Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Dev Cell ; 59(1): 91-107.e6, 2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38091997

RESUMO

Genomic regulation of cardiomyocyte differentiation is central to heart development and function. This study uses genetic loss-of-function human-induced pluripotent stem cell-derived cardiomyocytes to evaluate the genomic regulatory basis of the non-DNA-binding homeodomain protein HOPX. We show that HOPX interacts with and controls cardiac genes and enhancer networks associated with diverse aspects of heart development. Using perturbation studies in vitro, we define how upstream cell growth and proliferation control HOPX transcription to regulate cardiac gene programs. We then use cell, organoid, and zebrafish regeneration models to demonstrate that HOPX-regulated gene programs control cardiomyocyte function in development and disease. Collectively, this study mechanistically links cell signaling pathways as upstream regulators of HOPX transcription to control gene programs underpinning cardiomyocyte identity and function.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Animais , Humanos , Miócitos Cardíacos/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Peixe-Zebra/metabolismo , Diferenciação Celular/genética , Proliferação de Células
2.
Dev Cell ; 58(3): 224-238.e7, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36693371

RESUMO

Endothelial cells (ECs) line blood vessels and serve as a niche for hematopoietic stem and progenitor cells (HSPCs). Recent data point to tissue-specific EC specialization as well as heterogeneity; however, it remains unclear how ECs acquire these properties. Here, by combining live-imaging-based lineage-tracing and single-cell transcriptomics in zebrafish embryos, we identify an unexpected origin for part of the vascular HSPC niche. We find that islet1 (isl1)-expressing cells are the progenitors of the venous ECs that constitute the majority of the HSPC niche. These isl1-expressing cells surprisingly originate from the endoderm and differentiate into ECs in a process dependent on Bmp-Smad signaling and subsequently requiring npas4l (cloche) function. Single-cell RNA sequencing analyses show that isl1-derived ECs express a set of genes that reflect their distinct origin. This study demonstrates that endothelial specialization in the HSPC niche is determined at least in part by the origin of the ECs.


Assuntos
Células Endoteliais , Peixe-Zebra , Animais , Endoderma , Células-Tronco Hematopoéticas/fisiologia , Endotélio
3.
Front Immunol ; 13: 981000, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36059461

RESUMO

The role of T cells in appendage regeneration remains unclear. In this study, we revealed an important role for regulatory T cells (Tregs), a subset of T cells that regulate tolerance and tissue repair, in the epimorphic regeneration of zebrafish caudal fin tissue. Upon amputation, fin tissue-resident Tregs infiltrate into the blastema, a population of progenitor cells that produce new fin tissues. Conditional genetic ablation of Tregs attenuates blastemal cell proliferation during fin regeneration. Blastema-infiltrating Tregs upregulate the expression of igf2a and igf2b, and pharmacological activation of IGF signaling restores blastemal proliferation in Treg-ablated zebrafish. These findings further extend our understandings of Treg function in tissue regeneration and repair.


Assuntos
Linfócitos T Reguladores , Peixe-Zebra , Animais , Proliferação de Células , Transdução de Sinais/genética , Linfócitos T Reguladores/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
4.
Zebrafish ; 19(1): 7-17, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35108124

RESUMO

Heme oxygenase function is highly conserved between vertebrates where it plays important roles in normal embryonic development and controls oxidative stress. Expression of the zebrafish heme oxygenase 1 genes is known to be responsive to oxidative stress suggesting a conserved physiological function. In this study, we generate a knockout allele of zebrafish hmox1a and characterize the effects of hmox1a and hmox1b loss on embryonic development. We find that loss of hmox1a or hmox1b causes developmental defects in only a minority of embryos, in contrast to Hmox1 gene deletions in mice that cause loss of most embryos. Using a tail wound inflammation assay we find a conserved role for hmox1a, but not hmox1b, in normal macrophage migration to the wound site. Together our results indicate that zebrafish hmox1a has clearly a partitioned role from hmox1b that is more consistent with conserved functions of mammalian Heme oxygenase 1.


Assuntos
Heme Oxigenase (Desciclizante) , Peixe-Zebra , Animais , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase (Desciclizante)/farmacologia , Macrófagos/metabolismo , Camundongos , Estresse Oxidativo , Peixe-Zebra/metabolismo
5.
Nat Commun ; 13(1): 952, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177649

RESUMO

Prevalence of Mycobacterium abscessus infections is increasing in patients with respiratory comorbidities. After initial colonisation, M. abscessus smooth colony (S) variants can undergo an irreversible genetic switch into highly inflammatory, rough colony (R) variants, often associated with a decline in pulmonary function. Here, we use an adult zebrafish model of chronic infection with R and S variants to study M. abscessus pathogenesis in the context of fully functioning host immunity. We show that infection with an R variant causes an inflammatory immune response that drives necrotic granuloma formation through host TNF signalling, mediated by the tnfa, tnfr1 and tnfr2 gene products. T cell-dependent immunity is stronger against the R variant early in infection, and regulatory T cells associate with R variant granulomas and limit bacterial growth. In comparison, an S variant proliferates to high burdens but appears to be controlled by TNF-dependent innate immunity early during infection, resulting in delayed granuloma formation. Thus, our work demonstrates the applicability of adult zebrafish to model persistent M. abscessus infection, and illustrates differences in the immunopathogenesis induced by R and S variants during granulomatous infection.


Assuntos
Granuloma/imunologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium abscessus/patogenicidade , Infecção Persistente/imunologia , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Granuloma/microbiologia , Granuloma/patologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Ativação Linfocitária , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/imunologia , Infecção Persistente/microbiologia , Infecção Persistente/patologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
6.
FEBS J ; 289(3): 671-681, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34544203

RESUMO

Iron homeostasis is essential for both sides of the host-pathogen interface. Restricting access of iron slows bacterial growth while iron is also a necessary cofactor for host immunity. Haem oxygenase 1 (HMOX1) is a critical regulator of iron homeostasis that catalyses the liberation of iron during degradation of haem. It is also a stress-responsive protein that can be rapidly upregulated and confers protection to the host. Although a protective role of HMOX1 has been demonstrated in a variety of diseases, the role of HMOX1 in Mycobacterium tuberculosis infection is equivocal across experiments with different host-pathogen combinations. Here, we use the natural host-pathogen pairing of the zebrafish-Mycobacterium marinum infection platform to study the role of zebrafish haem oxygenase in mycobacterial infection. We identify zebrafish Hmox1a as the relevant functional paralog of mammalian HMOX1 and demonstrate a conserved role for Hmox1a in protecting the host from M. marinum infection. Using genetic and chemical tools, we show zebrafish Hmox1a protects the host against M. marinum infection by reducing infection-induced iron accumulation and ferrostatin-sensitive cell death.


Assuntos
Heme Oxigenase-1/genética , Ferro/metabolismo , Tuberculose/genética , Proteínas de Peixe-Zebra/genética , Animais , Morte Celular/genética , Cicloexilaminas/metabolismo , Modelos Animais de Doenças , Heme/genética , Homeostase , Interações Hospedeiro-Patógeno/genética , Humanos , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Fenilenodiaminas/metabolismo , Tuberculose/microbiologia , Peixe-Zebra/genética , Peixe-Zebra/microbiologia
8.
Cell Rep ; 37(1): 109775, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34610312

RESUMO

Motile cilia defects impair cerebrospinal fluid (CSF) flow and can cause brain and spine disorders. The development of ciliated cells, their impact on CSF flow, and their function in brain and axial morphogenesis are not fully understood. We have characterized motile ciliated cells within the zebrafish brain ventricles. We show that the ventricles undergo restructuring through development, involving a transition from mono- to multiciliated cells (MCCs) driven by gmnc. MCCs co-exist with monociliated cells and generate directional flow patterns. These ciliated cells have different developmental origins and are genetically heterogenous with respect to expression of the Foxj1 family of ciliary master regulators. Finally, we show that cilia loss from the tela choroida and choroid plexus or global perturbation of multiciliation does not affect overall brain or spine morphogenesis but results in enlarged ventricles. Our findings establish that motile ciliated cells are generated by complementary and sequential transcriptional programs to support ventricular development.


Assuntos
Encéfalo/metabolismo , Cílios/metabolismo , Epêndima/metabolismo , Animais , Animais Geneticamente Modificados/metabolismo , Encéfalo/citologia , Encéfalo/patologia , Linhagem da Célula , Líquido Cefalorraquidiano/fisiologia , Cílios/patologia , Embrião não Mamífero/metabolismo , Epêndima/citologia , Epêndima/patologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Edição de Genes , Morfogênese , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coluna Vertebral/crescimento & desenvolvimento , Coluna Vertebral/metabolismo , Telencéfalo/citologia , Telencéfalo/metabolismo , Telencéfalo/patologia , Tubulina (Proteína)/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Science ; 372(6538): 201-205, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33833125

RESUMO

Cardiac regeneration requires dedifferentiation and proliferation of mature cardiomyocytes, but the mechanisms underlying this plasticity remain unclear. Here, we identify a potent cardiomyogenic role for Krüppel-like factor 1 (Klf1/Eklf), which is induced in adult zebrafish myocardium upon injury. Myocardial inhibition of Klf1 function does not affect heart development, but it severely impairs regeneration. Transient Klf1 activation is sufficient to expand mature myocardium in uninjured hearts. Klf1 directs epigenetic reprogramming of the cardiac transcription factor network, permitting coordinated cardiomyocyte dedifferentiation and proliferation. Myocardial expansion is supported by Klf1-induced rewiring of mitochondrial metabolism from oxidative respiration to anabolic pathways. Our findings establish Klf1 as a core transcriptional regulator of cardiomyocyte renewal in adult zebrafish hearts.


Assuntos
Reprogramação Celular , Coração/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Miócitos Cardíacos/fisiologia , Regeneração , Proteínas de Peixe-Zebra/metabolismo , Animais , Cardiomegalia Induzida por Exercícios , Desdiferenciação Celular , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Glicólise , Coração/embriologia , Ventrículos do Coração/citologia , Fatores de Transcrição Kruppel-Like/genética , Desenvolvimento Muscular , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Via de Pentose Fosfato , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
10.
Methods Mol Biol ; 2158: 63-69, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32857366

RESUMO

The zebrafish (Danio rerio) possesses a spectacular capacity for cardiac regeneration. Zebrafish have been used in cardiac regeneration research for nearly two decades, contributing to the identification of signals and cellular mechanisms as potential targets for human heart repair. Investigations into cardiac regeneration in zebrafish have been facilitated by multiple methods of inducing cardiac tissue damage. Among the established methods, cardiac resection injury is a relatively simple, yet robust approach traditionally used to induce cardiac tissue damage in a reproducible manner. Here, we describe a detailed protocol to perform a cardiac resection injury in adult zebrafish and discuss potential complications for researchers who are new to this technique.


Assuntos
Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Modelos Animais de Doenças , Traumatismos Cardíacos/patologia , Coração/fisiologia , Remodelação Ventricular , Animais , Proliferação de Células , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/cirurgia , Peixe-Zebra
11.
Sci Rep ; 10(1): 19085, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33154446

RESUMO

Germline loss-of-function variation in TNFAIP3, encoding A20, has been implicated in a wide variety of autoinflammatory and autoimmune conditions, with acquired somatic missense mutations linked to cancer progression. Furthermore, human sequence data reveals that the A20 locus contains ~ 400 non-synonymous coding variants, which are largely uncharacterised. The growing number of A20 coding variants with unknown function, but potential clinical impact, poses a challenge to traditional mouse-based approaches. Here we report the development of a novel functional genomics approach that utilizes a new A20-deficient zebrafish (Danio rerio) model to investigate the impact of TNFAIP3 genetic variants in vivo. A20-deficient zebrafish are hyper-responsive to microbial immune activation and exhibit spontaneous early lethality. Ectopic addition of human A20 rescued A20-null zebrafish from lethality, while missense mutations at two conserved A20 residues, S381A and C243Y, reversed this protective effect. Ser381 represents a phosphorylation site important for enhancing A20 activity that is abrogated by its mutation to alanine, or by a causal C243Y mutation that triggers human autoimmune disease. These data reveal an evolutionarily conserved role for TNFAIP3 in limiting inflammation in the vertebrate linage and show how this function is controlled by phosphorylation. They also demonstrate how a zebrafish functional genomics pipeline can be utilized to investigate the in vivo significance of medically relevant human TNFAIP3 gene variants.


Assuntos
Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/genética , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados , Doenças Autoimunes/etiologia , Doenças Autoimunes/genética , Sequência Conservada , Evolução Molecular , Variação Genética , Humanos , Inflamação/etiologia , Inflamação/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Modelos Animais , Modelos Genéticos , Mutação de Sentido Incorreto , NF-kappa B/metabolismo , Fosforilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/deficiência , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/deficiência
12.
Science ; 370(6517)2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-33154111

RESUMO

Interactions of transcription factors (TFs) with DNA regulatory sequences, known as enhancers, specify cell identity during animal development. Unlike TFs, the origin and evolution of enhancers has been difficult to trace. We drove zebrafish and mouse developmental transcription using enhancers from an evolutionarily distant marine sponge. Some of these sponge enhancers are located in highly conserved microsyntenic regions, including an Islet enhancer in the Islet-Scaper region. We found that Islet enhancers in humans and mice share a suite of TF binding motifs with sponges, and that they drive gene expression patterns similar to those of sponge and endogenous Islet enhancers in zebrafish. Our results suggest the existence of an ancient and conserved, yet flexible, genomic regulatory syntax that has been repeatedly co-opted into cell type-specific gene regulatory networks across the animal kingdom.


Assuntos
Sequência Conservada , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Homeodomínio LIM/metabolismo , Poríferos/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Imunoprecipitação da Cromatina , Humanos , Camundongos , Peixe-Zebra/genética
13.
J Clin Invest ; 130(8): 4006-4018, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32568216

RESUMO

Ligand-dependent activation of Hedgehog (Hh) signaling in cancer occurs without mutations in canonical pathway genes. Consequently, the genetic basis of Hh pathway activation in adult solid tumors, such as small-cell lung cancer (SCLC), is unknown. Here we show that combined inactivation of Trp53 and Rb1, a defining genetic feature of SCLC, leads to hypersensitivity to Hh ligand in vitro, and during neural tube development in vivo. This response is associated with the aberrant formation of primary cilia, an organelle essential for canonical Hh signaling through smoothened, a transmembrane protein targeted by small-molecule Hh inhibitors. We further show that loss of both Trp53 and Rb1 disables transcription of genes in the autophagic machinery necessary for the degradation of primary cilia. In turn, we also demonstrate a requirement for Kif3a, a gene essential for the formation of primary cilia, in a mouse model of SCLC induced by conditional deletion of both Trp53 and Rb1 in the adult airway. Our results provide a mechanistic framework for therapeutic targeting of ligand-dependent Hh signaling in human cancers with somatic mutations in both TP53 and RB1.


Assuntos
Autofagia , Proteínas Hedgehog/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas de Ligação a Retinoblastoma/metabolismo , Transdução de Sinais , Carcinoma de Pequenas Células do Pulmão/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Hedgehog/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Proteínas de Ligação a Retinoblastoma/genética , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/patologia , Proteína Supressora de Tumor p53/genética
14.
Curr Opin Immunol ; 63: 7-13, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31765917

RESUMO

Zebrafish can efficiently regenerate complex tissue structures with a highly developed innate and adaptive immune system, which provides a model to investigate the roles of immune cells in tissue repair and regeneration. Two groups recently reported zebrafish mutants deficient in a forkhead box P3 (FOXP3) ortholog, which helped reveal the conserved immunosuppressive function of zebrafish FOXP3 in vivo. Zebrafish FOXP3 defines the development of a subset of T cell lineage with the conserved gene expression profile of mammalian regulatory T cells (Tregs). In damaged organs, zebrafish Tregs rapidly migrate to the injury site, where they promote the proliferation of regeneration precursor cells by producing tissue-specific regenerative factors through a distinct mechanism from the canonical anti-inflammatory pathway. These findings illuminate the potential for using zebrafish as an effective model in Treg research and demonstrate organ-specific roles for Tregs in maintaining proregenerative capacity that could potentially be harnessed for use in diverse regeneration therapies.


Assuntos
Organogênese/imunologia , Regeneração/imunologia , Linfócitos T Reguladores/imunologia , Peixe-Zebra/imunologia , Peixe-Zebra/fisiologia , Animais , Fatores de Transcrição Forkhead/imunologia , Proteínas de Peixe-Zebra/imunologia
15.
Int J Mol Sci ; 20(20)2019 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-31635133

RESUMO

Regenerative capacity varies greatly between species. Mammals are limited in their ability to regenerate damaged cells, tissues and organs compared to organisms with robust regenerative responses, such as zebrafish. The regeneration of zebrafish tissues including the heart, spinal cord and retina requires foxp3a+ zebrafish regulatory T cells (zTregs). However, it remains unclear whether the muted regenerative responses in mammals are due to impaired recruitment and/or function of homologous mammalian regulatory T cell (Treg) populations. Here, we explore the possibility of enhancing zTreg recruitment with pharmacological interventions using the well-characterized zebrafish tail amputation model to establish a high-throughput screening platform. Injury-infiltrating zTregs were transgenically labelled to enable rapid quantification in live animals. We screened the NIH Clinical Collection (727 small molecules) for modulators of zTreg recruitment to the regenerating tissue at three days post-injury. We discovered that the dopamine agonist pramipexole, a drug currently approved for treating Parkinson's Disease, specifically enhanced zTreg recruitment after injury. The dopamine antagonist SCH-23390 blocked pramipexole activity, suggesting that peripheral dopaminergic signaling may regulate zTreg recruitment. Similar pharmacological approaches for enhancing mammalian Treg recruitment may be an important step in developing novel strategies for tissue regeneration in humans.


Assuntos
Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Regeneração , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/fisiologia , Peixe-Zebra/fisiologia , Animais , Dopamina/metabolismo , Pramipexol/farmacologia , Transdução de Sinais
16.
NPJ Regen Med ; 3: 6, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29507774

RESUMO

Stimulating regeneration of complex tissues and organs after injury to effect complete structural and functional repair, is an attractive therapeutic option that would revolutionize clinical medicine. Compared to many metazoan phyla that show extraordinary regenerative capacity, which in some instances persists throughout life, regeneration in mammalians, particularly humans, is limited or absent. Here we consider recent insights in the elucidation of molecular mechanisms of regeneration that have come from studies of tissue homeostasis and injury repair in mammalian tissues that span the spectrum from little or no self-renewal, to those showing active cell turnover throughout life. These studies highlight the diversity of factors that constrain regeneration, including immune responses, extracellular matrix composition, age, injury type, physiological adaptation, and angiogenic and neurogenic capacity. Despite these constraints, much progress has been made in elucidating key molecular mechanisms that may provide therapeutic targets for the development of future regenerative therapies, as well as previously unidentified developmental paradigms and windows-of-opportunity for improved regenerative repair.

17.
Dev Cell ; 43(6): 659-672.e5, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29257949

RESUMO

The attenuation of ancestral pro-regenerative pathways may explain why humans do not efficiently regenerate damaged organs. Vertebrate lineages that exhibit robust regeneration, including the teleost zebrafish, provide insights into the maintenance of adult regenerative capacity. Using established models of spinal cord, heart, and retina regeneration, we discovered that zebrafish Treg-like (zTreg) cells rapidly homed to damaged organs. Conditional ablation of zTreg cells blocked organ regeneration by impairing precursor cell proliferation. In addition to modulating inflammation, infiltrating zTreg cells stimulated regeneration through interleukin-10-independent secretion of organ-specific regenerative factors (Ntf3: spinal cord; Nrg1: heart; Igf1: retina). Recombinant regeneration factors rescued the regeneration defects associated with zTreg cell depletion, whereas Foxp3a-deficient zTreg cells infiltrated damaged organs but failed to express regenerative factors. Our data delineate organ-specific roles for Treg cells in maintaining pro-regenerative capacity that could potentially be harnessed for diverse regenerative therapies.


Assuntos
Regeneração/fisiologia , Linfócitos T Reguladores/fisiologia , Peixe-Zebra/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Coração/fisiologia , Modelos Animais , Regeneração Nervosa/fisiologia , Organogênese/imunologia , Organogênese/fisiologia , Retina/fisiologia , Medula Espinal/fisiologia , Proteínas de Peixe-Zebra/fisiologia
18.
N Engl J Med ; 377(6): 544-552, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28792876

RESUMO

BACKGROUND: Congenital malformations can be manifested as combinations of phenotypes that co-occur more often than expected by chance. In many such cases, it has proved difficult to identify a genetic cause. We sought the genetic cause of cardiac, vertebral, and renal defects, among others, in unrelated patients. METHODS: We used genomic sequencing to identify potentially pathogenic gene variants in families in which a person had multiple congenital malformations. We tested the function of the variant by using assays of in vitro enzyme activity and by quantifying metabolites in patient plasma. We engineered mouse models with similar variants using the CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 system. RESULTS: Variants were identified in two genes that encode enzymes of the kynurenine pathway, 3-hydroxyanthranilic acid 3,4-dioxygenase (HAAO) and kynureninase (KYNU). Three patients carried homozygous variants predicting loss-of-function changes in the HAAO or KYNU proteins (HAAO p.D162*, HAAO p.W186*, or KYNU p.V57Efs*21). Another patient carried heterozygous KYNU variants (p.Y156* and p.F349Kfs*4). The mutant enzymes had greatly reduced activity in vitro. Nicotinamide adenine dinucleotide (NAD) is synthesized de novo from tryptophan through the kynurenine pathway. The patients had reduced levels of circulating NAD. Defects similar to those in the patients developed in the embryos of Haao-null or Kynu-null mice owing to NAD deficiency. In null mice, the prevention of NAD deficiency during gestation averted defects. CONCLUSIONS: Disruption of NAD synthesis caused a deficiency of NAD and congenital malformations in humans and mice. Niacin supplementation during gestation prevented the malformations in mice. (Funded by the National Health and Medical Research Council of Australia and others.).


Assuntos
3-Hidroxiantranilato 3,4-Dioxigenase/genética , Anormalidades Congênitas/genética , Suplementos Nutricionais , Hidrolases/genética , NAD/deficiência , Niacina/uso terapêutico , 3-Hidroxiantranilato 3,4-Dioxigenase/metabolismo , Canal Anal/anormalidades , Animais , Anormalidades Congênitas/prevenção & controle , Modelos Animais de Doenças , Esôfago/anormalidades , Feminino , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/prevenção & controle , Humanos , Hidrolases/metabolismo , Rim/anormalidades , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Mutação , NAD/biossíntese , NAD/genética , Análise de Sequência de DNA , Coluna Vertebral/anormalidades , Traqueia/anormalidades
19.
Elife ; 62017 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-28513431

RESUMO

Despite the extensive use of zebrafish as a model organism in developmental biology and regeneration research, genetic techniques enabling conditional analysis of gene function are limited. In this study, we generated Zwitch, a Cre-dependent invertible gene-trap cassette, enabling the establishment of conditional alleles in zebrafish by generating intronic insertions via in vivo homologous recombination. To demonstrate the utility of Zwitch, we generated a conditional sonic hedgehog a (shha) allele. Homozygous shha mutants developed normally; however, shha mutant embryos globally expressing Cre exhibited strong reductions in endogenous shha and shha target gene mRNA levels and developmental defects associated with null shha mutations. Analyzing a conditional shha mutant generated using an epicardium-specific inducible Cre driver revealed unique roles for epicardium-derived Shha in myocardial proliferation during heart development and regeneration. Zwitch will extend the utility of zebrafish in organ development and regeneration research and might be applicable to other model organisms.


Assuntos
Marcação de Genes , Coração/embriologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Morfogênese , Recombinação Genética
20.
Dev Comp Immunol ; 73: 156-162, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28365195

RESUMO

Regulatory T (Treg) cells play a central role in the suppression of excessive immune responses against both self and non-self antigens. The development and function of Treg cells are controlled by a master regulatory gene encoding the forkhead box P3 (FOXP3) protein in mammals. However, little is known regarding the functions of Treg cells and FOXP3 in non-mammalian vertebrates. In this study, we generated mutant zebrafish lacking a functional FOXP3 ortholog, and demonstrated a significant reduction in survival accompanied by a marked increase in inflammatory gene expression, mononuclear cell infiltration, and T cell proliferation in peripheral tissues. Our findings indicate that the zebrafish FOXP3 protein may have an evolutionally conserved role in the control of immune tolerance, illuminating the potential of the zebrafish as a novel model for investigating the development and functions of Treg cells.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Tolerância a Antígenos Próprios/imunologia , Linfócitos T Reguladores/imunologia , Proteínas de Peixe-Zebra/imunologia , Peixe-Zebra/imunologia , Animais , Animais Geneticamente Modificados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA