Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Methods Mol Biol ; 2453: 7-42, 2022.
Article in English | MEDLINE | ID: mdl-35622318

ABSTRACT

Immunoglobulin (IG) clonality assessment is a widely used supplementary test for the diagnosis of suspected lymphoid malignancies. The specific rearrangements of the immunoglobulin (IG) heavy and light chain genes act as a unique hallmark of a B-cell lymphoma, a feature that is used in clonality assessment. The widely used BIOMED-2/EuroClonality IG clonality assay, visualized by GeneScanning or heteroduplex analysis, has an unprecedented high detection rate because of the complementarity of this approach. However, the BIOMED-2/EuroClonality clonality assays have been developed for the assessment of specimens with optimal DNA quality. Further improvements for the assessment of samples with suboptimal DNA quality, such as from formalin-fixed paraffin-embedded (FFPE) specimens or specimens with a limited tumor burden, are required. The EuroClonality-NGS Working Group recently developed a next-generation sequencing (NGS)-based clonality assay for the detection of the IG heavy and kappa light chain rearrangements, using the same complementary approach as in the conventional assay. By employing next-generation sequencing, both the sensitivity and specificity of the clonality assay have increased, which not only is very useful for diagnostic clonality testing but also allows robust comparison of clonality patterns in a patient with multiple lymphoma's that have suboptimal DNA quality. Here, we describe the protocols for IG-NGS clonality assessment that are compatible for Ion Torrent and Illumina sequencing platforms including pre-analytical DNA isolation, the analytical phase, and the post-analytical data analysis.


Subject(s)
Gene Rearrangement , Genes, Immunoglobulin , High-Throughput Nucleotide Sequencing , Lymphoma, B-Cell , Sequence Analysis, DNA , Clone Cells/immunology , DNA/genetics , DNA/isolation & purification , Gene Rearrangement/genetics , Gene Rearrangement/immunology , Genes, Immunoglobulin/genetics , Genes, Immunoglobulin/immunology , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Sequence Analysis, DNA/methods
2.
Mol Immunol ; 141: 79-86, 2022 01.
Article in English | MEDLINE | ID: mdl-34837777

ABSTRACT

Our previous study had shown that member 13 (Hspa13) of heat shock protein family A (Hsp70) promotes plasma cell (PC) production and antibody secretion. To further explore Hspa13 expression and function, we combined single-cell RNA-sequencing and antigen receptor lineage (BCR) analysis to characterize sheep red cell‒primed splenocytes. The single-cell transcriptional profiles revealed that Hspa13 is specifically and highly expressed in PCs. These results suggest that Hspa13 is a novel PC-specific marker. In terms of its function, we found that the CD19cre-mediated conditional knock-out (cKO) of Hspa13 reduced the expression of Ebi3 and IL-10 in PCs. Ebi3 and IL-10 are important factors in IL-4‒secreting type 2 helper T cell (Th2) activation and differentiation. As expected, we found that the Hspa13 cKO reduced IL‒4-expressing follicular helper T (Tfh2) cells. Finally, the single-cell antigen receptor analysis demonstrated that the Hspa13 cKO reduced the Aicda-mediated antibody class-switching recombination (CSR) and somatic hypermutation (SHM) in germinal centers (GCs) B cells. Altogether, the single-cell atlas of splenocytes revealed a critical indirect role for the novel PC-specific marker Hspa13 in CSR and SHM in GC B cells by promoting Ebi3 and IL-10 expression in PCs to induce IL-4-expressing Tfh2 cells. Further exploration of Hspa13 expression and function will provide valuable clues for how to use Hspa13 in the treatment of autoimmune diseases.


Subject(s)
Antibodies/immunology , Germinal Center/immunology , HSP70 Heat-Shock Proteins/immunology , Recombination, Genetic/immunology , Somatic Hypermutation, Immunoglobulin/immunology , Animals , Antigens, CD19/immunology , Biomarkers/blood , Cell Differentiation/immunology , Gene Rearrangement/immunology , Mice , Mice, Knockout , Sheep , Th2 Cells/immunology , Transcription, Genetic/immunology
3.
Gut ; 69(9): 1667-1676, 2020 09.
Article in English | MEDLINE | ID: mdl-31907296

