Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(4)2021 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33672712

RESUMO

Drought and salinity can result in cell dehydration and water unbalance in plants, which seriously diminish plant growth and development. Cellular water homeostasis maintained by aquaporin is one of the important strategies for plants to cope with these two stresses. In this study, a stress-induced aquaporin, ZxPIP1;3, belonging to the PIP1 subgroup, was identified from the succulent xerophyte Zygophyllum xanthoxylum. The subcellular localization showed that ZxPIP1;3-GFP was located in the plasma membrane. The overexpression of ZxPIP1;3 in Arabidopsis prompted plant growth under favorable condition. In addition, it also conferred salt and drought tolerance with better water status as well as less ion toxicity and membrane injury, which led to more efficient photosynthesis and improved growth vigor via inducing stress-related responsive genes. This study reveals the molecular mechanisms of xerophytes' stress tolerance and provides a valuable candidate that could be used in genetic engineering to improve crop growth and stress tolerance.


Assuntos
Adaptação Fisiológica , Aquaporinas/genética , Regulação da Expressão Gênica de Plantas , Desenvolvimento Vegetal , Proteínas de Plantas/genética , Estresse Fisiológico , Zygophyllum/crescimento & desenvolvimento , Zygophyllum/genética , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/genética , Sequência de Aminoácidos , Aquaporinas/metabolismo , Arabidopsis/genética , Secas , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Pressão Osmótica , Desenvolvimento Vegetal/efeitos dos fármacos , Desenvolvimento Vegetal/genética , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Cloreto de Sódio/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Zygophyllum/efeitos dos fármacos
2.
BMC Genomics ; 19(1): 435, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29866045

RESUMO

BACKGROUND: Lung cancer is a very heterogeneous disease that can be pathologically classified into different subtypes including small-cell lung carcinoma (SCLC), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC) and large-cell carcinoma (LCC). Although much progress has been made towards the oncogenic mechanism of each subtype, transcriptional circuits mediating the upstream signaling pathways and downstream functional consequences remain to be systematically studied. RESULTS: Here we trained a one-class support vector machine (OC-SVM) model to establish a general transcription factor (TF) regulatory network containing 325 TFs and 18724 target genes. We then applied this network to lung cancer subtypes and identified those deregulated TFs and downstream targets. We found that the TP63/SOX2/DMRT3 module was specific to LUSC, corresponding to squamous epithelial differentiation and/or survival. Moreover, the LEF1/MSC module was specifically activated in LUAD and likely to confer epithelial-to-mesenchymal transition, known important for cancer malignant progression and metastasis. The proneural factor, ASCL1, was specifically up-regulated in SCLC which is known to have a neuroendocrine phenotype. Also, ID2 was differentially regulated between SCLC and LUSC, with its up-regulation in SCLC linking to energy supply for fast mitosis and its down-regulation in LUSC linking to the attenuation of immune response. We further described the landscape of TF regulation among the three major subtypes of lung cancer, highlighting their functional commonalities and specificities. CONCLUSIONS: Our approach uncovered the landscape of transcriptional deregulation in lung cancer, and provided a useful resource of TF regulatory network for future studies.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Transcrição Gênica , Redes Reguladoras de Genes , Humanos , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
3.
DNA Res ; 23(6): 571-580, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27559081

RESUMO

There is growing recognition that small open reading frames (sORFs) encoding peptides shorter than 100 amino acids are an important class of functional elements in the eukaryotic genome, with several already identified to play critical roles in growth, development, and disease. However, our understanding of their biological importance has been hindered owing to the significant technical challenges limiting their annotation. Here we combined ultra-deep sequencing of ribosome-associated poly-adenylated RNAs with rigorous conservation analysis to identify a comprehensive population of translated sORFs during early Drosophila embryogenesis. In total, we identify 399 sORFs, including those previously annotated but without evidence of translational capacity, those found within transcripts previously classified as non-coding, and those not previously known to be transcribed. Further, we find, for the first time, evidence for translation of many sORFs with different isoforms, suggesting their regulation is as complex as longer ORFs. Furthermore, many sORFs are found not associated with ribosomes in late-stage Drosophila S2 cells, suggesting that many of the translated sORFs may have stage-specific functions during embryogenesis. These results thus provide the first comprehensive annotation of the sORFs present during early Drosophila embryogenesis, a necessary basis for a detailed delineation of their function in embryogenesis and other biological processes.


Assuntos
Sequência Conservada , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Fases de Leitura Aberta , RNA Mensageiro/genética , Animais , Drosophila/embriologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Anotação de Sequência Molecular , RNA Mensageiro/química , Ribossomos/metabolismo , Análise de Sequência de RNA
4.
Dev Biol ; 416(2): 312-23, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27339292

RESUMO

The Drosophila larval ovary morphogenesis mainly involves coordinated development of somatic and germ cell lineages that is essential for forming a correct number of niche-germline stem cell (GSC) units (ovarioles) in the adult ovary. Ecdysone, Insulin, Activin, Dpp and EGFR signaling pathways form a regulatory network that orchestrates ovarian soma and germ line throughout larval development. Identification and characterization of additional genes or machineries involved in this process will provide more insights into the underlying mechanisms. Here, we show that the core microRNA (miRNA) pathway components Drosha and Pasha are required for coordinated development of somatic and germ cell precursors in the larval ovary. Drosha or pasha mutants display defective proliferation of primordial germ cells (PGCs), the precursors of GSCs prior to late third larval instar (LL3) and promoted PGC differentiation at LL3. In the mean time, loss of Drosha or Pasha function perturbs somatic precursor development, causing defects in formation of terminal filaments (TFs), a major composition of the GSC niche at LL3, as well as in TF precursor accumulation at early larval stages. Comparative analysis of the mutant phenotypes reveals that three other key miRNA pathway components, Dicer-1 (Dcr-1), Loquacious (Loqs) and Argonaute-1 (Ago-1) have similar effects as Drosha and Pasha indicated above, suggesting a role of the canonical miRNA pathway in the ovary development. Furthermore, genome-wide screening and genetic studies identify a set of Drosha-controlled miRNAs including miR-8, miR-14, miR-33, miR-184, miR-317 and let-7-C that function in this gonadogenesis. Taken together, this study provides the first ever demonstration that miRNA-mediated regulation is involved in the Drosophila larval ovary morphogenesis.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , MicroRNAs/genética , Ovário/crescimento & desenvolvimento , Proteínas de Ligação a RNA/fisiologia , Ribonuclease III/fisiologia , Animais , Diferenciação Celular , Citoesqueleto/ultraestrutura , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/crescimento & desenvolvimento , Células Germinativas Embrionárias/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Larva , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Microscopia de Fluorescência , Organogênese , Ovário/citologia , Interferência de RNA , Proteínas de Ligação a RNA/genética , Ribonuclease III/deficiência , Ribonuclease III/genética , Nicho de Células-Tronco
5.
PLoS One ; 9(7): e103473, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25068272

RESUMO

In the Drosophila oogenesis, germline stem cells (GSCs) continuously self-renew and differentiate into daughter cells for consecutive germline lineage commitment. This developmental process has become an in vivo working platform for studying adult stem cell fate regulation. An increasing number of studies have shown that while concerted actions of extrinsic signals from the niche and intrinsic regulatory machineries control GSC self-renewal and germline differentiation, epigenetic regulation is implicated in the process. Here, we report that Brahma (Brm), the ATPase subunit of the Drosophila SWI/SNF chromatin-remodeling complexes, is required for maintaining GSC fate. Removal or knockdown of Brm function in either germline or niche cells causes a GSC loss, but does not disrupt normal germline differentiation within the germarium evidenced at the molecular and morphological levels. There are two Drosophila SWI/SNF complexes: the Brm-associated protein (BAP) complex and the polybromo-containing BAP (PBAP) complex. More genetic studies reveal that mutations in polybromo/bap180, rather than gene encoding Osa, the BAP complex-specific subunit, elicit a defect in GSC maintenance reminiscent of the brm mutant phenotype. Further genetic interaction test suggests a functional association between brm and polybromo in controlling GSC self-renewal. Taken together, studies in this paper provide the first demonstration that Brm in the form of the PBAP complex functions in the GSC fate regulation.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Drosophila/genética , Óvulo/metabolismo , Transativadores/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Microscopia Confocal , Mutação , Ovário/citologia , Ligação Proteica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
6.
Dev Biol ; 384(1): 1-12, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24120347

RESUMO

Maintenance of adult stem cells is largely dependent on the balance between their self-renewal and differentiation. The Drosophila ovarian germline stem cells (GSCs) provide a powerful in vivo system for studying stem cell fate regulation. It has been shown that maintaining the GSC population involves both genetic and epigenetic mechanisms. Although the role of epigenetic regulation in this process is evident, the underlying mechanisms remain to be further explored. In this study, we find that Enoki mushroom (Enok), a Drosophila putative MYST family histone acetyltransferase controls GSC maintenance in the ovary at multiple levels. Removal or knockdown of Enok in the germline causes a GSC maintenance defect. Further studies show that the cell-autonomous role of Enok in maintaining GSCs is not dependent on the BMP/Bam pathway. Interestingly, molecular studies reveal an ectopic expression of Bruno, an RNA binding protein, in the GSCs and their differentiating daughter cells elicited by the germline Enok deficiency. Misexpression of Bruno in GSCs and their immediate descendants results in a GSC loss that can be exacerbated by incorporating one copy of enok mutant allele. These data suggest a role for Bruno in Enok-controlled GSC maintenance. In addition, we observe that Enok is required for maintaining GSCs non-autonomously. Compromised expression of enok in the niche cells impairs the niche maintenance and BMP signal output, thereby causing defective GSC maintenance. This is the first demonstration that the niche size control requires an epigenetic mechanism. Taken together, studies in this paper provide new insights into the GSC fate regulation.


Assuntos
Proteínas de Drosophila/genética , Drosophila/embriologia , Drosophila/genética , Células Germinativas/metabolismo , Histona Acetiltransferases/genética , Proteínas de Ligação a RNA/genética , Nicho de Células-Tronco/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Embrião não Mamífero/metabolismo , Epigenômica , Feminino , Histona Acetiltransferases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/genética
7.
Dev Biol ; 379(2): 167-81, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23624310

RESUMO

The Drosophila ovarian germline stem cells (GSCs) constantly experience self-renewal and differentiation, ensuring the female fertility throughout life. The balance between GSC self-renewal and differentiation is exquisitely regulated by the stem cell niche, the stem cells themselves and systemic factors. Increasing evidence has shown that the GSC regulation also involves epigenetic mechanisms including chromatin remodeling and histone modification. Here, we find that dBre1, an E3 ubiquitin ligase, functions in controlling GSC self-renewal and germ cell differentiation via distinct mechanisms. Removal or knock down of dBre1 function in the germline or somatic niche cell lineage leads to a gradual GSC loss and disruption of H3K4 trimethylation in the Drosophila ovary. Further studies suggest that the defective GSC maintenance is attributable to compromised BMP signaling emitted from the stem cell niche and impaired adhesion of GSCs to their niche. On the other hand, dBre1-RNAi expression in escort cells causes a loss of H3K4 trimethylation and accumulation of spectrosome-containing single germ cells in the germarium. Reducing dpp or dally levels suppresses the germ cell differentiation defects, indicating that dBre1 limits BMP signaling activities for the differentiation control. Strikingly, all phenotypes observed in dBre1 mutant ovaries can be mimicked by RNAi-based reduced expression of dSet1, a Drosophila H3K4 trimethylase. Moreover, genetic studies favor that dBre1 interacts with dSet1 in controlling GSC maintenance and germ cell differentiation. Taken together, we identify a dBre1/dSet1-dependent pathway for the H3K4 methylation involved in the cell fate regulation in the Drosophila ovary.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Diferenciação Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Células Germinativas/fisiologia , Ovário/citologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Primers do DNA/genética , Epigênese Genética/fisiologia , Feminino , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metilação , Microscopia de Fluorescência , Ovário/embriologia , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Ubiquitina-Proteína Ligases/genética
8.
Fly (Austin) ; 5(2): 81-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21245664

RESUMO

The epithelial follicle cell layer over the egg chamber in Drosophila ovary undergoes patterning and morphogenesis at oogenesis. These developmental processes are essential for constructing the eggshell and establishing the body axes of the egg and resultant embryo, thereby being crucial for the egg development. We have previously shown that lethal(2)giant larvae (lgl), a Drosophila neoplastic tumor suppressor gene (nTSG) is required for the posterior follicle cell (PFC) fate induction during antero-posterior pattern formation of the follicular epithelium. In this report, we further characterize lgl in this epithelium patterning and the morphogenetic changes of specified border cells. Genetic interactions of lgl with discs large (dlg) and scribble (scrib), another two nTSGs in specifying the PFC fate reveal a cooperative role of this group of genes. Meanwhile, we find that loss of lgl function causes failure of follicle cells at the anterior to differentiate properly. The clonal analysis further indicates that lgl is necessary not only for the border cell differentiation, but also for control of the collective border cell migration via presumably modulating the apico-basal polarity and cell adhesion. Overall, we identify Lgl as an essential factor in regulating differentiation and morphogenetic movement of the ovarian epithelial follicle cells.


Assuntos
Diferenciação Celular/genética , Movimento Celular/genética , Proteínas de Drosophila/fisiologia , Drosophila/genética , Morfogênese/genética , Folículo Ovariano/crescimento & desenvolvimento , Proteínas Supressoras de Tumor/fisiologia , Animais , Drosophila/citologia , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epitélio/metabolismo , Feminino , Folículo Ovariano/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
10.
BMC Dev Biol ; 9: 60, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19948068

RESUMO

BACKGROUND: Proper patterning of the follicle cell epithelium over the egg chamber is essential for the Drosophila egg development. Differentiation of the epithelium into several distinct cell types along the anterior-posterior axis requires coordinated activities of multiple signaling pathways. Previously, we reported that lethal(2)giant larvae (lgl), a Drosophila tumor suppressor gene, is required in the follicle cells for the posterior follicle cell (PFC) fate induction at mid-oogenesis. Here we explore the role of another two tumor suppressor genes, scribble (scrib) and discs large (dlg), in the epithelial patterning. RESULTS: We found that removal of scrib or dlg function from the follicle cells at posterior terminal of the egg chamber causes a complete loss of the PFC fate. Aberrant specification and differentiation of the PFCs in the mosaic clones can be ascribed to defects in coordinated activation of the EGFR, JAK and Notch signaling pathways in the multilayered cells. Meanwhile, the clonal analysis revealed that loss-of-function mutations in scrib/dlg at the anterior domains result in a partially penetrant phenotype of defective induction of the stretched and centripetal cell fate, whereas specification of the border cell fate can still occur in the most anterior region of the mutant clones. Further, we showed that scrib genetically interacts with dlg in regulating posterior patterning of the epithelium. CONCLUSION: In this study we provide evidence that scrib and dlg function differentially in anterior and posterior patterning of the follicular epithelium at oogenesis. Further genetic analysis indicates that scrib and dlg act in a common pathway to regulate PFC fate induction. This study may open another window for elucidating role of scrib/dlg in controlling epithelial polarity and cell proliferation during development.


Assuntos
Proteínas de Drosophila/fisiologia , Epitélio/embriologia , Epitélio/metabolismo , Proteínas de Membrana/fisiologia , Oogênese/fisiologia , Folículo Ovariano/embriologia , Folículo Ovariano/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Drosophila , Proteínas de Drosophila/genética , Feminino , Proteínas de Membrana/genética , Oogênese/genética , Folículo Ovariano/citologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/genética
11.
Cell Biochem Biophys ; 55(3): 117-26, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19649745

RESUMO

S100A11, as a member of S100 protein family, while featuring the common identities as the other EF-hand Ca(2+)-binding family members, has its own individual characteristics. S100A11 is widely expressed in multiple tissues, and is located in cytoplasm, nucleus, and even cell periphery. S100A11 exists as a non-covalent homodimer with an antiparallel conformation. Ca(2+) binding to S100A11 would trigger conformational changes which would expose the hydrophobic cleft of S100A11 and facilitate its interaction with target proteins. Since S100A11 appears to lack enzymatic activity, in this article, corresponding to a variety of its target proteins, we systematically describe the biological roles of S100A11 and its possible mechanism in the processes of inflammation, regulation of enzyme activity, and cell growth regulation. As a dual cell growth mediator, S100A11 acts as either a tumor suppressor or promoter in many different types of tumors and would play respective roles in influencing the proliferation of the cancer cells. We intend to illustrate the biological function of the S100 protein, and shed light on the further research, which will provide us with a better understanding of it.


Assuntos
Proteínas S100/química , Proteínas S100/metabolismo , Proteínas S100/farmacologia , Relação Estrutura-Atividade , Animais , Anticarcinógenos/química , Anticarcinógenos/metabolismo , Anticarcinógenos/farmacologia , Cálcio/metabolismo , Carcinógenos/química , Carcinógenos/metabolismo , Carcinógenos/farmacologia , Proliferação de Células/efeitos dos fármacos , Motivos EF Hand , Humanos , Inflamação/metabolismo
12.
Protein Pept Lett ; 16(8): 977-83, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19689425

RESUMO

Reversible acetylation on lysine residues, a crucial post-translational modification (PTM) for both histone and non-histone proteins, governs many central cellular processes. Due to limited data and lack of a clear acetylation consensus sequence, little research has focused on prediction of lysine acetylation sites. Incorporating almost all currently available lysine acetylation information, and using the support vector machine (SVM) method along with coding schema for protein sequence coupling patterns, we propose here a novel lysine acetylation prediction algorithm: LysAcet. When compared with other methods or existing tools, LysAcet is the best predictor of lysine acetylation, with K-fold (5- and 10-) and jackknife cross-validation accuracies of 75.89%, 76.73%, and 77.16%, respectively. LysAcet's superior predictive accuracy is attributed primarily to the use of sequence coupling patterns, which describe the relative position of two amino acids. LysAcet contributes to the limited PTM prediction research on lysine epsilon-acetylation, and may serve as a complementary in-silicon approach for exploring acetylation on proteomes. An online web server is freely available at http://www.biosino.org/LysAcet/.


Assuntos
Algoritmos , Lisina/metabolismo , Proteínas/metabolismo , Acetilação , Inteligência Artificial , Modelos Biológicos
13.
Genes Dev ; 22(18): 2578-90, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18794353

RESUMO

Morphogens are secreted signaling molecules that form concentration gradients and control cell fate in developing tissues. During development, it is essential that morphogen range is strictly regulated in order for correct cell type specification to occur. One of the best characterized morphogens is Drosophila Decapentaplegic (Dpp), a BMP signaling molecule that patterns the dorsal ectoderm of the embryo by activating the Mad and Medea (Med) transcription factors. We demonstrate that there is a spatial and temporal expansion of the expression patterns of Dpp target genes in SUMO pathway mutant embryos. We identify Med as the primary SUMOylation target in the Dpp pathway, and show that failure to SUMOylate Med leads to the increased Dpp signaling range observed in the SUMO pathway mutant embryos. Med is SUMO modified in the nucleus, and we provide evidence that SUMOylation triggers Med nuclear export. Hence, Med SUMOylation provides a mechanism by which nuclei can continue to monitor the presence of extracellular Dpp signal to activate target gene expression for an appropriate duration. Overall, our results identify an unusual strategy for regulating morphogen range that, rather than impacting on the morphogen itself, targets an intracellular transducer.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Transdução de Sinais , Proteína Smad4/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Mutação , Processamento de Proteína Pós-Traducional
14.
Cell Res ; 18(3): 372-84, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18268543

RESUMO

The intricately regulated differentiation of the somatic follicle cell lineages into distinct subpopulations with specific functions plays an essential role in Drosophila egg development. At early oogenesis, induction of the stalk cells generates the first anteroposterior (AP) asymmetry in the egg chamber by inducing the posterior localization of the oocyte. Later, the properly specified posterior follicle cells signal to polarize the oocyte along the AP and dorsoventral (DV) axes at mid-oogenesis. Here, we show that lethal(2)giant larvae (lgl), a Drosophila tumor suppressor gene, is required in the follicle cells for the differentiation of both stalk cells and posterior follicle cells. Loss-of-function mutations in lgl cause oocyte mispositioning in the younger one of the fused chambers, due to lack of the stalk. Removal of lgl function from the posterior follicle cells using the FLP/FRT system results in loss of the oocyte polarity that is elicited by the failure of those posterior cells to differentiate normally. Thus, we provide the first demonstration that lgl is implicated in the formation of the initial AP asymmetry and the patterning of the AP and DV axes in the oocyte by acting in the specification of a subset of somatic follicle cells.


Assuntos
Polaridade Celular/fisiologia , Proteínas de Drosophila/metabolismo , Oócitos/metabolismo , Oogênese/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Masculino , Mutação , Oócitos/citologia , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Proteínas Supressoras de Tumor/genética
15.
Int J Dev Biol ; 49(7): 873-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16172984

RESUMO

In an effort to identify the role of Rab11, a small GTP binding protein, during Drosophila differentiation, phenotypic manifestations associated with different alleles of Rab11 were studied. The phenotypes ranged from eye-defects, bristle abnormalities and sterility to lethality during various developmental stages. In this paper, our focus is targeted on eye defects caused by Rab11 mutations. A novel P-element insertion in the Rab11 locus, Rab11mo, displayed characteristic retinal anomalies, which could be reverted by P-element excision and expression of Rab11+ transgenes. During larval development, Rab11 is widely synthesized in photoreceptor cells and localizes to the rhabdomeres and lamina neuropil in adult eyes. Photoreceptors and associated bristles failed to be formed in homozygous clones generated in Rab11EP(3)3017 eyes. Decreased levels of Rab11 protein and increased cell death in Rab11mo third-instar larval eye-antennal discs suggest that the retinal defects originate during larval development. Our data indicate a requirement for Rab11 in ommatidial differentiation during Drosophila eye development.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Olho/crescimento & desenvolvimento , Olho/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Alelos , Animais , Animais Geneticamente Modificados , Morte Celular , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Fotorreceptoras/metabolismo , Proteínas rab de Ligação ao GTP/genética
16.
Development ; 130(23): 5705-16, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14534137

RESUMO

Genetic evidence suggests that the Drosophila ectoderm is patterned by a spatial gradient of bone morphogenetic protein (BMP). Here we compare patterns of two related cellular responses, both signal-dependent phosphorylation of the BMP-regulated R-SMAD, MAD, and signal-dependent changes in levels and sub-cellular distribution of the co-SMAD Medea. Our data demonstrate that nuclear accumulation of the co-SMAD Medea requires a BMP signal during blastoderm and gastrula stages. During this period, nuclear co-SMAD responses occur in three distinct patterns. At the end of blastoderm, a broad dorsal domain of weak SMAD response is detected. During early gastrulation, this domain narrows to a thin stripe of strong SMAD response at the dorsal midline. SMAD response levels continue to rise in the dorsal midline region during gastrulation, and flanking plateaus of weak responses are detected in dorsolateral cells. Thus, the thresholds for gene expression responses are implicit in the levels of SMAD responses during gastrulation. Both BMP ligands, DPP and Screw, are required for nuclear co-SMAD responses during these stages. The BMP antagonist Short gastrulation (SOG) is required to elevate peak responses at the dorsal midline as well as to depress responses in dorsolateral cells. The midline SMAD response gradient can form in embryos with reduced dpp gene dosage, but the peak level is reduced. These data support a model in which weak BMP activity during blastoderm defines the boundary between ventral neurogenic ectoderm and dorsal ectoderm. Subsequently, BMP activity creates a step gradient of SMAD responses that patterns the amnioserosa and dorsomedial ectoderm.


Assuntos
Padronização Corporal , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Anticorpos/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/fisiologia , Dosagem de Genes , Ligantes , Morfogênese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína Smad4 , Transativadores/genética , Transativadores/imunologia , Fatores de Transcrição/genética
17.
Gene Expr Patterns ; 3(2): 143-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12711540

RESUMO

The tumour suppressor gene scribble (scrib) is required for epithelial polarity and growth control in Drosophila, and encodes two protein isoforms. Here, we report the pattern of Scrib1 synthesis in pole cells and embryonic gonads. We found that Scrib1 synthesis became strongly enhanced in pole cells at the time of gonad formation and was also detectable in cortical domains of gonadal mesodermal cells adjacent to pole cells. Scrib1 synthesis in mesodermal cells was independent of pole cells and occurred in agametic valois and capsuléen embryonic gonads. In contrast, Scrib1 synthesis in pole cells required contact with gonadal mesodermal cells as revealed by the absence of Scrib1 in wunen or tinman-zinc finger homeodomain-1 pseudo-gonads made only of aggregated pole cells.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Gônadas/embriologia , Proteínas de Membrana/genética , Animais , RNA Helicases DEAD-box , Proteínas de Drosophila/biossíntese , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Perfilação da Expressão Gênica , Células Germinativas , Gônadas/metabolismo , Proteínas de Membrana/biossíntese , RNA Helicases/metabolismo
18.
Nature ; 417(6889): 664-7, 2002 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-12050670

RESUMO

The Wnt family of secreted glycoproteins mediate cell cell interactions during cell growth and differentiation in both embryos and adults. Canonical Wnt signalling by way of the beta-catenin pathway is transduced by two receptor families. Frizzled proteins and lipoprotein-receptor-related proteins 5 and 6 (LRP5/6) bind Wnts and transmit their signal by stabilizing intracellular beta-catenin. Wnt/beta-catenin signalling is inhibited by the secreted protein Dickkopf1 (Dkk1), a member of a multigene family, which induces head formation in amphibian embryos. Dkk1 has been shown to inhibit Wnt signalling by binding to and antagonizing LRP5/6. Here we show that the transmembrane proteins Kremen1 and Kremen2 are high-affinity Dkk1 receptors that functionally cooperate with Dkk1 to block Wnt/beta-catenin signalling. Kremen2 forms a ternary complex with Dkk1 and LRP6, and induces rapid endocytosis and removal of the Wnt receptor LRP6 from the plasma membrane. The results indicate that Kremen1 and Kremen2 are components of a membrane complex modulating canonical Wnt signalling through LRP6 in vertebrates.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Transativadores , Proteínas de Peixe-Zebra , Animais , Animais Geneticamente Modificados , Linhagem Celular , Membrana Celular/metabolismo , Drosophila melanogaster/genética , Endocitose , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Substâncias Macromoleculares , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Testes de Precipitina , Ligação Proteica , Transporte Proteico , Proteínas/genética , Receptores de LDL/metabolismo , Deleção de Sequência/genética , Proteínas Wnt , beta Catenina
19.
Chromosoma ; 111(1): 13-21, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12068919

RESUMO

The Drosophila gene dMBD2/3 encodes a protein with significant homologies to the mammalian methyl-DNA binding proteins MBD2 and MBD3. These proteins are essential components of chromatin complexes involved in epigenetic gene regulation. Because the available in vitro data on dMBD2/3 are conflicting we have started an in vivo characterization of dMBD2/3. We detected expression of two isoforms specifically during embryonic development. Staining of whole embryos combined with high-resolution confocal microscopy revealed a highly regulated spatial distribution. During the syncytial blastoderm stage, dMBD2/3 formed speckles that localized to the cytoplasm. Shortly after, during the cellular blastoderm stage, the protein entered the nucleus and formed bright foci that associated with DNA. This rapid transition coincided with the activation of the embryonic genome. A similar observation was made during activation of the spermatocyte genome as dMBD2/3 formed distinct foci associated with the activated Y chromosome. Our results indicate that dMBD2/3 forms specialized nuclear compartments to keep certain genes epigenetically silenced during genome activation.


Assuntos
Cromossomos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Animais , Drosophila/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Immunoblotting , Masculino , Microscopia Confocal , Ligação Proteica , Espermatócitos/metabolismo , Cromossomo Y/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...