Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(7)2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37047441

ABSTRACT

Primary immunodeficiency (PID) disorders, also commonly referred to as inborn errors of immunity, are a heterogenous group of human genetic diseases characterized by defects in immune cell development and/or function. Since these disorders are generally uncommon and occur on a variable background profile of potential genetic and environmental modifiers, animal models are critical to provide mechanistic insights as well as to create platforms to underpin therapeutic development. This review aims to review the relevance of zebrafish as an alternative genetic model for PIDs. It provides an overview of the conservation of the zebrafish immune system and details specific examples of zebrafish models for a multitude of specific human PIDs across a range of distinct categories, including severe combined immunodeficiency (SCID), combined immunodeficiency (CID), multi-system immunodeficiency, autoinflammatory disorders, neutropenia and defects in leucocyte mobility and respiratory burst. It also describes some of the diverse applications of these models, particularly in the fields of microbiology, immunology, regenerative biology and oncology.


Subject(s)
Immunologic Deficiency Syndromes , Pelvic Inflammatory Disease , Primary Immunodeficiency Diseases , Severe Combined Immunodeficiency , Female , Animals , Humans , Zebrafish/genetics , Models, Genetic , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Primary Immunodeficiency Diseases/genetics , Severe Combined Immunodeficiency/genetics
2.
Methods Mol Biol ; 2580: 355-377, 2023.
Article in English | MEDLINE | ID: mdl-36374469

ABSTRACT

While the zebrafish has for some time been regarded as a powerful model organism with which to study early events in hematopoiesis, recent evidence suggests that it also ideal for unraveling the molecular requirements for T cell development in the thymus. Like mammals, zebrafish possess an adaptive immune system, comprising B lymphocytes as well as both the γδ and αß lineages of T cells, which develop in the thymus. Moreover, the molecular processes underlying T cell development in zebrafish appear to be remarkably conserved. Thus, findings in the zebrafish model will be of high relevance to the equivalent processes in mammals. Finally, molecular processes can be interrogated in zebrafish far more rapidly than is possible in mammals because the zebrafish possesses many unique advantages. Here, we describe these unique attributes and the methods by which they can be exploited to investigate the role of novel genes in T cell development.


Subject(s)
Hematopoiesis , Zebrafish , Animals , Zebrafish/genetics , Cell Differentiation , Hematopoiesis/genetics , T-Lymphocytes , Mammals
3.
Front Immunol ; 13: 928252, 2022.
Article in English | MEDLINE | ID: mdl-35967429

ABSTRACT

Newborn screening for severe combined immunodeficiency (SCID) has not only accelerated diagnosis and improved treatment for affected infants, but also led to identification of novel genes required for human T cell development. A male proband had SCID newborn screening showing very low T cell receptor excision circles (TRECs), a biomarker for thymic output of nascent T cells. He had persistent profound T lymphopenia, but normal numbers of B and natural killer (NK) cells. Despite an allogeneic hematopoietic stem cell transplant from his brother, he failed to develop normal T cells. Targeted resequencing excluded known SCID genes; however, whole exome sequencing (WES) of the proband and parents revealed a maternally inherited X-linked missense mutation in MED14 (MED14V763A), a component of the mediator complex. Morpholino (MO)-mediated loss of MED14 function attenuated T cell development in zebrafish. Moreover, this arrest was rescued by ectopic expression of cDNA encoding the wild type human MED14 ortholog, but not by MED14V763A , suggesting that the variant impaired MED14 function. Modeling of the equivalent mutation in mouse (Med14V769A) did not disrupt T cell development at baseline. However, repopulation of peripheral T cells upon competitive bone marrow transplantation was compromised, consistent with the incomplete T cell reconstitution experienced by the proband upon transplantation with bone marrow from his healthy male sibling, who was found to have the same MED14V763A variant. Suspecting that the variable phenotypic expression between the siblings was influenced by further mutation(s), we sought to identify genetic variants present only in the affected proband. Indeed, WES revealed a mutation in the L1 cell adhesion molecule (L1CAMQ498H); however, introducing that mutation in vivo in mice did not disrupt T cell development. Consequently, immunodeficiency in the proband may depend upon additional, unidentified gene variants.


Subject(s)
Lymphopenia , Severe Combined Immunodeficiency , Animals , Humans , Infant , Infant, Newborn , Lymphopenia/genetics , Male , Mice , Neonatal Screening , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , T-Lymphocytes , Zebrafish
4.
Front Biosci (Landmark Ed) ; 27(4): 110, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35468669

ABSTRACT

BACKGROUND: Colony-stimulating factor 3 (CSF3), more commonly known as granulocyte colony-stimulating factor (G-CSF), acts via a specific cell surface receptor CSF3R (or G-CSFR) to regulate hematopoiesis, with a particularly key role in the myeloid cell lineage where it impacts the development and function of neutrophilic granulocytes. Zebrafish possess a conserved CSF3R homologue, Csf3r, which is involved in both steady-state and emergency myelopoiesis, as well as regulating early myeloid cell migration. Two CSF3 proteins have been identified in zebrafish, Csf3a and Csf3b. METHODS: This study investigated the roles of the Csf3a and Csf3b ligands as well as the downstream Janus kinase (JAK) and phosphatidylinositol 3-kinase (PI3K) pathways in mediating the effects of Csf3r in early myeloid cell development and function using gene knockdown and pharmacologic approaches. RESULTS: This study revealed that both Csf3a and Csf3b contribute to the developmental and emergency production of early myeloid cells, but Csf3a is responsible for the developmental migration of early neutrophils whereas Csf3b plays the major role in their wounding-induced migration, differentially participated in these responses, as did several downstream signaling pathways. Both JAK and PI3K signaling were required for developmental production and migration of early myeloid cells, but PI3K signaling was required for emergency production and initial migration in response to wounding, while JAK signaling mediated retention at the site of wounding. CONCLUSIONS: This study has revealed both distinct and overlapping functions for Csf3a and Csf3b and the downstream JAK and PI3K signaling pathways in early myeloid cell production and function.


Subject(s)
Phosphatidylinositol 3-Kinases , Zebrafish , Animals , Granulocyte Colony-Stimulating Factor/genetics , Janus Kinases/metabolism , Myeloid Cells , Phosphatidylinositol 3-Kinases/metabolism , Zebrafish/genetics , Zebrafish/metabolism
5.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216498

ABSTRACT

The IL-2 family of cytokines act via receptor complexes that share the interleukin-2 receptor gamma common (IL-2Rγc) chain to play key roles in lymphopoiesis. Inactivating IL-2Rγc mutations results in severe combined immunodeficiency (SCID) in humans and other species. This study sought to generate an equivalent zebrafish SCID model. The zebrafish il2rga gene was targeted for genome editing using TALENs and presumed loss-of-function alleles analyzed with respect to immune cell development and impacts on intestinal microbiota and tumor immunity. Knockout of zebrafish Il-2rγc.a resulted in a SCID phenotype, including a significant reduction in T cells, with NK cells also impacted. This resulted in dysregulated intestinal microbiota and defective immunity to tumor xenotransplants. Collectively, this establishes a useful zebrafish SCID model.


Subject(s)
Severe Combined Immunodeficiency/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Base Sequence , Gastrointestinal Microbiome/physiology , Interleukin Receptor Common gamma Subunit , Killer Cells, Natural/metabolism , Lymphopoiesis/physiology , Models, Animal , Phenotype , T-Lymphocytes/metabolism
6.
J Immunol ; 208(4): 870-880, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35046107

ABSTRACT

Ribosomal proteins are thought to primarily facilitate biogenesis of the ribosome and its ability to synthesize protein. However, in this study, we show that Rpl22-like1 (Rpl22l1) regulates hematopoiesis without affecting ribosome biogenesis or bulk protein synthesis. Conditional loss of murine Rpl22l1 using stage or lineage-restricted Cre drivers impairs development of several hematopoietic lineages. Specifically, Tie2-Cre-mediated ablation of Rpl22l1 in hemogenic endothelium impairs the emergence of embryonic hematopoietic stem cells. Ablation of Rpl22l1 in late fetal liver progenitors impairs the development of B lineage progenitors at the pre-B stage and development of T cells at the CD44-CD25+ double-negative stage. In vivo labeling with O-propargyl-puromycin revealed that protein synthesis at the stages of arrest was not altered, indicating that the ribosome biogenesis and function were not generally compromised. The developmental arrest was associated with p53 activation, suggesting that the arrest may be p53-dependent. Indeed, development of both B and T lymphocytes was rescued by p53 deficiency. p53 induction was not accompanied by DNA damage as indicated by phospho-γH2AX induction or endoplasmic reticulum stress, as measured by phosphorylation of EIF2α, thereby excluding the known likely p53 inducers as causal. Finally, the developmental arrest of T cells was not rescued by elimination of the Rpl22l1 paralog, Rpl22, as we had previously found for the emergence of hematopoietic stem cells. This indicates that Rpl22 and Rpl22l1 play distinct and essential roles in supporting B and T cell development.


Subject(s)
Cell Differentiation/genetics , Lymphopoiesis/genetics , Protein Biosynthesis , Ribosomal Proteins/deficiency , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Lineage/genetics , Cell Lineage/immunology , Cell Plasticity/genetics , Cell Plasticity/immunology , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Front Immunol ; 12: 788278, 2021.
Article in English | MEDLINE | ID: mdl-34887873

ABSTRACT

B-cell lymphoma/leukemia 11B (BCL11B) is a C2H2 zinc finger transcription factor that is critically important for regulating the development and function of a variety of systems including the central nervous system, the skin, and the immune system. Germline heterozygous variants are associated with a spectrum of clinical disorders, including severe combined immunodeficiency as well as neurological, craniofacial, and dermal defects. Of these individuals, ~50% present with severe allergic disease. Here, we report the detailed clinical and laboratory workup of one of the most severe BCL11B-dependent atopic cases to date. Leveraging a zebrafish model, we were able to confirm a strong T-cell defect in the patient. Based on these data, we classify germline BCL11B-dependent atopic disease as a novel primary atopic disorder.


Subject(s)
Germ-Line Mutation , Hypersensitivity/genetics , Primary Immunodeficiency Diseases/genetics , Repressor Proteins/genetics , T-Lymphocytes/immunology , Tumor Suppressor Proteins/genetics , Adolescent , Animals , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Hypersensitivity/diagnosis , Hypersensitivity/immunology , Phenotype , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/metabolism , Repressor Proteins/metabolism , Severity of Illness Index , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
J Immunol ; 197(1): 11-8, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27317733

ABSTRACT

Cytokines represent essential mediators of cell-cell communication with particularly important roles within the immune system. These secreted factors are produced in response to developmental and/or environmental cues and act via cognate cytokine receptors on target cells, stimulating specific intracellular signaling pathways to facilitate appropriate cellular responses. This review describes the evolution of cytokine receptor signaling, focusing on the class I and class II receptor families and the downstream JAK-STAT pathway along with its key negative regulators. Individual components generated over a long evolutionary time frame coalesced to form an archetypal signaling pathway in bilateria that was expanded extensively during early vertebrate evolution to establish a substantial "core" signaling network, which has subsequently undergone limited diversification within discrete lineages. The evolution of cytokine receptor signaling parallels that of the immune system, particularly the emergence of adaptive immunity, which has likely been a major evolutionary driver.


Subject(s)
Biological Evolution , Cytokines/immunology , Receptors, Cytokine/immunology , Signal Transduction , Suppressor of Cytokine Signaling Proteins/immunology , Adaptive Immunity , Animals , Humans , Signal Transduction/immunology
9.
J Immunol ; 196(1): 135-43, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26590317

ABSTRACT

The IL-2 receptor γ common (IL-2Rγc) chain is the shared subunit of the receptors for the IL-2 family of cytokines, which mediate signaling through JAK3 and various downstream pathways to regulate lymphopoiesis. Inactivating mutations in human IL-2Rγc result in SCID, a primary immunodeficiency characterized by greatly reduced numbers of lymphocytes. This study used bioinformatics, expression analysis, gene ablation, and specific pharmacologic inhibitors to investigate the function of two putative zebrafish IL-2Rγc paralogs, il-2rγc.a and il-2rγc.b, and downstream signaling components during early lymphopoiesis. Expression of il-2rγc.a commenced at 16 h post fertilization (hpf) and rose steadily from 4-6 d postfertilization (dpf) in the developing thymus, with il-2rγc.a expression also confirmed in adult T and B lymphocytes. Transcripts of il-2rγc.b were first observed from 8 hpf, but waned from 16 hpf before reaching maximal expression at 6 dpf, but this was not evident in the thymus. Knockdown of il-2rγc.a, but not il-2rγc.b, substantially reduced embryonic lymphopoiesis without affecting other aspects of hematopoiesis. Specific targeting of zebrafish Jak3 exerted a similar effect on lymphopoiesis, whereas ablation of zebrafish Stat5.1 and pharmacologic inhibition of PI3K and MEK also produced significant but smaller effects. Ablation of il-2rγc.a was further demonstrated to lead to an absence of mature T cells, but not B cells in juvenile fish. These results indicate that conserved IL-2Rγc signaling via JAK3 plays a key role during early zebrafish lymphopoiesis, which can be potentially targeted to generate a zebrafish model of human SCID.


Subject(s)
Janus Kinase 3/genetics , Lymphopoiesis/immunology , Receptors, Interleukin-2/immunology , Severe Combined Immunodeficiency/immunology , Zebrafish Proteins/immunology , Animals , B-Lymphocytes/immunology , Cell Line , Disease Models, Animal , Gene Knockdown Techniques , HEK293 Cells , Humans , Lymphopoiesis/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , Morpholinos/genetics , Phosphoinositide-3 Kinase Inhibitors , Receptors, Interleukin-2/genetics , STAT5 Transcription Factor/genetics , Severe Combined Immunodeficiency/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , Zebrafish , Zebrafish Proteins/genetics
10.
Brief Funct Genomics ; 15(4): 322-30, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26654901

ABSTRACT

Zebrafish is a powerful model for the study of vertebrate development, being amenable to a wide range of genetic and other manipulations to probe the molecular basis of development and its perturbation in disease. Over recent years, genome editing approaches have become increasingly used as an efficient and sophisticated approach to precisely engineer the zebrafish genome, which has further enhanced the utility of this organism. This review provides a practical overview of genome editing and its application in zebrafish research, including alternate strategies for introducing and screening for specific genetic changes.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Zebrafish/genetics , Animals , Genome
11.
J Immunol ; 192(12): 5739-48, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24835394

ABSTRACT

Cytokine-inducible SH2 domain-containing protein (CISH), a member of the suppressor of cytokine signaling family of negative feedback regulators, is induced by cytokines that activate STAT5 and can inhibit STAT5 signaling in vitro. However, demonstration of a definitive in vivo role for CISH during development has remained elusive. This study employed expression analysis and morpholino-mediated knockdown in zebrafish in concert with bioinformatics and biochemical approaches to investigate CISH function. Two zebrafish CISH paralogs were identified, cish.a and cish.b, with high overall conservation (43-46% identity) with their mammalian counterparts. The cish.a gene was maternally derived, with transcripts present throughout embryogenesis, and increasing at 4-5 d after fertilization, whereas cish.b expression commenced at 8 h after fertilization. Expression of cish.a was regulated by the JAK2/STAT5 pathway via conserved tetrameric STAT5 binding sites (TTCN3GAA) in its promoter. Injection of morpholinos targeting cish.a, but not cish.b or control morpholinos, resulted in enhanced embryonic erythropoiesis, myelopoiesis, and lymphopoiesis, including a 2- 3-fold increase in erythrocytic markers. This occurred concomitantly with increased activation of STAT5. This study indicates that CISH functions as a conserved in vivo target and regulator of STAT5 in the control of embryonic hematopoiesis.


Subject(s)
Embryo, Nonmammalian/immunology , Hematopoiesis/immunology , STAT5 Transcription Factor/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Zebrafish Proteins/immunology , Zebrafish/immunology , Animals , Base Sequence , Hematopoiesis/genetics , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Molecular Sequence Data , STAT5 Transcription Factor/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...