Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Annu Rev Immunol ; 36: 221-246, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29328786

ABSTRACT

Researchers are intensifying efforts to understand the mechanisms by which changes in metabolic states influence differentiation programs. An emerging objective is to define how fluctuations in metabolites influence the epigenetic states that contribute to differentiation programs. This is because metabolites such as S-adenosylmethionine, acetyl-CoA, α-ketoglutarate, 2-hydroxyglutarate, and butyrate are donors, substrates, cofactors, and antagonists for the activities of epigenetic-modifying complexes and for epigenetic modifications. We discuss this topic from the perspective of specialized CD4+ T cells as well as effector and memory T cell differentiation programs. We also highlight findings from embryonic stem cells that give mechanistic insight into how nutrients processed through pathways such as glycolysis, glutaminolysis, and one-carbon metabolism regulate metabolite levels to influence epigenetic events and discuss similar mechanistic principles in T cells. Finally, we highlight how dysregulated environments, such as the tumor microenvironment, might alter programming events.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/immunology , Energy Metabolism , Epigenesis, Genetic , Animals , Biomarkers , Gene Expression Regulation, Developmental , Humans , Neoplasms/etiology , Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Nat Immunol ; 24(1): 8-9, 2023 01.
Article in English | MEDLINE | ID: mdl-36596891
3.
Nat Immunol ; 18(11): 1249-1260, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28892471

ABSTRACT

Interleukin 2 (IL-2) promotes Foxp3+ regulatory T (Treg) cell responses, but inhibits T follicular helper (TFH) cell development. However, it is not clear how IL-2 affects T follicular regulatory (TFR) cells, a cell type with properties of both Treg and TFH cells. Using an influenza infection model, we found that high IL-2 concentrations at the peak of the infection prevented TFR cell development by a Blimp-1-dependent mechanism. However, once the immune response resolved, some Treg cells downregulated CD25, upregulated Bcl-6 and differentiated into TFR cells, which then migrated into the B cell follicles to prevent the expansion of self-reactive B cell clones. Thus, unlike its effects on conventional Treg cells, IL-2 inhibits TFR cell responses.


Subject(s)
Interleukin-2/pharmacology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Cell Movement/genetics , Cell Movement/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling/methods , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Interleukin-2/administration & dosage , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
4.
Immunity ; 50(3): 542-544, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893582

ABSTRACT

Committing to a differentiation pathway means leaving alternative pathways behind. Adoue et al. (2019) report that the H3K9-methyltrasferase Setdb1 plays a role in inhibiting the Th1 program in committed Th2 cells, and mechanistically, its role might relate to the selective targeting of endogenous retroviruses adjacent to Th1 enhancers.


Subject(s)
Endogenous Retroviruses , Cell Differentiation , Cell Lineage , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Th2 Cells
5.
Immunity ; 51(1): 155-168.e5, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31248780

ABSTRACT

Genetic variation influences how the genome is interpreted in individuals and in mouse strains used to model immune responses. We developed approaches to utilize next-generation sequencing datasets to identify sequence variation in genes and enhancer elements in congenic and backcross mouse models. We defined genetic variation in the widely used B6-CD45.2 and B6.SJL-CD45.1 congenic model, identifying substantial differences in SJL genetic content retained in B6.SJL-CD45.1 strains on the basis of the vendor source of the mice. Genes encoding PD-1, CD62L, Bcl-2, cathepsin E, and Cxcr4 were within SJL genetic content in at least one vendor source of B6.SJL-CD45.1 mice. SJL genetic content affected enhancer elements, gene regulation, protein expression, and amino acid content in CD4+ T helper 1 cells, and mice infected with influenza showed reduced expression of Cxcr4 on B6.SJL-CD45.1 T follicular helper cells. These findings provide information on experimental variables and aid in creating approaches that account for genetic variables.


Subject(s)
Cathepsin E/metabolism , Enhancer Elements, Genetic/genetics , Immunity/genetics , Receptors, CXCR4/metabolism , Th1 Cells/immunology , Animals , Cathepsin E/genetics , Commerce , Gene Expression Regulation , Genetic Background , Genetic Variation , Germinal Center/immunology , High-Throughput Nucleotide Sequencing , Inbreeding , Leukocyte Common Antigens/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Models, Animal , Receptors, CXCR4/genetics
6.
Immunity ; 50(5): 1172-1187.e7, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31076359

ABSTRACT

Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.


Subject(s)
B-Lymphocytes/immunology , Interferon-gamma/immunology , Receptors, Interferon/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibody-Producing Cells/immunology , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cells, Cultured , Chromatin/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Positive Regulatory Domain I-Binding Factor 1/biosynthesis , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Box Domain Proteins/genetics , Interferon gamma Receptor
7.
Nat Immunol ; 15(10): 957-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25194422

ABSTRACT

Despite the increasing knowledge of the molecular events that induce the glycolysis pathway in effector T cells, very little is known about the transcriptional mechanisms that dampen the glycolysis program in quiescent cell populations such as memory T cells. Here we found that the transcription factor Bcl-6 directly repressed genes encoding molecules involved in the glycolysis pathway, including Slc2a1, Slc2a3, Pkm and Hk2, in type 1 helper T cells (TH1 cells) exposed to low concentrations of interleukin 2 (IL-2). Thus, Bcl-6 had a role opposing the IL-2-sensitive glycolytic transcriptional program that the transcription factors c-Myc and HIF-1α promote in effector T cells. Additionally, the TH1 lineage-specifying factor T-bet functionally antagonized the Bcl-6-dependent repression of genes encoding molecules in the glycolysis pathway, which links the molecular balance of these two factors to regulation of the metabolic gene program.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , Glycolysis/genetics , Metabolic Networks and Pathways/genetics , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , Reverse Transcriptase Polymerase Chain Reaction
8.
Trends Immunol ; 44(7): 493-495, 2023 07.
Article in English | MEDLINE | ID: mdl-37270301

ABSTRACT

Genomic studies are transforming knowledge about the epigenetic, transcription factor, and 3D landscapes of the genome. However, comprehensive information is lacking about the effector domains used by transcription factors to influence gene expression. Addressing this gap, DelRosso et al. developed a high-throughput screen to discover effector domains in human regulatory factors.


Subject(s)
Gene Expression Regulation , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Genome , Genomics
9.
Immunity ; 47(2): 251-267.e7, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813658

ABSTRACT

Despite considerable research connecting cellular metabolism with differentiation decisions, the underlying mechanisms that translate metabolite-sensitive activities into unique gene programs are still unclear. We found that aspects of the interleukin-2 (IL-2)-sensitive effector gene program in CD4+ and CD8+ T cells in type 1 conditions (Th1) were regulated by glutamine and alpha-ketoglutarate (αKG)-induced events, in part through changes in DNA and histone methylation states. We further identified a mechanism by which IL-2- and αKG-sensitive metabolic changes regulated the association of CCCTC-binding factor (CTCF) with select genomic sites. αKG-sensitive CTCF sites were often associated with loci containing IL-2- and αKG-sensitive genome organization patterns and gene expression in T cells. IL-2- and αKG-sensitive CTCF sites in T cells were also associated with genes from developmental pathways that had αKG-sensitive expression in embryonic stem cells. The data collectively support a mechanism wherein CTCF serves to translate αKG-sensitive metabolic changes into context-dependent differentiation gene programs.


Subject(s)
Cell Differentiation , Interleukin-2/metabolism , Ketoglutaric Acids/metabolism , Repressor Proteins/metabolism , Th1 Cells/immunology , Animals , CCCTC-Binding Factor , Cell Differentiation/genetics , Cells, Cultured , Cellular Microenvironment , DNA Methylation , Female , Gene Expression Regulation , Glutamine/metabolism , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics
10.
Trends Immunol ; 43(11): 851-854, 2022 11.
Article in English | MEDLINE | ID: mdl-36182546

ABSTRACT

The criminalization of women's healthcare in many USA states has created uncertainty about women's access to evidence-based medical care and will affect the physical, mental, and emotional health and well-being of women. This article is intended to start a discussion on this complex topic in the immunology community.


Subject(s)
Delivery of Health Care , Female , Humans , United States , Women's Health Services , Health Equity , Gender Equity
11.
Nat Immunol ; 13(4): 405-11, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22406686

ABSTRACT

The transcription factors T-bet and Bcl-6 are required for the establishment of a T helper type 1 cell (T(H)1 cell) and follicular helper T cell (T(FH) cell) gene-expression profile, respectively. Here we found that high concentrations of interleukin 2 (IL-2) inhibited Bcl-6 expression in polarized T(H)1 cells. Mechanistically, the low concentrations of Bcl-6 normally found in effector T(H)1 cells did not repress its target genes because a T-bet-Bcl-6 complex masked the Bcl-6 DNA-binding domain. T(H)1 cells increased their Bcl-6/T-bet ratio in response to limiting IL-2 conditions, which allowed excess Bcl-6 to repress its direct target Prdm1 (which encodes the transcriptional repressor Blimp-1). The Bcl-6-dependent repression of Blimp-1 effectively induced a partial T(FH) profile because Blimp-1 directly repressed a subset of T(FH) signature genes, including Cxcr5. Thus, IL-2-signaling regulates the Bcl-6-Blimp-1 axis in T(H)1 cells to maintain flexibility with a T(FH) cell-like gene profile.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation/immunology , Proto-Oncogene Proteins c-bcl-6/genetics , T-Lymphocyte Subsets/cytology , Th1 Cells/cytology , Animals , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Profiling , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6/immunology , Proto-Oncogene Proteins c-bcl-6/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Transfection
12.
Trends Immunol ; 42(12): 1077-1087, 2021 12.
Article in English | MEDLINE | ID: mdl-34740529

ABSTRACT

Model organisms such as mice are important for basic research and serve as valuable tools in preclinical translational studies. A challenge with translating findings from mice to humans is identifying and separating evolutionarily conserved mechanisms in the immune system from those diverging between species. A significant emphasis has been placed on defining conserved gene regulation principles, with divergent mechanisms often overlooked. We put forward the perspective that both conserved and divergent mechanisms that regulate gene expression programs are of equal importance. With recent advances and availability of datasets, immunologists should take a closer look at the role for genetic diversity in altering gene expression programs between mouse and human immune cells.


Subject(s)
Gene Expression Regulation , Immune System , Animals , Humans , Mice
13.
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
14.
Trends Immunol ; 41(4): 269-271, 2020 04.
Article in English | MEDLINE | ID: mdl-32169284

ABSTRACT

A new study by Fasolino et al. defines how genetic variation in a mouse model of type 1 diabetes mellitus (T1DM) affects long-distance genomic interactions. The research has widespread implications for understanding how genetic diversity impacts disease susceptibility, and raises important concepts about mechanisms that can be influenced by genetic diversity between individuals.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , Chromatin , Diabetes Mellitus, Type 1/genetics , Genetic Variation , Humans , Mice , T-Lymphocytes
15.
J Autoimmun ; 118: 102593, 2021 03.
Article in English | MEDLINE | ID: mdl-33508637

ABSTRACT

BACKGROUND: IgA nephropathy is thought to be an autoimmune disease wherein galactose-deficient IgA1 (Gd-IgA1) is recognized by IgG autoantibodies, resulting in formation and renal accumulation of nephritogenic immune complexes. Although this hypothesis is supported by recent findings that, in renal immunodeposits of IgA nephropathy patients, IgG is enriched for Gd-IgA1-specific autoantibodies, experimental proof is still lacking. METHODS: IgG isolated from sera of IgA nephropathy patients or produced as a recombinant IgG (rIgG) was mixed with human Gd-IgA1 to form immune complexes. IgG from healthy individuals served as a control. Nude and SCID mice were injected with human IgG and Gd-IgA1, in immune complexes or individually, and their presence in kidneys was ascertained by immunofluorescence. Pathologic changes in the glomeruli were evaluated by quantitative morphometry and exploratory transcriptomic profiling was performed by RNA-Seq. RESULTS: Immunodeficient mice injected with Gd-IgA1 mixed with IgG autoantibodies from patients with IgA nephropathy, but not Gd-IgA1 mixed with IgG from healthy individuals, displayed IgA, IgG, and mouse complement C3 glomerular deposits and mesangioproliferative glomerular injury with hematuria and proteinuria. Un-complexed Gd-IgA1 or IgG did not induce pathological changes. Moreover, Gd-IgA1-rIgG immune complexes injected into immunodeficient mice induced histopathological changes characteristic of human disease. Exploratory transcriptome profiling of mouse kidney tissues indicated that these immune complexes altered gene expression of multiple pathways, in concordance with the changes observed in kidney biopsies of patients with IgA nephropathy. CONCLUSIONS: This study provides the first in vivo evidence for a pathogenic role of IgG autoantibodies specific for Gd-IgA1 in the pathogenesis of IgA nephropathy.


Subject(s)
Autoantibodies/immunology , Glomerulonephritis, IGA/immunology , Immunoglobulin G/immunology , Animals , Antigen-Antibody Complex/administration & dosage , Antigen-Antibody Complex/immunology , Autoantibodies/blood , Disease Models, Animal , Glomerulonephritis, IGA/blood , Glomerulonephritis, IGA/pathology , Humans , Immunoglobulin A/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Mice
16.
Immunol Rev ; 300(1): 5-8, 2021 03.
Article in English | MEDLINE | ID: mdl-33638253
17.
J Immunol ; 198(11): 4244-4254, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28468969

ABSTRACT

CK2 is a highly conserved and pleiotropic serine/threonine kinase that promotes many prosurvival and proinflammatory signaling pathways, including PI3K/Akt/mTOR and JAK/STAT. These pathways are essential for CD4+ T cell activation and polarization, but little is known about how CK2 functions in T cells. In this article, we demonstrate that CK2 expression and kinase activity are induced upon CD4+ T cell activation. Targeting the catalytic activity of CK2 using the next-generation small molecule inhibitor CX-4945 in vitro significantly and specifically inhibited mouse and human Th17 cell differentiation while promoting the generation of Foxp3+ regulatory T cells (Tregs). These findings were associated with suppression of PI3K/Akt/mTOR activation and STAT3 phosphorylation upon CX-4945 treatment. Furthermore, we demonstrate that CX-4945 treatment inhibits the maturation of Th17 cells into inflammatory IFN-γ-coproducing effector cells. The Th17/Treg axis and maturation of Th17 cells are major contributing factors to the pathogenesis of many autoimmune disorders, including multiple sclerosis. Using a murine model of multiple sclerosis, experimental autoimmune encephalomyelitis, we demonstrate that in vivo administration of CX-4945 targets Akt/mTOR signaling in CD4+ T cells and the Th17/Treg axis throughout disease. Importantly, CX-4945 treatment after disease initiation significantly reduced disease severity, which was associated with a significant decrease in the frequency of pathogenic IFN-γ+ and GM-CSF+ Th17 cells in the CNS. Our data implicate CK2 as a regulator of the Th17/Treg axis and Th17 cell maturation and suggest that CK2 could be targeted for the treatment of Th17 cell-driven autoimmune disorders.


Subject(s)
Casein Kinase II/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Differentiation , Class I Phosphatidylinositol 3-Kinases , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Naphthyridines/pharmacology , Phenazines , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/physiology , Th1 Cells/immunology , Th17 Cells/physiology
20.
Immunity ; 31(4): 551-64, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818655

ABSTRACT

How cell type-specific differences in chromatin conformation are achieved and their contribution to gene expression are incompletely understood. Here we identify a cryptic upstream orchestrator of interferon-gamma (IFNG) transcription, which is embedded within the human IL26 gene, compromised of a single CCCTC-binding factor (CTCF) binding site and retained in all mammals, even surviving near-complete evolutionary deletion of the equivalent gene encoding IL-26 in rodents. CTCF and cohesins occupy this element in vivo in a cell type-nonspecific manner. This element is juxtaposed to two other sites located within the first intron and downstream of Ifng, where CTCF, cohesins, and the transcription factor T-bet bind in a T helper 1 (Th1) cell-specific manner. These interactions, close proximity of other elements within the locus to each other and to the gene encoding interferon-gamma, and robust murine Ifng expression are dependent on CTCF and T-bet. The results demonstrate that cooperation between architectural (CTCF) and transcriptional enhancing (T-bet) factors and the elements to which they bind is required for proper Th1 cell-specific expression of Ifng.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/metabolism , Repressor Proteins/metabolism , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Animals , CCCTC-Binding Factor , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin/immunology , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/immunology , Chromosomal Proteins, Non-Histone/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Introns/genetics , Introns/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Repressor Proteins/genetics , Repressor Proteins/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/metabolism , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL