Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Curr Biol ; 33(3): 517-532.e5, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36640763

RESUMO

The development of neuronal connectivity requires stabilization of dynamic axonal branches at sites of synapse formation. Models that explain how axonal branching is coupled to synaptogenesis postulate molecular regulators acting in a spatiotemporally restricted fashion to ensure branching toward future synaptic partners while also stabilizing the emerging synaptic contacts between such partners. We investigated this question using neuronal circuit development in the Drosophila brain as a model system. We report that epidermal growth factor receptor (EGFR) activity is required in presynaptic axonal branches during two distinct temporal intervals to regulate circuit wiring in the developing Drosophila visual system. EGFR is required early to regulate primary axonal branching. EGFR activity is then independently required at a later stage to prevent degradation of the synaptic active zone protein Bruchpilot (Brp). Inactivation of EGFR results in a local increase of autophagy in presynaptic branches and the translocation of active zone proteins into autophagic vesicles. The protection of synaptic material during this later interval of wiring ensures the stabilization of terminal branches, circuit connectivity, and appropriate visual behavior. Phenotypes of EGFR inactivation can be rescued by increasing Brp levels or downregulating autophagy. In summary, we identify a temporally restricted molecular mechanism required for coupling axonal branching and synaptic stabilization that contributes to the emergence of neuronal wiring specificity.


Assuntos
Proteínas de Drosophila , Animais , Proteínas de Drosophila/metabolismo , Axônios/fisiologia , Drosophila/genética , Receptores ErbB/metabolismo , Autofagia , Sinapses/fisiologia , Receptores de Peptídeos de Invertebrados/metabolismo
2.
Life Sci Alliance ; 6(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36720500

RESUMO

FTSJ1 is a conserved human 2'-O-methyltransferase (Nm-MTase) that modifies several tRNAs at position 32 and the wobble position 34 in the anticodon loop. Its loss of function has been linked to X-linked intellectual disability (XLID), and more recently to cancers. However, the molecular mechanisms underlying these pathologies are currently unclear. Here, we report a novel FTSJ1 pathogenic variant from an X-linked intellectual disability patient. Using blood cells derived from this patient and other affected individuals carrying FTSJ1 mutations, we performed an unbiased and comprehensive RiboMethSeq analysis to map the ribose methylation on all human tRNAs and identify novel targets. In addition, we performed a transcriptome analysis in these cells and found that several genes previously associated with intellectual disability and cancers were deregulated. We also found changes in the miRNA population that suggest potential cross-regulation of some miRNAs with these key mRNA targets. Finally, we show that differentiation of FTSJ1-depleted human neural progenitor cells into neurons displays long and thin spine neurites compared with control cells. These defects are also observed in Drosophila and are associated with long-term memory deficits. Altogether, our study adds insight into FTSJ1 pathologies in humans and flies by the identification of novel FTSJ1 targets and the defect in neuron morphology.


Assuntos
Deficiência Intelectual , Ribose , Humanos , Metilação , Deficiência Intelectual/genética , Metiltransferases/genética , RNA de Transferência/genética , RNA de Transferência/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética
3.
Elife ; 112022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36214451

RESUMO

The differentiation of oligodendroglia from oligodendrocyte precursor cells (OPCs) to complex and extensive myelinating oligodendrocytes (OLs) is a multistep process that involves large-scale morphological changes with significant strain on the cytoskeleton. While key chromatin and transcriptional regulators of differentiation have been identified, their target genes responsible for the morphological changes occurring during OL myelination are still largely unknown. Here, we show that the regulator of focal adhesion, Tensin3 (Tns3), is a direct target gene of Olig2, Chd7, and Chd8, transcriptional regulators of OL differentiation. Tns3 is transiently upregulated and localized to cell processes of immature OLs, together with integrin-ß1, a key mediator of survival at this transient stage. Constitutive <i>Tns3</i> loss of function leads to reduced viability in mouse and humans, with surviving knockout mice still expressing Tns3 in oligodendroglia. Acute deletion of <i>Tns3</i> in vivo, either in postnatal neural stem cells (NSCs) or in OPCs, leads to a twofold reduction in OL numbers. We find that the transient upregulation of Tns3 is required to protect differentiating OPCs and immature OLs from cell death by preventing the upregulation of p53, a key regulator of apoptosis. Altogether, our findings reveal a specific time window during which transcriptional upregulation of Tns3 in immature OLs is required for OL differentiation likely by mediating integrin-ß1 survival signaling to the actin cytoskeleton as OL undergo the large morphological changes required for their terminal differentiation.


Assuntos
Adesões Focais , Proteína Supressora de Tumor p53 , Humanos , Animais , Camundongos , Adesões Focais/metabolismo , Proteína Supressora de Tumor p53/genética , Oligodendroglia/metabolismo , Diferenciação Celular/genética , Camundongos Knockout , Fatores de Transcrição/metabolismo , Cromatina/metabolismo , Integrinas/metabolismo
4.
Elife ; 102021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34515635

RESUMO

The Amyloid Precursor Protein (APP) and its homologues are transmembrane proteins required for various aspects of neuronal development and activity, whose molecular function is unknown. Specifically, it is unclear whether APP acts as a receptor, and if so what its ligand(s) may be. We show that APP binds the Wnt ligands Wnt3a and Wnt5a and that this binding regulates APP protein levels. Wnt3a binding promotes full-length APP (flAPP) recycling and stability. In contrast, Wnt5a promotes APP targeting to lysosomal compartments and reduces flAPP levels. A conserved Cysteine-Rich Domain (CRD) in the extracellular portion of APP is required for Wnt binding, and deletion of the CRD abrogates the effects of Wnts on flAPP levels and trafficking. Finally, loss of APP results in increased axonal and reduced dendritic growth of mouse embryonic primary cortical neurons. This phenotype can be cell-autonomously rescued by full length, but not CRD-deleted, APP and regulated by Wnt ligands in a CRD-dependent manner.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Receptores Wnt/metabolismo , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/citologia , Células Cultivadas , Clonagem Molecular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Deleção de Genes , Regulação da Expressão Gênica/fisiologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Corpos Pedunculados/citologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Transporte Proteico , Receptores Wnt/genética , Transdução de Sinais
5.
Sci Adv ; 7(26)2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34162555

RESUMO

The identity of the cell of origin is a key determinant of cancer subtype, progression, and prognosis. Group 3 medulloblastoma (MB) is a malignant childhood brain cancer with poor prognosis and few candidates as putative cell of origin. We overexpressed the group 3 MB genetic drivers MYC and Gfi1 in different candidate cells of origin in the postnatal mouse cerebellum. We found that S100b+ cells are competent to initiate group 3 MB, and we observed that S100b+ cells have higher levels of Notch1 pathway activity compared to Math1+ cells. We found that additional activation of Notch1 in Math1+ and Sox2+ cells was sufficient to induce group 3 MB upon MYC/Gfi1 expression. Together, our data suggest that the Notch1 pathway plays a critical role in group 3 MB initiation.

6.
J Hematol Oncol ; 11(1): 108, 2018 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-30144809

RESUMO

BACKGROUND: The CNOT3 protein is a subunit of the CCR4-NOT complex, which is involved in mRNA degradation. We recently identified CNOT3 loss-of-function mutations in patients with T-cell acute lymphoblastic leukemia (T-ALL). METHODS: Here, we use different Drosophila melanogaster eye cancer models to study the potential tumor suppressor function of Not3, the CNOT3 orthologue, and other members of the CCR4-NOT complex. RESULTS: Our data show that knockdown of Not3, the structural components Not1/Not2, and the deadenylases twin/Pop2 all result in increased tumor formation. In addition, overexpression of Not3 could reduce tumor formation. Not3 downregulation has a mild but broad effect on gene expression and leads to increased levels of genes involved in DNA replication and ribosome biogenesis. CycB upregulation also contributes to the Not3 tumor phenotype. Similar findings were obtained in human T-ALL cell lines, pointing out the conserved function of Not3. CONCLUSIONS: Together, our data establish a critical role for Not3 and the entire CCR4-NOT complex as tumor suppressor.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/patogenicidade , Neoplasias Oculares/genética , Genes Supressores de Tumor/fisiologia , Proteínas de Ligação a RNA/metabolismo , Ribonucleases/metabolismo , Animais , Modelos Animais de Doenças , Neoplasias Oculares/metabolismo , Humanos , Ligação Proteica
7.
EMBO Mol Med ; 7(4): 423-37, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25693964

RESUMO

Loss of function of the FMR1 gene leads to fragile X syndrome (FXS), the most common form of intellectual disability. The loss of FMR1 function is usually caused by epigenetic silencing of the FMR1 promoter leading to expansion and subsequent methylation of a CGG repeat in the 5' untranslated region. Very few coding sequence variations have been experimentally characterized and shown to be causal to the disease. Here, we describe a novel FMR1 mutation and reveal an unexpected nuclear export function for the C-terminus of FMRP. We screened a cohort of patients with typical FXS symptoms who tested negative for CGG repeat expansion in the FMR1 locus. In one patient, we identified a guanine insertion in FMR1 exon 15. This mutation alters the open reading frame creating a short novel C-terminal sequence, followed by a stop codon. We find that this novel peptide encodes a functional nuclear localization signal (NLS) targeting the patient FMRP to the nucleolus in human cells. We also reveal an evolutionarily conserved nuclear export function associated with the endogenous C-terminus of FMRP. In vivo analyses in Drosophila demonstrate that a patient-mimetic mutation alters the localization and function of Dfmrp in neurons, leading to neomorphic neuronal phenotypes.


Assuntos
Núcleo Celular , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Mutação , Sinais de Localização Nuclear , Expansão das Repetições de Trinucleotídeos , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Drosophila melanogaster , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Humanos , Masculino , Camundongos , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico/genética
8.
Elife ; 3: e01699, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24755286

RESUMO

Axonal branching allows a neuron to connect to several targets, increasing neuronal circuit complexity. While axonal branching is well described, the mechanisms that control it remain largely unknown. We find that in the Drosophila CNS branches develop through a process of excessive growth followed by pruning. In vivo high-resolution live imaging of developing brains as well as loss and gain of function experiments show that activation of Epidermal Growth Factor Receptor (EGFR) is necessary for branch dynamics and the final branching pattern. Live imaging also reveals that intrinsic asymmetry in EGFR localization regulates the balance between dynamic and static filopodia. Elimination of signaling asymmetry by either loss or gain of EGFR function results in reduced dynamics leading to excessive branch formation. In summary, we propose that the dynamic process of axon branch development is mediated by differential local distribution of signaling receptors. DOI: http://dx.doi.org/10.7554/eLife.01699.001.


Assuntos
Axônios/fisiologia , Plasticidade Neuronal , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Drosophila , Proteínas de Drosophila/metabolismo , Receptores ErbB/metabolismo , Imagem Óptica , Receptores de Peptídeos de Invertebrados/metabolismo
9.
PLoS One ; 8(11): e81791, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24303071

RESUMO

We recently reported that duplication of the E3 ubiquitin ligase HUWE1 results in intellectual disability (ID) in male patients. However, the underlying molecular mechanism remains unknown. We used Drosophila melanogaster as a model to investigate the effect of increased HUWE1 levels on the developing nervous system. Similar to the observed levels in patients we overexpressed the HUWE1 mRNA about 2-fold in the fly. The development of the mushroom body and neuromuscular junctions were not altered, and basal neurotransmission was unaffected. These data are in agreement with normal learning and memory in the courtship conditioning paradigm. However, a disturbed branching phenotype at the axon terminals of the dorsal cluster neurons (DCN) was detected. Interestingly, overexpression of HUWE1 was found to decrease the protein levels of dishevelled (dsh) by 50%. As dsh as well as Fz2 mutant flies showed the same disturbed DCN branching phenotype, and the constitutive active homolog of ß-catenin, armadillo, could partially rescue this phenotype, our data strongly suggest that increased dosage of HUWE1 compromises the Wnt/ß-catenin pathway possibly by enhancing the degradation of dsh.


Assuntos
Axônios/metabolismo , Deficiência Intelectual/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Expressão Gênica , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Aprendizagem , Memória , Corpos Pedunculados/metabolismo , Corpos Pedunculados/fisiopatologia , Junção Neuromuscular/metabolismo , Neurônios/metabolismo , Transmissão Sináptica , Proteínas Supressoras de Tumor
10.
Nat Genet ; 45(2): 186-90, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23263491

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


Assuntos
Exoma/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Ribossômicas/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Drosophila melanogaster , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação/genética , Polirribossomos/genética , Interferência de RNA , Proteína Ribossômica L10 , Saccharomyces cerevisiae , Alinhamento de Sequência
11.
Nat Commun ; 1: 18, 2010 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-20975679

RESUMO

Intestinal cells are constantly produced from a stem cell reservoir that gives rise to proliferating transient amplifying cells, which subsequently differentiate into one of the four principal cell types. Signalling pathways, including the Notch signalling pathway, coordinate these differentiation processes and their deregulation may cause cancer. Pharmacological inhibition through γ-secretase inhibitors or genetic inactivation of the Notch signalling pathway results in the complete loss of proliferating crypt progenitors due to their conversion into post-mitotic goblet cells. The basic helix-loop-helix transcription factor Math1 is essential for intestinal secretory cell differentiation. Because of the critical roles of both Math1 and Notch signalling in intestinal homeostasis and neoplastic transformation, we sought to determine the genetic hierarchy regulating the differentiation of intestinal stem cells into secretory cells. In this paper, we demonstrate that the conversion of intestinal stem cells into goblet cells upon inhibition of the Notch signalling pathway requires Math1.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Intestinos/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Dibenzazepinas/farmacologia , Células Caliciformes/citologia , Células Caliciformes/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Técnicas In Vitro , Camundongos , Receptores Notch/genética , Receptores Notch/metabolismo , Células-Tronco/efeitos dos fármacos
12.
PLoS Biol ; 8(7): e1000435, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20668662

RESUMO

A comprehensive systems-level understanding of developmental programs requires the mapping of the underlying gene regulatory networks. While significant progress has been made in mapping a few such networks, almost all gene regulatory networks underlying cell-fate specification remain unknown and their discovery is significantly hampered by the paucity of generalized, in vivo validated tools of target gene and functional enhancer discovery. We combined genetic transcriptome perturbations and comprehensive computational analyses to identify a large cohort of target genes of the proneural and tumor suppressor factor Atonal, which specifies the switch from undifferentiated pluripotent cells to R8 photoreceptor neurons during larval development. Extensive in vivo validations of the predicted targets for the proneural factor Atonal demonstrate a 50% success rate of bona fide targets. Furthermore we show that these enhancers are functionally conserved by cloning orthologous enhancers from Drosophila ananassae and D. virilis in D. melanogaster. Finally, to investigate cis-regulatory cross-talk between Ato and other retinal differentiation transcription factors (TFs), we performed motif analyses and independent target predictions for Eyeless, Senseless, Suppressor of Hairless, Rough, and Glass. Our analyses show that cisTargetX identifies the correct motif from a set of coexpressed genes and accurately predicts target genes of individual TFs. The validated set of novel Ato targets exhibit functional enrichment of signaling molecules and a subset is predicted to be coregulated by other TFs within the retinal gene regulatory network.


Assuntos
Drosophila melanogaster/genética , Elementos Facilitadores Genéticos/genética , Perfilação da Expressão Gênica , Genes de Insetos/genética , Genoma/genética , Retina/metabolismo , Sensação/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sítios de Ligação , Sequência Conservada , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes/genética , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fosfoproteínas/metabolismo , Reprodutibilidade dos Testes , Retina/citologia , Retina/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo , Transcrição Gênica
13.
J Cell Biol ; 187(1): 91-100, 2009 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-19786578

RESUMO

Merkel cells (MCs) are located in the touch-sensitive area of the epidermis and mediate mechanotransduction in the skin. Whether MCs originate from embryonic epidermal or neural crest progenitors has been a matter of intense controversy since their discovery >130 yr ago. In addition, how MCs are maintained during adulthood is currently unknown. In this study, using lineage-tracing experiments, we show that MCs arise through the differentiation of epidermal progenitors during embryonic development. In adults, MCs undergo slow turnover and are replaced by cells originating from epidermal stem cells, not through the proliferation of differentiated MCs. Conditional deletion of the Atoh1/Math1 transcription factor in epidermal progenitors results in the absence of MCs in all body locations, including the whisker region. Our study demonstrates that MCs arise from the epidermis by an Atoh1-dependent mechanism and opens new avenues for study of MC functions in sensory perception, neuroendocrine signaling, and MC carcinoma.


Assuntos
Células Epidérmicas , Homeostase , Células de Merkel/citologia , Células de Merkel/fisiologia , Envelhecimento , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Caderinas/metabolismo , Diferenciação Celular , Linhagem da Célula , Epiderme/metabolismo , Epiderme/ultraestrutura , Técnica Direta de Fluorescência para Anticorpo , Imuno-Histoquímica , Integrases/genética , Integrases/metabolismo , Células de Merkel/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Crista Neural/citologia , Crista Neural/embriologia , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Pele/citologia , Pele/embriologia , Pele/metabolismo , Pele/ultraestrutura , Células-Tronco/citologia , Fatores de Tempo , Vibrissas/citologia , Vibrissas/embriologia , Vibrissas/metabolismo
14.
PLoS Biol ; 7(2): e39, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19243219

RESUMO

Colon cancer accounts for more than 10% of all cancer deaths annually. Our genetic evidence from Drosophila and previous in vitro studies of mammalian Atonal homolog 1 (Atoh1, also called Math1 or Hath1) suggest an anti-oncogenic function for the Atonal group of proneural basic helix-loop-helix transcription factors. We asked whether mouse Atoh1 and human ATOH1 act as tumor suppressor genes in vivo. Genetic knockouts in mouse and molecular analyses in the mouse and in human cancer cell lines support a tumor suppressor function for ATOH1. ATOH1 antagonizes tumor formation and growth by regulating proliferation and apoptosis, likely via activation of the Jun N-terminal kinase signaling pathway. Furthermore, colorectal cancer and Merkel cell carcinoma patients show genetic and epigenetic ATOH1 loss-of-function mutations. Our data indicate that ATOH1 may be an early target for oncogenic mutations in tissues where it instructs cellular differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinoma de Célula de Merkel/genética , Neoplasias Colorretais/genética , Genes Supressores de Tumor/fisiologia , Neoplasias Cutâneas/genética , Animais , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma de Célula de Merkel/metabolismo , Carcinoma de Célula de Merkel/patologia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Análise Mutacional de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Proteínas Quinases JNK Ativadas por Mitógeno , Masculino , Camundongos , Camundongos Knockout , Mutação , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
15.
PLoS Biol ; 7(2): e40, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19243220

RESUMO

The acquisition of terminal cell fate and onset of differentiation are instructed by cell type-specific master control genes. Loss of differentiation is frequently observed during cancer progression, but the underlying causes and mechanisms remain poorly understood. We tested the hypothesis that master regulators of differentiation may be key regulators of tumor formation. Using loss- and gain-of-function analyses in Drosophila, we describe a critical anti-oncogenic function for the atonal transcription factor in the fly retina, where atonal instructs tissue differentiation. In the tumor context, atonal acts by regulating cell proliferation and death via the JNK stress response pathway. Combined with evidence that atonal's mammalian homolog, ATOH1, is a tumor suppressor gene, our data support a critical, evolutionarily conserved, function for ato in oncogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Transformação Celular Neoplásica/genética , Drosophila , Neoplasias Oculares/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Proteínas do Tecido Nervoso/genética , Animais , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/genética , Diferenciação Celular/genética , Drosophila/embriologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neoplasias Oculares/metabolismo , Inativação Gênica , Proteínas do Tecido Nervoso/metabolismo , Organismos Geneticamente Modificados , Retina/citologia , Retina/embriologia
16.
PLoS Biol ; 4(11): e348, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17032066

RESUMO

The precise number and pattern of axonal connections generated during brain development regulates animal behavior. Therefore, understanding how developmental signals interact to regulate axonal extension and retraction to achieve precise neuronal connectivity is a fundamental goal of neurobiology. We investigated this question in the developing adult brain of Drosophila and find that it is regulated by crosstalk between Wnt, fibroblast growth factor (FGF) receptor, and Jun N-terminal kinase (JNK) signaling, but independent of neuronal activity. The Rac1 GTPase integrates a Wnt-Frizzled-Disheveled axon-stabilizing signal and a Branchless (FGF)-Breathless (FGF receptor) axon-retracting signal to modulate JNK activity. JNK activity is necessary and sufficient for axon extension, whereas the antagonistic Wnt and FGF signals act to balance the extension and retraction required for the generation of the precise wiring pattern.


Assuntos
Encéfalo/metabolismo , Encéfalo/fisiologia , Neurônios/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Axônios/metabolismo , Proteínas Desgrenhadas , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Imuno-Histoquímica , MAP Quinase Quinase 4/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Fenótipo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transgenes , Proteínas Wnt/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA