Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
J Neurochem ; 163(6): 478-499, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36153691

RESUMO

The developmental process of central nervous system (CNS) myelin sheath formation is characterized by well-coordinated cellular activities ultimately ensuring rapid and synchronized neural communication. During this process, myelinating CNS cells, namely oligodendrocytes (OLGs), undergo distinct steps of differentiation, whereby the progression of earlier maturation stages of OLGs represents a critical step toward the timely establishment of myelinated axonal circuits. Given the complexity of functional integration, it is not surprising that OLG maturation is controlled by a yet fully to be defined set of both negative and positive modulators. In this context, we provide here first evidence for a role of lysophosphatidic acid (LPA) signaling via the G protein-coupled receptor LPA6 as a negative modulatory regulator of myelination-associated gene expression in OLGs. More specifically, the cell surface accessibility of LPA6 was found to be restricted to the earlier maturation stages of differentiating OLGs, and OLG maturation was found to occur precociously in Lpar6 knockout mice. To further substantiate these findings, a novel small molecule ligand with selectivity for preferentially LPA6 and LPA6 agonist characteristics was functionally characterized in vitro in primary cultures of rat OLGs and in vivo in the developing zebrafish. Utilizing this approach, a negative modulatory role of LPA6 signaling in OLG maturation could be corroborated. During development, such a functional role of LPA6 signaling likely serves to ensure timely coordination of circuit formation and myelination. Under pathological conditions as seen in the major human demyelinating disease multiple sclerosis (MS), however, persistent LPA6 expression and signaling in OLGs can be seen as an inhibitor of myelin repair. Thus, it is of interest that LPA6 protein levels appear elevated in MS brain samples, thereby suggesting that LPA6 signaling may represent a potential new druggable pathway suitable to promote myelin repair in MS.


Assuntos
Oligodendroglia , Peixe-Zebra , Camundongos , Animais , Ratos , Humanos , Oligodendroglia/metabolismo , Bainha de Mielina/metabolismo , Neurogênese/fisiologia , Diferenciação Celular/fisiologia , Receptores de Ácidos Lisofosfatídicos
2.
PLoS One ; 16(1): e0244794, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33439865

RESUMO

Understanding how fate specification of distinct cell-types from multipotent progenitors occurs is a fundamental question in embryology. Neural crest stem cells (NCSCs) generate extraordinarily diverse derivatives, including multiple neural, skeletogenic and pigment cell fates. Key transcription factors and extracellular signals specifying NCSC lineages remain to be identified, and we have only a little idea of how and when they function together to control fate. Zebrafish have three neural crest-derived pigment cell types, black melanocytes, light-reflecting iridophores and yellow xanthophores, which offer a powerful model for studying the molecular and cellular mechanisms of fate segregation. Mitfa has been identified as the master regulator of melanocyte fate. Here, we show that an Mitf-related transcription factor, Tfec, functions as master regulator of the iridophore fate. Surprisingly, our phenotypic analysis of tfec mutants demonstrates that Tfec also functions in the initial specification of all three pigment cell-types, although the melanocyte and xanthophore lineages recover later. We show that Mitfa represses tfec expression, revealing a likely mechanism contributing to the decision between melanocyte and iridophore fate. Our data are consistent with the long-standing proposal of a tripotent progenitor restricted to pigment cell fates. Moreover, we investigate activation, maintenance and function of tfec in multipotent NCSCs, demonstrating for the first time its role in the gene regulatory network forming and maintaining early neural crest cells. In summary, we build on our previous work to characterise the gene regulatory network governing iridophore development, establishing Tfec as the master regulator driving iridophore specification from multipotent progenitors, while shedding light on possible cellular mechanisms of progressive fate restriction.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Crista Neural/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Melanócitos/citologia , Melanócitos/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Mutagênese , Crista Neural/citologia , Pigmentação/genética , RNA Guia de Cinetoplastídeos/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
J Ultrasound Med ; 40(1): 175-181, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32557791

RESUMO

When evaluating patients with hip pain, clinicians may be trained to both evaluate for a hip effusion and perform ultrasound-guided arthrocentesis to evaluate the etiology of the effusion. We present a novel 3-dimensional-printed hip arthrocentesis model, which can be used to train clinicians to perform both tasks under ultrasound guidance. Our model uses a combination of a 3-dimensional-printed hip joint, as well as readily available materials such as an infant Ambu (Ballerup, Denmark) bag, syringe, intravenous line kit, and silicone. We present our experience so that others may use and adapt our model for their training purposes.


Assuntos
Artrocentese , Artralgia , Articulação do Quadril/diagnóstico por imagem , Articulação do Quadril/cirurgia , Humanos , Ultrassonografia , Ultrassonografia de Intervenção
4.
Development ; 147(21)2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-32541011

RESUMO

A crucial step in eye development is the closure of the choroid fissure (CF), a transient structure in the ventral optic cup through which vasculature enters the eye and ganglion cell axons exit. Although many factors have been identified that function during CF closure, the molecular and cellular mechanisms mediating this process remain poorly understood. Failure of CF closure results in colobomas. Recently, MITF was shown to be mutated in a subset of individuals with colobomas, but how MITF functions during CF closure is unknown. To address this issue, zebrafish with mutations in mitfa and tfec, two members of the Mitf family of transcription factors, were analyzed and their functions during CF closure determined. mitfa;tfec mutants possess severe colobomas and our data demonstrate that Mitf activity is required within cranial neural crest cells (cNCCs) during CF closure. In the absence of Mitf function, cNCC migration and localization in the optic cup are perturbed. These data shed light on the cellular mechanisms underlying colobomas in individuals with MITF mutations and identify a novel role for Mitf function in cNCCs during CF closure.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Corioide/citologia , Corioide/embriologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Crista Neural/citologia , Crânio/citologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Coloboma/patologia , Embrião de Mamíferos/citologia , Humanos , Mutação/genética , Crista Neural/metabolismo , Epitélio Pigmentado da Retina/embriologia
5.
Cancer Res ; 79(22): 5769-5784, 2019 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-31582381

RESUMO

The melanocyte-inducing transcription factor (MITF)-low melanoma transcriptional signature is predictive of poor outcomes for patients, but little is known about its biological significance, and animal models are lacking. Here, we used zebrafish genetic models with low activity of Mitfa (MITF-low) and established that the MITF-low state is causal of melanoma progression and a predictor of melanoma biological subtype. MITF-low zebrafish melanomas resembled human MITF-low melanomas and were enriched for stem and invasive (mesenchymal) gene signatures. MITF-low activity coupled with a p53 mutation was sufficient to promote superficial growth melanomas, whereas BRAFV600E accelerated MITF-low melanoma onset and further promoted the development of MITF-high nodular growth melanomas. Genetic inhibition of MITF activity led to rapid regression; recurrence occurred following reactivation of MITF. At the regression site, there was minimal residual disease that was resistant to loss of MITF activity (termed MITF-independent cells) with very low-to-no MITF activity or protein. Transcriptomic analysis of MITF-independent residual disease showed enrichment of mesenchymal and neural crest stem cell signatures similar to human therapy-resistant melanomas. Single-cell RNA sequencing revealed MITF-independent residual disease was heterogeneous depending on melanoma subtype. Further, there was a shared subpopulation of residual disease cells that was enriched for a neural crest G0-like state that preexisted in the primary tumor and remained present in recurring melanomas. These findings suggest that invasive and stem-like programs coupled with cellular heterogeneity contribute to poor outcomes for MITF-low melanoma patients and that MITF-independent subpopulations are an important therapeutic target to achieve long-term survival outcomes. SIGNIFICANCE: This study provides a useful model for MITF-low melanomas and MITF-independent cell populations that can be used to study the mechanisms that drive these tumors as well as identify potential therapeutic options.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/22/5769/F1.large.jpg.


Assuntos
Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , Neoplasia Residual/genética , Transcrição Gênica/genética , Peixe-Zebra/genética , Animais , Resistência a Medicamentos/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Melanócitos/patologia , Melanoma/patologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neoplasia Residual/patologia , Crista Neural/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Células-Tronco/patologia
6.
Pigment Cell Melanoma Res ; 32(5): 724-727, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30896066

RESUMO

Although xanthophores are found widely among poikilothermic species, the developmental and biochemical pathways underlying differentiation of these pteridine- and carotenoid-containing cells remain murky. I have identified a recessive zebrafish mutant, camembert (cmm), which displays defective xanthophore pigmentation during embryonic and larval stages with cells appearing grayish rather than yellow, but as an adult appears to have normally pigmented xanthophores and wild-type stripe pattern. Examination of molecular markers reveals that xanthophores are present in typical numbers and position in cmm embryos; however, the localization of transcripts for the gene GTP cyclohydrolase 2 (gch2), encoding a critical protein in the pteridine biosynthetic pathway, is strikingly altered. RT-PCR analysis indicates that gch2 transcripts in mutant embryos skip an exon or retain the intron upstream and that no correctly spliced mRNA is made. Sequencing of genomic DNA reveals that the skipped exon is intact, but the retained intron contains a deletion of approximately 180 base pairs, just upstream of the splice acceptor. Microinjection of a gch2 BAC clone rescues yellow pigmentation in camembert larvae, confirming that the pigmentation defect is due to mutation of gch2.


Assuntos
Embrião não Mamífero/metabolismo , GTP Cicloidrolase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Larva/crescimento & desenvolvimento , Melanóforos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Embrião não Mamífero/citologia , GTP Cicloidrolase/genética , Larva/genética , Larva/metabolismo , Pigmentação , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
7.
PLoS Genet ; 14(10): e1007402, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30286071

RESUMO

Multipotent neural crest (NC) progenitors generate an astonishing array of derivatives, including neuronal, skeletal components and pigment cells (chromatophores), but the molecular mechanisms allowing balanced selection of each fate remain unknown. In zebrafish, melanocytes, iridophores and xanthophores, the three chromatophore lineages, are thought to share progenitors and so lend themselves to investigating the complex gene regulatory networks (GRNs) underlying fate segregation of NC progenitors. Although the core GRN governing melanocyte specification has been previously established, those guiding iridophore and xanthophore development remain elusive. Here we focus on the iridophore GRN, where mutant phenotypes identify the transcription factors Sox10, Tfec and Mitfa and the receptor tyrosine kinase, Ltk, as key players. Here we present expression data, as well as loss and gain of function results, guiding the derivation of an initial iridophore specification GRN. Moreover, we use an iterative process of mathematical modelling, supplemented with a Monte Carlo screening algorithm suited to the qualitative nature of the experimental data, to allow for rigorous predictive exploration of the GRN dynamics. Predictions were experimentally evaluated and testable hypotheses were derived to construct an improved version of the GRN, which we showed produced outputs consistent with experimentally observed gene expression dynamics. Our study reveals multiple important regulatory features, notably a sox10-dependent positive feedback loop between tfec and ltk driving iridophore specification; the molecular basis of sox10 maintenance throughout iridophore development; and the cooperation between sox10 and tfec in driving expression of pnp4a, a key differentiation gene. We also assess a candidate repressor of mitfa, a melanocyte-specific target of sox10. Surprisingly, our data challenge the reported role of Foxd3, an established mitfa repressor, in iridophore regulation. Our study builds upon our previous systems biology approach, by incorporating physiologically-relevant parameter values and rigorous evaluation of parameter values within a qualitative data framework, to establish for the first time the core GRN guiding specification of the iridophore lineage.


Assuntos
Cromatóforos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Crista Neural/metabolismo , Biologia de Sistemas/métodos , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Linhagem da Célula/genética , Cromatóforos/citologia , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Mutação , Crista Neural/citologia , Crista Neural/embriologia , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
J Soc Work Pract Addict ; 17(1-2): 114-134, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-31588200

RESUMO

There has been a rapid increase in the development of technological innovations to reduce the escalation and impact of alcohol problems among adolescents and adults. Technology-based interventions offer the possibility of reaching individuals who otherwise might not seek treatment, (e.g., those in remote areas, those not perceiving a need for treatment, or others who may resist treatment). This article describes four case examples of technology-based interventions for risky drinking: 1) a freely available and interactive website that provides individualized feedback and information on risky drinking patterns; 2) a brief intervention for adolescents that provides individualized feedback to teens regarding their alcohol use; 3) a computer-delivered screening and brief intervention for alcohol use among pregnant women, and 4) a simulation program for training social workers in screening and brief intervention. These case examples highlight how technology may have a role in addressing the Alcohol Misuse Grand Challenge.

9.
Methods Mol Biol ; 1451: 143-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27464806

RESUMO

Melanoma is the most lethal form of skin cancer with high mortality rates. Most melanoma cases have activating mutations in BRAF (V600E) and the selective inhibitors of BRAF(V600E) have been successfully used in patients. However, after initial tumor regression, the majority of patients develop drug resistance resulting in tumor regrowth. It is therefore important to understand the mechanisms underlying these processes. We have recently described the role of the master melanocyte transcription factor MITF in tumor growth, regression, and recurrence. Here, we describe protocols to study regression and recurrence in vivo, as well as for histology and immunohistochemistry, using a temperature-sensitive zebrafish model of human melanoma.


Assuntos
Melanoma/patologia , Neoplasias/patologia , Peixe-Zebra/metabolismo , Animais , Humanos , Imuno-Histoquímica , Melanoma/genética , Melanoma/metabolismo , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Neoplasias/genética , Proteínas Proto-Oncogênicas B-raf/genética , Peixe-Zebra/genética
11.
J Neurosci ; 35(32): 11399-414, 2015 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-26269646

RESUMO

During development, oligodendrocytes (OLGs), the myelinating cells of the CNS, undergo a stepwise progression during which OLG progenitors, specified from neural stem/progenitor cells, differentiate into fully mature myelinating OLGs. This progression along the OLG lineage is characterized by well synchronized changes in morphology and gene expression patterns. The latter have been found to be particularly critical during the early stages of the lineage, and they have been well described to be regulated by epigenetic mechanisms, especially by the activity of the histone deacetylases HDAC1 and HDAC2. The data presented here identify the extracellular factor autotaxin (ATX) as a novel upstream signal modulating HDAC1/2 activity and gene expression in cells of the OLG lineage. Using the zebrafish as an in vivo model system as well as rodent primary OLG cultures, this functional property of ATX was found to be mediated by its lysophospholipase D (lysoPLD) activity, which has been well characterized to generate the lipid signaling molecule lysophosphatidic acid (LPA). More specifically, the lysoPLD activity of ATX was found to modulate HDAC1/2 regulated gene expression during a time window coinciding with the transition from OLG progenitor to early differentiating OLG. In contrast, HDAC1/2 regulated gene expression during the transition from neural stem/progenitor to OLG progenitor appeared unaffected by ATX and its lysoPLD activity. Thus, together, our data suggest that an ATX-LPA-HDAC1/2 axis regulates OLG differentiation specifically during the transition from OLG progenitor to early differentiating OLG and via a molecular mechanism that is evolutionarily conserved from at least zebrafish to rodent. SIGNIFICANCE STATEMENT: The formation of the axon insulating and supporting myelin sheath by differentiating oligodendrocytes (OLGs) in the CNS is considered an essential step during vertebrate development. In addition, loss and/or dysfunction of the myelin sheath has been associated with a variety of neurologic diseases in which repair is limited, despite the presence of progenitor cells with the potential to differentiate into myelinating OLGs. This study characterizes the autotaxin-lysophosphatidic acid signaling axis as a modulator of OLG differentiation in vivo in the developing zebrafish and in vitro in rodent OLGs in culture. These findings provide novel insight into the regulation of developmental myelination, and they are likely to lead to advancing studies related to the stimulation of myelin repair under pathologic conditions.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Lisofosfolipídeos/metabolismo , Oligodendroglia/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Acetilação , Animais , Células Cultivadas , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/citologia , Rombencéfalo/citologia , Rombencéfalo/metabolismo , Transdução de Sinais/fisiologia , Peixe-Zebra
13.
J Invest Dermatol ; 134(1): 133-140, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23831555

RESUMO

The microphthalmia-associated transcription factor (MITF) is the "master melanocyte transcription factor" with a complex role in melanoma. MITF protein levels vary between and within clinical specimens, and amplifications and gain- and loss-of-function mutations have been identified in melanoma. How MITF functions in melanoma development and the effects of targeting MITF in vivo are unknown because MITF levels have not been directly tested in a genetic animal model. Here, we use a temperature-sensitive mitf zebrafish mutant to conditionally control endogenous MITF activity. We show that low levels of endogenous MITF activity are oncogenic with BRAF(V600E) to promote melanoma that reflects the pathology of the human disease. Remarkably, abrogating MITF activity in BRAF(V600E)mitf melanoma leads to dramatic tumor regression marked by melanophage infiltration and increased apoptosis. These studies are significant because they show that targeting MITF activity is a potent antitumor mechanism, but also show that caution is required because low levels of wild-type MITF activity are oncogenic.


Assuntos
Melanoma/metabolismo , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Proteínas de Peixe-Zebra/genética , Animais , Apoptose/fisiologia , Modelos Animais de Doenças , Genótipo , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Mutação Puntual , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
14.
Genome Biol ; 14(10): R113, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24148783

RESUMO

BACKGROUND: Melanoma is the most deadly form of skin cancer. Expression of oncogenic BRAF or NRAS, which are frequently mutated in human melanomas, promote the formation of nevi but are not sufficient for tumorigenesis. Even with germline mutated p53, these engineered melanomas present with variable onset and pathology, implicating additional somatic mutations in a multi-hit tumorigenic process. RESULTS: To decipher the genetics of these melanomas, we sequence the protein coding exons of 53 primary melanomas generated from several BRAF(V600E) or NRAS(Q61K) driven transgenic zebrafish lines. We find that engineered zebrafish melanomas show an overall low mutation burden, which has a strong, inverse association with the number of initiating germline drivers. Although tumors reveal distinct mutation spectrums, they show mostly C > T transitions without UV light exposure, and enrichment of mutations in melanogenesis, p53 and MAPK signaling. Importantly, a recurrent amplification occurring with pre-configured drivers BRAF(V600E) and p53-/- suggests a novel path of BRAF cooperativity through the protein kinase A pathway. CONCLUSION: This is the first analysis of a melanoma mutational landscape in the absence of UV light, where tumors manifest with remarkably low mutation burden and high heterogeneity. Genotype specific amplification of protein kinase A in cooperation with BRAF and p53 mutation suggests the involvement of melanogenesis in these tumors. This work is important for defining the spectrum of events in BRAF or NRAS driven melanoma in the absence of UV light, and for informed exploitation of models such as transgenic zebrafish to better understand mechanisms leading to human melanoma formation.


Assuntos
Heterogeneidade Genética , Melanoma/genética , Mutação , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Amplificação de Genes , Técnicas de Inativação de Genes , Homozigoto , Mutação INDEL , Melanoma/patologia , Mutação/efeitos da radiação , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Deleção de Sequência , Raios Ultravioleta
16.
J Neurosci ; 33(8): 3424-33, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23426670

RESUMO

The CA1 region of the hippocampus receives distinct patterns of afferent input to distal (near subiculum) and proximal (near CA2) zones. Specifically, distal CA1 receives a direct projection from cells in the lateral entorhinal cortex that are sensitive to objects, whereas proximal CA1 is innervated by cells in the medial entorhinal cortex that are responsive to space. This suggests that neurons in different areas along the proximodistal axis of CA1 of the hippocampus will be functionally distinct. The current experiment investigated this possibility by monitoring behavior-induced cell activity across the CA1 axis using Arc mRNA imaging methods that compared adult and old rats in two conditions: (1) exploration of the same environment containing the same objects twice (AA) or (2) exploration of two different environments that contained identical objects (AB). The hypothesis was that CA1 place cells should show field remapping in the condition in which environments were changed, but the extent of remapping was expected to differ between proximal and distal regions and between age groups. In fact, neurons in the proximal region of CA1 in adult animals exhibited a greater degree of remapping than did distal CA1 cells when the environment changed, suggesting that cells receiving input from the medial entorhinal cortex are more sensitive to spatial context. However, in old rats, there were no differences in remapping across the proximodistal CA1 axis. Together, these data suggest that distal and proximal CA1 may be functionally distinct and differentially vulnerable to normative aging processes.


Assuntos
Envelhecimento/fisiologia , Região CA1 Hipocampal/fisiologia , Proteínas do Citoesqueleto/genética , Comportamento Exploratório/fisiologia , Genes Precoces/fisiologia , Proteínas do Tecido Nervoso/genética , Transcrição Gênica/fisiologia , Envelhecimento/genética , Animais , Mapeamento Encefálico/métodos , Masculino , Aprendizagem em Labirinto/fisiologia , Imagem Molecular/métodos , RNA Mensageiro/biossíntese , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344
17.
PLoS One ; 7(11): e49357, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23139843

RESUMO

Otx and Mitf transcription factors have been implicated in the development of the retinal pigmented epithelium (RPE), but the relationship between these factors and their specific roles in the development of the RPE have not been fully defined. The role of the three Otx transcription factors (Otx1a, Otx1b, and Otx2) and two Mitf transcription factors (Mitfa and Mitfb) in the development of the zebrafish RPE was explored in these experiments. The loss of Otx activity through morpholino knockdown produced variable eye defects, ranging from delayed RPE pigmentation to severe coloboma, depending on the combination of Otx factors that were targeted. Expression analysis through in situ hybridization demonstrates that otx transcription factors are necessary for the proper expression of mitfa and mitfb while Mitf transcription factors are not required for the expression of otx genes. Surprisingly, the loss of Mitf activity in mitfa, mitfb, or double mitf mutant zebrafish had no effect on RPE pigmentation or development. Moreover, histological analysis revealed that retinal lamination is unaffected in mitf mutants, as well as in otx morphants, even in regions lacking RPE. Otx and Mitf combined loss of function experiments suggest that mitfa and mitfb may still influence zebrafish RPE development. This is further supported by the ability of mitfa to induce pigmentation in the zebrafish retina when misexpressed. These findings suggest that one or more Otx targets in addition to mitfa and mitfb, possibly another mitf family member, are necessary for development of the RPE in zebrafish.


Assuntos
Epitélio Pigmentado da Retina/embriologia , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Linhagem da Célula/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Morfolinos/farmacologia , Mutação/genética , Fenótipo , Pigmentação/efeitos dos fármacos , Pigmentação/genética , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia , Fatores de Transcrição/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Glia ; 60(10): 1605-18, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22821873

RESUMO

During development, progenitors that are committed to differentiate into oligodendrocytes, the myelinating cells of the central nervous system (CNS), are generated within discrete regions of the neuroepithelium. More specifically, within the developing spinal cord and hindbrain ventrally located progenitor cells that are characterized by the expression of the transcription factor olig2 give temporally rise to first motor neurons and then oligodendrocyte progenitors. The regulation of this temporal neuron-glial switch has been found complex and little is known about the extrinsic factors regulating it. Our studies described here identified a zebrafish ortholog to mammalian atx, which displays evolutionarily conserved expression pattern characteristics. Most interestingly, atx was found to be expressed by cells of the cephalic floor plate during a time period when ventrally-derived oligodendrocyte progenitors arise in the developing hindbrain of the zebrafish. Knock-down of atx expression resulted in a delay and/or inhibition of the timely appearance of oligodendrocyte progenitors and subsequent developmental stages of the oligodendrocyte lineage. This effect of atx knock-down was not accompanied by changes in the number of olig2-positive progenitor cells, the overall morphology of the axonal network or the number of somatic abducens motor neurons. Thus, our studies identified Atx as an extrinsic factor that is likely secreted by cells from the floor plate and that is involved in regulating specifically the progression of olig2-positive progenitor cells into lineage committed oligodendrocyte progenitors.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Oligodendroglia/fisiologia , Diester Fosfórico Hidrolases/metabolismo , Rombencéfalo/citologia , Rombencéfalo/crescimento & desenvolvimento , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Diferenciação Celular/genética , Clonagem Molecular , Embrião não Mamífero , Lateralidade Funcional/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Morfolinos/farmacologia , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/efeitos dos fármacos , Diester Fosfórico Hidrolases/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
19.
Methods Cell Biol ; 104: 195-208, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21924164

RESUMO

On the strengths of forward genetics and embryology, the zebrafish Danio rerio has become an ideal system for the study of early vertebrate development. However, additional tools will be needed to perform more sophisticated analyses and to successfully carry this model into new areas of study such as adult physiology, cancer, and aging. As improved tools make transgenesis more and more efficient, the stage has been set for precise modification of the zebrafish genome such as are done in other model organisms. Genome engineering strategies employing site-specific recombinase (SSR) systems such as Cre/lox and Flp/FRT have become invaluable to the study of gene function in the mouse and Drosophila and are now being exploited in zebrafish as well. My laboratory has begun to use another such SSR, the integrase encoded by the Streptomyces bacteriophage PhiC31, for manipulation of the zebrafish genome. The PhiC31 integrase promotes recombination between an attachment site in the phage (attP) and another on the bacterial chromosome (attB). Here I describe strategies using the PhiC31 integrase to mediate recombination of transgenes containing attP and attB sites in cis to excise elements with spatial and temporal specificity. The feasibility of the intramolecular recombination approach having been established, I discuss prospects for employing PhiC31 integrase for intermolecular recombination, i.e., transgene integration at defined sites in the genome.


Assuntos
Bacteriófagos/enzimologia , Genoma , Integrases/genética , Mutagênese Insercional/métodos , Proteínas Virais/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , DNA Nucleotidiltransferases/genética , DNA Nucleotidiltransferases/metabolismo , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genes Reporter , Vetores Genéticos , Integrases/metabolismo , Larva/genética , Larva/metabolismo , Masculino , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Virais/metabolismo
20.
Dev Dyn ; 240(11): 2529-38, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21932325

RESUMO

The MiT family comprises four genes in mammals: Mitf, Tfe3, Tfeb, and Tfec, which encode transcription factors of the basic-helix-loop-helix/leucine zipper class. Mitf is well-known for its essential role in the development of melanocytes, however the functions of the other members of this family, and of interactions between them, are less well understood. We have now characterized the complete set of MiT genes from zebrafish, which totals six instead of four. The zebrafish genome contain two mitf (mitfa and mitfb), two tfe3 (tfe3a and tfe3b), and single tfeb and tfec genes; this distribution is shared with other teleosts. We present here the sequence and embryonic expression patterns for the zebrafish tfe3b, tfeb, and tfec genes, and identify a new isoform of tfe3a. These findings will assist in elucidating the roles of the MiT gene family over the course of vertebrate evolution.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fator de Transcrição Associado à Microftalmia/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Mapeamento Cromossômico , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Larva/genética , Larva/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Dados de Sequência Molecular , Família Multigênica , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...