Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.628
Filter
2.
Am J Surg Pathol ; 46(1): 58-70, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34265801

ABSTRACT

Follicular lymphoma and classic Hodgkin lymphoma can be associated in composite and/or sequential lymphomas. Common IGH and BCL2 rearrangements have already been identified between both contingents of these entities, but mutation profiles have not yet been investigated. The main objective of this study was to analyze the transdifferentiation process that may occur between Hodgkin and follicular contingents in sequential and composite lymphomas to better characterize these entities. From 2004 to 2020, a retrospective multicentric study was performed, including 9 composite and 13 sequential lymphomas. Clinical data were retrospectively collected. Fluorescent in situ hybridization of BCL2 and BCL6 rearrangements, polymerase chain reaction of IGH and IGK rearrangements, next-generation sequencing of IGK rearrangement, and targeted next-generation sequencing (TNGS) on a panel of genes frequently mutated in lymphomas were performed on each contingent of composite and sequential lymphomas. For TNGS, each contingent was isolated by laser capture microdissection. Clinical presentation and evolution were more aggressive in sequential than composite lymphomas. By fluorescent in situ hybridization, common rearrangements of BCL6 and BCL2 were identified between both contingents. Similarly, a common clonal relationship was established by evaluating IGH and IGK rearrangement by polymerase chain reaction or next-generation sequencing. By TNGS, the same pathogenic variants were identified in both contingents in the following genes: CREBBP, KMT2D, BCL2, EP300, SF3B1, SOCS1, ARID1A, and BCOR. Specific pathogenic variants for each contingent were also identified: XPO1 for Hodgkin lymphoma contingent and FOXO1, TNFRSF14 for follicular lymphoma contingent. This study reinforces the hypothesis of a transdifferentiation process between Hodgkin and follicular contingent of sequential/composite lymphomas.


Subject(s)
B-Lymphocytes/immunology , Biomarkers, Tumor/genetics , Cell Plasticity , Hodgkin Disease/immunology , Lymphoma, Follicular/immunology , Aged , Aged, 80 and over , B-Lymphocytes/pathology , DNA Mutational Analysis , Female , France , Gene Rearrangement, B-Lymphocyte, Heavy Chain , High-Throughput Nucleotide Sequencing , Hodgkin Disease/drug therapy , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Humans , Immunoglobulin Heavy Chains , Immunoglobulins/genetics , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Male , Middle Aged , Mutation , Phenotype , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Retrospective Studies
5.
PLoS One ; 15(11): e0241634, 2020.
Article in English | MEDLINE | ID: mdl-33180881

ABSTRACT

AIM: Lymphoplasmacytic lymphoma (LPL) is an indolent mature B-cell-neoplasm with involvement of the bone marrow. At least 90% of LPLs carry MYD88-L265P mutation and some of them (~10%) transform into diffuse large B-cell-lymphoma (DLBCL). MATERIAL AND METHODS: Over the past 15 years we have collected 7 cases where the both LPL and DLBCL were diagnosed in the same patient. Clinical records, analytical data and histopathological specimens were reviewed. FISH studies on paraffin-embedded tissue for MYC, BCL2 and BCL6 genes were performed, as well as MYD88-L265P mutation and IGH rearrangement analysis by PCR. A mutational study was done by massive next generation sequencing (NGS). RESULTS: There were 4 women and 3 men between 36-91 years of age. Diagnoses were made simultaneously in 4 patients. In two cases the LPL appeared before the DLBCL and in the remaining case the high-grade component was discovered 5 years before the LPL. In 6 cases both samples shared the MYD88-L265P mutation. IGH rearrangement analysis showed overlapping features in two of 6 cases tested. Mutational study was evaluable in three cases for both samples showing shared and divergent mutations. CONCLUSIONS: These data suggest different mechanisms of DLBCL development in LPL patients.


Subject(s)
Genetic Heterogeneity , Lymphoma, Large B-Cell, Diffuse/genetics , Adult , Aged , Aged, 80 and over , Clonal Evolution , Disease Progression , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-myc/genetics
6.
J Hematol Oncol ; 13(1): 127, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32972438

ABSTRACT

The clinical impact of minimal residual disease detection at early time points or during follow-ups has been shown to accurately predict relapses among patients with lymphomas, mainly in follicular and diffuse large B cell lymphoma. The field of minimal residual disease testing in mantle cell lymphoma is still evolving but has great impact in determining the prognosis. Flow cytometry and polymerase chain reaction-based testing are most commonly used methods in practice; however, these methods are not sensitive enough to detect the dynamic changes that underline lymphoma progression. Newer methods using next-generation sequencing, such as ClonoSeq, are being incorporated in clinical trials. Other techniques under evolution include CAPP-seq and anchored multiplex polymerase chain reaction-based methods. This review article aims to provide a comprehensive update on the status of minimal residual disease detection and its prognostic effect in mantle cell patients. The role of circulating tumor DNA-based minimal residual disease detection in lymphomas is also discussed.


Subject(s)
Lymphoma, Mantle-Cell/pathology , Biomarkers, Tumor , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 14/ultrastructure , Cyclin D1/genetics , DNA, Neoplasm/blood , Flow Cytometry/methods , Forecasting , Gene Rearrangement, B-Lymphocyte, Heavy Chain , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Heavy Chains/genetics , Liquid Biopsy , Lymphoma, Mantle-Cell/diagnosis , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/therapy , Multiplex Polymerase Chain Reaction , Neoplasm, Residual , Prognosis , Translocation, Genetic
8.
Cancer Genet ; 242: 8-14, 2020 04.
Article in English | MEDLINE | ID: mdl-32058318

ABSTRACT

Acute lymphoblastic leukaemia (ALL) is the most common childhood malignancy with the majority of patients being classified as B-cell lineage (B-ALL). The sub-classification of B-ALL is based on genomic architecture. Recent studies have demonstrated the capability of SNP-microarrays to detect genomic changes in B-ALL which cannot be observed by conventional cytogenetic methods. In current clinical trials, B-ALL patients at high risk of relapse are mainly identified by adverse cancer genomics and/or poor response to early therapy. To test the hypothesis that inclusion of SNP-microarrays in frontline diagnostics could more efficiently and accurately identify adverse genomic factors than conventional techniques, we evaluated the Australian high-risk B-ALL cohort enrolled on AIEOP-BFM ALL 2009 study (n = 33). SNP-microarray analysis identified additional aberrations in 97% of patients (32/33) compared to conventional techniques. This changed the genomic risk category of 24% (8/33) of patients. Additionally, 27% (9/33) of patients exhibited a 'hyperdiploid' genome, which is generally associated with a good genomic risk and favourable outcomes. An enrichment of IKZF1 deletions was observed with one third of the cohort affected. Our findings suggest the current classification system could be improved and highlights the need to use more sensitive techniques such as SNP-microarray for cytogenomic risk stratification in B-ALL.


Subject(s)
Ikaros Transcription Factor/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Australia , Child , Child, Preschool , Chromosome Aberrations , Chromosome Banding , Core Binding Factor Alpha 2 Subunit/genetics , Female , Fusion Proteins, bcr-abl/genetics , Gene Deletion , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Loss of Heterozygosity , Male , Neoplasm, Residual , Oncogene Proteins, Fusion/genetics , Polyploidy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/classification , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prospective Studies , Risk Assessment , Sequence Deletion
9.
Br J Haematol ; 189(5): 853-859, 2020 06.
Article in English | MEDLINE | ID: mdl-32064595

ABSTRACT

In chronic lymphocytic leukaemia (CLL), caution is warranted regarding the clinical implications of immunoglobulin variable heavy chain region (IGHV) rearrangements with a 'borderline' (BL) percentage of mutations (i.e. 97-97·9% IGHV identity). We analysed the IGHV mutational status in 759 untreated CLL patients (cohort 1). BL-CLL (n = 36, 5%) showed a time to first treatment (TFT) similar to that of M-CLL (n = 338) and significantly longer than that of UM-CLL (n = 385), despite the enrichment in subset #2 cases. In fact, CLLs belonging to subset #2 (n = 15/759, 2%) were significantly more frequent among BL-CLLs (n = 5/36, 14%), with a brief TFT. TFT of BL-CLL remained comparable to that of M-CLL also considering the 327 CLL patients evaluated at diagnosis. These findings were then validated in an independent cohort 2 of 759 newly diagnosed CLL patients (BL-CLL: n = 11, 1·4%) and in all newly diagnosed patients from cohorts 1 and 2 (n = 1 086, 84% stage A; BL-CLL: n = 47, 4·3%). BL-CLL at diagnosis showed a biological profile comparable to that of M-CLL with a low frequency of unfavourable prognostic markers, except for a significant enrichment in subset #2. Our data suggest that the prognosis of BL-CLL is good and similar to that of M-CLL, with the exception of subset #2 cases.


Subject(s)
Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Somatic Hypermutation, Immunoglobulin , ADP-ribosyl Cyclase 1/analysis , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/analysis , Female , Humans , Immunoglobulin Variable Region/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Membrane Glycoproteins/analysis , Middle Aged , Mutation , Prognosis , Retrospective Studies , Time-to-Treatment
10.
Am J Hematol ; 94(12): 1364-1373, 2019 12.
Article in English | MEDLINE | ID: mdl-31571261

ABSTRACT

Minimal residual disease (MRD) tracking, by next generation sequencing of immunoglobulin sequences, is moving towards clinical implementation in multiple myeloma. However, there is only sparse information available to address whether clonal sequences remain stable for tracking over time, and to what extent light chain sequences are sufficiently unique for tracking. Here, we analyzed immunoglobulin repertoires from 905 plasma cell myeloma and healthy control samples, focusing on the third complementarity determining region (CDR3). Clonal heavy and/or light chain expression was identified in all patients at baseline, with one or more subclones related to the main clone in 3.2%. In 45 patients with 101 sequential samples, the dominant clonal CDR3 sequences remained identical over time, despite differential clonal evolution by whole exome sequencing in 49% of patients. The low frequency of subclonal CDR3 variants, and absence of evolution over time in active multiple myeloma, indicates that tumor cells at this stage are not under selective pressure to undergo antibody affinity maturation. Next, we establish somatic hypermutation and non-templated insertions as the most important determinants of light chain clonal uniqueness, identifying a potentially trackable sequence in the majority of patients. Taken together, we show that dominant clonal sequences identified at baseline are reliable biomarkers for long-term tracking of the malignant clone, including both IGH and the majority of light chain clones.


Subject(s)
Complementarity Determining Regions/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Gene Rearrangement, B-Lymphocyte, Light Chain , High-Throughput Nucleotide Sequencing , Multiple Myeloma/pathology , Biomarkers, Tumor , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Clinical Trials as Topic/statistics & numerical data , Clonal Evolution , Clone Cells/pathology , Genes, Immunoglobulin , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Multiple Myeloma/genetics , Neoplasm, Residual/diagnosis , Neoplasm, Residual/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Somatic Hypermutation, Immunoglobulin , VDJ Exons
11.
J Immunol ; 203(7): 1687-1692, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31484734

ABSTRACT

B cell clonal expansion is vital for adaptive immunity. High-throughput BCR sequencing enables investigating this process but requires computational inference to identify clonal relationships. This inference usually relies on only the BCR H chain, as most current protocols do not preserve H:L chain pairing. The extent to which paired L chains aids inference is unknown. Using human single-cell paired BCR datasets, we assessed the ability of H chain-based clonal clustering to identify clones. Of the expanded clones identified, <20% grouped cells expressing inconsistent L chains. H chains from these misclustered clones contained more distant junction sequences and shared fewer V segment mutations than the accurate clones. This suggests that additional H chain information could be leveraged to refine clonal relationships. Conversely, L chains were insufficient to refine H chain-based clonal clusters. Overall, the BCR H chain alone is sufficient to identify clonal relationships with confidence.


Subject(s)
B-Lymphocytes/immunology , Databases, Genetic , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Immunoglobulin Heavy Chains/immunology , Receptors, Antigen, B-Cell/immunology , B-Lymphocytes/cytology , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Receptors, Antigen, B-Cell/genetics
12.
Hematol Oncol ; 37(4): 368-374, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31325190

ABSTRACT

In 2009, the four laboratories of the Fondazione Italiana Linfomi (FIL) minimal residual disease (MRD) Network started a collaborative effort to harmonize and standardize their methodologies at the national level, performing quality control (QC) rounds for follicular lymphoma (FL) and mantle cell lymphoma (MCL) MRD assessment. In 16 QC rounds between 2010 and 2017, the four laboratories received 208 bone marrow (BM) samples (126 FL; 82 MCL); 187 were analyzed, according to the EuroMRD Consortium guidelines, by both nested (NEST) polymerase chain reaction (PCR) and real-time quantitative (RQ) PCR for BCL2/IGH MBR or IGHV rearrangements. Here, we aimed at analyzing the samples that challenged the interlaboratory reproducibility and data interpretation. Overall, 156/187 BM samples (83%) were concordantly classified as NEST+/RQ+ or NEST-/RQ- by all the four laboratories. The remaining 31 samples (17%) resulted alternatively positive and negative in the interlaboratory evaluations, independently of the method and the type of rearrangement, and were defined "borderline" (brd) samples: 12 proved NEST brd/RQ brd, 7 NEST-/RQ brd, 10 NEST brd/RQ positive not quantifiable (PNQ), and 2 NEST brd/RQ-. Results did not change even increasing the number of replicates/sample. In 6/31 brd samples, droplet digital PCR (ddPCR) was tested and showed no interlaboratory discordance. Despite the high interlaboratory reproducibility in the MRD analysis obtained and maintained by the QC round strategy, samples with the lowest MRD levels can still represent a challenge: 17% (31/187) of our samples showed discordant results in interlaboratory assessments, with 6.4% (12/187) remained brd even applying the two methods. Thus, although representing a minority, brd samples are still problematic, especially when a clinically oriented interpretation of MRD results is required. Alternative, novel methods such as ddPCR and next-generation sequencing have the potential to overcome the current limitations.


Subject(s)
Bone Marrow Examination , Bone Marrow/pathology , Laboratory Proficiency Testing , Lymphoma, Non-Hodgkin/pathology , Polymerase Chain Reaction , Bone Marrow Examination/standards , Clone Cells , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin , Genes, bcl-2 , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Italy/epidemiology , Lymphoma, Non-Hodgkin/genetics , Neoplasm, Residual , Oncogene Proteins, Fusion/analysis , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards , Proto-Oncogene Proteins c-bcl-2/genetics , Quality Assurance, Health Care , Reproducibility of Results , Translocation, Genetic
13.
Blood Cancer J ; 9(7): 52, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31209206

ABSTRACT

Follicular lymphoma (FL) is a heterogeneous disease whose pathogenesis remains partially unknown. Around 20% of FL patients experience early progression or treatment-refractory disease and 2-3% of patients per year experience histological transformation (HT) into a more aggressive lymphoma (tFL). Here, we evaluate the immunoglobulin heavy chain variable (IGHV) gene usage and mutational status in 187 FL cases to assess its impact on clinical outcome and histological transformation. The IGHV gene repertoire was remarkably biased in FL. The IGHV4-34 (14%), IGHV3-23 (14%), IGHV3-48 (10%), IGHV3-30 (9%) and IGHV3-21 (7%) genes accounted for more than half of the whole cohort. IGHV3-48 was overrepresented in cases of tFL (19%) compared with non-transformed FL at 5 years (5%, P = 0.05). Patients with the IGHV3-48 gene were significantly more likely to have had HT after 10 years than those who used other genes (71% vs. 25%, P < 0.05), irrespective of the therapy they received. Moreover, IGHV3-30 was also overrepresented in cases of FL (9%) and tFL (13%) compared with diffuse large B-cell lymphoma in which it was nearly absent. In conclusion, our results indicate a role for antigen selection in the development of FL, while the use of IGHV3-48 could help predict histological transformation.


Subject(s)
Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Variable Region , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Female , Humans , Lymphoma, Follicular/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Middle Aged , Prognosis , Young Adult
14.
Methods Mol Biol ; 1956: 61-75, 2019.
Article in English | MEDLINE | ID: mdl-30779030

ABSTRACT

Normal and malignant B cells carry rearranged immunoglobulin (Ig) variable region genes, which due to their practically limitless diversity represent ideal clonal markers for these cells. We describe here an approach to isolate single cells from frozen tissue sections by microdissection using a laser-based method. From the isolated cells, rearranged IgH and Igκ genes are amplified in a semi-nested PCR approach, using a collection of V gene subgroup-specific primers recognizing nearly all V genes together with primers for the J genes. By sequence analysis of V region genes from distinct cells, the clonal relationship of the B lineage cells can unequivocally be determined and related to the histological distribution of the cells. The approach is also useful to determine V, D, and J gene usage. Moreover, the presence and pattern of somatic Ig V gene mutations give valuable insight into the stage of differentiation of the B cells.


Subject(s)
Gene Rearrangement, B-Lymphocyte, Heavy Chain , Hodgkin Disease/genetics , Immunoglobulin Variable Region/genetics , Laser Capture Microdissection/methods , Lymphoma, B-Cell/genetics , Polymerase Chain Reaction/methods , Single-Cell Analysis/methods , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Genes, Immunoglobulin , Hodgkin Disease/pathology , Humans , Lymphoma, B-Cell/pathology
15.
Virchows Arch ; 474(3): 353-363, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30680453

ABSTRACT

Immunoglobulin light chain-derived (AL) amyloidosis may occur as a systemic disease usually with dismal prognosis and a localized variant with favorable outcome. We report 29 patients with AL amyloidosis and associated lymphoplasmacytic infiltrate spatially related to amyloid deposits. In 17 cases, the amyloid deposits were classified as ALλ and 12 as ALκ Histopathology in all cases showed relatively sparse plasma cells and B cells without tumor or sheet formation by the lymphoplasmacytic infiltrate. The B cells predominantly showed an immunophenotype of the marginal zone. In situ, hybridization revealed 17 cases with λ- and 10 with κ light chain restricted plasma cells, which was concordant with the AL subtype in each case. Clonal immunoglobulin heavy variable gene (IGHV) or κ light chain rearrangement was found in 23/29 interpretable cases. A single case harbored a MYD88L265P-mutation. Taken together, we detected 27 (93%) cases of AL amyloidosis with an associated light chain restricted and predominantly molecularly clonal plasma cell population. Clinical data were available in 18 patients. Five patients suffered from systemic lymphoma and two from systemic AL amyloidosis. The remaining cases were classified as localized with regard to both, the AL amyloidosis and the light chain restricted plasma cell population. To the best of our knowledge, we herein present the largest cohort of AL amyloidosis associated with a light chain restricted and predominantly molecularly clonal plasma cell population, which we designate as a distinct disease entity: "AL amyloidosis with a localized B cell neoplasia of undetermined significance".


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Light-chain Amyloidosis/immunology , Lymphoma, B-Cell/immunology , Plasma Cells/immunology , Waldenstrom Macroglobulinemia/immunology , Adult , Aged , Aged, 80 and over , B-Lymphocytes/pathology , Biomarkers, Tumor/genetics , DNA Mutational Analysis , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Gene Rearrangement, B-Lymphocyte, Light Chain , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light-chain Amyloidosis/genetics , Immunoglobulin Light-chain Amyloidosis/pathology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Immunoglobulin lambda-Chains/genetics , Immunoglobulin lambda-Chains/immunology , Immunohistochemistry , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Middle Aged , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Phenotype , Plasma Cells/pathology , Prospective Studies , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/pathology
16.
J Immunol ; 201(6): 1765-1774, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30097530

ABSTRACT

The formation of nontemplated (N) regions during Ig gene rearrangement is a major contributor to Ab diversity. To gain insights into the mechanisms behind this, we studied the nucleotide composition of N regions within 29,962 unique human VHDJH rearrangements and 8728 unique human DJH rearrangements containing exactly one identifiable D gene segment and thus two N regions, N1 and N2. We found a distinct decreasing content of cytosine (C) and increasing content of guanine (G) across each N region, suggesting that N regions are typically generated by concatenation of two 3' overhangs synthesized by addition of nucleoside triphosphates with a preference for dCTP. This challenges the general assumption that the terminal deoxynucleotidyl transferase favors dGTP in vivo. Furthermore, we found that the G and C gradients depended strongly on whether the germline gene segments were trimmed or not. Our data show that C-enriched N addition preferentially happens at trimmed 3' ends of VH, D, and JH gene segments, indicating a dependency of the transferase mechanism upon the nuclease mechanism.


Subject(s)
DNA Nucleotidylexotransferase/immunology , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Immunoglobulin Heavy Chains , Immunoglobulin Variable Region , Adolescent , Adult , Child , Child, Preschool , Cytosine/immunology , DNA Nucleotidylexotransferase/genetics , Female , Guanosine/genetics , Guanosine/immunology , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Male
18.
Mol Cell Biol ; 38(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29760281

ABSTRACT

Accessibility of antigen receptor loci to RAG is correlated with the presence of H3K4me3, which binds to a plant homeodomain (PHD) in the RAG-2 subunit and promotes V(D)J recombination. A point mutation in the PHD, W453A, eliminates binding of H3K4me3 and impairs recombination. The debilitating effect of the W453A mutation is ameliorated by second-site mutations that locate an inhibitory domain in the interval from residues 352 through 405 of RAG-2. Disruption of the inhibitory domain stimulates V(D)J recombination within extrachromosomal substrates and at endogenous antigen receptor loci. Association of RAG-1 and RAG-2 with chromatin at the IgH locus in B cell progenitors is dependent on recognition of H3K4me3 by the PHD. Strikingly, disruption of the inhibitory domain permits association of RAG with the IgH locus in the absence of H3K4me3 binding. Thus, the inhibitory domain acts as a gate that prohibits RAG from accessing the IgH locus unless RAG-2 is engaged by H3K4me3.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , VDJ Recombinases/metabolism , Adaptive Immunity , Allosteric Regulation , Amino Acid Substitution , Animals , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin Heavy Chain , HEK293 Cells , Histone Code , Humans , Mice , Models, Immunological , NIH 3T3 Cells , Point Mutation , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Protein Domains
19.
Methods Mol Biol ; 1792: 1-14, 2018.
Article in English | MEDLINE | ID: mdl-29797248

ABSTRACT

A quantitative allele-specific polymerase chain reaction in combination with an extreme limiting dilution approach (ELDA qASO-PCR) enables the detection of tumor cells in patients with multiple myeloma (MM) in bone marrow (BM) samples and in peripheral blood (PB) with a sensitivity of <10-6. The two-step procedure of patient-specific tumor cell identification via the immunoglobulin heavy chain (IgH) and kappa/lambda light chain (k/λ LC) locus, followed by tumor cells quantification by ELDA qASO-PCR allows for the application of this method to the majority of MM patients, including those with Bence Jones proteinuria.


Subject(s)
Alleles , Neoplasms/diagnosis , Neoplasms/genetics , Neoplastic Cells, Circulating/metabolism , Polymerase Chain Reaction , Base Sequence , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Humans , Immunoglobulin Heavy Chains/genetics , Neoplasm, Residual/diagnosis , Neoplasm, Residual/genetics , Neoplastic Cells, Circulating/pathology , Polymerase Chain Reaction/methods , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...