ABSTRACT

BACKGROUND: Inflammatory hepatocellular adenomas (IHCAs) are benign liver tumours characterised by an activation of the janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway caused by oncogenic activating mutations. However, a subset of IHCA lacks of identified mutation explaining the inflammatory phenotype. METHODS: 657 hepatocellular adenomas developed in 504 patients were analysed for gene expression of 17 genes and for mutations in seven genes by sequencing. 22 non-mutated IHCAs were analysed by whole-exome and/or RNA sequencing. RESULTS: We identified 296 IHCA (45%), 81% of them were mutated in either IL6ST (61%), FRK (8%), STAT3 (5%), GNAS (3%) or JAK1 (2%). Among non-mutated IHCA, RNA sequencing identified recurrent chromosome rearrangement involving ROS1, FRK or IL6 genes. ROS1 fusions were identified in 8 IHCA, involving C-terminal part of genes highly expressed in the liver (PLG, RBP4, APOB) fused with exon 33-35 to 43 of ROS1 including the tyrosine kinase domain. In two cases a truncated ROS1 transcript from exon 36 to 43 was identified. ROS1 rearrangements were validated by fluorescence in situ hybridisation (FISH) and led to ROS1 overexpression. Among the 5 IHCA with FRK rearrangements, 5 different partners were identified (MIA3, MIA2, LMO7, PLEKHA5, SEC16B) fused to a common region in FRK that included exon 3-8. No overexpression of FRK transcript was detected but the predicted chimeric proteins lacked the auto-inhibitory SH2-SH3 domains. In two IHCA, we identified truncated 3'UTR of IL6 associated with overexpression of the transcript. CONCLUSION: Recurrent chromosomal alterations involving ROS1, FRK or IL6 genes lead to activation of the JAK/STAT pathway in IHCAs.


Subject(s)
Adenoma, Liver Cell , Cytokine Receptor gp130/genetics , Liver Neoplasms , Neoplasm Proteins/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Adenoma, Liver Cell/genetics , Adenoma, Liver Cell/immunology , Adenoma, Liver Cell/pathology , Adult , Female , Gene Expression Profiling/statistics & numerical data , Gene Rearrangement/immunology , Humans , Inflammation/genetics , Janus Kinases/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Male , Mutation , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
4.
J Exp Med ; 215(11): 2901-2918, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30327417

ABSTRACT

MHCII in antigen-presenting cells (APCs) is a key regulator of adaptive immune responses. Expression of MHCII genes is controlled by the transcription coactivator CIITA, itself regulated through cell type-specific promoters. Here we show that the transcription factor NFAT5 is needed for expression of Ciita and MHCII in macrophages, but not in dendritic cells and other APCs. NFAT5-deficient macrophages showed defective activation of MHCII-dependent responses in CD4+ T lymphocytes and attenuated capacity to elicit graft rejection in vivo. Ultrasequencing analysis of NFAT5-immunoprecipitated chromatin uncovered an NFAT5-regulated region distally upstream of Ciita This region was required for CIITA and hence MHCII expression, exhibited NFAT5-dependent characteristics of active enhancers such as H3K27 acetylation marks, and required NFAT5 to interact with Ciita myeloid promoter I. Our results uncover an NFAT5-regulated mechanism that maintains CIITA and MHCII expression in macrophages and thus modulates their T lymphocyte priming capacity.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , Nuclear Proteins/immunology , Trans-Activators/immunology , Transcription Factors/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Gene Rearrangement/immunology , Histocompatibility Antigens Class II/genetics , Macrophages/cytology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics
5.
Front Immunol ; 9: 2249, 2018.
Article in English | MEDLINE | ID: mdl-30349529

ABSTRACT

Discussion of the antibody repertoire usually emphasizes diversity, but a conspicuous feature of the light chain repertoire is its lack of diversity. The diversity of reported allelic variants of germline light chain genes is also limited, even in well-studied species. In this review, the implications of this lack of diversity are considered. We explore germline and rearranged light chain genes in a variety of species, with a particular focus on human and mouse genes. The importance of the number, organization and orientation of the genes for the control of repertoire development is discussed, and we consider how primary rearrangements and receptor editing together shape the expressed light chain repertoire. The resulting repertoire is dominated by just a handful of IGKV and IGLV genes. It has been hypothesized that an important function of the light chain is to guard against self-reactivity, and the role of secondary rearrangements in this process could explain the genomic organization of the light chain genes. It could also explain why the light chain repertoire is so limited. Heavy and light chain genes may have co-evolved to ensure that suitable light chain partners are usually available for each heavy chain that forms early in B cell development. We suggest that the co-evolved loci of the house mouse often became separated during the inbreeding of laboratory mice, resulting in new pairings of loci that are derived from different sub-species of the house mouse. A resulting vulnerability to self-reactivity could explain at least some mouse models of autoimmune disease.


Subject(s)
Antibodies/immunology , Gene Rearrangement/immunology , Genes, Immunoglobulin Light Chain/immunology , Immunoglobulin Light Chains/immunology , Mice, Inbred Strains/immunology , Receptors, Immunologic/immunology , Self Tolerance/immunology , Animals , Antibodies/genetics , Gene Rearrangement/genetics , Genes, Immunoglobulin Light Chain/genetics , Genetic Variation/genetics , Genetic Variation/immunology , Immunoglobulin Light Chains/genetics , Mice, Inbred Strains/classification , Mice, Inbred Strains/genetics , Receptors, Immunologic/genetics , Self Tolerance/genetics , Species Specificity
6.
Mol Immunol ; 99: 30-38, 2018 07.
Article in English | MEDLINE | ID: mdl-29679865

ABSTRACT

Variable lymphocyte receptors B (VLRBs) are non-immunoglobulin components of the humoral immune system in jawless vertebrates including hagfish (Eptatretus burgeri) and lamprey (Petromyzon marinus). Hagfish VLRBs consist of leucine rich repeat (LRR) modules with a superhydrophobic C-terminal tail, the latter of which leads to extremely low expression levels in recombinant protein technology. Here, we present an artificially oligomerized VLRB (arVLRB) that conjugates via the C4bp oligomerization domain derived from human C4b-binding protein (hC4bp) rather than the superhydrophobic tail. The resulting arVLRB had a tightly multimerized form with seven monomeric VLRB arms and showed high expression and secretion levels in a mammalian expression system. To isolate antigen-specific arVLRB, we constructed large VLRB libraries from hagfish immunized with the fish pathogen, viral hemorrhagic septicemia virus (VHSV). The selected arVLRBs were found to recognize various types of antigens, including the recombinant target protein, purified viruses, and progeny viruses, with high antigen binding abilities and specificities. We also performed in vitro affinity maturation of the arVLRBs through LRRCT mutagenesis, and found that this enhanced their antigen-binding properties by at least 125-fold. Our epitope mapping analysis revealed that 37DWDTPL42, which is located in a region conserved among the glycoproteins of all VHSV isolates, is the recognition epitope of the arVLRBs. Thus, our newly developed arVLRB could prove useful in the development of universal diagnostic tools and/or therapeutic agents for the virus. Together, our novel findings provide valuable insights into hagfish VLRB and its potential use as a novel alternative to conventional antibodies for biotechnological applications.


Subject(s)
Glycoproteins/immunology , Hagfishes/immunology , Hemorrhagic Septicemia, Viral/immunology , Lymphocytes/immunology , Novirhabdovirus/immunology , Adaptive Immunity/immunology , Animals , Antibodies/immunology , Complement C4b-Binding Protein/immunology , Epitopes/immunology , Gene Rearrangement/immunology , Humans , Immunization/methods , Mammals/immunology , Petromyzon/immunology
7.
Int J Mol Sci ; 18(9)2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28867784

ABSTRACT

Lymphocytes are endowed with unique and specialized enzymatic mutagenic properties that allow them to diversify their antigen receptors, which are crucial sensors for pathogens and mediators of adaptive immunity. During lymphocyte development, the antigen receptors expressed by B and T lymphocytes are assembled in an antigen-independent fashion by ordered variable gene segment recombinations (V(D)J recombination), which is a highly ordered and regulated process that requires the recombination activating gene products 1 & 2 (RAG1, RAG2). Upon activation by antigen, B lymphocytes undergo additional diversifications of their immunoglobulin B-cell receptors. Enzymatically induced somatic hypermutation (SHM) and immunoglobulin class switch recombination (CSR) improves the affinity for antigen and shape the effector function of the humoral immune response, respectively. The activation-induced cytidine deaminase (AID) enzyme is crucial for both SHM and CSR. These processes have evolved to both utilize as well as evade different DNA repair and DNA damage response pathways. The delicate balance between enzymatic mutagenesis and DNA repair is crucial for effective immune responses and the maintenance of genomic integrity. Not surprisingly, disturbances in this balance are at the basis of lymphoid malignancies by provoking the formation of oncogenic mutations and chromosomal aberrations. In this review, we discuss recent mechanistic insight into the regulation of RAG1/2 and AID expression and activity in lymphocytes and the complex interplay between these mutagenic enzymes and DNA repair and DNA damage response pathways, focusing on the base excision repair and mismatch repair pathways. We discuss how disturbances of this interplay induce genomic instability and contribute to oncogenesis.


Subject(s)
DNA Repair/genetics , Immunity, Humoral/genetics , Somatic Hypermutation, Immunoglobulin/genetics , V(D)J Recombination/genetics , B-Lymphocytes/immunology , Cytidine Deaminase/genetics , DNA Damage/genetics , DNA Damage/immunology , DNA Repair/immunology , Gene Rearrangement/genetics , Gene Rearrangement/immunology , Humans , Mutagenesis/genetics , Mutagenesis/immunology , Somatic Hypermutation, Immunoglobulin/immunology , T-Lymphocytes/immunology , V(D)J Recombination/immunology
8.
J Immunol ; 193(3): 1440-50, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973444

ABSTRACT

Activation-induced cytidine deaminase (AID) is essential for class-switch recombination (CSR) and somatic hypermutation (SHM) of Ig genes. The AID C terminus is required for CSR, but not for S-region DNA double-strand breaks (DSBs) during CSR, and it is not required for SHM. AID lacking the C terminus (ΔAID) is a dominant negative (DN) mutant, because human patients heterozygous for this mutant fail to undergo CSR. In agreement, we show that ΔAID is a DN mutant when expressed in AID-sufficient mouse splenic B cells. To have DN function, ΔAID must have deaminase activity, suggesting that its ability to induce DSBs is important for the DN function. Supporting this hypothesis, Msh2-Msh6 have been shown to contribute to DSB formation in S regions, and we find in this study that Msh2 is required for the DN activity, because ΔAID is not a DN mutant in msh2(-/-) cells. Our results suggest that the DNA DSBs induced by ΔAID are unable to participate in CSR and might interfere with the ability of full-length AID to participate in CSR. We propose that ΔAID is impaired in its ability to recruit nonhomologous end joining repair factors, resulting in accumulation of DSBs that undergo aberrant resection. Supporting this hypothesis, we find that the S-S junctions induced by ΔAID have longer microhomologies than do those induced by full-length AID. In addition, our data suggest that AID binds Sµ regions in vivo as a monomer.


Subject(s)
Cytidine Deaminase/physiology , DNA Mismatch Repair/immunology , Gene Rearrangement/immunology , Animals , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Mismatch Repair/genetics , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Peptide Fragments/genetics , Primary Cell Culture
9.
Fish Shellfish Immunol ; 36(1): 206-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24239597

ABSTRACT

The interleukin 1 receptor-associated kinase 4 (IRAK4) is an essential factor for TLR-mediated activation of the host's immune functions subsequent to pathogen contact. We have characterized the respective cDNA and gene sequences from three salmonid species, salmon, rainbow trout and maraena whitefish. The gene from salmon is structured into eleven exons, as is the mammalian homologue, while exons have been fused in the genes from the two other salmonid species. Rainbow trout expresses also a pseudogene at low levels. Its basic structure resembles more closely the primordial gene than the functional copy does. The N-terminal death domain and the C-terminal protein kinase domain of the factors are better conserved throughout evolution than the linker domain. The deduced amino acid sequences of the factors from all three species group together in an evolutionary tree of IRAK4 factors. Scrutinizing expression and function of IRAK4 from rainbow trout, we found its highest expression in head kidney and spleen and lowest expression in muscle tissue. Infecting fish with Aeromonas salmonicida did not modulate its expression during 72 h of observation. Expression of a GFP-tagged trout IRAK4 revealed, expectedly, its cytoplasmic localization in human HEK-293 cells. However, this factor significantly quenched in a dose-dependent fashion not only the pathogen-induced stimulation of NF-κB factors in the HEK-293 reconstitution system of TLR2 signaling, but also the basal NF-κB levels in unstimulated control cells. Our data unexpectedly imply that IRAK4 is involved in establishing threshold levels of active NF-κB in resting cells.


Subject(s)
Aeromonas salmonicida/immunology , Fish Diseases/microbiology , Gram-Negative Bacterial Infections/veterinary , Interleukin-1 Receptor-Associated Kinases/immunology , Phylogeny , Salmonidae , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Fish Diseases/genetics , Fish Diseases/immunology , Gene Rearrangement/immunology , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , HEK293 Cells , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Molecular Sequence Data , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment , Sequence Analysis, DNA , Signal Transduction/immunology , Toll-Like Receptors/immunology
10.
PLoS One ; 8(2): e57877, 2013.
Article in English | MEDLINE | ID: mdl-23469094

ABSTRACT

VH replacement occurs through RAG-mediated recombination between the cryptic recombination signal sequence (cRSS) near the 3' end of a rearranged VH gene and the 23-bp RSS from an upstream unrearranged VH gene. Due to the location of the cRSS, VH replacement leaves a short stretch of nucleotides from the previously rearranged VH gene at the newly formed V-D junction, which can be used as a marker to identify VH replacement products. To determine the contribution of VH replacement products to mouse antibody repertoire, we developed a Java-based VH Replacement Footprint Analyzer (VHRFA) program and analyzed 17,179 mouse IgH gene sequences from the NCBI database to identify VH replacement products. The overall frequency of VH replacement products in these IgH genes is 5.29% based on the identification of pentameric VH replacement footprints at their V-D junctions. The identified VH replacement products are distributed similarly in IgH genes using most families of VH genes, although different families of VH genes are used differentially. The frequencies of VH replacement products are significantly elevated in IgH genes derived from several strains of autoimmune prone mice and in IgH genes encoding autoantibodies. Moreover, the identified VH replacement footprints in IgH genes from autoimmune prone mice or IgH genes encoding autoantibodies preferentially encode positively charged amino acids. These results revealed a significant contribution of VH replacement products to the diversification of antibody repertoire and potentially, to the generation of autoantibodies in mice.


Subject(s)
Antibody Diversity/genetics , Computational Biology , Gene Rearrangement/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Animals , Autoantibodies/genetics , Autoantibodies/immunology , Autoimmunity/genetics , Databases, Genetic , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Mice , Mutation/immunology
11.
PLoS Genet ; 8(4): e1002675, 2012.
Article in English | MEDLINE | ID: mdl-22570620

ABSTRACT

Class-switch recombination (CSR), induced by activation-induced cytidine deaminase (AID), can be divided into two phases: DNA cleavage of the switch (S) regions and the joining of the cleaved ends of the different S regions. Here, we show that the DSIF complex (Spt4 and Spt5), a transcription elongation factor, is required for CSR in a switch-proficient B cell line CH12F3-2A cells, and Spt4 and Spt5 carry out independent functions in CSR. While neither Spt4 nor Spt5 is required for transcription of S regions and AID, expression array analysis suggests that Spt4 and Spt5 regulate a distinct subset of transcripts in CH12F3-2A cells. Curiously, Spt4 is critically important in suppressing cryptic transcription initiating from the intronic Sµ region. Depletion of Spt5 reduced the H3K4me3 level and DNA cleavage at the Sα region, whereas Spt4 knockdown did not perturb the H3K4me3 status and S region cleavage. H3K4me3 modification level thus correlated well with the DNA breakage efficiency. Therefore we conclude that Spt5 plays a role similar to the histone chaperone FACT complex that regulates H3K4me3 modification and DNA cleavage in CSR. Since Spt4 is not involved in the DNA cleavage step, we suspected that Spt4 might be required for DNA repair in CSR. We examined whether Spt4 or Spt5 is essential in non-homologous end joining (NHEJ) and homologous recombination (HR) as CSR utilizes general repair pathways. Both Spt4 and Spt5 are required for NHEJ and HR as determined by assay systems using synthetic repair substrates that are actively transcribed even in the absence of Spt4 and Spt5. Taken together, Spt4 and Spt5 can function independently in multiple transcription-coupled steps of CSR.


Subject(s)
Chromatin , Chromosomal Proteins, Non-Histone , DNA Repair , Homologous Recombination , Immunoglobulin Class Switching , Immunoglobulins , Transcriptional Elongation Factors , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Culture Techniques , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/immunology , Cytidine Deaminase/genetics , DNA Cleavage , DNA End-Joining Repair/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Rearrangement/immunology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Homologous Recombination/genetics , Homologous Recombination/immunology , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulins/genetics , Immunoglobulins/metabolism , Ku Autoantigen , Mice , Protein Processing, Post-Translational , Signal Transduction , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/immunology
12.
Ann Hematol ; 91(8): 1251-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22453306

ABSTRACT

The M protein of POEMS syndrome is essentially λ light chain restricted. Several studies have demonstrated the restrict usage of immunoglobulin λ light chain variable region (IGLV) genes in patients with POEMS syndrome. However, these studies only included a limited number of cases, and it is not clear whether the clinical features are influenced by the IGLV gene in POEMS syndrome. Here we demonstrated that the clonal IGLV genes were strictly derived from IGLV 1-40 (11 patients, 36.7 %) and IGLV 1-44 (19, 63.3 %) gene in 30 patients with POEMS syndrome. We further evaluated the relationship between clinical features and IGLV genes. Our study showed that patients with IGLV 1-44 were older than those with IGLV 1-40, and patients with IGLV 1-40 had more severe neuropathy, hypertrichosis, and papilledema. It was suggested that the IGLV gene influenced clinical characteristics in POEMS syndrome.


Subject(s)
Immunoglobulin Variable Region/genetics , Immunoglobulin lambda-Chains/genetics , POEMS Syndrome/diagnosis , POEMS Syndrome/genetics , Adult , Aged , Cohort Studies , DNA Mutational Analysis/methods , Diagnosis, Differential , Female , Gene Rearrangement/genetics , Gene Rearrangement/immunology , Humans , Immunoglobulin Light Chains/analysis , Immunoglobulin Light Chains/genetics , Immunoglobulin Variable Region/analysis , Immunoglobulin lambda-Chains/analysis , Male , Middle Aged , POEMS Syndrome/immunology , Prognosis , Young Adult
13.
Eur J Immunol ; 42(3): 760-70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22105329

ABSTRACT

In humanized mice, the T-cell repertoire is derived from genetically identical human progenitors in distinct animals. Thus, careful comparison of the T-cell repertoires of humanized mice with those of humans may reveal the contribution of genetic determinism on T-cell repertoire formation. Here, we performed a comprehensive assessment of the distribution of V-J combinations of the human ß chain of the T-cell receptor (hTRBV) in NOD.SCID.γc(-/-) (NSG) humanized mice. We observed that numerous V-J combinations were equally distributed in the thymus and in the periphery of humanized mice compared with human references. A global analysis of the data, comparing repertoire perturbation indices in humanized NSG mice and unrelated human PBMCs, reveals that 50% of the hTRBV families significantly overlapped. Using multivariate ranking and bootstrap analyses, we found that 18% of all possible V-J combinations contributed close to 50% of the expressed diversity, with significant over-representation of BV5-J1.1+1.2 and BV6-J1.1+1.2 rearrangements. Finally, comparison of CD3(-) and CD3(+) thymocyte repertoires indicated that the observed V-J combination overlap was already present before TCR-MHC selection in the thymus. Altogether, our results show that half of the T-cell repertoire combinatorial diversity in humans is genetically determined.


Subject(s)
Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn , DNA/chemistry , DNA/genetics , Flow Cytometry , Gene Rearrangement/genetics , Gene Rearrangement/immunology , Humans , Immunoglobulin Joining Region/genetics , Immunoglobulin Joining Region/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Linear Models , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multivariate Analysis , Polymerase Chain Reaction , Specific Pathogen-Free Organisms , V(D)J Recombination/genetics , V(D)J Recombination/immunology
14.
Blood ; 118(12): 3331-9, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21725051

ABSTRACT

One-third of all splenic marginal zone lymphomas (SMZL) use the IgH VH1-02 gene. These cases are usually not associated with hepatitis C virus infection. Of interest, the rearranged VH1-02 genes display similar complementarity determining regions 3, a finding confirmed by our study. The latter suggests that these SMZL may produce antibodies with similar reactivity. We produced recombinant antibodies from 5 SMZL cases with VH1-02 gene rearrangement to study the binding reactivity of these antibodies. Surprisingly, the recombinant antibodies demonstrated poly- and self-reactivity as demonstrated by their reactivity with nuclear, cytoplasmic, as well as membranous antigens expressed by human cells and by reactivity with human serum. This polyreactivity was specific as demonstrated by ELISA. The antibodies did not react with proteins on the cell surface that are induced by apoptosis as shown for antibodies produced by chronic lymphatic leukemia with VH1-02 gene rearrangement. The results indicate that a common subset of SMZL arises from polyreactive B cells, a subset of marginal zone B cells that are important in the immunologic defense against infection.


Subject(s)
Antibodies/metabolism , Gene Rearrangement/immunology , Immunoglobulin Light Chains/metabolism , Immunoglobulin Variable Region/metabolism , Lymphoma, B-Cell, Marginal Zone/immunology , Recombinant Proteins/metabolism , Spleen/immunology , Splenic Neoplasms/immunology , Antibodies/genetics , Antibodies/immunology , Antibody Specificity , Blood Proteins/immunology , Blood Proteins/metabolism , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Female , HEK293 Cells , Hepacivirus/growth & development , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/virology , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunohistochemistry , Immunophenotyping , Isoantigens/immunology , Isoantigens/metabolism , Karyotyping , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/pathology , Male , Middle Aged , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/metabolism , Spleen/pathology , Splenic Neoplasms/genetics , Splenic Neoplasms/pathology , Transfection
15.
Scand J Immunol ; 72(1): 44-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20591075

ABSTRACT

IgG4-related sclerosing sialadenitis is currently considered as an autoimmune disease distinct from Sjogren's syndrome (SS) and responds extremely well to steroid therapy. To further elucidate the characteristics of IgG4-related sclerosing sialadenitis, we analysed VH fragments of IgH genes and their somatic hypermutation in SS (n = 3) and IgG4-related sclerosing sialadenitis (n = 3), using sialolithiasis (n = 3) as a non-autoimmune control. DNA was extracted from the affected inflammatory lesions. After PCR amplification of rearranged IgH genes, at least 50 clones per case (more than 500 clones in total) were sequenced for VH fragments. Monoclonal IgH rearrangement was not detected in any cases examined. When compared with sialolithiasis, there was no VH family or VH fragment specific to SS or IgG4-related sclerosing sialadenitis. However, rates of unmutated VH fragments in SS (30%) and IgG4-related sclerosing sialadenitis (39%) were higher than that in sialolithiasis (14%) with statistical significance (P = 0.0005 and P < 0.0001, respectively). This finding suggests that some autoantibodies encoded by germline or less mutated VH genes may fail to be eliminated and could play a role in the development of SS and IgG4-related sclerosing sialadenitis.


Subject(s)
Gene Rearrangement/immunology , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/immunology , Sialadenitis/immunology , Sjogren's Syndrome/immunology , Somatic Hypermutation, Immunoglobulin/immunology , Aged , Biopsy , Cloning, Molecular , DNA/chemistry , DNA/genetics , Female , Gene Rearrangement/genetics , Humans , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction , Sialadenitis/genetics , Sjogren's Syndrome/genetics , Somatic Hypermutation, Immunoglobulin/genetics
16.
Dev Comp Immunol ; 34(5): 579-89, 2010 May.
Article in English | MEDLINE | ID: mdl-20056120

ABSTRACT

The reptiles are the last major taxon of jawed vertebrates in which immunoglobulin light chain isotypes have not been well characterized. Using the recently released genome sequencing data, we show in this study that the reptile Anolis carolinensis expresses both lambda and kappa light chain genes. The genomic organization of both gene loci is structurally similar to their respective counterparts in mammals. The identified lambda locus contains three constant region genes each preceded by a joining gene segment, and a total of 37 variable gene segments. In contrast, the kappa locus contains only a single constant region gene, and two joining gene segments with a single family of 14 variable gene segments located upstream. Analysis of junctions of the recombined VJ transcripts reveals a paucity of N and P nucleotides in both expressed lambda and kappa sequences. These results help us to understand the generation of the immunoglobulin repertoire in reptiles and immunoglobulin evolution in vertebrates.


Subject(s)
Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/genetics , Lizards/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Evolution, Molecular , Gene Expression/immunology , Gene Rearrangement/immunology , Genome , Immunoglobulin kappa-Chains/immunology , Immunoglobulin kappa-Chains/metabolism , Immunoglobulin lambda-Chains/immunology , Immunoglobulin lambda-Chains/metabolism , Lizards/immunology , Molecular Sequence Data , Phylogeny
17.
Nat Immunol ; 11(2): 171-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19946273

ABSTRACT

STAT5 and interleukin 7 (IL-7) signaling are thought to control B lymphopoiesis by regulating the expression of key transcription factors and by activating variable (V(H)) gene segments at the immunoglobulin heavy-chain (Igh) locus. Using conditional mutagenesis to delete the gene encoding the transcription factor STAT5, we demonstrate that the development of pro-B cells was restored by transgenic expression of the prosurvival protein Bcl-2, which compensated for loss of the antiapoptotic protein Mcl-1. Expression of the genes encoding the B cell-specification factor EBF1 and the B cell-commitment protein Pax5 as well as V(H) gene recombination were normal in STAT5- or IL-7 receptor alpha-chain (IL-7Ralpha)-deficient pro-B cells rescued by Bcl-2. STAT5-expressing pro-B cells contained little or no active chromatin at most V(H) genes. In contrast, rearrangements of the immunoglobulin-kappa light-chain locus (Igk) were more abundant in STAT5- or IL-7Ralpha-deficient pro-B cells. Hence, STAT5 and IL-7 signaling control cell survival and the developmental ordering of immunoglobulin gene rearrangements by suppressing premature Igk recombination in pro-B cells.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation/genetics , Lymphoid Progenitor Cells/cytology , Lymphopoiesis/genetics , STAT5 Transcription Factor/genetics , Signal Transduction/immunology , Animals , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/immunology , Gene Rearrangement/genetics , Gene Rearrangement/immunology , Genes, Immunoglobulin/genetics , Genes, Immunoglobulin/immunology , Interleukin-7/genetics , Interleukin-7/immunology , Lymphoid Progenitor Cells/immunology , Lymphopoiesis/immunology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/immunology
18.
J Immunol Methods ; 352(1-2): 111-7, 2010 Jan 31.
Article in English | MEDLINE | ID: mdl-19919841

ABSTRACT

Current techniques to peripherally assess thymic function are: the signal-joint T-cell receptor excision circle (sj-TREC) level measurement and the naive T cell and CD31+ TREC-rich subset determination. However, all of them are indirect approaches and none could be considered a direct recent thymic emigrant (RTE) marker. To overcome their limitations, Dion et al. (2004) described the sj/beta-TREC ratio that allows the peripheral quantification of the double negative to double positive intrathymic proliferation step. Nevertheless, the protocol described is expensive, sample and time-consuming, thus, limiting its usefulness. In this study, we describe a simplified protocol that reduces from 33 to 9 the amount of PCR reaction needed but maintaining the sensitivity and reproducibility of the original technique. In addition, we corroborated the effectiveness of our technique as an accurate thymic output-related marker by correlating the peripheral sj/beta-TREC ratio with a direct measurement of thymic function as the percentage of double positive thymocytes (r=0.601, p<0.001).


Subject(s)
Antigens, Differentiation/immunology , Gene Rearrangement/immunology , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/metabolism , Antigens, CD/biosynthesis , Antigens, Differentiation/genetics , Cell Differentiation , Cell Proliferation , Female , Fetal Blood/cytology , Gene Rearrangement/genetics , Humans , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Reproducibility of Results , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology
19.
Nat Immunol ; 10(8): 817-21, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19621044

ABSTRACT

During B cell and T cell development, the lymphoid-specific proteins RAG-1 and RAG-2 act together to initiate the assembly of antigen receptor genes through a series of site-specific somatic DNA rearrangements that are collectively called variable-diversity-joining (V(D)J) recombination. In the past 20 years, a great deal has been learned about the enzymatic activities of the RAG-1-RAG-2 complex. Recent studies have identified several new and exciting regulatory functions of the RAG-1-RAG-2 complex. Here we discuss some of these functions and suggest that the RAG-1-RAG-2 complex nucleates a specialized subnuclear compartment that we call the 'V(D)J recombination factory'.


Subject(s)
DNA-Binding Proteins/immunology , Gene Rearrangement/immunology , Homeodomain Proteins/immunology , Models, Biological , Nuclear Proteins/immunology , Recombination, Genetic , VDJ Recombinases/immunology , Animals , B-Lymphocytes/immunology , Chromatin/metabolism , DNA Damage/immunology , DNA Repair/immunology , Histones/metabolism , Humans , Protein Binding
20.
Arch Immunol Ther Exp (Warsz) ; 57(2): 117-28, 2009.
Article in English | MEDLINE | ID: mdl-19333728

ABSTRACT

The recent discovery that natural killer T (NKT) cell nuclei are totipotent opens a novel avenue for further understanding NKT cell function in normal and diseased states. The progeny of a cloned mouse harboring the in-frame rearranged Valpha14-Jalpha18 T cell receptor in one allele showed a significant increase in NKT cell number compared with wild-type or littermate control mice that possessed a different TCR. Importantly, NKT cells from such progeny produced both interferon-gamma and interleukin-4, a hallmark of NKT cells. In these progeny, NKT cell development appeared to be instructively, rather than permissively, determined. Using embryonic stem cells prepared via the somatic cell nuclear transfer of NKT nuclei, relatively mature NKT cells were induced under conditions permissible for T cell induction. Furthermore, these NKT cells matured autonomously upon injection into mice, resulting in an antigen-specific adjuvant effect.


Subject(s)
Gene Rearrangement/physiology , Natural Killer T-Cells/physiology , Regeneration/physiology , Totipotent Stem Cells/physiology , Animals , Cloning, Organism , Gene Rearrangement/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Natural Killer T-Cells/immunology , Nuclear Transfer Techniques , Totipotent Stem Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...