Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Sci Immunol ; 9(93): eadh5318, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489350

ABSTRACT

Recombination activating gene (RAG) expression increases as thymocytes transition from the CD4-CD8- double-negative (DN) to the CD4+CD8+ double-positive (DP) stage, but the physiological importance and mechanism of transcriptional up-regulation are unknown. Here, we show that a DP-specific component of the recombination activating genes antisilencer (DPASE) provokes elevated RAG expression in DP thymocytes. Mouse DP thymocytes lacking the DPASE display RAG expression equivalent to that in DN thymocytes, but this supports only a partial Tcra repertoire due to inefficient secondary Vα-Jα rearrangement. These data indicate that RAG up-regulation is required for a replete Tcra repertoire and that RAG expression is fine-tuned during lymphocyte development to meet the requirements of distinct antigen receptor loci. We further show that transcription factor RORγt directs RAG up-regulation in DP thymocytes by binding to the DPASE and that RORγt influences the Tcra repertoire by binding to the Tcra enhancer. These data, together with prior work showing RORγt to control Tcra rearrangement by regulating DP thymocyte proliferation and survival, reveal RORγt to orchestrate multiple pathways that support formation of the Tcra repertoire.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3 , Thymocytes , Animals , Mice , Gene Expression , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, Antigen, T-Cell, alpha-beta , Transcription Factors/genetics , Genes, RAG-1/genetics
2.
Front Immunol ; 14: 1188738, 2023.
Article in English | MEDLINE | ID: mdl-37483636

ABSTRACT

V(D)J recombination of antigen receptor loci is a highly developmentally regulated process. During T lymphocyte development, recombination of the Tcra gene occurs in CD4+CD8+ double positive (DP) thymocytes and requires the Tcra enhancer (Eα). E proteins are known regulators of DP thymocyte development and have three identified binding sites in Eα. To understand the contribution of E proteins to Eα function, mutants lacking one or two of the respective binding sites were generated. The double-binding site mutant displayed a partial block at the positive selection stage of αß T cell development. Further investigation revealed loss of germline transcription within the Tcra locus at the Jα array, along with dysregulated primary and impaired secondary Vα-Jα rearrangement. Eα E protein binding increases Tcra locus accessibility and regulates TCRα recombination, thus directly promoting Tcra repertoire diversity.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta , Thymocytes , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics , V(D)J Recombination/genetics , Transcription Factors/genetics , Enhancer Elements, Genetic
3.
Methods Mol Biol ; 2580: 261-282, 2023.
Article in English | MEDLINE | ID: mdl-36374463

ABSTRACT

Quantitative real-time PCR and next-generation sequencing (NGS) are invaluable techniques to analyze T cell receptor (Tcr) gene rearrangements in mouse lymphocyte populations. Although these approaches are powerful, they also have limitations that must be accounted for in experimental design and data interpretation. Here, we provide relevant background required for understanding these limitations and then outline established quantitative real-time PCR and NGS methods that can be used for analysis of mouse Tcra and Tcrb gene rearrangements in mice.


Subject(s)
Gene Rearrangement , Receptors, Antigen, T-Cell, alpha-beta , Mice , Animals , Receptors, Antigen, T-Cell, alpha-beta/genetics , Polymerase Chain Reaction
4.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-34910107

ABSTRACT

The Tcra repertoire is generated by multiple rounds of Vα-Jα rearrangement. However, Tcrd recombination precedes Tcra recombination within the complex Tcra-Tcrd locus. Here, by ablating Tcrd recombination, we report that Tcrd rearrangement broadens primary Vα use to diversify the Tcra repertoire in mice. We reveal that use of Trav15-dv6 family V gene segments in Tcrd recombination imparts diversity in the Tcra repertoire by instigating use of central and distal Vα segments. Moreover, disruption of the regions containing these genes and their cis-regulatory elements identifies the Trav15-dv6 family as being responsible for driving central and distal Vα recombinations beyond their roles as substrates for Tcrd recombination. Our study demonstrates an indispensable role for Tcrd recombination in general, and the Trav15-dv6 family in particular, in the generation of a combinatorially diverse Tcra repertoire.


Subject(s)
Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Animals , Female , High-Throughput Nucleotide Sequencing , Mice , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymocytes/metabolism , V(D)J Recombination
5.
Nat Commun ; 12(1): 99, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397934

ABSTRACT

CD4 and CD8 mark helper and cytotoxic T cell lineages, respectively, and serve as coreceptors for MHC-restricted TCR recognition. How coreceptor expression is matched with TCR specificity is central to understanding CD4/CD8 lineage choice, but visualising coreceptor gene activity in individual selection intermediates has been technically challenging. It therefore remains unclear whether the sequence of coreceptor gene expression in selection intermediates follows a stereotypic pattern, or is responsive to signaling. Here we use single cell RNA sequencing (scRNA-seq) to classify mouse thymocyte selection intermediates by coreceptor gene expression. In the unperturbed thymus, Cd4+Cd8a- selection intermediates appear before Cd4-Cd8a+ selection intermediates, but the timing of these subsets is flexible according to the strength of TCR signals. Our data show that selection intermediates discriminate MHC class prior to the loss of coreceptor expression and suggest a model where signal strength informs the timing of coreceptor gene activity and ultimately CD4/CD8 lineage choice.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histocompatibility Antigens/metabolism , Lymphocyte Activation/genetics , Mice, Inbred C57BL , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transcription Factors/metabolism
6.
Nucleic Acids Res ; 48(17): 9621-9636, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32853367

ABSTRACT

The regulation of T cell receptor Tcra gene rearrangement has been extensively studied. The enhancer Eα plays an essential role in Tcra rearrangement by establishing a recombination centre in the Jα array and a chromatin hub for interactions between Vα and Jα genes. But the mechanism of the Eα and its downstream CTCF binding site (here named EACBE) in dynamic chromatin regulation is unknown. The Hi-C data showed that the EACBE is located at the sub-TAD boundary which separates the Tcra-Tcrd locus and the downstream region including the Dad1 gene. The EACBE is required for long-distance regulation of the Eα on the proximal Vα genes, and its deletion impaired the Tcra rearrangement. We also noticed that the EACBE and Eα regulate the genes in the downstream sub-TAD via asymmetric chromatin extrusion. This study provides a new insight into the role of CTCF binding sites at TAD boundaries in gene regulation.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Enhancer Elements, Genetic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Binding Sites , CCCTC-Binding Factor/genetics , Chromatin/genetics , Gene Expression Regulation , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Homeodomain Proteins/genetics , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Mutant Strains , Thymus Gland/cytology
7.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32793983

ABSTRACT

In this issue of JEM, Wu et al. (https://doi.org/10.1084/jem.20200412) provide new insights into allelic exclusion. They demonstrate that Vß-to-DßJß rearrangement occurs stochastically on two competing Tcrb alleles, with suboptimal Vß recombination signal sequences limiting synchronous rearrangements and essential for allelic exclusion.


Subject(s)
Protein Sorting Signals , Receptors, Antigen, T-Cell, alpha-beta , Alleles , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombination, Genetic
8.
J Exp Med ; 216(1): 231-243, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30545902

ABSTRACT

Expression of Rag1 and Rag2 is tightly regulated in developing T cells to mediate TCR gene assembly. Here we have investigated the molecular mechanisms governing the assembly and disassembly of a transcriptionally active RAG locus chromatin hub in CD4+CD8+ thymocytes. Rag1 and Rag2 gene expression in CD4+CD8+ thymocytes depends on Rag1 and Rag2 promoter activation by a distant antisilencer element (ASE). We identify GATA3 and E2A as critical regulators of the ASE, and Runx1 and E2A as critical regulators of the Rag1 promoter. We reveal hierarchical assembly of a transcriptionally active chromatin hub containing the ASE and RAG promoters, with Rag2 recruitment and expression dependent on assembly of a functional ASE-Rag1 framework. Finally, we show that signal-dependent down-regulation of RAG gene expression in CD4+CD8+ thymocytes depends on Ikaros and occurs with disassembly of the RAG locus chromatin hub. Our results provide important new insights into the molecular mechanisms that orchestrate RAG gene expression in developing T cells.


Subject(s)
DNA-Binding Proteins/biosynthesis , Gene Expression Regulation/physiology , Genetic Loci/physiology , Homeodomain Proteins/biosynthesis , Thymocytes/metabolism , Transcription, Genetic/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , DNA-Binding Proteins/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Homeodomain Proteins/genetics , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Mice , Response Elements/physiology , Thymocytes/cytology
9.
Cell Rep ; 25(7): 1729-1740.e6, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428344

ABSTRACT

Tcrb locus V(D)J recombination is regulated by positioning at the nuclear periphery. Here, we used DamID to profile Tcrb locus interactions with the nuclear lamina at high resolution. We identified a lamina-associated domain (LAD) border composed of several CTCF-binding elements that segregates active non-LAD from inactive LAD regions of the locus. Deletion of the LAD border causes an enhancer-dependent spread of histone H3 lysine 27 acetylation from the active recombination center into recombination center-proximal LAD chromatin. This is associated with a disruption to nuclear lamina association, increased chromatin looping to the recombination center, and increased transcription and recombination of recombination center-proximal gene segments. Our results show that a LAD and LAD border are critical components of Tcrb locus gene regulation and suggest that LAD borders may generally function to constrain the activity of nearby enhancers.


Subject(s)
Genetic Loci , Nuclear Lamina/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombination, Genetic/genetics , Transcription, Genetic , Animals , Cell Line , Chromatin/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Mice, Inbred C57BL , Models, Biological , Transcriptional Activation/genetics , V(D)J Recombination/genetics
10.
Front Immunol ; 9: 425, 2018.
Article in English | MEDLINE | ID: mdl-29593713

ABSTRACT

CCCTC-binding factor (CTCF) is largely responsible for the 3D architecture of the genome, in concert with the action of cohesin, through the creation of long-range chromatin loops. Cohesin is hypothesized to be the main driver of these long-range chromatin interactions by the process of loop extrusion. Here, we performed ChIP-seq for CTCF and cohesin in two stages each of T and B cell differentiation and examined the binding pattern in all six antigen receptor (AgR) loci in these lymphocyte progenitors and in mature T and B cells, ES cells, and fibroblasts. The four large AgR loci have many bound CTCF sites, most of which are only occupied in lymphocytes, while only the CTCF sites at the end of each locus near the enhancers or J genes tend to be bound in non-lymphoid cells also. However, despite the generalized lymphocyte restriction of CTCF binding in AgR loci, the Igκ locus is the only locus that also shows significant lineage-specificity (T vs. B cells) and developmental stage-specificity (pre-B vs. pro-B) in CTCF binding. We show that cohesin binding shows greater lineage- and stage-specificity than CTCF at most AgR loci, providing more specificity to the loops. We also show that the culture of pro-B cells in IL7, a common practice to expand the number of cells before ChIP-seq, results in a CTCF-binding pattern resembling pre-B cells, as well as other epigenetic and transcriptional characteristics of pre-B cells. Analysis of the orientation of the CTCF sites show that all sites within the large V portions of the Igh and TCRß loci have the same orientation. This suggests either a lack of requirement for convergent CTCF sites creating loops, or indicates an absence of any loops between CTCF sites within the V region portion of those loci but only loops to the convergent sites at the D-J-enhancer end of each locus. The V region portions of the Igκ and TCRα/δ loci, by contrast, have CTCF sites in both orientations, providing many options for creating CTCF-mediated convergent loops throughout the loci. CTCF/cohesin loops, along with transcription factors, drives contraction of AgR loci to facilitate the creation of a diverse repertoire of antibodies and T cell receptors.


Subject(s)
B-Lymphocytes/physiology , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Immunoglobulins/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/physiology , Animals , CCCTC-Binding Factor/genetics , Cell Differentiation , Cell Lineage , Cells, Cultured , Chromatin/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Genes, RAG-1 , Genetic Loci/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Protein Binding , Cohesins
11.
Immunohorizons ; 2(11): 377-383, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30957077

ABSTRACT

The architectural protein CTCF regulates the conformation and recombination of antigen receptor loci. To study the importance of CTCF in Tcrb locus repertoire formation, we created a conditional knockout mouse line that deletes Ctcf early during thymocyte development. We observed an incomplete block in thymocyte development at the double-negative to double-positive transition, resulting in greatly lowered thymic cellularity. The Tcrb repertoire was altered with a decrease in recombination of Vß gene segments in close proximity to a CTCF binding element (CBE), resulting in an overall repertoire that was skewed in favor of Vß gene segments with no nearby CBE. Therefore we show that CTCF functions to diversify the Tcrb repertoire.

12.
Cell Rep ; 19(10): 2157-2173, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28591585

ABSTRACT

Adaptive immunity depends on diverse T cell receptor repertoires generated by variable, diversity, and joining (V[D]J) recombination. Here, we define the principles by which combinatorial diversity is generated in the murine Tcra repertoire. Tcra and Tcrd gene segments share the Tcra-Tcrd locus, with interspersed Vα and Vδ segments undergoing Vδ-Dδ-Jδ rearrangement in CD4-CD8- thymocytes and then multiple rounds of Vα-Jα rearrangement in CD4+CD8+ thymocytes. We document stepwise, highly coordinated proximal-to-distal progressions of Vα and Jα use on individual Tcra alleles, limiting combinatorial diversity. This behavior is supported by an extended chromatin conformation in CD4+CD8+ thymocytes, with only nearby Vα and Jα segments contacting each other. Tcrd rearrangements can use distal Vδ segments due to a contracted Tcra-Tcrd conformation in CD4-CD8- thymocytes. These rearrangements expand the Tcra repertoire by truncating the Vα array to permit otherwise disfavored Vα-Jα combinations. Therefore, recombination events at two developmental stages with distinct chromatin conformations synergize to promote Tcra repertoire diversity.


Subject(s)
Models, Genetic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Thymocytes , V(D)J Recombination , Animals , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology
14.
J Immunol ; 197(8): 3188-3197, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27613698

ABSTRACT

Chromatin looping mediated by the CCCTC binding factor (CTCF) regulates V(D)J recombination at Ag receptor loci. CTCF-mediated looping can influence recombination signal sequence (RSS) accessibility by regulating enhancer activation of germline promoters. CTCF-mediated looping has also been shown to limit directional tracking of the RAG recombinase along chromatin, and to regulate long-distance interactions between RSSs, independent of the RAG recombinase. However, in all prior instances in which CTCF-mediated looping was shown to influence V(D)J recombination, it was not possible to fully resolve the relative contributions to the V(D)J recombination phenotype of changes in accessibility, RAG tracking, and RAG-independent long-distance interactions. In this study, to assess mechanisms by which CTCF-mediated looping can impact V(D)J recombination, we introduced an ectopic CTCF binding element (CBE) immediately downstream of Eδ in the murine Tcra-Tcrd locus. The ectopic CBE impaired inversional rearrangement of Trdv5 in the absence of measurable effects on Trdv5 transcription and chromatin accessibility. The ectopic CBE also limited directional RAG tracking from the Tcrd recombination center, demonstrating that a single CBE can impact the distribution of RAG proteins along chromatin. However, such tracking cannot account for Trdv5-to-Trdd2 inversional rearrangement. Rather, the defect in Trdv5 rearrangement could only be attributed to a reconfigured chromatin loop organization that limited RAG-independent contacts between the Trdv5 and Trdd2 RSSs. We conclude that CTCF can regulate V(D)J recombination by segregating RSSs into distinct loop domains and inhibiting RSS synapsis, independent of any effects on transcription, RSS accessibility, and RAG tracking.


Subject(s)
Gene Rearrangement, delta-Chain T-Cell Antigen Receptor/genetics , Repressor Proteins/immunology , V(D)J Recombination/genetics , Animals , Binding Sites , CCCTC-Binding Factor , Female , Gene Rearrangement, delta-Chain T-Cell Antigen Receptor/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , V(D)J Recombination/immunology
15.
J Exp Med ; 213(9): 1921-36, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27526713

ABSTRACT

T cell antigen receptor δ (Tcrd) variable region exons are assembled by RAG-initiated V(D)J recombination events in developing γδ thymocytes. Here, we use linear amplification-mediated high-throughput genome-wide translocation sequencing (LAM-HTGTS) to map hundreds of thousands of RAG-initiated Tcrd D segment (Trdd1 and Trdd2) rearrangements in CD4(-)CD8(-) double-negative thymocyte progenitors differentiated in vitro from bone marrow-derived hematopoietic stem cells. We find that Trdd2 joins directly to Trdv, Trdd1, and Trdj segments, whereas Trdd1 joining is ordered with joining to Trdd2, a prerequisite for further rearrangement. We also find frequent, previously unappreciated, Trdd1 and Trdd2 rearrangements that inactivate Tcrd, including sequential rearrangements from V(D)J recombination signal sequence fusions. Moreover, we find dozens of RAG off-target sequences that are generated via RAG tracking both upstream and downstream from the Trdd2 recombination center across the Tcrd loop domain that is bounded by the upstream INT1-2 and downstream TEA elements. Disruption of the upstream INT1-2 boundary of this loop domain allows spreading of RAG on- and off-target activity to the proximal Trdv domain and, correspondingly, shifts the Tcrd V(D)J recombination landscape by leading to predominant V(D)J joining to a proximal Trdv3 pseudogene that lies just upstream of the normal boundary.


Subject(s)
Homeodomain Proteins/physiology , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/physiology , V(D)J Recombination , Amino Acid Transport Systems, Basic/genetics , Animals , Cells, Cultured , DNA Breaks, Double-Stranded , Mice
17.
Trends Genet ; 32(5): 253-255, 2016 05.
Article in English | MEDLINE | ID: mdl-27012539

ABSTRACT

Immunoglobulin heavy-chain locus V(D)J recombination requires a 3D chromatin organization which permits widely distributed variable (V) gene segments to contact distant diversity (D) and joining (J) gene segments. A recent study has identified key nodes in the locus interactome, paving the way for new molecular insights into how the locus is configured for recombination.


Subject(s)
Immunoglobulins/genetics , Recombination, Genetic , Transcription, Genetic/immunology , VDJ Recombinases/immunology , Animals , Chromatin/immunology , Enhancer Elements, Genetic , Humans , Mice , Transcription, Genetic/genetics , VDJ Recombinases/genetics
18.
J Immunol ; 196(6): 2572-82, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26843327

ABSTRACT

Yin Yang 1 (YY1) is a zinc finger protein that functions as a transcriptional activator or repressor and participates in multiple biological processes, including development and tumorigenesis. To investigate the role of YY1 in developing T cells, we used mouse models that depleted YY1 at two distinct stages of thymocyte development. When YY1 was depleted in CD4(-)CD8(-) double-negative thymocytes, development to the CD4(+)CD8(+) double-positive stage was impaired, due to increased apoptosis that prevented expansion of post-ß-selection thymocytes. When YY1 was depleted in double-positive thymocytes, they underwent increased cell-autonomous apoptosis in vitro and displayed a shorter lifespan in vivo, as judged by their ability to undergo secondary Vα-to-Jα recombination. Mechanistically, we found that the increased apoptosis in YY1-deficient thymocytes was attributed to overexpression of p53, because concurrent loss of p53 completely rescued the developmental defects of YY1-deficient thymocytes. These results indicated that YY1 functions as a critical regulator of thymocyte survival and that it does so by suppressing the expression of p53.


Subject(s)
Gene Expression Regulation/immunology , Lymphopoiesis/immunology , Thymocytes/immunology , Tumor Suppressor Protein p53/biosynthesis , YY1 Transcription Factor/immunology , Animals , Blotting, Western , Cell Separation , Cell Survival/immunology , Disease Models, Animal , Flow Cytometry , Mice , Mice, Knockout , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Thymocytes/cytology , Transcription, Genetic , Tumor Suppressor Protein p53/immunology
19.
Nat Commun ; 7: 10553, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26822034

ABSTRACT

Variable, diversity and joining (V(D)J) recombination and immunoglobulin class switch recombination (CSR) are key processes in adaptive immune responses that naturally generate DNA double-strand breaks (DSBs) and trigger a DNA repair response. It is unclear whether this response is associated with distinct survival signals that protect T and B cells. Glycogen synthase kinase 3ß (GSK3ß) is a constitutively active kinase known to promote cell death. Here we show that phosphorylation of GSK3ß on Ser(389) by p38 MAPK (mitogen-activated protein kinase) is induced selectively by DSBs through ATM (ataxia telangiectasia mutated) as a unique mechanism to attenuate the activity of nuclear GSK3ß and promote survival of cells undergoing DSBs. Inability to inactivate GSK3ß through Ser(389) phosphorylation in Ser(389)Ala knockin mice causes a decrease in the fitness of cells undergoing V(D)J recombination and CSR. Preselection-Tcrß repertoire is impaired and antigen-specific IgG antibody responses following immunization are blunted in Ser(389)GSK3ß knockin mice. Thus, GSK3ß emerges as an important modulator of the adaptive immune response.


Subject(s)
DNA Breaks, Double-Stranded , Gene Expression Regulation, Enzymologic/physiology , Glycogen Synthase Kinase 3/metabolism , Lymphocytes/physiology , Animals , Cells, Cultured , DNA Repair , Gene Knock-In Techniques , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mutation , Phosphorylation
20.
Methods Mol Biol ; 1323: 179-202, 2016.
Article in English | MEDLINE | ID: mdl-26294409

ABSTRACT

PCR on genomic DNA isolated from lymphocyte populations is an invaluable technique to analyze T cell receptor (TCR) α and ß gene rearrangements. Although this approach is powerful, it also has limitations that must be accounted for in experimental design and data interpretation. Here, we provide background required for understanding these limitations, and then outline standard PCR methods that can be used for analysis of TCRα and ß gene rearrangements in mice.


Subject(s)
Gene Rearrangement, T-Lymphocyte , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/metabolism , Animals , Blotting, Southern , Cell Differentiation/genetics , Genetic Loci , Mice , Polymerase Chain Reaction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL