Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
3.
Commun Biol ; 7(1): 31, 2024 01 05.
Article in English | MEDLINE | ID: mdl-38182651

ABSTRACT

The stability of cellular phenotypes in developing organisms depends on error-free transmission of epigenetic and genetic information during mitosis. Methylation of cytosine residues in genomic DNA is a key epigenetic mark that modulates gene expression and prevents genome instability. Here, we report on a genetic test of the relationship between DNA replication and methylation in the context of the developing vertebrate organism instead of cell lines. Our analysis is based on the identification of hypomorphic alleles of dnmt1, encoding the DNA maintenance methylase Dnmt1, and pole1, encoding the catalytic subunit of leading-strand DNA polymerase epsilon holoenzyme (Pole). Homozygous dnmt1 mutants exhibit genome-wide DNA hypomethylation, whereas the pole1 mutation is associated with increased DNA methylation levels. In dnmt1/pole1 double-mutant zebrafish larvae, DNA methylation levels are restored to near normal values, associated with partial rescue of mutant-associated transcriptional changes and phenotypes. Hence, a balancing antagonism between DNA replication and maintenance methylation buffers against replicative errors contributing to the robustness of vertebrate development.


Subject(s)
DNA Methylation , Zebrafish , Animals , Zebrafish/genetics , Alleles , DNA , Epigenesis, Genetic
4.
Eur J Immunol ; 53(12): e2350725, 2023 12.
Article in English | MEDLINE | ID: mdl-37724048

ABSTRACT

In mammals, T-cell development depends on the activity of the Foxn1 transcription factor in the thymic epithelium; mutations in the vertebrate-specific Foxn1 gene are associated with profound T-cell lymphopenia and fatal immunodeficiency. Here, we examined the extent of T-cell development in teleosts lacking a functional foxn1 gene. In zebrafish carrying a deleterious internal deletion of foxn1, reduced but robust lymphopoietic activity is maintained in the mutant thymus. Moreover, pseudogenization or loss of foxn1 in the genomes of deep-sea anglerfishes is independent of the presence or absence of the canonical signatures of the T-cell lineage. Thus, in contrast to the situation in mammals, the teleost thymus can support foxn1-independent lymphopoiesis, most likely through the activity of the Foxn4, an ancient metazoan paralog of Foxn1. Our results imply that during the early stages of vertebrate evolution, genetic control of thymopoiesis was functionally redundant and thus robust; in mammals, the genetic network was reorganized to become uniquely dependent on the FOXN1 transcription factor.


Subject(s)
Gene Regulatory Networks , Zebrafish , Mice , Animals , Mice, Transgenic , Zebrafish/genetics , T-Lymphocytes , Thymus Gland , Transcription Factors/genetics , Forkhead Transcription Factors/genetics , Epithelial Cells , Mammals/genetics , Zebrafish Proteins/genetics
5.
BMC Immunol ; 24(1): 20, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37480016

ABSTRACT

BACKGROUND: The anglerfish, belonging to the teleost order Lophiiformes, are a diverse and species-rich group of fish that are known to exhibit a number of unique morphological, reproductive and immunological adaptations. Work to date has identified the loss of specific adaptive immune components in two of the five Lophiiformes sub-orders (Lophioidei and Ceratioidei), while no anomalies have been identified to date in two other sub-orders, Antennaroidei and Chaunacoidei. The immunogenome of the fifth sub-order, Ogcocephaloidei has not yet been investigated, and we have therefore used whole genome shotgun sequencing, combined with RNA-seq, to survey the adaptive immune capabilities of the polka-dot batfish, O. cubifrons, as a representative of this as yet unexplored sub-order. RESULTS: We find that the O. cubifrons genome encodes the core genes needed to mount adaptive T and B cell responses. These genes include those necessary for rearranging and editing antigen receptors, the antigen receptors themselves; as well as the co-receptors, signalling molecules, and antigen presenting molecules (both class I and class II) needed for B cell and T cell development and activation. CONCLUSIONS: From an immune perspective, the polka-dot batfish has a canonical complement of adaptive immune genes, and does not exhibit any of the adaptive immune changes previously identified in monkfish and oceanic anglerfish.


Subject(s)
B-Lymphocytes , Animals , Cell Differentiation
6.
Nature ; 619(7968): 193-200, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37344590

ABSTRACT

Lymphocytes of vertebrate adaptive immune systems acquired the capability to assemble, from split genes in the germline, billions of functional antigen receptors1-3. These receptors show specificity; unlike the broadly tuned receptors of the innate system, antibodies (Ig) expressed by B cells, for instance, can accurately distinguish between the two enantiomers of organic acids4, whereas T cell receptors (TCRs) reliably recognize single amino acid replacements in their peptide antigens5. In developing lymphocytes, antigen receptor genes are assembled from a comparatively small set of germline-encoded genetic elements in a process referred to as V(D)J recombination6,7. Potential self-reactivity of some antigen receptors arising from the quasi-random somatic diversification is suppressed by several robust control mechanisms8-12. For decades, scientists have puzzled over the evolutionary origin of somatically diversifying antigen receptors13-16. It has remained unclear how, at the inception of this mechanism, immunologically beneficial expanded receptor diversity was traded against the emerging risk of destructive self-recognition. Here we explore the hypothesis that in early vertebrates, sequence microhomologies marking the ends of recombining elements became the crucial targets of selection determining the outcome of non-homologous end joining-based repair of DNA double-strand breaks generated during RAG-mediated recombination. We find that, across the main clades of jawed vertebrates, TCRα repertoire diversity is best explained by species-specific extents of such sequence microhomologies. Thus, selection of germline sequence composition of rearranging elements emerges as a major factor determining the degree of diversity of somatically generated antigen receptors.


Subject(s)
Evolution, Molecular , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Receptors, Antigen, T-Cell, alpha-beta , V(D)J Recombination , Animals , Receptors, Antigen, T-Cell, alpha-beta/genetics , V(D)J Recombination/genetics , Vertebrates/classification , Vertebrates/genetics , DNA End-Joining Repair , DNA Breaks, Double-Stranded , Genes, RAG-1 , Species Specificity , Sequence Homology , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/genetics , Lymphocytes/metabolism
7.
Sci Rep ; 12(1): 21401, 2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36496511

ABSTRACT

The zinc finger transcription factor Ikaros1 (Ikzf1) is required for lymphoid development in mammals. Four zinc fingers constitute its DNA binding domain and two zinc fingers are present in the C-terminal protein interaction module. We describe the phenotypes of zebrafish homozygous for two distinct mutant ikzf1 alleles. The IT325 variant lacks the C-terminal two zinc fingers, whereas the fr105 variant retains only the first zinc finger of the DNA binding domain. An intact ikzf1 gene is required for larval T cell development, whereas low levels of adult lymphoid development recover in the mutants. By contrast, the mutants exhibit a signature of increased myelopoiesis at larval and adult stages. Both mutations stimulate erythroid differentiation in larvae, indicating that the C-terminal zinc fingers negatively regulate the extent of red blood cell production. An unexpected differential effect of the two mutants on adult erythropoiesis suggests a direct requirement of an intact DNA binding domain for entry of progenitors into the red blood cell lineage. Collectively, our results reinforce the biological differences between larval and adult haematopoiesis, indicate a stage-specific function of ikzf1 in regulating the hierarchical bifurcations of differentiation, and assign distinct functions to the DNA binding domain and the C-terminal zinc fingers.


Subject(s)
Transcription Factors , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Transcription Factors/metabolism , Zinc Fingers/genetics , Cell Differentiation/genetics , Erythropoiesis/genetics , DNA/metabolism , Mammals/metabolism
8.
Commun Biol ; 4(1): 1201, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34671088

ABSTRACT

To capture the global gene network regulating the differentiation of immature T cells in an unbiased manner, large-scale forward genetic screens in zebrafish were conducted and combined with genetic interaction analysis. After ENU mutagenesis, genetic lesions associated with failure of T cell development were identified by meiotic recombination mapping, positional cloning, and whole genome sequencing. Recessive genetic variants in 33 genes were identified and confirmed as causative by additional experiments. The mutations affected T cell development but did not perturb the development of an unrelated cell type, growth hormone-expressing somatotrophs, providing an important measure of cell-type specificity of the genetic variants. The structure of the genetic network encompassing the identified components was established by a subsequent genetic interaction analysis, which identified many instances of positive (alleviating) and negative (synthetic) genetic interactions. Several examples of synthetic lethality were subsequently phenocopied using combinations of small molecule inhibitors. These drugs not only interfered with normal T cell development, but also elicited remission in a model of T cell acute lymphoblastic leukaemia. Our findings illustrate how genetic interaction data obtained in the context of entire organisms can be exploited for targeted interference with specific cell types and their malignant derivatives.


Subject(s)
Gene Regulatory Networks , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Synthetic Lethal Mutations , T-Lymphocytes/metabolism , Animals , Disease Models, Animal , Epistasis, Genetic , Phenotype , Zebrafish
9.
Sci Adv ; 7(1)2021 01.
Article in English | MEDLINE | ID: mdl-33523858

ABSTRACT

The rules underlying the structure of antigen receptor repertoires are not yet fully defined, despite their enormous importance for the understanding of adaptive immunity. With current technology, the large antigen receptor repertoires of mice and humans cannot be comprehensively studied. To circumvent the problems associated with incomplete sampling, we have studied the immunogenetic features of one of the smallest known vertebrates, the cyprinid fish Paedocypris sp. "Singkep" ("minifish"). Despite its small size, minifish has the key genetic facilities characterizing the principal vertebrate lymphocyte lineages. As described for mammals, the frequency distributions of immunoglobulin and T cell receptor clonotypes exhibit the features of fractal systems, demonstrating that self-similarity is a fundamental property of antigen receptor repertoires of vertebrates, irrespective of body size. Hence, minifish achieve immunocompetence via a few thousand lymphocytes organized in robust scale-free networks, thereby ensuring immune reactivity even when cells are lost or clone sizes fluctuate during immune responses.


Subject(s)
Receptors, Antigen, T-Cell , Vertebrates , Adaptive Immunity , Animals , Fishes , Mammals , Receptors, Antigen, T-Cell/genetics
10.
Open Biol ; 11(2): 200383, 2021 02.
Article in English | MEDLINE | ID: mdl-33622100

ABSTRACT

In vertebrates, the development of lymphocytes from undifferentiated haematopoietic precursors takes place in so-called primary lymphoid organs, such as the thymus. Therein, lymphocytes undergo a complex differentiation and selection process that culminates in the generation of a pool of mature T cells that collectively express a self-tolerant repertoire of somatically diversified antigen receptors. Throughout this entire process, the microenvironment of the thymus in large parts dictates the sequence and outcome of the lymphopoietic activity. In vertebrates, direct genetic evidence in some species and circumstantial evidence in others suggest that the formation of a functional thymic microenvironment is controlled by members of the Foxn1/4 family of transcription factors. In teleost fishes, both Foxn1 and Foxn4 contribute to thymopoietic activity, whereas Foxn1 is both necessary and sufficient in the mammalian thymus. The evolutionary history of Foxn1/4 genes suggests that an ancient Foxn4 gene lineage gave rise to the Foxn1 genes in early vertebrates, raising the question of the thymopoietic capacity of the ancestor common to all vertebrates. Recent attempts to reconstruct the early events in the evolution of thymopoietic tissues by replacement of the mouse Foxn1 gene by Foxn1-like genes isolated from various chordate species suggest a plausible scenario. It appears that the primordial thymus was a bi-potent lymphoid organ, supporting both B cell and T cell development; however, during the course of vertebrate, evolution B cell development was gradually diminished converting the thymus into a site specialized in T cell development.


Subject(s)
Lymphopoiesis , Stem Cell Niche , Thymus Gland/cytology , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Receptors, Antigen/genetics , Receptors, Antigen/metabolism , Thymus Gland/immunology
11.
Nat Commun ; 11(1): 4505, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32908148

ABSTRACT

Evidence for transgenerational inheritance of epigenetic information in vertebrates is scarce. Aberrant patterns of DNA methylation in gametes may set the stage for transmission into future generations. Here, we describe a viable hypomorphic allele of dnmt1 in zebrafish that causes widespread demethylation of CpG dinucleotides in sperm and somatic tissues. We find that homozygous mutants are essentially normal, with the exception of drastically impaired lymphopoiesis, affecting both larval and adult phases of T cell development. The phenotype of impaired larval (but not adult) T cell development is transmitted to subsequent generations by genotypically wildtype fish. We further find that about 200 differentially methylated regions in sperm DNA of transmitting and non-transmitting males, including hypermethylated sites associated with runx3 and rptor genes, whose reduced activities are associated with impaired larval T cell development. Our results indicate a particular sensitivity of larval T cell development to transgenerationally inherited epimutations.


Subject(s)
Cell Differentiation/genetics , Genes, Recessive , Larva/growth & development , Lymphopoiesis/genetics , T-Lymphocytes/physiology , Alleles , Animals , Animals, Genetically Modified , Core Binding Factor Alpha 3 Subunit/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Epigenesis, Genetic , Female , Genetics , Larva/cytology , Male , Mutation , Regulatory-Associated Protein of mTOR/genetics , Spermatozoa/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
iScience ; 23(7): 101260, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32585597

ABSTRACT

DNA methylation is a universal epigenetic mechanism involved in regulation of gene expression and genome stability. The DNA maintenance methylase DNMT1 ensures that DNA methylation patterns are faithfully transmitted to daughter cells during cell division. Because loss of DNMT1 is lethal, a pan-organismic analysis of DNMT1 function is lacking. We identified new recessive dnmt1 alleles in medaka and zebrafish and, guided by the structures of mutant proteins, generated a recessive variant of mouse Dnmt1. Each of the three missense mutations studied here distorts the catalytic pocket and reduces enzymatic activity. Because all three DNMT1 mutant animals are viable, it was possible to examine their phenotypes throughout life. The consequences of genome-wide hypomethylation of DNA of somatic tissues in the Dnmt1 mutants are surprisingly mild but consistently affect the development of the lymphoid lineage. Our findings indicate that developing lymphocytes in vertebrates are sensitive to perturbations of DNA maintenance methylation.

13.
Proc Natl Acad Sci U S A ; 117(27): 15799-15808, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571908

ABSTRACT

The transcriptome of eukaryotic cells is constantly monitored for errors to avoid the production of undesired protein variants. The evolutionarily conserved nonsense-mediated mRNA decay (NMD) pathway degrades aberrant mRNAs, but also functions in the regulation of transcript abundance in response to changed physiological states. Here, we describe a zebrafish mutant of upf1, encoding the central component of the NMD machinery. Fish homozygous for the upf1t20450 allele (Y163X) survive until day 10 after fertilization, presenting with impaired T cell development as one of the most conspicuous features of the mutant phenotype. Analysis of differentially expressed genes identified dysregulation of the pre-mRNA splicing pathway, accompanied by perturbed autoregulation of canonical splicing activators (SRSF) and repressors (HNRNP). In upf1-deficient mutants, NMD-susceptible transcripts of ribosomal proteins that are known for their role as noncanonical splicing regulators were greatly increased, most notably, rpl10a When the levels of NMD-susceptible rpl10a transcripts were artificially increased in zebrafish larvae, T cell development was significantly impaired, suggesting that perturbed autoregulation of rpl10a splicing contributes to failing T cell development in upf1 deficiency. Our results identify an extraribosomal tissue-specific function to rpl10a in the immune system, and thus exemplify the advantages of the zebrafish model to study the effects of upf1-deficiency in the context of a vertebrate organism.


Subject(s)
Glutathione/analogs & derivatives , Nonsense Mediated mRNA Decay/genetics , RNA Splicing/genetics , RNA-Binding Proteins/genetics , T-Lymphocytes/immunology , Zebrafish Proteins/genetics , Animals , Codon, Nonsense/genetics , Fertilization/genetics , Gene Expression Regulation, Developmental/genetics , Glutathione/genetics , Homozygote , Humans , Nonsense Mediated mRNA Decay/immunology , RNA, Messenger/genetics , Transcription Factors/genetics , Transcriptome/genetics , Zebrafish/genetics
14.
Sci Immunol ; 5(45)2020 03 13.
Article in English | MEDLINE | ID: mdl-32169953

ABSTRACT

The antibodies of jawless vertebrates consist of leucine-rich repeat arrays encoded by somatically assembled VLRB genes. It is unknown how the incomplete germline VLRB loci are converted into functional antibody genes during B lymphocyte development in lampreys. In Lampetra planeri larvae lacking the cytidine deaminase CDA2 gene, VLRB assembly fails, whereas the T lineage-associated VLRA and VLRC antigen receptor gene assemblies occur normally. Thus, CDA2 acts in a B cell lineage-specific fashion to support the somatic diversification of VLRB antibody genes. CDA2 is closely related to activation-induced cytidine deaminase (AID), which is essential for the elaboration of immunoglobulin gene repertoires in jawed vertebrates. Our results thus identify a convergent mechanism of antigen receptor gene assembly and diversification that independently evolved in the two sister branches of vertebrates.


Subject(s)
Antibodies, Monoclonal/genetics , Cytidine Deaminase/genetics , Lampreys/genetics , Receptors, Antigen/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , Lampreys/immunology , Lampreys/metabolism , Receptors, Antigen/immunology , Receptors, Antigen/metabolism
15.
Proc Natl Acad Sci U S A ; 116(52): 26759-26767, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31822609

ABSTRACT

In mammals, T cell development critically depends on the IL-7 cytokine signaling pathway. Here we describe the identification of the zebrafish ortholog of mammalian IL-7 based on chromosomal localization, deduced protein sequence, and expression patterns. To examine the biological role of il7 in teleosts, we generated an il7 allele lacking most of its coding exons using CRISPR/Cas9-based mutagenesis. il7-deficient animals are viable and exhibit no obvious signs of immune disorder. With respect to intrathymic T cell development, il7 deficiency is associated with only a mild reduction of thymocyte numbers, contrasting with a more pronounced impairment of T cell development in il7r-deficient fish. Genetic interaction studies between il7 and il7r mutants, and il7 and crlf2(tslpr) mutants suggest the contribution of additional, as-yet unidentified cytokines to intrathymic T cell development. Such activities were also ascertained for other cytokines, such as il2 and il15, collectively indicating that in contrast to the situation in mammals, T cell development in the thymus of teleosts is driven by a degenerate multicomponent network of γc cytokines; this explains why deficiencies of single components have little detrimental effect. In contrast, the dependence on a single cytokine in the mammalian thymus has catastrophic consequences in cases of congenital deficiencies in genes affecting the IL-7 signaling pathway. We speculate that the transition from a degenerate to a nonredundant cytokine network supporting intrathymic T cell development emerged as a consequence of repurposing evolutionarily ancient constitutive cytokine pathways for regulatory functions in the mammalian peripheral immune system.

16.
Proc Natl Acad Sci U S A ; 115(14): E3211-E3220, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29555777

ABSTRACT

Cytidine deaminases of the AID/APOBEC family catalyze C-to-U nucleotide transitions in mRNA or DNA. Members of the APOBEC3 branch are involved in antiviral defense, whereas AID contributes to diversification of antibody repertoires in jawed vertebrates via somatic hypermutation, gene conversion, and class switch recombination. In the extant jawless vertebrate, the lamprey, two members of the AID/APOBEC family are implicated in the generation of somatic diversity of the variable lymphocyte receptors (VLRs). Expression studies linked CDA1 and CDA2 genes to the assembly of VLRA/C genes in T-like cells and the VLRB genes in B-like cells, respectively. Here, we identify and characterize several CDA1-like genes in the larvae of different lamprey species and demonstrate that these encode active cytidine deaminases. Structural comparisons of the CDA1 variants highlighted substantial differences in surface charge; this observation is supported by our finding that the enzymes require different conditions and substrates for optimal activity in vitro. Strikingly, we also found that the number of CDA-like genes present in individuals of the same species is variable. Nevertheless, irrespective of the number of different CDA1-like genes present, all lamprey larvae have at least one functional CDA1-related gene encoding an enzyme with predicted structural and chemical features generally comparable to jawed vertebrate AID. Our findings suggest that, similar to APOBEC3 branch expansion in jawed vertebrates, the AID/APOBEC family has undergone substantial diversification in lamprey, possibly indicative of multiple distinct biological roles.


Subject(s)
APOBEC-1 Deaminase/genetics , Cytidine Deaminase/classification , Cytidine Deaminase/genetics , DNA Copy Number Variations , Lampreys/genetics , Lymphocytes/immunology , Receptors, Antigen/genetics , APOBEC-1 Deaminase/chemistry , APOBEC-1 Deaminase/immunology , Amino Acid Sequence , Animals , Cytidine Deaminase/chemistry , Cytidine Deaminase/immunology , High-Throughput Nucleotide Sequencing , Protein Conformation , Receptors, Antigen/classification , Sequence Homology , Whole Genome Sequencing
17.
Annu Rev Immunol ; 36: 19-42, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29144837

ABSTRACT

Adaptive immunity in jawless fishes is based on antigen recognition by three types of variable lymphocyte receptors (VLRs) composed of variable leucine-rich repeats, which are differentially expressed by two T-like lymphocyte lineages and one B-like lymphocyte lineage. The T-like cells express either VLRAs or VLRCs of yet undefined antigen specificity, whereas the VLRB antibodies secreted by B-like cells bind proteinaceous and carbohydrate antigens. The incomplete VLR germline genes are assembled into functional units by a gene conversion-like mechanism that employs flanking variable leucine-rich repeat sequences as templates in association with lineage-specific expression of cytidine deaminases. B-like cells develop in the hematopoietic typhlosole and kidneys, whereas T-like cells develop in the thymoid, a thymus-equivalent region at the gill fold tips. Thus, the dichotomy between T-like and B-like cells and the presence of dedicated lymphopoietic tissues emerge as ancestral vertebrate features, whereas the somatic diversification of structurally distinct antigen receptor genes evolved independently in jawless and jawed vertebrates.


Subject(s)
Adaptive Immunity , Biological Evolution , Vertebrates/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Lineage , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Humans , Immunity, Innate , Multigene Family , Receptors, Antigen, B-Cell/chemistry , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vertebrates/metabolism
18.
Sci Rep ; 7: 44145, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266617

ABSTRACT

T cells are an evolutionarily conserved feature of the adaptive immune systems of vertebrates. Comparative studies using evolutionarily distant species hold great promise for unraveling the genetic landscape underlying this process. To this end, we used ENU mutagenesis to generate mutant zebrafish with specific aberrations in early T cell development. Here, we describe the identification of a recessive missense mutation in the transcriptional regulator zbtb17 (Q562K), which affects the ninth zinc finger module of the protein. Homozygous mutant fish exhibit an early block of intrathymic T cell development, as a result of impaired thymus colonization owing to reduced expression of the gene encoding the homing receptor ccr9a, and inefficient T cell differentiation owing to reduced expression of socs1a. Our results reveal the zbtb17-socs1 axis as an evolutionarily conserved central regulatory module of early T cell development of vertebrates.


Subject(s)
Cell Differentiation , Mutation, Missense , T-Lymphocytes/immunology , Transcription Factors , Zebrafish Proteins , Zebrafish , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , T-Lymphocytes/cytology , Transcription Factors/genetics , Transcription Factors/immunology , Zebrafish/genetics , Zebrafish/immunology , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology
19.
Cell Rep ; 17(9): 2259-2270, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27880902

ABSTRACT

Lymphocytes represent basic components of vertebrate adaptive immune systems, suggesting the utility of non-mammalian models to define the molecular basis of their development and differentiation. Our forward genetic screens in zebrafish for recessive mutations affecting early T cell development revealed several major genetic pathways. The identification of lineage-specific transcription factors and specific components of cytokine signaling and DNA replication and/or repair pathways known from studies of immunocompromised mammals provided an evolutionary cross-validation of the screen design. Unexpectedly, however, genes encoding proteins required for pre-mRNA processing were enriched in the collection of mutants identified here. In both zebrafish and mice, deficiency of the splice regulator TNPO3 impairs intrathymic T cell differentiation, illustrating the evolutionarily conserved and cell-type-specific functions of certain pre-mRNA-processing factors for T cell development.


Subject(s)
Genetic Testing , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Zebrafish/genetics , Alternative Splicing/genetics , Animals , Epistasis, Genetic , Gene Expression Regulation, Developmental , Larva/genetics , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Organ Specificity/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Transcriptome/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Karyopherins/deficiency , beta Karyopherins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL