Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 150(6): 1135-46, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22980977

RESUMO

DNA methylation at the 5 position of cytosine (5-mC) is a key epigenetic mark that is critical for various biological and pathological processes. 5-mC can be converted to 5-hydroxymethylcytosine (5-hmC) by the ten-eleven translocation (TET) family of DNA hydroxylases. Here, we report that "loss of 5-hmC" is an epigenetic hallmark of melanoma, with diagnostic and prognostic implications. Genome-wide mapping of 5-hmC reveals loss of the 5-hmC landscape in the melanoma epigenome. We show that downregulation of isocitrate dehydrogenase 2 (IDH2) and TET family enzymes is likely one of the mechanisms underlying 5-hmC loss in melanoma. Rebuilding the 5-hmC landscape in melanoma cells by reintroducing active TET2 or IDH2 suppresses melanoma growth and increases tumor-free survival in animal models. Thus, our study reveals a critical function of 5-hmC in melanoma development and directly links the IDH and TET activity-dependent epigenetic pathway to 5-hmC-mediated suppression of melanoma progression, suggesting a new strategy for epigenetic cancer therapy.


Assuntos
Citosina/análogos & derivados , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Nevo/genética , 5-Metilcitosina/análogos & derivados , Citosina/metabolismo , Proteínas de Ligação a DNA/genética , Dioxigenases , Estudo de Associação Genômica Ampla , Humanos , Isocitrato Desidrogenase/genética , Melanócitos/metabolismo , Melanoma/patologia , Nevo/patologia , Proteínas Proto-Oncogênicas/genética
2.
Cell ; 137(4): 736-48, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19450519

RESUMO

During vertebrate embryogenesis, hematopoietic stem cells (HSCs) arise in the aorta-gonads-mesonephros (AGM) region. We report here that blood flow is a conserved regulator of HSC formation. In zebrafish, chemical blood flow modulators regulated HSC development, and silent heart (sih) embryos, lacking a heartbeat and blood circulation, exhibited severely reduced HSCs. Flow-modifying compounds primarily affected HSC induction after the onset of heartbeat; however, nitric oxide (NO) donors regulated HSC number even when treatment occurred before the initiation of circulation, and rescued HSCs in sih mutants. Morpholino knockdown of nos1 (nnos/enos) blocked HSC development, and its requirement was shown to be cell autonomous. In the mouse, Nos3 (eNos) was expressed in HSCs in the AGM. Intrauterine Nos inhibition or embryonic Nos3 deficiency resulted in a reduction of hematopoietic clusters and transplantable murine HSCs. This work links blood flow to AGM hematopoiesis and identifies NO as a conserved downstream regulator of HSC development.


Assuntos
Fenômenos Fisiológicos Sanguíneos , Hematopoese , Células-Tronco Hematopoéticas/citologia , Animais , Embrião de Mamíferos/metabolismo , Embrião não Mamífero/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Peixe-Zebra
3.
Mol Carcinog ; 59(9): 1052-1063, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32562448

RESUMO

Melanoma is an aggressive, deadly skin cancer derived from melanocytes, a neural crest cell derivative. Melanoma cells mirror the developmental program of neural crest cells in that they exhibit the same gene expression patterns and utilize similar cellular mechanisms, including increased cell proliferation, epithelial-mesenchymal transition, and migration. Here we studied the role of neural crest regulator PRDM1 in melanoma onset and progression. In development, Prdm1a functions to promote neural crest progenitor fate, and in melanoma, we found that PRDM1 has reduced copy number and is recurrently deleted in both zebrafish and humans. When examining expression of neural crest and melanocyte development genes, we show that sox10 progenitor expression is high in prdm1a-/- mutants, while more differentiated melanocyte markers are reduced, suggesting that normally Prdm1a is required for differentiation. Data mining of human melanoma datasets indicates that high PRDM1 expression in human melanoma is correlated with better patient survival and decreased PRDM1 expression is common in metastatic tumors. When one copy of prdm1a is lost in the zebrafish melanoma model Tg(mitfa:BRAFV600E );p53-/- ;prdm1a+/- , melanoma onset occurs more quickly, and the tumors that form have a larger area with increased expression of sox10. These data demonstrate a novel role for PRDM1 as a tumor suppressor in melanoma.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Melanócitos/patologia , Melanoma/patologia , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Animais , Diferenciação Celular , Células Cultivadas , Progressão da Doença , Humanos , Melanócitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Prognóstico , Taxa de Sobrevida , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
4.
Nature ; 471(7339): 513-7, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21430779

RESUMO

The most common mutation in human melanoma, BRAF(V600E), activates the serine/threonine kinase BRAF and causes excessive activity in the mitogen-activated protein kinase pathway. BRAF(V600E) mutations are also present in benign melanocytic naevi, highlighting the importance of additional genetic alterations in the genesis of malignant tumours. Such changes include recurrent copy number variations that result in the amplification of oncogenes. For certain amplifications, the large number of genes in the interval has precluded an understanding of the cooperating oncogenic events. Here we have used a zebrafish melanoma model to test genes in a recurrently amplified region of chromosome 1 for the ability to cooperate with BRAF(V600E) and accelerate melanoma. SETDB1, an enzyme that methylates histone H3 on lysine 9 (H3K9), was found to accelerate melanoma formation significantly in zebrafish. Chromatin immunoprecipitation coupled with massively parallel DNA sequencing and gene expression analyses uncovered genes, including HOX genes, that are transcriptionally dysregulated in response to increased levels of SETDB1. Our studies establish SETDB1 as an oncogene in melanoma and underscore the role of chromatin factors in regulating tumorigenesis.


Assuntos
Variações do Número de Cópias de DNA/genética , Amplificação de Genes/genética , Histona-Lisina N-Metiltransferase/genética , Melanoma/genética , Melanoma/patologia , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Idade de Início , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados , Transformação Celular Neoplásica/genética , Imunoprecipitação da Cromatina , Cromossomos Humanos Par 1/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Genes Homeobox/genética , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Melanócitos/citologia , Melanócitos/enzimologia , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/enzimologia , Nevo/enzimologia , Oncogenes/genética , Proteínas Proto-Oncogênicas B-raf/química , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Peixe-Zebra/genética
5.
Adv Exp Med Biol ; 916: 3-19, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27165347

RESUMO

Over a decade has elapsed since the first genetically-engineered zebrafish cancer model was described. During this time remarkable progress has been made. Sophisticated genetic tools have been built to generate oncogene expressing cancers and characterize multiple models of solid and blood tumors. These models have led to unique insights into mechanisms of tumor initiation and progression. New drug targets have been identified, particularly through the functional analysis of cancer genomes. Now in the second decade, zebrafish cancer models are poised for even faster growth as they are used in high-throughput genetic analyses to elucidate key mechanisms underlying critical cancer phenotypes.


Assuntos
Modelos Animais de Doenças , Neoplasias/genética , Oncogenes , Animais , Peixe-Zebra
6.
Sci Adv ; 10(11): eadh9547, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38489372

RESUMO

Solid tumors, especially those with aberrant MYCN activation, often harbor an immunosuppressive microenvironment to fuel malignant growth and trigger treatment resistance. Despite this knowledge, there are no effective strategies to tackle this problem. We found that chemokine-like factor (CKLF) is highly expressed by various solid tumor cells and transcriptionally up-regulated by MYCN. Using the MYCN-driven high-risk neuroblastoma as a model system, we demonstrated that as early as the premalignant stage, tumor cells secrete CKLF to attract CCR4-expressing CD4+ cells, inducing immunosuppression and tumor aggression. Genetic depletion of CD4+ T regulatory cells abolishes the immunorestrictive and protumorigenic effects of CKLF. Our work supports that disrupting CKLF-mediated cross-talk between tumor and CD4+ suppressor cells represents a promising immunotherapeutic approach to battling MYCN-driven tumors.


Assuntos
Quimiocinas , Proteínas com Domínio MARVEL , Proteína Proto-Oncogênica N-Myc , Neuroblastoma , Humanos , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas com Domínio MARVEL/metabolismo , Proteína Proto-Oncogênica N-Myc/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neuroblastoma/terapia , Microambiente Tumoral
7.
Cancer Discov ; 13(10): 2128-2130, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37794841

RESUMO

SUMMARY: GABA signaling by melanoma cells was found by Tagore and colleagues to trigger keratinocyte-driven growth of melanomas. This study reveals new roles for nonneuronal signaling by a neurotransmitter in regulating tumor initiation and outgrowth. See related article by Tagore et al., p. 2270 (4).


Assuntos
Melanoma , Humanos , Melanoma/genética , Melanoma/patologia , Queratinócitos , Transformação Celular Neoplásica , Transdução de Sinais , Ácido gama-Aminobutírico
8.
Elife ; 122023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-37021774

RESUMO

Tissue-resident stem and progenitor cells are present in many adult organs, where they are important for organ homeostasis and repair in response to injury. However, the signals that activate these cells and the mechanisms governing how these cells renew or differentiate are highly context-dependent and incompletely understood, particularly in non-hematopoietic tissues. In the skin, melanocyte stem and progenitor cells are responsible for replenishing mature pigmented melanocytes. In mammals, these cells reside in the hair follicle bulge and bulb niches where they are activated during homeostatic hair follicle turnover and following melanocyte destruction, as occurs in vitiligo and other skin hypopigmentation disorders. Recently, we identified melanocyte progenitors in adult zebrafish skin. To elucidate mechanisms governing melanocyte progenitor renewal and differentiation we analyzed individual transcriptomes from thousands of melanocyte lineage cells during the regeneration process. We identified transcriptional signatures for progenitors, deciphered transcriptional changes and intermediate cell states during regeneration, and analyzed cell-cell signaling changes to discover mechanisms governing melanocyte regeneration. We identified KIT signaling via the RAS/MAPK pathway as a regulator of melanocyte progenitor direct differentiation and asymmetric division. Our findings show how activation of different subpopulations of mitfa-positive cells underlies cellular transitions required to properly reconstitute the melanocyte pigmentary system following injury.


Assuntos
Melanócitos , Peixe-Zebra , Animais , Peixe-Zebra/fisiologia , Melanócitos/metabolismo , Pele , Células-Tronco/metabolismo , Folículo Piloso , Transdução de Sinais , Diferenciação Celular , Mamíferos
9.
medRxiv ; 2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-38106222

RESUMO

Polycystic kidney disease (PKD) is an important cause of end stage renal disease, but treatment options are limited. While later stages of the disease have been extensively studied, mechanisms driving the initial conversion of renal tubules into cysts are not understood. To identify factors that promote the initiation of cysts we deleted polycystin-2 ( Pkd2 ) in mice and surveyed transcriptional changes before and immediately after cysts developed. We identified 74 genes which we term cyst initiation candidates (CICs). To identify conserved changes with relevance to human disease we compared these murine CICs to single cell transcriptomic data derived from patients with PKD and from healthy controls. Tumor-associated calcium signal transducer 2 ( Tacstd2 ) stood out as an epithelial-expressed gene whose levels were elevated prior to cystic transformation and further increased with disease progression. Human tissue biopsies and organoids show that TACSTD2 protein is low in normal kidney cells but is elevated in cyst lining cells. While TACSTD2 has not been studied in PKD, it has been studied in cancer where it is highly expressed in solid tumors while showing minimal expression in normal tissue. This property is being exploited by antibody drug conjugates that target TACSTD2 for the delivery of cytotoxic drugs. Our finding that Tacstd2 is highly expressed in cysts, but not normal tissue, suggests that it should be explored as a candidate for drug development in PKD. More immediately, our work suggests that PKD patients undergoing TACSTD2 treatment for cancer should be monitored for kidney effects. One Sentence Summary: The oncogene, tumor-associated calcium signal transducer 2 (Tacstd2) mRNA increased in abundance shortly after Pkd2 loss and may be a driver of cyst initiation in polycystic kidney disease.

10.
J Invest Dermatol ; 142(3 Pt A): 499-506.e1, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35184798

RESUMO

Skin diseases affect nearly one third of the world's population. Disease types range from oncologic to inflammatory, and outcomes can be as severe as death and disfigurement. Although many skin diseases have been modeled in murine models, the advantages of zebrafish models have led to recent increasing use in modeling human disease. Their rapid development, comparable skin architecture, tractable genetics, unparalleled optical properties, and straightforward drug screens make them an excellent model to study skin disease. In this review, we discuss the attributes of the zebrafish model system as well as current zebrafish models for dermatologic diseases, including melanoma, squamous cell carcinoma, vitiligo, epidermal bullosa, psoriasis, and wounding.


Assuntos
Melanoma , Vitiligo , Animais , Modelos Animais de Doenças , Humanos , Melanoma/patologia , Camundongos , Projetos de Pesquisa , Peixe-Zebra
11.
Nat Commun ; 13(1): 4109, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35840569

RESUMO

Melanomas and other solid tumors commonly have increased ploidy, with near-tetraploid karyotypes being most frequently observed. Such karyotypes have been shown to arise through whole-genome doubling events that occur during early stages of tumor progression. The generation of tetraploid cells via whole-genome doubling is proposed to allow nascent tumor cells the ability to sample various pro-tumorigenic genomic configurations while avoiding the negative consequences that chromosomal gains or losses have in diploid cells. Whereas a high prevalence of whole-genome doubling events has been established, the means by which whole-genome doubling arises is unclear. Here, we find that BRAFV600E, the most common mutation in melanomas, can induce whole-genome doubling via cytokinesis failure in vitro and in a zebrafish melanoma model. Mechanistically, BRAFV600E causes decreased activation and localization of RhoA, a critical cytokinesis regulator. BRAFV600E activity during G1/S phases of the cell cycle is required to suppress cytokinesis. During G1/S, BRAFV600E activity causes inappropriate centriole amplification, which is linked in part to inhibition of RhoA and suppression of cytokinesis. Together these data suggest that common abnormalities of melanomas linked to tumorigenesis - amplified centrosomes and whole-genome doubling events - can be induced by oncogenic BRAF and other mutations that increase RAS/MAPK pathway activity.


Assuntos
Melanoma , Proteínas Proto-Oncogênicas B-raf , Animais , Linhagem Celular Tumoral , Citocinese/genética , Melanoma/genética , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Tetraploidia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
12.
Nat Commun ; 13(1): 3732, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35768444

RESUMO

Melanoma is commonly driven by activating mutations in the MAP kinase BRAF; however, oncogenic BRAF alone is insufficient to promote melanomagenesis. Instead, its expression induces a transient proliferative burst that ultimately ceases with the development of benign nevi comprised of growth-arrested melanocytes. The tumor suppressive mechanisms that restrain nevus melanocyte proliferation remain poorly understood. Here we utilize cell and murine models to demonstrate that oncogenic BRAF leads to activation of the Hippo tumor suppressor pathway, both in melanocytes in vitro and nevus melanocytes in vivo. Mechanistically, we show that oncogenic BRAF promotes both ERK-dependent alterations in the actin cytoskeleton and whole-genome doubling events, which independently reduce RhoA activity to promote Hippo activation. We also demonstrate that functional impairment of the Hippo pathway enables oncogenic BRAF-expressing melanocytes to bypass nevus formation and rapidly form melanomas. Our data reveal that the Hippo pathway enforces the stable arrest of nevus melanocytes and represents a critical barrier to melanoma development.


Assuntos
Melanoma , Nevo , Neoplasias Cutâneas , Animais , Melanócitos/metabolismo , Melanoma/patologia , Camundongos , Mutação , Nevo/genética , Nevo/metabolismo , Nevo/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/patologia
13.
Cell Rep ; 39(12): 110995, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35732120

RESUMO

Dysregulated cellular metabolism is a cancer hallmark for which few druggable oncoprotein targets have been identified. Increased fatty acid (FA) acquisition allows cancer cells to meet their heightened membrane biogenesis, bioenergy, and signaling needs. Excess FAs are toxic to non-transformed cells but surprisingly not to cancer cells. Molecules underlying this cancer adaptation may provide alternative drug targets. Here, we demonstrate that diacylglycerol O-acyltransferase 1 (DGAT1), an enzyme integral to triacylglyceride synthesis and lipid droplet formation, is frequently up-regulated in melanoma, allowing melanoma cells to tolerate excess FA. DGAT1 over-expression alone transforms p53-mutant zebrafish melanocytes and co-operates with oncogenic BRAF or NRAS for more rapid melanoma formation. Antagonism of DGAT1 induces oxidative stress in melanoma cells, which adapt by up-regulating cellular reactive oxygen species defenses. We show that inhibiting both DGAT1 and superoxide dismutase 1 profoundly suppress tumor growth through eliciting intolerable oxidative stress.


Assuntos
Diacilglicerol O-Aciltransferase , Melanoma , Animais , Diacilglicerol O-Aciltransferase/genética , Diacilglicerol O-Aciltransferase/metabolismo , Proteínas Oncogênicas/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio , Triglicerídeos , Peixe-Zebra/metabolismo
14.
Pigment Cell Melanoma Res ; 34(2): 280-287, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33283422

RESUMO

Melanoma arises from the melanocyte lineage and is the most aggressive and lethal form of skin cancer. There are several genetic, genomic, and cellular changes associated with melanoma initiation. Here, we discuss these alterations and the melanoma cells of origin in which they are proposed to promote melanomagenesis.


Assuntos
Carcinogênese/patologia , Regulação Neoplásica da Expressão Gênica , Melanócitos/patologia , Melanoma/patologia , Neoplasias Cutâneas/patologia , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Humanos , Melanócitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo
15.
Dev Cell ; 56(20): 2783-2784, 2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34699785

RESUMO

In this issue of Developmental Cell, Campbell et al. (2021) show that melanoma cells with distinct invasive or proliferative gene signatures can form heterotypic clusters that extravasate collectively and readily seed the growth of metastatic lesions. These findings highlight interactions between heterogenous tumor cells as being critical for metastasis.


Assuntos
Melanoma , Contagem de Células , Movimento Celular , Humanos
16.
Cancer Cell ; 39(5): 610-631, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33545064

RESUMO

There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.


Assuntos
Modelos Animais de Doenças , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Microambiente Tumoral/imunologia , Animais , Humanos , Imunidade/imunologia , Imunoterapia/métodos , Melanoma/patologia , Neoplasias Cutâneas/patologia
17.
Genes Chromosomes Cancer ; 48(2): 155-70, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18973135

RESUMO

The zebrafish is emerging as a prominent model system for studying the genetics of human development and disease. Genetic alterations that underlie each mutant model can exist in the form of single base changes, balanced chromosomal rearrangements, or genetic imbalances. To detect genetic imbalances in an unbiased genome-wide fashion, array comparative genomic hybridization (CGH) can be used. We have developed a 5-Mb resolution array CGH platform specifically for the zebrafish. This platform contains 286 bacterial artificial chromosome (BAC) clones, enriched for orthologous sequences of human oncogenes and tumor suppressor genes. Each BAC clone has been end-sequenced and cytogenetically assigned to a specific location within the zebrafish genome, allowing for ease of integration of array CGH data with the current version of the genome assembly. This platform has been applied to three zebrafish cancer models. Significant genomic imbalances were detected in each model, identifying different regions that may potentially play a role in tumorigenesis. Hence, this platform should be a useful resource for genetic dissection of additional zebrafish developmental and disease models as well as a benchmark for future array CGH platform development.


Assuntos
Hibridização Genômica Comparativa/métodos , Genes Supressores de Tumor , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Oncogenes , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Cromossomos Artificiais Bacterianos , Modelos Animais de Doenças , Humanos , Hibridização in Situ Fluorescente , Melanoma/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Reprodutibilidade dos Testes , Rabdomiossarcoma/genética , Peixe-Zebra/metabolismo
18.
Cells ; 9(5)2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32455885

RESUMO

Melanoma is the deadliest form of skin cancer and one of few cancers with a growing incidence. A thorough understanding of its pathogenesis is fundamental to developing new strategies to combat mortality and morbidity. Zebrafish-due in large part to their tractable genetics, conserved pathways, and optical properties-have emerged as an excellent system to model melanoma. Zebrafish have been used to study melanoma from a single tumor initiating cell, through metastasis, remission, and finally into relapse. In this review, we examine seminal zebrafish studies that have advanced our understanding of melanoma.


Assuntos
Melanoma/patologia , Peixe-Zebra/fisiologia , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença , Melanócitos/patologia , Melanoma/genética , Crista Neural/patologia
19.
Dev Biol ; 320(1): 161-74, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18585699

RESUMO

Developmental signaling pathways hold the keys to unlocking the promise of adult tissue regeneration, and to inhibiting carcinogenesis. Patients with mutations in the Adenomatous Polyposis Coli (APC) gene are at increased risk of developing hepatoblastoma, an embryonal form of liver cancer, suggesting that Wnt affects hepatic progenitor cells. To elucidate the role of APC loss and enhanced Wnt activity in liver development, we examined APC mutant and wnt inducible transgenic zebrafish. APC(+/-) embryos developed enlarged livers through biased induction of hepatic gene programs and increased proliferation. Conversely, APC(-/-) embryos formed no livers. Blastula transplantations determined that the effects of APC loss were cell autonomous. Induction of wnt modulators confirmed biphasic consequences of wnt activation: endodermal pattern formation and gene expression required suppression of wnt signaling in early somitogenesis; later, increased wnt activity altered endodermal fate by enhancing liver growth at the expense of pancreas formation; these effects persisted into the larval stage. In adult APC(+/-) zebrafish, increased wnt activity significantly accelerated liver regeneration after partial hepatectomy. Similarly, liver regeneration was significantly enhanced in APC(Min/+) mice, indicating the conserved effect of Wnt pathway activation in liver regeneration across vertebrate species. These studies reveal an important and time-dependent role for wnt signaling during liver development and regeneration.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Fígado/embriologia , Mutação/genética , Transdução de Sinais , Proteínas Wnt/metabolismo , Peixe-Zebra/embriologia , Animais , Apoptose , Padronização Corporal , Linhagem da Célula , Proliferação de Células , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Endoderma/citologia , Endoderma/embriologia , Hepatectomia , Hepatócitos/citologia , Fígado/citologia , Regeneração Hepática , Fenótipo , Células-Tronco/citologia , Fatores de Tempo , beta Catenina/metabolismo
20.
Dev Cell ; 6(4): 563-76, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15068795

RESUMO

The class A and class B synMuv genes are functionally redundant negative regulators of a Ras signaling pathway that induces C. elegans vulval development. A number of class B synMuv genes encode components of an Rb and histone deacetylase complex that likely acts to repress transcription of genes required for vulval induction. We discovered a new class of synMuv genes that acts redundantly with both the A and B classes of genes in vulval cell-fate determination. These new class C synMuv genes encode TRRAP, MYST family histone acetyltransferase, and Enhancer of Polycomb homologs, which form a putative C. elegans Tip60/NuA4-like histone acetyltransferase complex. A fourth gene with partial class C synMuv properties encodes a homolog of the mammalian SWI/SNF family ATPase p400. Our findings indicate that the coordinated action of two chromatin-modifying complexes, one with histone deacetylase and the other with histone acetyltransferase activity, is important in regulating Ras signaling and specifying cell fates during C. elegans development.


Assuntos
Acetiltransferases/genética , Acetiltransferases/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Caenorhabditis elegans/isolamento & purificação , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/genética , Proteínas Cromossômicas não Histona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Transativadores/metabolismo , Proteínas ras/metabolismo , Acetiltransferases/isolamento & purificação , Proteínas Adaptadoras de Transdução de Sinal , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/isolamento & purificação , Animais , Caenorhabditis elegans/citologia , Proteínas de Caenorhabditis elegans/genética , Linhagem da Célula/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/isolamento & purificação , DNA Complementar/análise , DNA Complementar/genética , Feminino , Histona Acetiltransferases , Histona Desacetilases/genética , Histona Desacetilases/isolamento & purificação , Lisina Acetiltransferase 5 , Substâncias Macromoleculares , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Mutação/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Transativadores/genética , Transativadores/isolamento & purificação , Fatores de Transcrição , Vulva/citologia , Vulva/crescimento & desenvolvimento , Vulva/metabolismo , Proteínas ras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA