Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nature ; 630(8015): 189-197, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811728

ABSTRACT

In developing B cells, V(D)J recombination assembles exons encoding IgH and Igκ variable regions from hundreds of gene segments clustered across Igh and Igk loci. V, D and J gene segments are flanked by conserved recombination signal sequences (RSSs) that target RAG endonuclease1. RAG orchestrates Igh V(D)J recombination upon capturing a JH-RSS within the JH-RSS-based recombination centre1-3 (RC). JH-RSS orientation programmes RAG to scan upstream D- and VH-containing chromatin that is presented in a linear manner by cohesin-mediated loop extrusion4-7. During Igh scanning, RAG robustly utilizes only D-RSSs or VH-RSSs in convergent (deletional) orientation with JH-RSSs4-7. However, for Vκ-to-Jκ joining, RAG utilizes Vκ-RSSs from deletional- and inversional-oriented clusters8, inconsistent with linear scanning2. Here we characterize the Vκ-to-Jκ joining mechanism. Igk undergoes robust primary and secondary rearrangements9,10, which confounds scanning assays. We therefore engineered cells to undergo only primary Vκ-to-Jκ rearrangements and found that RAG scanning from the primary Jκ-RC terminates just 8 kb upstream within the CTCF-site-based Sis element11. Whereas Sis and the Jκ-RC barely interacted with the Vκ locus, the CTCF-site-based Cer element12 4 kb upstream of Sis interacted with various loop extrusion impediments across the locus. Similar to VH locus inversion7, DJH inversion abrogated VH-to-DJH joining; yet Vκ locus or Jκ inversion allowed robust Vκ-to-Jκ joining. Together, these experiments implicated loop extrusion in bringing Vκ segments near Cer for short-range diffusion-mediated capture by RC-based RAG. To identify key mechanistic elements for diffusional V(D)J recombination in Igk versus Igh, we assayed Vκ-to-JH and D-to-Jκ rearrangements in hybrid Igh-Igk loci generated by targeted chromosomal translocations, and pinpointed remarkably strong Vκ and Jκ RSSs. Indeed, RSS replacements in hybrid or normal Igk and Igh loci confirmed the ability of Igk-RSSs to promote robust diffusional joining compared with Igh-RSSs. We propose that Igk evolved strong RSSs to mediate diffusional Vκ-to-Jκ joining, whereas Igh evolved weaker RSSs requisite for modulating VH joining by RAG-scanning impediments.


Subject(s)
Immunoglobulin Heavy Chains , Immunoglobulin Joining Region , Immunoglobulin Variable Region , Immunoglobulin kappa-Chains , V(D)J Recombination , Animals , Female , Male , Mice , Alleles , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromatin/chemistry , Cohesins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Joining Region/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulin Variable Region/genetics , V(D)J Recombination/genetics
2.
Trends Genet ; 40(6): 471-479, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643034

ABSTRACT

Enhancers are the key regulators of other DNA-based processes by virtue of their unique ability to generate nucleosome-depleted regions in a highly regulated manner. Enhancers regulate cell-type-specific transcription of tRNA genes by RNA polymerase III (Pol III). They are also responsible for the binding of the origin replication complex (ORC) to DNA replication origins, thereby regulating origin utilization, replication timing, and replication-dependent chromosome breaks. Additionally, enhancers regulate V(D)J recombination by increasing access of the recombination-activating gene (RAG) recombinase to target sites and by generating non-coding enhancer RNAs and localized regions of trimethylated histone H3-K4 recognized by the RAG2 PHD domain. Thus, enhancers represent the first step in decoding the genome, and hence they regulate biological processes that, unlike RNA polymerase II (Pol II) transcription, do not have dedicated regulatory proteins.


Subject(s)
DNA Replication , Enhancer Elements, Genetic , RNA Polymerase III , Transcription, Genetic , V(D)J Recombination , Animals , Humans , DNA Replication/genetics , Gene Expression Regulation/genetics , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Transcription, Genetic/genetics , V(D)J Recombination/genetics
3.
Brief Bioinform ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38483254

ABSTRACT

The extraordinary diversity of T cells and B cells is critical for body maintenance. This diversity has an important role in protecting against tumor formation. In humans, the T-cell receptor (TCR) repertoire is generated through a striking stochastic process called V(D)J recombination, in which different gene segments are assembled and modified, leading to extensive variety. In ovarian cancer (OC), an unfortunate 80% of cases are detected late, leading to poor survival outcomes. However, when detected early, approximately 94% of patients live longer than 5 years after diagnosis. Thus, early detection is critical for patient survival. To determine whether the TCR repertoire obtained from peripheral blood is associated with tumor status, we collected blood samples from 85 women with or without OC and obtained TCR information. We then used machine learning to learn the characteristics of samples and to finally predict, over a set of unseen samples, whether the person is with or without OC. We successfully stratified the two groups, thereby associating the peripheral blood TCR repertoire with the formation of OC tumors. A careful study of the origin of the set of T cells most informative for the signature indicated the involvement of a specific invariant natural killer T (iNKT) clone and a specific mucosal-associated invariant T (MAIT) clone. Our findings here support the proposition that tumor-relevant signal is maintained by the immune system and is coded in the T-cell repertoire available in peripheral blood. It is also possible that the immune system detects tumors early enough for repertoire technologies to inform us near the beginning of tumor formation. Although such detection is made by the immune system, we might be able to identify it, using repertoire data from peripheral blood, to offer a pragmatic way to search for early signs of cancer with minimal patient burden, possibly with enhanced sensitivity.


Subject(s)
Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/genetics , B-Lymphocytes , Machine Learning , V(D)J Recombination , Receptors, Antigen, T-Cell/genetics
4.
J Immunol ; 213(5): 651-662, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39007649

ABSTRACT

The expressed Ab repertoire is a critical determinant of immune-related phenotypes. Ab-encoding transcripts are distinct from other expressed genes because they are transcribed from somatically rearranged gene segments. Human Abs are composed of two identical H and L chain polypeptides derived from genes in IGH locus and one of two L chain loci. The combinatorial diversity that results from Ab gene rearrangement and the pairing of different H and L chains contributes to the immense diversity of the baseline Ab repertoire. During rearrangement, Ab gene selection is mediated by factors that influence chromatin architecture, promoter/enhancer activity, and V(D)J recombination. Interindividual variation in the composition of the Ab repertoire associates with germline variation in IGH, implicating polymorphism in Ab gene regulation. Determining how IGH variants directly mediate gene regulation will require integration of these variants with other functional genomic datasets. In this study, we argue that standard approaches using short reads have limited utility for characterizing regulatory regions in IGH at haplotype resolution. Using simulated and chromatin immunoprecipitation sequencing reads, we define features of IGH that limit use of short reads and a single reference genome, namely 1) the highly duplicated nature of the DNA sequence in IGH and 2) structural polymorphisms that are frequent in the population. We demonstrate that personalized diploid references enhance performance of short-read data for characterizing mappable portions of the locus, while also showing that long-read profiling tools will ultimately be needed to fully resolve functional impacts of IGH germline variation on expressed Ab repertoires.


Subject(s)
Gene Expression Regulation , Humans , Immunoglobulin Heavy Chains/genetics , Haplotypes , V(D)J Recombination/genetics , Genes, Immunoglobulin
5.
Nucleic Acids Res ; 52(9): 5048-5066, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38412274

ABSTRACT

Two DNA repair pathways, non-homologous end joining (NHEJ) and alternative end joining (A-EJ), are involved in V(D)J recombination and chromosome translocation. Previous studies reported distinct repair mechanisms for chromosome translocation, with NHEJ involved in humans and A-EJ in mice predominantly. NHEJ depends on DNA-PKcs, a critical partner in synapsis formation and downstream component activation. While DNA-PKcs inhibition promotes chromosome translocations harboring microhomologies in mice, its synonymous effect in humans is not known. We find partial DNA-PKcs inhibition in human cells leads to increased translocations and the continued involvement of a dampened NHEJ. In contrast, complete DNA-PKcs inhibition substantially increased microhomology-mediated end joining (MMEJ), thus bridging the two different translocation mechanisms between human and mice. Similar to a previous study on Ku70 deletion, DNA-PKcs deletion in G1/G0-phase mouse progenitor B cell lines, significantly impairs V(D)J recombination and generated higher rates of translocations as a consequence of dysregulated coding and signal end joining. Genetic DNA-PKcs inhibition suppresses NHEJ entirely, with repair phenotypically resembling Ku70-deficient A-EJ. In contrast, we find DNA-PKcs necessary in generating the near-exclusive MMEJ associated with Lig4 deficiency. Our study underscores DNA-PKcs in suppressing illegitimate chromosome rearrangement while also contributing to MMEJ in both species.


Subject(s)
Chromosome Aberrations , DNA End-Joining Repair , DNA-Activated Protein Kinase , Animals , Humans , Mice , Cell Line , DNA Ligase ATP/genetics , DNA Ligase ATP/metabolism , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Translocation, Genetic , V(D)J Recombination
6.
Front Immunol ; 15: 1345467, 2024.
Article in English | MEDLINE | ID: mdl-38504980

ABSTRACT

The vast diversity of mammalian adaptive antigen receptors allows for robust and efficient immune responses against a wide number of pathogens. The antigen receptor repertoire is built during the recombination of B and T cell receptor (BCR, TCR) loci and hypermutation of BCR loci. V(D)J recombination rearranges these antigen receptor loci, which are organized as an array of separate V, (D), and J gene segments. Transcription activation at the recombining locus leads to changes in the local three-dimensional architecture, which subsequently contributes to which gene segments are utilized for recombination. The endogenous retrovirus (ERV) mouse mammary tumor provirus 8 (Mtv8) resides on mouse chromosome 6 interposed within the large array of light chain kappa V gene segments. As ERVs contribute to changes in genomic architecture by driving high levels of transcription of neighboring genes, it was suggested that Mtv8 could influence the BCR repertoire. We generated Mtv8-deficient mice to determine if the ERV influences V(D)J recombination to test this possibility. We find that Mtv8 does not influence the BCR repertoire.


Subject(s)
Receptors, Antigen, T-Cell , V(D)J Recombination , Animals , Mice , Immunoglobulins/genetics , Mammals , Receptors, Antigen, T-Cell/genetics , V(D)J Recombination/genetics
7.
Appl Immunohistochem Mol Morphol ; 32(7): 305-308, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38695552

ABSTRACT

INTRODUCTION: Multiplex polymerase chain reaction (PCR) and next-generation sequencing (NGS) can both be used to identify a neoplastic clonotype by targeting CDR3 and assessing rearrangements in IgH, IgK, IgL, TCR-ß, and TCR-gamma loci. The clonotypic sequence can be robustly used to track minimal residual disease (MRD). The ability to track MRD by NGS in mixed phenotype acute leukemia (MPAL) is unknown and warrants investigation. METHODS: Institutional Review Board (IRB) approval was obtained. Central Moffitt Cancer Center (MCC) database was searched to locate any patients with MPAL from over 600,000 entries. Patient charts were manually curated to identify those with clonoSEQ data, and clinical data was procured from the electronic medical record (EMR). RESULTS: Twenty-nine patients with MPAL were identified. Only 2 patients with clonoSEQ testing were found. Both demonstrated a B/myeloid phenotype, and both were bilineal. NGS (clonoSEQ) identified 4 dominant (IGH) (patient A; 8/2019) and 2 dominant sequences (patient B; 10/2019), respectively. In both patients, clonoSEQ testing successfully tracked minimal residual disease and mirrored clinical disease burden. CONCLUSIONS: This report is the first to confirm the utility of NGS-based MRD tracking in patients with MPAL and shows increased sensitivity of NGS over MRD flow cytometry.


Subject(s)
High-Throughput Nucleotide Sequencing , Neoplasm, Residual , Humans , Male , Female , Middle Aged , Adult , Aged , Phenotype , V(D)J Recombination
8.
Sci Immunol ; 9(95): eade5705, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38787962

ABSTRACT

Inborn errors of T cell development present a pediatric emergency in which timely curative therapy is informed by molecular diagnosis. In 11 affected patients across four consanguineous kindreds, we detected homozygosity for a single deleterious missense variant in the gene NudC domain-containing 3 (NUDCD3). Two infants had severe combined immunodeficiency with the complete absence of T and B cells (T -B- SCID), whereas nine showed classical features of Omenn syndrome (OS). Restricted antigen receptor gene usage by residual T lymphocytes suggested impaired V(D)J recombination. Patient cells showed reduced expression of NUDCD3 protein and diminished ability to support RAG-mediated recombination in vitro, which was associated with pathologic sequestration of RAG1 in the nucleoli. Although impaired V(D)J recombination in a mouse model bearing the homologous variant led to milder immunologic abnormalities, NUDCD3 is absolutely required for healthy T and B cell development in humans.


Subject(s)
Severe Combined Immunodeficiency , V(D)J Recombination , Humans , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Animals , Mice , V(D)J Recombination/immunology , V(D)J Recombination/genetics , Male , Female , Infant , B-Lymphocytes/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , T-Lymphocytes/immunology , Child, Preschool , Mutation, Missense
9.
Sci Adv ; 10(31): eadn4682, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39083600

ABSTRACT

G0-G1 phase alternative end joining (A-EJ) is a recently defined mutagenic pathway characterized by resected deletion and translocation joints that are predominantly direct and are distinguished from A-EJ in cycling cells that rely much more on microhomology-mediated end joining (MMEJ). Using chemical and genetic approaches, we systematically evaluate potential A-EJ factors and DNA damage response (DDR) genes to support this mechanism by mapping the repair fates of RAG1/2-initiated double-strand breaks in the context of Igκ locus V-J recombination and chromosome translocation. Our findings highlight a polymerase theta-independent Parp1-XRCC1/LigIII axis as central A-EJ components, supported by 53BP1 in the context of an Ataxia-telangiectasia mutated (ATM)-activated DDR. Mechanistically, we demonstrate varied changes in short-range resection, MMEJ, and translocation, imposed by compromising specific DDR activities, which include polymerase alpha, Ataxia-telangiectasia and Rad3-related (ATR), DNA2, and Mre11. This study advances our understanding of DNA damage repair within the 53BP1 regulatory domain and the RAG1/2 postcleavage complex.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA End-Joining Repair , Tumor Suppressor p53-Binding Protein 1 , V(D)J Recombination , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Animals , Humans , DNA Breaks, Double-Stranded , Mice , DNA Damage , Translocation, Genetic
10.
Elife ; 122024 Jul 26.
Article in English | MEDLINE | ID: mdl-39056282

ABSTRACT

The evolutionary conservation of non-core RAG regions suggests significant roles that might involve quantitative or qualitative alterations in RAG activity. Off-target V(D)J recombination contributes to lymphomagenesis and is exacerbated by RAG2' C-terminus absence in Tp53-/- mice thymic lymphomas. However, the genomic stability effects of non-core regions from both Rag1c/c and Rag2c/c in BCR-ABL1+ B-lymphoblastic leukemia (BCR-ABL1+ B-ALL), the characteristics, and mechanisms of non-core regions in suppressing off-target V(D)J recombination remain unclear. Here, we established three mouse models of BCR-ABL1+ B-ALL in mice expressing full-length RAG (Ragf/f), core RAG1 (Rag1c/c), and core RAG2 (Rag2c/c). The Ragc/c (Rag1c/c and Rag2c/c) leukemia cells exhibited greater malignant tumor characteristics compared to Ragf/f cells. Additionally, Ragc/c cells showed higher frequency of off-target V(D)J recombination and oncogenic mutations than Ragf/f. We also revealed decreased RAG cleavage accuracy in Ragc/c cells and a smaller recombinant size in Rag1c/c cells, which could potentially exacerbate off-target V(D)J recombination in Ragc/c cells. In conclusion, these findings indicate that the non-core RAG regions, particularly the non-core region of RAG1, play a significant role in preserving V(D)J recombination precision and genomic stability in BCR-ABL1+ B-ALL.


Subject(s)
DNA-Binding Proteins , Fusion Proteins, bcr-abl , Homeodomain Proteins , V(D)J Recombination , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , V(D)J Recombination/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Carcinogenesis/genetics , Humans , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
11.
Leuk Res ; 143: 107521, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38879405

ABSTRACT

Early T-cell Precursor Acute Lymphoblastic Leukemia (ETP-ALL), T-Lymphoid/Myeloid Mixed Phenotype Acute Leukemia (T/M-MPAL), and Acute Myeloid Leukemia with minimal differentiation (AML-M0) are immature acute leukemias (AL) that present overlapping T-cell lymphoid and myeloid features at different degrees, with impact to disease classification. An interesting strategy to assess lymphoid lineage commitment and maturation is the analysis of V(D)J gene segment recombination, which can be applied to investigate leukemic cells in immature AL. Herein, we revisited 19 ETP-ALL, 8 T/M-MPAL, and 12 AML-M0 pediatric patients to characterize V(D)J rearrangement (V(D)J-r) profiles associated with other somatic alterations. V(D)J-r were identified in 74 %, 25 %, and 25 % of ETP-ALL, T/M-MPAL, and AML-M0, respectively. Forty-six percent of ETP-ALL harbored ≥ 3 V(D)J-r, while there was no more than one V(D)J-r per patient in AML-M0 and T/M-MPAL. TCRD was the most rearranged locus in ETPALL, but it was not rearranged in other AL. In ETP-ALL, N/KRAS mutations were associated with absence of V(D)J-r, while NF1 deletion was most frequent in patients with ≥ 3 V(D)J-r. Relapse and death occurred mainly in patients harboring one or no rearranged locus. Molecular characterization of V(D)J-r in our cohort indicates a distinct profile of ETP-ALL, compared to T/M-MPAL and AML-M0. Our findings also suggest that the clinical outcome of ETP-ALL patients may be affected by blast cell maturity, inferred from the number of rearranged TCR loci.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Child , Child, Preschool , Male , Adolescent , Female , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , V(D)J Recombination/genetics , Infant , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Mutation , Gene Rearrangement
12.
Immunol Res ; 72(3): 438-449, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38240953

ABSTRACT

Recombination-activating gene 1 (RAG1) is a vital player in V(D)J recombination, a fundamental process in primary B cell and T cell receptor diversification of the adaptive immune system. Current vertebrate RAG evolved from RAG transposon; however, it has been modified to play a crucial role in the adaptive system instead of being irreversibly silenced by CpG methylation. By interrogating a range of publicly available datasets, the current study investigated whether RAG1 has retained a disproportionate level of its original CpG dinucleotides compared to other genes, thereby rendering it more exposed to methylation-mediated mutation. Here, we show that 57.57% of RAG1 pathogenic mutations and 51.6% of RAG1 disease-causing mutations were associated with CpG methylation, a percentage that was significantly higher than that of its RAG2 cofactor alongside the whole genome. The CpG scores and densities for all RAG ancestors suggested that RAG transposon was CpG denser. The percentage of the ancestral CpG of RAG1 and RAG2 were 6% and 4.2%, respectively, with no preference towards CG containing codons. Furthermore, CpG loci of RAG1 in sperms were significantly higher methylated than that of RAG2. In conclusion, RAG1 has been exposed to CpG mediated methylation mutagenesis more than RAG2 and the whole genome, presumably due to its late entry to the genome later with an initially higher CpG content.


Subject(s)
CpG Islands , DNA Methylation , Evolution, Molecular , Homeodomain Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , CpG Islands/genetics , Humans , Animals , Mutagenesis , Transposases/genetics , Transposases/metabolism , Mutation , V(D)J Recombination/genetics , DNA Transposable Elements/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
13.
Leuk Lymphoma ; 65(8): 1100-1109, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38608254

ABSTRACT

We describe two types of IGH::BCL2 breakpoints involving the 5' region of BCL2 (5' BCL2). One was ins(14;18)(q32;q21q21) observed in 2 follicular lymphoma (FL) cases, in which IGH was cleaved at 3' of IGHD and 5' of IGHJ and BCL2 was cleaved at 5' BCL2 and downstream regions, and a 281- or 201-kilobase pair fragment containing the BCL2 protein-coding sequences was invertedly inserted into IGH. In another type observed in 2 FL and 2 chronic lymphocytic leukemia (CLL) cases, breakage and reunion occurred within the switch region associated with IGHM (Sµ) and 5' BCL2, creating IGH Sµ::5' BCL2 fusion sequences on der(18)t(14;18)(q32;q21). The former is considered to be mediated by VDJ-recombination, while the latter by the class switch recombination process. There were no particular features in FL or CLL cases with IGH::5' BCL2 breakpoints compared with those with t(14;18)(q32;q21)/IGH::BCL2 involving the 3' breakpoint cluster regions.


Subject(s)
Chromosome Breakpoints , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 18 , Immunoglobulin Heavy Chains , Proto-Oncogene Proteins c-bcl-2 , Translocation, Genetic , Humans , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 18/genetics , Immunoglobulin Heavy Chains/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Immunoglobulin Class Switching/genetics , V(D)J Recombination/genetics , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Middle Aged , Male , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Female , In Situ Hybridization, Fluorescence , Aged
14.
Front Immunol ; 14: 1330153, 2023.
Article in English | MEDLINE | ID: mdl-38406579

ABSTRACT

Introduction: Analysis of an individual's immunoglobulin (IG) gene repertoire requires the use of high-quality germline gene reference sets. When sets only contain alleles supported by strong evidence, AIRR sequencing (AIRR-seq) data analysis is more accurate and studies of the evolution of IG genes, their allelic variants and the expressed immune repertoire is therefore facilitated. Methods: The Adaptive Immune Receptor Repertoire Community (AIRR-C) IG Reference Sets have been developed by including only human IG heavy and light chain alleles that have been confirmed by evidence from multiple high-quality sources. To further improve AIRR-seq analysis, some alleles have been extended to deal with short 3' or 5' truncations that can lead them to be overlooked by alignment utilities. To avoid other challenges for analysis programs, exact paralogs (e.g. IGHV1-69*01 and IGHV1-69D*01) are only represented once in each set, though alternative sequence names are noted in accompanying metadata. Results and discussion: The Reference Sets include less than half the previously recognised IG alleles (e.g. just 198 IGHV sequences), and also include a number of novel alleles: 8 IGHV alleles, 2 IGKV alleles and 5 IGLV alleles. Despite their smaller sizes, erroneous calls were eliminated, and excellent coverage was achieved when a set of repertoires comprising over 4 million V(D)J rearrangements from 99 individuals were analyzed using the Sets. The version-tracked AIRR-C IG Reference Sets are freely available at the OGRDB website (https://ogrdb.airr-community.org/germline_sets/Human) and will be regularly updated to include newly observed and previously reported sequences that can be confirmed by new high-quality data.


Subject(s)
Genes, Immunoglobulin , Immunoglobulins , Humans , Immunoglobulins/genetics , Alleles , V(D)J Recombination/genetics , Germ Cells
15.
Inmunología (1987) ; 32(2): 57-69, abr.-jun. 2013. ilus
Article in Spanish | IBECS (Spain) | ID: ibc-112121

ABSTRACT

La recombinación V(D)J consiste en el ensamblaje de los segmentos génicos presentes en los genes de las cadenas variables de los receptores de antígeno para generar la diversidad del reconocimiento antigénico en linfocitos. El conocimiento de su regulación en condiciones normales es esencial para entender los casos en que este proceso se desregula, dando lugar a transformaciones leucémicas. La recombinación V(D)J se inicia por acción de una endonucleasa específica presente exclusivamente en linfocitos inmaduros. Según el «modelo de accesibilidad» propuesto hace más de 25 años, la recombinación V(D)J está regulada a través del control de la accesibilidad de esta endonucleasa a sus sitios de corte en el ADN, de acuerdo con unos programas de diferenciación celular muy definidos. En esta revisión se resumen los hallazgos descubiertos en este campo en los últimos años, tales como el importante papel que tiene la conformación génica y la posición de estos genes en el núcleo celular, así como aquellos que muy recientemente han permitido la validación definitiva del «modelo de accesibilidad» (AU)


V(D)J recombination is the assembly of gene segments at the antigen receptor loci in order to generate antigen receptor diversity in T and B lymphocytes. Detailed knowledge of how V(D)J recombination is normally regulated during lymphocyte development is essential to understand the cases of dysregulation of this process that result in leukemic transformation. V(D)J recombination is triggered by action of a specific endonuclease which is exclusively expressed in immature lymphocytes. According to the “accessibility model” proposed more than 25 years ago, DNA cleavage by this endonuclease is very strictly controlled during cell differentiation by regulating its accessibility to chromatin. This review summarizes the advances in the field over the last few years, including the important role of the genomic conformation and position of the antigen receptor loci within the nucleus, as well as those that have recently culminated with the validation of the “accessibility model” to control this process (AU)


Subject(s)
Humans , V(D)J Recombination/immunology , Receptors, Lymphocyte Homing/immunology , Leukemia/immunology , Receptors, Antigen, T-Cell/immunology , Immunoglobulins/immunology , Chromatin/immunology , Transcription Factors/immunology , Cell Differentiation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL