotoxic phenotype and function of mouse CD27+Ly6C+ cells and human Vδ2+ cells, while IL-27 is dispensable for mouse CD27+Ly6C- cell and human Vδ1+ cell functions. These data reveal increased complexity within IFNγ-producing γδ-T cells, comprising immature and terminally differentiated subsets, that offer new insights into unconventional T-cell biology.


Subject(s)
Antigens, Ly , Receptors, Antigen, T-Cell, gamma-delta , Tumor Necrosis Factor Receptor Superfamily, Member 7 , Animals , Mice , Antigens, Ly/metabolism , Antigens, Ly/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Humans , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Interferon-gamma/metabolism , Interferon-gamma/immunology , Interleukin-27/metabolism , Interleukin-27/genetics , Cell Differentiation/immunology , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
2.
Eur J Immunol ; 51(9): 2164-2177, 2021 09.
Article in English | MEDLINE | ID: mdl-34224140

ABSTRACT

T cells of the gamma delta (γδ) lineage are evolutionary conserved from jawless to cartilaginous and bony fish to mammals and represent the "swiss army knife" of the immune system capable of antigen-dependent or independent responses, memory, antigen presentation, regulation of other lymphocytes, tissue homeostasis, and mucosal barrier maintenance, to list a few. Over the last 10 years, γδ T cells that produce the cytokine IL-17 (γδT17) have taken a leading position in our understanding of how our immune system battles infection, inflicts tissue damage during inflammation, and gets rewired by the tumor microenvironment. A lot of what we know about γδT17 cells stems from mouse models, however, increasing evidence implicates these cells in numerous human diseases. Herein, we aim to give an overview of the most common mouse models that have been used to study the role of γδT17 cells in infection, inflammation, and cancer, while at the same time we will evaluate evidence for their importance in humans. We hope and believe that in the next 10 years, means to take advantage of the protective and destructive properties of γδ T and in particular γδT17 cells will be part of our standard immunotherapy toolkit.


Subject(s)
Interleukin-17/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Th17 Cells/immunology , Animals , Autoimmune Diseases/immunology , Bacterial Infections/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Mice , Mycoses/immunology , Tumor Microenvironment/immunology , Virus Diseases/immunology
3.
Eur J Immunol ; 51(12): 3186-3193, 2021 12.
Article in English | MEDLINE | ID: mdl-34624928

ABSTRACT

Interleukin (IL)-17-producing gamma delta (γδ) T (γδT17) cells are an essential part of innate type 3 immunity against numerous pathogens. At the same time, a large body of evidence from mouse models and human clinical studies suggests that γδT17 cells contribute to the pathogenesis of many inflammatory diseases as well as cancer. It is therefore relevant to elucidate their immunobiology in detail and identify molecules and pathways that can regulate their function. Herein, we investigated the importance of the type I interferon (IFN) signaling system in γδT17 homeostasis and activation. We found that the IFN alpha receptor 1 (IFNAR1) was critical to maintain their normal homeostasis and to promote their activation during cutaneous inflammation. However, this did not require γδT17-intrinsic expression of IFNAR1. In contrast, expression of IFNAR1 by γδT17 cells was required in order to suppress IL-17 production during viral infection. Our data delineate direct from indirect IFNAR1 signaling and reveal an important immunoregulatory role for both tonic and inducible type I IFN in γδT17 cells.


Subject(s)
Interferon Type I/immunology , Lymphocyte Activation , Receptor, Interferon alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Interferon Type I/genetics , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction/genetics
4.
Eur J Immunol ; 51(8): 2097-2099, 2021 08.
Article in English | MEDLINE | ID: mdl-33960415

ABSTRACT

SMAC antagonization of cIAP1/2 in TH 17 cells upregulates cell adhesion and cytoskeleton genes through the NIK-RelB and p52 axis. SMAC also increases the homotypic interactions of TH 17 cells through a non-canonical NF-κB- and integrin-mediated mechanism resulting in increased ability of TH 17 cells to withstand shear stress.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , Th17 Cells/metabolism , Baculoviral IAP Repeat-Containing 3 Protein/antagonists & inhibitors , Cell Adhesion/physiology , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lymphocyte Activation/physiology
5.
Immunity ; 39(6): 1082-1094, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24315996

ABSTRACT

γδ T cells rapidly secrete inflammatory cytokines at barrier sites that aid in protection from pathogens, but mechanisms limiting inflammatory damage remain unclear. We found that retinoid-related orphan receptor gamma-t (RORγt) and interleukin-7 (IL-7) influence γδ T cell homeostasis and function by regulating expression of the inhibitory receptor, B and T lymphocyte attenuator (BTLA). The transcription factor RORγt, via its activating function-2 domain, repressed Btla transcription, whereas IL-7 increased BTLA levels on the cell surface. BTLA expression limited γδ T cell numbers and sustained normal γδ T cell subset frequencies by restricting IL-7 responsiveness and expansion of the CD27(-)RORγt(+) population. BTLA also negatively regulated IL-17 and TNF production in CD27(-) γδ T cells. Consequently, BTLA-deficient mice exhibit enhanced disease in a γδ T cell-dependent model of dermatitis, whereas BTLA agonism reduced inflammation. Therefore, by coordinating expression of BTLA, RORγt and IL-7 balance suppressive and activation stimuli to regulate γδ T cell homeostasis and inflammatory responses.


Subject(s)
Homeostasis , Inflammation , Receptors, Immunologic/metabolism , T-Lymphocytes/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Flow Cytometry , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Immunologic/genetics , T-Lymphocytes/immunology
6.
Eur J Immunol ; 50(6): 873-879, 2020 06.
Article in English | MEDLINE | ID: mdl-32092146

ABSTRACT

IL-17-producing gamma delta (γδT17) cells are innate lymphocytes critical for antibacterial protection at barrier surfaces such as the skin but also highly pathogenic during inflammation. It is therefore important to understand the cellular and molecular mechanisms that could counter-balance overt γδT17 cell activation. Immune checkpoint receptors (ICRs) deliver inhibitory signals to activated lymphocytes and have been implicated as negative regulators of mouse γδT17 cells. In this report, we investigated the cytokine signals that induce ICR expression on γδT17 cells and studied the in vivo role of the Src-homology-2 phosphatases 1 and 2 (SHP-1 and SHP-2) in the context of γδT17-induced psoriasis. We found that surface expression of ICRs can be induced by cytokines; however, SHP-1 or SHP-2 could not inhibit γδT17 responses. In this regard, conditional deletion of SHP-1, SHP-2, or both did no impact γδT17 cell development, expansion, cytokine production, or skin pathology.


Subject(s)
Lymphocyte Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Skin/immunology , Th17 Cells/immunology , Animals , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Skin/pathology , Th17 Cells/pathology
7.
Eur J Immunol ; 50(10): 1525-1536, 2020 10.
Article in English | MEDLINE | ID: mdl-32383212

ABSTRACT

Initiation of adaptive immunity to particulate antigens in lymph nodes largely depends on their presentation by migratory dendritic cells (DCs). DC subsets differ in their capacity to induce specific types of immunity, allowing subset-specific DC-targeting to influence vaccination and therapy outcomes. Faithful drug design, however, requires exact understanding of subset-specific versus global activation mechanisms. cDC1, the subset of DCs that excel in supporting immunity toward viruses, intracellular bacteria, and tumors, express uniquely high levels of the pattern recognition receptor TLR3. Using various murine genetic models, we show here that both, the cDC1 and cDC2 subsets of cDCs are activated and migrate equally well in response to TLR3 stimulation in a cell extrinsic and TNF-α dependent manner, but that cDC1 show a unique requirement for type I interferon signaling. Our findings reveal common and differing pathways regulating DC subset migration, offering important insights for the design of DC-based vaccination and therapy approaches.


Subject(s)
Dendritic Cells/immunology , Intestines/immunology , Toll-Like Receptor 3/metabolism , Animals , Cancer Vaccines , Cell Movement , Cells, Cultured , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Toll-Like Receptor 3/immunology
8.
EMBO Rep ; 20(11): e48647, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31549795

ABSTRACT

The transcription factors STAT3 and STAT4 are essential for lymphocyte differentiation and function. Interleukin (IL)-17 producing γδ T (γδT17) cells are innate lymphocytes important for anti-bacterial and inflammatory responses at barrier surfaces. Herein, we examine the role of STAT3 and STAT4 in regulating the homeostasis, activation, and pathogenicity of γδT17 cells. We show that STAT3 sustains γδT17 numbers in the skin but not in the lymph nodes, while STAT4 deficiency does not affect their homeostasis. Similarly, STAT3 but not STAT4 is essential for IL-23-induced IL-22 production by γδT17 cells. Concomitantly, mice lacking STAT3 expression in γδT17 cells develop significantly reduced psoriasis-like inflammation. STAT3-deficient γδT17 cells fail to expand and to upregulate IL-17A, IL-17F, and IL-22 in response to psoriatic stimuli. Although STAT4-deficient animals develop psoriasis-like disease, γδT17 cells in these mice are defective in IL-17F production. Collectively, our data demonstrate for the first time a critical role for STAT3 in orchestrating the homeostasis and pathogenicity of γδT17 cells and provide evidence for the requirement of STAT4 for optimal cytokine responses during inflammation.


Subject(s)
Dermatitis/etiology , Dermatitis/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , STAT3 Transcription Factor/metabolism , STAT4 Transcription Factor/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Biopsy , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Immunomodulation , Immunophenotyping , Inflammation Mediators/metabolism , Lymphocyte Count , Mice , Psoriasis/etiology , Psoriasis/metabolism
9.
Immunol Rev ; 260(1): 86-101, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24942684

ABSTRACT

The intestine presents a huge surface area to the outside environment, a property that is of critical importance for its key functions in nutrient digestion, absorption, and waste disposal. As such, the intestine is constantly exposed to dietary and microbial-derived foreign antigens, to which immune cells within the mucosa must suitably respond to maintain intestinal integrity, while also providing the ability to mount effective immune responses to potential pathogens. Dendritic cells (DCs) are sentinel immune cells that play a central role in the initiation and differentiation of adaptive immune responses. In the intestinal mucosa, DCs are located diffusely throughout the intestinal lamina propria, within gut-associated lymphoid tissues, including Peyer's patches and smaller lymphoid aggregates, as well as in intestinal-draining lymph nodes, including mesenteric lymph nodes. The recognition that dietary nutrients and microbial communities in the intestine influence both mucosal and systemic immune cell development and function as well as immune-mediated disease has led to an explosion of literature in mucosal immunology in recent years and a growing interest in the functionality of intestinal DCs. In the current review, we discuss recent findings from our group and others that have provided important insights regarding murine and human intestinal lamina propria DCs and highlighted marked developmental and functional heterogeneity within this compartment. A thorough understanding of the role these subsets play in the regulation of intestinal immune homeostasis and inflammation will help to define novel strategies for the treatment of intestinal pathologies and contribute to improved rational design of mucosal vaccines.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Mucosal/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Antigens/immunology , Antigens, CD/metabolism , CD11b Antigen/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Humans , Integrin alpha Chains/metabolism , Lymphocyte Activation/immunology , Mice , Phenotype , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription Factors/metabolism , Transcription, Genetic
10.
J Immunol ; 191(2): 828-36, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23761635

ABSTRACT

Lymphocyte activation is regulated by costimulatory and inhibitory receptors, of which both B and T lymphocyte attenuator (BTLA) and CD160 engage herpesvirus entry mediator (HVEM). Notably, it remains unclear how HVEM functions with each of its ligands during immune responses. In this study, we show that HVEM specifically activates CD160 on effector NK cells challenged with virus-infected cells. Human CD56(dim) NK cells were costimulated specifically by HVEM but not by other receptors that share the HVEM ligands LIGHT, Lymphotoxin-α, or BTLA. HVEM enhanced human NK cell activation by type I IFN and IL-2, resulting in increased IFN-γ and TNF-α secretion, and tumor cell-expressed HVEM activated CD160 in a human NK cell line, causing rapid hyperphosphorylation of serine kinases ERK1/2 and AKT and enhanced cytolysis of target cells. In contrast, HVEM activation of BTLA reduced cytolysis of target cells. Together, our results demonstrate that HVEM functions as a regulator of immune function that activates NK cells via CD160 and limits lymphocyte-induced inflammation via association with BTLA.


Subject(s)
Antigens, CD/metabolism , Killer Cells, Natural/immunology , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , CD56 Antigen/metabolism , Cell Line , Enzyme Activation , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Inflammation , Interferon Type I/metabolism , Interferon-gamma/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Lymphotoxin-alpha/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
J Immunol ; 191(9): 4611-8, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24078690

ABSTRACT

Innate lymphoid cells encompass a diverse array of lymphocyte subsets with unique phenotype that initiate inflammation and provide host defenses in specific microenvironments. In this study, we identify a rare human CD4(+)CD3(-) innate-like lymphoid population with high TNF expression that is enriched in blood from patients with rheumatoid arthritis. These CD4(+)CD3(-) cells belong to the T cell lineage, but the lack of AgR at the cell surface renders them nonresponsive to TCR-directed stimuli. By developing a culture system that sustains survival, we show that CD4(+)CD3(-) innate-like T cells display IL-7-dependent induction of surface lymphotoxin-αß, demonstrating their potential to modify tissue microenvironments. Furthermore, expression of CCR6 on the CD4(+)CD3(-) population defines a CD127(high) subset that is highly responsive to IL-7. This CD4(+)CD3(-) population is enriched in the peripheral blood from rheumatoid arthritis patients, suggesting a link to their involvement in chronic inflammatory disease.


Subject(s)
Arthritis, Rheumatoid/metabolism , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Tumor Necrosis Factors/metabolism , Adult , Aged , Animals , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Female , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Inflammation , Interleukin-7/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphotoxin alpha1, beta2 Heterotrimer/immunology , Male , Mice , Mice, Knockout , Middle Aged , Receptors, CCR6/metabolism
12.
Immunol Rev ; 244(1): 134-48, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22017436

ABSTRACT

CD4(+) effector and memory T cells play a pivotal role in the development of both normal and pathogenic immune responses. This review focuses on the molecular and cellular mechanisms that regulate their development, with particular focus on the tumor necrosis factor superfamily members OX40 (TNFRSF4) and CD30 (TNFRSF8). We discuss the evidence that in mice, these molecular signaling pathways act synergistically to regulate the development of both effector and memory CD4(+) T cells but that the cells that regulate memory versus effector function are distinct, effectively allowing the independent regulation of the memory and effector CD4(+) T-cell pools.


Subject(s)
B-Lymphocytes/immunology , Immunity, Innate , Immunologic Memory , Ki-1 Antigen/immunology , Lymphoid Tissue/immunology , Receptors, OX40/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmunity , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Communication , Gene Expression/immunology , Humans , Ki-1 Antigen/genetics , Ki-1 Antigen/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Knockout , Receptors, OX40/genetics , Receptors, OX40/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology , Tumor Necrosis Factors/metabolism
13.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G338-46, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24875104

ABSTRACT

Adenosine is a purine metabolite that can mediate anti-inflammatory responses in the digestive tract through the A(2A) adenosine receptor (A(2A)AR). We examined the role of this receptor in the control of inflammation in the adoptive transfer model of colitis. Infection of A(2A)AR(-/-) mice with Helicobacter hepaticus increased colonic inflammation scores compared with uninfected A(2A)AR controls. Comparison of T cell subsets in wild-type and A(2A)AR(-/-) mice revealed differences in markers associated with activated helper T (Th) cells and regulatory T (Treg) cells. Previous studies showed that expression of A(2A)AR on CD45RB(HI) and CD45RB(LO) Th cells is essential for the proper regulation of colonic inflammation. Adoptive transfer of CD45RB(HI) with CD45RB(LO) from wild-type mice into RAG1(-/-)/A(2A)AR(-/-) mice induced severe disease within 3 wk, although transfer of the same subsets into RAG1(-/-) mice does not induce colitis. This suggests that the presence of A(2A)AR on recipient cells is also important for controlling colitis. To investigate the role of A(2A)AR in myeloid cells, chimeric recipients were generated by injection of bone marrow from RAG1(-/-) or RAG1(-/-)/A(2A)AR(-/-) mice into irradiated RAG1(-/-) mice. After adoptive transfer, these recipients did not develop colitis, regardless of A(2A)AR expression by the donor. Together, our results suggest that the control of inflammation in vivo is dependent on A(2A)AR signaling through multiple cell types that collaborate in the regulation of colitis by responding to extracellular adenosine.


Subject(s)
Adenosine/metabolism , Colitis/prevention & control , Colon/metabolism , Lymph Nodes/metabolism , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , Biomarkers/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colon/immunology , Colon/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Helicobacter hepaticus/pathogenicity , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation Mediators/metabolism , Leukocyte Common Antigens/metabolism , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors
14.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37440178

ABSTRACT

Early-life cues shape the immune system during adulthood. However, early-life signaling pathways and their temporal functions are not well understood. Herein, we demonstrate that the cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1/2), which are E3 ubiquitin ligases, sustain interleukin (IL)-17-producing γ δ T cells (γδT17) and group 3 innate lymphoid cells (ILC3) during late neonatal and prepubescent life. We show that cell-intrinsic deficiency of cIAP1/2 at 3-4 wk of life leads to downregulation of the transcription factors cMAF and RORγt and failure to enter the cell cycle, followed by progressive loss of γδT17 cells and ILC3 during aging. Mice deficient in cIAP1/2 have severely reduced γδT17 cells and ILC3, present with suboptimal γδT17 responses in the skin, lack intestinal isolated lymphoid follicles, and cannot control intestinal bacterial infection. Mechanistically, these effects appear to be dependent on overt activation of the non-canonical NF-κB pathway. Our data identify cIAP1/2 as early-life molecular switches that establish effective type 3 immunity during aging.


Subject(s)
Immunity, Innate , Ubiquitin , Mice , Animals , Lymphocytes , Interleukins/metabolism , Aging
15.
Eur J Immunol ; 41(6): 1563-72, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469096

ABSTRACT

Here, we identify cells within human adult secondary lymphoid tissues that are comparable in phenotype and location to the lymphoid tissue inducer (LTi) cells that persist in the adult mouse. Identified as CD117(+) CD3(-) CD56(-) cells, like murine LTi cells, they lack expression of many common lineage markers and express CD127, OX40L and TRANCE. These cells were detected at the interface between the B- and T- zones, as well as at the subcapsular sinus in LNs, the location where LTi cells reside in murine spleen and LNs. Furthermore, like murine LTi cells, these cells expressed high levels of IL-22 and upregulated IL-22 expression upon IL-23 stimulation. Importantly, these cells were not an NK cell subset since they showed no expression of IFN-γ and perforin. Interestingly, a subset of the CD117(+) CD3(-) CD56(-) OX40L(+) population expressed NKp46, again similar to recent findings in mice. Finally, these cells supported memory CD4(+) T-cell survival in an OX40L-dependent manner. Combined, these data indicate that the CD117(+) CD3(-) CD56(-) OX40L(+) cells in human secondary lymphoid tissues are comparable in phenotype, location and function to the LTi cells that persist within adult murine secondary lymphoid tissues.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interleukins/metabolism , OX40 Ligand/metabolism , Palatine Tonsil/cytology , Th17 Cells/metabolism , Animals , Antigens, CD/biosynthesis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Separation , Cell Survival , Cells, Cultured , Flow Cytometry , Humans , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukins/genetics , Interleukins/immunology , Lymph Nodes/cytology , Mice , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , OX40 Ligand/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Interleukin-22
16.
J Immunol ; 182(8): 4771-5, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342654

ABSTRACT

Lymphoid tissue inducer cells express a diverse array of tumor necrosis family ligands, including those that bind CD30 and the lymphotoxin beta receptor. Both of these signaling pathways have been linked with B/T segregation in the spleen. In this study, we have dissected a lymphotoxin-independent CD30-dependent signal for the induction of expression of the T zone chemokine, CCL21. Reduced expression of CCL21 due to CD30 deficiency was functionally significant: mice deficient in both lymphotoxin and CD30 (dKO) signals had significantly smaller accumulations of lymphocytes in their splenic white pulp areas, with no evidence of focal aggregation of T cells. Furthermore, recruitment of wild-type CD4 T cells was poor in dKO mice compared with both wild-type or lymphotoxin-deficient mice. Phylogeny suggests that CD30 signals predated those through the lymphotoxin beta receptor. We suggest that CD30 signals from lymphoid tissue inducer cells were a primitive mechanism to recruit and prime CD4 T cells. This would have been a stepping stone in the evolution of the highly organized lymphotoxin dependent B and T white pulp areas within which CD4-dependent memory Ab responses now develop.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chemokine CCL21/immunology , Ki-1 Antigen/immunology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , Animals , Female , Gene Expression Regulation , Ki-1 Antigen/deficiency , Ki-1 Antigen/genetics , Ki-1 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
Cancers (Basel) ; 13(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34572874

ABSTRACT

In recent years, checkpoint inhibitor (CPI) therapy has shown promising clinical responses across a broad range of cancers. However, many patients remain unresponsive and there is need for improvement. CPI therapy relies on antibody-mediated neutralization of immune inhibitory or checkpoint receptors (ICRs) that constitutively suppress leukocytes. In this regard, the clinical outcome of CPI therapy has primarily been attributed to modulating classical MHC-restricted αß T cell responses, yet, it will inevitably target most lymphoid (and many myeloid) populations. As such, unconventional non-MHC-restricted gamma delta (γδ) T, mucosal associated invariant T (MAIT) and natural killer T (NKT) cells express ICRs at steady-state and after activation and may thus be affected by CPI therapies. To which extent, however, remains unclear. These unconventional T cells are polyfunctional innate-like lymphocytes that play a key role in tumor immune surveillance and have a plethora of protective and pathogenic immune responses. The robust anti-tumor potential of γδ T, MAIT, and NKT cells has been established in a variety of preclinical cancer models and in clinical reports. In contrast, recent studies have documented a pro-tumor effect of innate-like T cell subsets that secrete pro-inflammatory cytokines. Consequently, understanding the mechanisms that regulate such T cells and their response to CPI is critical in designing effective cancer immunotherapies that favor anti-tumor immunity. In this Review, we will discuss the current understanding regarding the role of immune checkpoint regulation in γδ T, MAIT, and NKT cells and its importance in anti-cancer immunity.

18.
Cancers (Basel) ; 13(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34944832

ABSTRACT

γδ T cells are unique players in shaping immune responses, lying at the intersection between innate and adaptive immunity. Unlike conventional αß T cells, γδ T cells largely populate non-lymphoid peripheral tissues, demonstrating tissue specificity, and they respond to ligands in an MHC-independent manner. γδ T cells display rapid activation and effector functions, with a capacity for cytotoxic anti-tumour responses and production of inflammatory cytokines such as IFN-γ or IL-17. Their rapid cytotoxic nature makes them attractive cells for use in anti-cancer immunotherapies. However, upon transformation, γδ T cells can give rise to highly aggressive lymphomas. These rare malignancies often display poor patient survival, and no curative therapies exist. In this review, we discuss the diverse roles of γδ T cells in immune surveillance and response, with a particular focus on cancer immunity. We summarise the intriguing dichotomy between pro- and anti-tumour functions of γδ T cells in solid and haematological cancers, highlighting the key subsets involved. Finally, we discuss potential drivers of γδ T-cell transformation, summarising the main γδ T-cell lymphoma/leukaemia entities, their clinical features, recent advances in mapping their molecular and genomic landscapes, current treatment strategies and potential future targeting options.

19.
Eur J Immunol ; 39(1): 280-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19089813

ABSTRACT

In this report we describe a transplantation system where embryonic spleens are grafted into adult hosts. This model can be used to analyze the cellular and molecular requirements for the development and organization of splenic microenvironments. Whole embryonic day 15 (ED15) spleens, grafted under the kidney capsule of adult mice, were colonized by host-derived lymphocytes and DC and developed normal splenic architecture. Grafts were also able to form germinal centers in response to T-dependent antigen. Using this system we demonstrated that adult host-derived lymphotoxin (LT) alpha was sufficient for the development of ED15 LT alpha(-/-) grafts. Grafting of ED15 LT alpha(-/-) spleens into RAG(-/-) hosts followed by transfer of LT alpha(-/-) splenocytes revealed no requirement for lymphocyte-derived LT alpha in the induction of CCL21 or the development of T-zone stroma. These data suggest that interactions between adult lymphoid-tissue inducer-like cells and embryonic stromal cells initiated T-zone development. Furthermore, adult lymphoid tissue inducer-like cells were shown to develop from bone marrow-derived progenitors. The model described here demonstrates a method of transferring whole splenic microenvironments and dissecting the stromal and hematopoietic signals involved in spleen development and organization.


Subject(s)
Cell Communication/immunology , Models, Animal , Spleen/embryology , Spleen/immunology , T-Lymphocytes/immunology , Animals , Antigens, Viral, Tumor/immunology , Cell Differentiation/immunology , Chemokine CCL21/metabolism , Kidney/immunology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology , Spleen/transplantation
20.
Eur J Immunol ; 39(8): 2120-5, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19609980

ABSTRACT

Prior to acquiring a memory phenotype, antigen-activated CD8(+) T cells need to expand and then undergo a contraction phase. Utilizing two different antigenic stimuli, we provide evidence that the tumor necrosis factor receptors OX40 and CD30 integrate synergistic signals during the expansion phase to help maintain CD8(+) effectors. Thus, double deficiency in OX40 and CD30 leads to CD8(+) cell loss during expansion after immunization either with OVA or with murine CMV. Following their contraction, OX40- and CD30-deficient CD8(+) T cells persist normally in CMV-infected mice. In contrast, persistence after OVA challenge is dependent on OX40 and CD30. Collectively, our data define the important role of both OX40 and CD30 during CD8(+) T-cell activation, and show that long-term CD8 persistence after contraction is regulated not only by stimulatory receptors but also by the nature of the antigen or how the antigen is presented.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Ki-1 Antigen/physiology , Receptors, OX40/physiology , Signal Transduction , Adoptive Transfer , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Flow Cytometry , Immunization, Secondary , Immunologic Memory/immunology , Interferon-gamma/metabolism , Ki-1 Antigen/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, OX40/genetics
SELECTION OF CITATIONS
SEARCH DETAIL