Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Cell ; 185(19): 3501-3519.e20, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36041436

ABSTRACT

How intestinal microbes regulate metabolic syndrome is incompletely understood. We show that intestinal microbiota protects against development of obesity, metabolic syndrome, and pre-diabetic phenotypes by inducing commensal-specific Th17 cells. High-fat, high-sugar diet promoted metabolic disease by depleting Th17-inducing microbes, and recovery of commensal Th17 cells restored protection. Microbiota-induced Th17 cells afforded protection by regulating lipid absorption across intestinal epithelium in an IL-17-dependent manner. Diet-induced loss of protective Th17 cells was mediated by the presence of sugar. Eliminating sugar from high-fat diets protected mice from obesity and metabolic syndrome in a manner dependent on commensal-specific Th17 cells. Sugar and ILC3 promoted outgrowth of Faecalibaculum rodentium that displaced Th17-inducing microbiota. These results define dietary and microbiota factors posing risk for metabolic syndrome. They also define a microbiota-dependent mechanism for immuno-pathogenicity of dietary sugar and highlight an elaborate interaction between diet, microbiota, and intestinal immunity in regulation of metabolic disorders.


Subject(s)
Metabolic Syndrome , Microbiota , Animals , Diet, High-Fat , Dietary Sugars , Interleukin-17 , Intestinal Mucosa , Lipids , Mice , Mice, Inbred C57BL , Obesity , Th17 Cells
2.
Nat Immunol ; 13(6): 604-11, 2012 May 06.
Article in English | MEDLINE | ID: mdl-22561605

ABSTRACT

Antibody class defines function in B cell immunity, but how class is propagated into B cell memory remains poorly understood. Here we demonstrate that memory B cell subsets unexpectedly diverged across antibody class through differences in the effects of major transcriptional regulators. Conditional genetic deletion of the gene encoding the transcription factor T-bet selectively blocked the formation and antigen-specific response of memory B cells expressing immunoglobulin G2a (IgG2a) in vivo. Cell-intrinsic expression of T-bet regulated expression of the transcription factor STAT1, steady-state cell survival and transcription of IgG2a-containing B cell antigen receptors (BCRs). In contrast, the transcription factor RORα and not T-bet was expressed in IgA(+) memory B cells, with evidence that knockdown of RORα mRNA expression and chemical inhibition of transcriptional activity also resulted in lower survival and BCR expression of IgA(+) memory B cells. Thus, divergent transcriptional regulators dynamically maintain subset integrity to promote specialized immune function in class-specific memory B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Immunologic Memory/immunology , Receptor Tyrosine Kinase-like Orphan Receptors/immunology , T-Box Domain Proteins/immunology , Animals , B-Lymphocytes/classification , Flow Cytometry , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/chemistry , RNA, Messenger/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptors, Antigen, B-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/immunology , Specific Pathogen-Free Organisms , T-Box Domain Proteins/genetics , Transcription, Genetic/immunology
3.
J Immunol ; 209(4): 660-664, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35905999

ABSTRACT

Immune checkpoint blockade is limited by resistance to treatment, with many patients not achieving durable antitumor responses. Self-renewing (T cell factor 1+ [TCF1+]) CD8+ T cells have recently been implicated in efficacy of anti-programmed cell death protein 1 (anti-PD-1). Mice challenged with syngeneic tumors were treated with anti-PD-1 and/or a reversible inhibitor of PI3K δ, designed to promote T cell self-renewal. Growth of tumors in untreated mice was characterized by waning proportions of TCF1+ T cells, suggesting self-renewing T cells become limiting for successful immunotherapy. Higher proportions of TCF1+ T cells in tumor and blood correlated with better control of tumor growth. Combining anti-PD-1 and inhibitor of PI3K δ conferred superior protection compared with either monotherapy and was associated with higher frequency of TCF1+ T cells in tumor and blood compared with anti-PD-1 alone. These findings reveal predictive importance of self-renewing T cells in anti-tumor immunity and suggest that resistance-directed strategies to enhance T cell self-renewal could potentiate the efficacy of PD-1 blockade.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Cell Death , Immunotherapy , Mice , Phosphatidylinositol 3-Kinases , T Cell Transcription Factor 1
4.
Nat Immunol ; 12(7): 663-71, 2011 May 29.
Article in English | MEDLINE | ID: mdl-21623380

ABSTRACT

T cell exhaustion has a major role in failure to control chronic infection. High expression of inhibitory receptors, including PD-1, and the inability to sustain functional T cell responses contribute to exhaustion. However, the transcriptional control of these processes remains unclear. Here we demonstrate that the transcription factor T-bet regulated the exhaustion of CD8(+) T cells and the expression of inhibitory receptors. T-bet directly repressed transcription of the gene encoding PD-1 and resulted in lower expression of other inhibitory receptors. Although a greater abundance of T-bet promoted terminal differentiation after acute infection, high T-bet expression sustained exhausted CD8(+) T cells and repressed the expression of inhibitory receptors during chronic viral infection. Persistent antigenic stimulation caused downregulation of T-bet, which resulted in more severe exhaustion of CD8(+) T cells. Our observations suggest therapeutic opportunities involving higher T-bet expression during chronic infection.


Subject(s)
Antigens, Differentiation/immunology , Lymphocytic Choriomeningitis/immunology , T-Box Domain Proteins/immunology , Animals , Antigens, Differentiation/genetics , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Transcription, Genetic/immunology
5.
Immunity ; 36(1): 55-67, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22261438

ABSTRACT

Natural killer (NK) cells play critical roles defending against tumors and pathogens. We show that mice lacking both transcription factors Eomesodermin (Eomes) and T-bet failed to develop NK cells. Developmental stability of immature NK cells constitutively expressing the death ligand TRAIL depended on T-bet. Conversely, maturation characterized by loss of constitutive TRAIL expression and induction of Ly49 receptor diversity and integrin CD49b (DX5(+)) required Eomes. Mature NK cells from which Eomes was deleted reverted to phenotypic immaturity if T-bet was present or downregulated NK lineage antigens if T-bet was absent, despite retaining expression of Ly49 receptors. Fetal and adult hepatic hematopoiesis restricted Eomes expression and limited NK development to the T-bet-dependent, immature stage, whereas medullary hematopoiesis permitted Eomes-dependent NK maturation in adult mice. These findings reveal two sequential, genetically separable checkpoints of NK cell maturation, the progression of which is metered largely by the anatomic localization of hematopoiesis.


Subject(s)
Cell Cycle/genetics , Cell Differentiation , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , T-Box Domain Proteins/metabolism , Animals , Cell Lineage , Flow Cytometry , Gene Deletion , Mice , Mice, Knockout , Models, Immunological , Real-Time Polymerase Chain Reaction , T-Box Domain Proteins/genetics
6.
Immunity ; 37(3): 511-23, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22981537

ABSTRACT

Interferon-γ (IFN-γ) promotes a population of T-bet(+) CXCR3(+) regulatory T (Treg) cells that limit T helper 1 (Th1) cell-mediated pathology. Our studies demonstrate that interleukin-27 (IL-27) also promoted expression of T-bet and CXCR3 in Treg cells. During infection with Toxoplasma gondii, a similar population emerged that limited T cell responses and was dependent on IFN-γ in the periphery but on IL-27 at mucosal sites. Transfer of Treg cells ameliorated the infection-induced pathology observed in Il27(-/-) mice, and this was dependent on their ability to produce IL-10. Microarray analysis revealed that Treg cells exposed to either IFN-γ or IL-27 have distinct transcriptional profiles. Thus, IFN-γ and IL-27 have different roles in Treg cell biology and IL-27 is a key cytokine that promotes the development of Treg cells specialized to control Th1 cell-mediated immunity at local sites of inflammation.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-17/pharmacology , Salmonella Infections, Animal/immunology , T-Lymphocytes, Regulatory/drug effects , Toxoplasmosis, Animal/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toxoplasma/immunology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology
7.
Immunity ; 34(4): 492-504, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21497118

ABSTRACT

Polarized segregation of proteins in T cells is thought to play a role in diverse cellular functions including signal transduction, migration, and directed secretion of cytokines. Persistence of this polarization can result in asymmetric segregation of fate-determining proteins during cell division, which may enable a T cell to generate diverse progeny. Here, we provide evidence that a lineage-determining transcription factor, T-bet, underwent asymmetric organization in activated T cells preparing to divide and that it was unequally partitioned into the two daughter cells. This unequal acquisition of T-bet appeared to result from its asymmetric destruction during mitosis by virtue of concomitant asymmetric segregation of the proteasome. These results suggest a mechanism by which a cell may unequally localize cellular activities during division, thereby imparting disparity in the abundance of cell fate regulators in the daughter cells.


Subject(s)
Mitosis , Proteasome Endopeptidase Complex/metabolism , T-Box Domain Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Polarity , Cells, Cultured , Mice , Mice, Inbred C57BL , Phosphorylation , T-Box Domain Proteins/metabolism , T-Lymphocytes/enzymology
8.
J Immunol ; 200(6): 1977-1981, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29440505

ABSTRACT

The T-box transcription factors T-bet and Eomesodermin (Eomes) instruct discrete stages in NK cell development. However, their role in the immune response of mature NK cells against pathogens remains unexplored. We used an inducible deletion system to elucidate the cell-intrinsic role of T-bet and Eomes in mature NK cells during the course of mouse CMV infection. We show both T-bet and Eomes to be necessary for the expansion of virus-specific NK cells, with T-bet upregulation induced by IL-12 signaling and STAT4 binding to a conserved enhancer region upstream of the Tbx21 loci. Interestingly, our data suggest maintenance of virus-specific memory NK cell numbers and phenotype was dependent on T-bet, but not Eomes. These findings uncover a nonredundant and stage-specific influence of T-box transcription factors in the antiviral NK cell response.


Subject(s)
Immunologic Memory/immunology , Killer Cells, Natural/immunology , T-Box Domain Proteins/immunology , Animals , Cytomegalovirus Infections/immunology , Interleukin-12/immunology , Mice , STAT4 Transcription Factor/immunology , Up-Regulation/immunology
9.
J Immunol ; 200(4): 1513-1526, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29305435

ABSTRACT

Agonists to the TNF/TNFR costimulatory receptors CD134 (OX40) and CD137 (4-1BB) elicit antitumor immunity. Dual costimulation with anti-CD134 plus anti-CD137 is particularly potent because it programs cytotoxic potential in CD8+ and CD4+ T cells. Cytotoxicity in dual-costimulated CD4 T cells depends on the T-box transcription factor eomesodermin (Eomes), which we report is induced via a mechanism that does not rely on IL-2, in contrast to CD8+ CTL, but rather depends on the CD8 T cell lineage commitment transcription factor Runx3, which supports Eomes expression in mature CD8+ CTLs. Further, Eomes and Runx3 were indispensable for dual-costimulated CD4 T cells to mediate antitumor activity in an aggressive melanoma model. Runx3 is also known to be expressed in standard CD4 Th1 cells where it fosters IFN-γ expression; however, the CD4 T cell lineage commitment factor ThPOK represses transcription of Eomes and other CD8 lineage genes, such as Cd8a Hence, CD4 T cells can differentiate into Eomes+ cytotoxic CD4+CD8+ double-positive T cells by terminating ThPOK expression. In contrast, dual-costimulated CD4 T cells express Eomes, despite the continued expression of ThPOK and the absence of CD8α, indicating that Eomes is selectively released from ThPOK repression. Finally, although Eomes was induced by CD137 agonist, but not CD134 agonist, administered individually, CD137 agonist failed to induce CD134-/- CD4 T cells to express Eomes or Runx3, indicating that both costimulatory pathways are required for cytotoxic Th1 programming, even when only CD137 is intentionally engaged with a therapeutic agonist.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , T-Box Domain Proteins/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Cell Differentiation/immunology , Core Binding Factor Alpha 3 Subunit/immunology , Immunotherapy , Lymphocyte Activation/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Transgenic , Receptors, OX40/agonists , Receptors, OX40/immunology , Transcription Factors/immunology , Transcription Factors/metabolism
10.
Immunol Cell Biol ; 96(8): 863-871, 2018 09.
Article in English | MEDLINE | ID: mdl-29570858

ABSTRACT

Growth signals drive hematopoietic progenitor cells to proliferate and branch into divergent cell fates, but how unequal outcomes arise from a common progenitor is not fully understood. We used steady-state analysis of in vivo hematopoiesis and Fms-related tyrosine kinase 3 ligand (Flt3L)-induced in vitro differentiation of dendritic cells (DCs) to determine how growth signals regulate lineage bias. We found that Flt3L signaling induced anabolic activation and proliferation of DC progenitors, which was associated with DC differentiation. Perturbation of processes associated with quiescence and catabolism, including AMP-activated protein kinase signaling, fatty acid oxidation, or mitochondrial clearance increased development of cDC2 cells at the expense of cDC1 cells. Conversely, scavenging anabolism-associated reactive oxygen species skewed differentiation toward cDC1 cells. Sibling daughter cells of dividing DC progenitors exhibited unequal expression of the transcription factor interferon regulatory factor 8, which correlated with clonal divergence in FoxO3a signaling and population-level bifurcation of cell fate. We propose that unequal transmission of growth signals during cell division might support fate branches during proliferative expansion of progenitors.


Subject(s)
Dendritic Cells/physiology , Fatty Acids/metabolism , Hematopoietic Stem Cells/physiology , Interferon Regulatory Factors/metabolism , Membrane Proteins/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Hematopoiesis , Interferon Regulatory Factors/genetics , Lipid Metabolism , Metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Signal Transduction
11.
Immunity ; 31(1): 122-30, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19592277

ABSTRACT

The transcription factor KLF2 regulates T cell trafficking by promoting expression of the lipid-binding receptor S1P(1) and the selectin CD62L. Recently, it was proposed that KLF2 also represses the expression of chemokine receptors. We confirmed the upregulation of the chemokine receptor CXCR3 on KLF2-deficient T cells. However, we showed that this was a cell-nonautonomous effect, as revealed by CXCR3 upregulation on wild-type bystander cells in mixed bone-marrow chimeras with KLF2-deficient cells. Furthermore, KLF2-deficient T cells overproduced IL-4, leading to the upregulation of CXCR3 through an IL-4-receptor- and eomesodermin-dependent pathway. Consistent with the increased IL-4 production, we found high concentrations of serum IgE in mice with T cell-specific KLF2 deficiency. Our findings support a model where KLF2 regulates T cell trafficking by direct regulation of S1P(1) and CD62L and restrains spontaneous cytokine production in naive T cells.


Subject(s)
Interleukin-4/biosynthesis , Kruppel-Like Transcription Factors/metabolism , L-Selectin/metabolism , Receptors, Lysosphingolipid/metabolism , T-Lymphocytes/immunology , Animals , Immunoglobulin E/blood , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/metabolism , T-Box Domain Proteins/metabolism , Up-Regulation/immunology
12.
Immunity ; 31(2): 309-20, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19664943

ABSTRACT

T cell exhaustion is common during chronic infections and can prevent optimal immunity. Although recent studies have demonstrated the importance of inhibitory receptors and other pathways in T cell exhaustion, the underlying transcriptional mechanisms are unknown. Here, we define a role for the transcription factor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Blimp-1 repressed key aspects of normal memory CD8(+) T cell differentiation and promoted high expression of inhibitory receptors during chronic infection. These cardinal features of CD8(+) T cell exhaustion were corrected by conditionally deleting Blimp-1. Although high expression of Blimp-1 fostered aspects of CD8(+) T cell exhaustion, haploinsufficiency indicated that moderate Blimp-1 expression sustained some effector function during chronic viral infection. Thus, we identify Blimp-1 as a transcriptional regulator of CD8(+) T cell exhaustion during chronic viral infection and propose that Blimp-1 acts as a transcriptional rheostat balancing effector function and T cell exhaustion.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Transcription Factors/metabolism , Virus Diseases/immunology , Acute Disease , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Chronic Disease , Cytotoxicity, Immunologic/immunology , GPI-Linked Proteins , Granzymes/immunology , Granzymes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1 , Programmed Cell Death 1 Receptor , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family , Transcription Factors/genetics , Virus Diseases/genetics , Lymphocyte Activation Gene 3 Protein
13.
J Immunol ; 196(4): 1449-54, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26792802

ABSTRACT

Type 1 innate lymphocytes comprise two developmentally divergent lineages, type 1 helper innate lymphoid cells (hILC1s) and conventional NK cells (cNKs). All type 1 innate lymphocytes (ILCs) express the transcription factor T-bet, but cNKs additionally express Eomesodermin (Eomes). We show that deletion of Eomes alleles at the onset of type 1 ILC maturation using NKp46-Cre imposes a substantial block in cNK development. Formation of the entire lymphoid and nonlymphoid type 1 ILC compartment appears to require the semiredundant action of both T-bet and Eomes. To determine if Eomes is sufficient to redirect hILC1 development to a cNK fate, we generated transgenic mice that express Eomes when and where T-bet is expressed using Tbx21 locus control to drive expression of Eomes codons. Ectopic Eomes induces cNK-like properties across the lymphoid and nonlymphoid type 1 ILC compartments. Subsequent to their divergent lineage specification, hILC1s and cNKs thus possess substantial developmental plasticity.


Subject(s)
Killer Cells, Natural/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Lineage , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Mice, Transgenic , Th1 Cells/immunology
14.
J Immunol ; 197(4): 1017-22, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27430722

ABSTRACT

The role of Ab and B cells in preventing infection is established. In contrast, the role of B cell responses in containing chronic infections remains poorly understood. IgG2a (IgG1 in humans) can prevent acute infections, and T-bet promotes IgG2a isotype switching. However, whether IgG2a and B cell-expressed T-bet influence the host-pathogen balance during persisting infections is unclear. We demonstrate that B cell-specific loss of T-bet prevents control of persisting viral infection. T-bet in B cells controlled IgG2a production, as well as mucosal localization, proliferation, glycosylation, and a broad transcriptional program. T-bet controlled a broad antiviral program in addition to IgG2a because T-bet in B cells was important, even in the presence of virus-specific IgG2a. Our data support a model in which T-bet is a universal controller of antiviral immunity across multiple immune lineages.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , T-Box Domain Proteins/immunology , Animals , Cell Separation , Chronic Disease , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoglobulin G/immunology , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis
15.
Immunity ; 29(6): 971-85, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19100702

ABSTRACT

Although T lymphocytes are constitutively nonadherent cells, they undergo facultative polarity during migration and upon interaction with cells presenting cognate antigen, suggesting that cell polarity might be critical for target cell destruction. Using two-photon imaging of tumor-infiltrating T lymphocytes, we found that CD44, a receptor for extracellular matrix proteins and glycosaminoglycans, was crucial for interstitial T cell navigation and, consequently, efficient tumor cell screening. CD44 functioned as a critical regulator of intratumoral movement by stabilizing cell polarity in migrating T cells, but not during target cell interactions. Stable anterior-posterior asymmetry was maintained by CD44 independently of its extracellular domain. Instead, migratory polarity depended on the recruitment of ezrin, radixin, moesin (ERM) proteins by the intracellular domain of CD44 to the posterior cellular protrusion. Our results formally demonstrate that CD44-dependent T lymphocyte locomotion within target sites represents an essential immunologic checkpoint that determines the potency of T cell effector functions.


Subject(s)
Cell Movement/immunology , Cell Polarity/immunology , Hyaluronan Receptors/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Adhesion/immunology , Cell Communication/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Movement/genetics , Cytoskeletal Proteins/immunology , Cytoskeletal Proteins/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Neoplasms/metabolism , Protein Structure, Tertiary , T-Lymphocytes, Cytotoxic/metabolism
16.
J Immunol ; 194(3): 1131-40, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25556247

ABSTRACT

The transcription factor T-bet has been most prominently linked to NK and T cell production of IFN-γ, a cytokine required for the control of a diverse array of intracellular pathogens. Indeed, in mice challenged with the parasite Toxoplasma gondii, NK and T cell responses are characterized by marked increases of T-bet expression. Unexpectedly, T-bet(-/-) mice infected with T. gondii develop a strong NK cell IFN-γ response that controls parasite replication at the challenge site, but display high parasite burdens at secondary sites colonized by T. gondii and succumb to infection. The loss of T-bet had a modest effect on T cell production of IFN-γ but did not impact on the generation of parasite-specific T cells. However, the absence of T-bet resulted in lower T cell expression of CD11a, Ly6C, KLRG-1, and CXCR3 and fewer parasite-specific T cells at secondary sites of infection, associated with a defect in parasite control at these sites. Together, these data highlight T-bet-independent pathways to IFN-γ production and reveal a novel role for this transcription factor in coordinating the T cell responses necessary to control this infection in peripheral tissues.


Subject(s)
Disease Resistance/genetics , Disease Resistance/immunology , Immunity , Infections/genetics , Infections/immunology , T-Box Domain Proteins/genetics , Animals , Disease Models, Animal , Gene Expression , Genetic Predisposition to Disease , Immunity, Cellular , Immunity, Innate , Immunophenotyping , Infections/metabolism , Infections/parasitology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Knockout , Phenotype , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toxoplasma/immunology , Toxoplasmosis, Animal/genetics , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/metabolism
18.
J Immunol ; 193(3): 1013-6, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973450

ABSTRACT

Central memory (CM) CD8(+) T cells "remember" prior encounters because they maintain themselves through cell division in the absence of ongoing challenge (homeostatic self-renewal), as well as reproduce the CM fate while manufacturing effector cells during secondary Ag encounters (rechallenge self-renewal). We tested the consequence of conditional deletion of the bone marrow homing receptor CXCR4 on antiviral T cell responses. CXCR4-deficient CD8(+) T cells have impaired memory cell maintenance due to defective homeostatic proliferation. Upon rechallenge, however, CXCR4-deficient T cells can re-expand and renew the CM pool while producing secondary effector cells. The critical bone marrow-derived signals essential for CD8(+) T cell homeostatic self-renewal appear to be dispensable to yield self-renewing, functionally asymmetric cell fates during rechallenge.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Homeostasis/immunology , Immunologic Memory , Receptors, CXCR4/deficiency , Receptors, CXCR4/physiology , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/cytology , Clone Cells , Homeostasis/genetics , Humans , Immunologic Memory/genetics , Immunophenotyping , Mice , Mice, Knockout , Mice, Transgenic , Receptors, CXCR4/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism
19.
Proc Natl Acad Sci U S A ; 110(10): 4081-6, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431145

ABSTRACT

The cortical area map is initially patterned by transcription factor (TF) gradients in the neocortical primordium, which define a "protomap" in the embryonic ventricular zone (VZ). However, mechanisms that propagate regional identity from VZ progenitors to cortical plate (CP) neurons are unknown. Here we show that the VZ, subventricular zone (SVZ), and CP contain distinct molecular maps of regional identity, reflecting different gene expression gradients in radial glia progenitors, intermediate progenitors, and projection neurons, respectively. The "intermediate map" in the SVZ is modulated by Eomes (also known as Tbr2), a T-box TF. Eomes inactivation caused rostrocaudal shifts in SVZ and CP gene expression, with loss of corticospinal axons and gain of corticotectal projections. These findings suggest that cortical areas and connections are shaped by sequential maps of regional identity, propagated by the Pax6 → Eomes → Tbr1 TF cascade. In humans, PAX6, EOMES, and TBR1 have been linked to intellectual disability and autism.


Subject(s)
Cerebral Cortex/anatomy & histology , Cerebral Cortex/metabolism , T-Box Domain Proteins/metabolism , Animals , Autistic Disorder/genetics , Autistic Disorder/metabolism , Autistic Disorder/pathology , Body Patterning , Brain Mapping , Cerebral Cortex/growth & development , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/cytology , Neurons/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pregnancy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics
20.
Nat Rev Immunol ; 2(12): 933-44, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12461566

ABSTRACT

After encountering antigen, helper T (T(H)) cells undergo differentiation to effector cells, which can secrete high levels of interferon-gamma, interleukin-4 (IL-4), IL-10 and other immunomodulators. How T(H) cells acquire, and remember, new patterns of gene expression is an area of intensive investigation. The process is remarkably plastic, with cytokines being key regulators. Extrinsic signals seem to be integrated into cell-intrinsic programming, in what is becoming an intriguing story of regulated development. We summarize the latest insights into mechanisms that govern the lineage choices that are made during T(H)-cell responses to foreign pathogens.


Subject(s)
Cytokines/physiology , Lymphopoiesis , T-Lymphocytes, Helper-Inducer/immunology , Adjuvants, Immunologic/physiology , Animals , DNA-Binding Proteins/genetics , GATA3 Transcription Factor , Humans , Immunity, Active/genetics , Immunologic Memory/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-10/physiology , Interleukin-4/immunology , Parasitic Diseases/immunology , Signal Transduction , Th1 Cells/immunology , Th2 Cells/immunology , Trans-Activators/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL