Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(19)2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33019537

RESUMO

Awd, the Drosophila homologue of NME1/2 metastasis suppressors, plays key roles in many signaling pathways. Mosaic analysis of the null awdJ2A4 allele showed that loss of awd gene function blocks Notch signaling and the expression of its target genes including the Wingless (Wg/Wnt1) morphogen. We also showed that RNA interference (RNAi)-mediated awd silencing (awdi) in larval wing disc leads to chromosomal instability (CIN) and to Jun amino-terminal kinases (JNK)-mediated cell death. Here we show that this cell death is independent of p53 activity. Based on our previous finding showing that forced survival of awdi-CIN cells leads to aneuploidy without the hyperproliferative effect, we investigated the Wg expression in awdi wing disc cells. Interestingly, the Wg protein is expressed in its correct dorso-ventral domain but shows an altered cellular distribution which impairs its signaling. Further, we show that RNAi-mediated knock down of awd in wing discs does not affect Notch signaling. Thus, our analysis of the hypomorphic phenotype arising from awd downregulation uncovers a dose-dependent effect of Awd in Notch and Wg signaling.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Nucleosídeo NM23 Difosfato Quinases/genética , Núcleosídeo-Difosfato Quinase/genética , Asas de Animais/metabolismo , Via de Sinalização Wnt/genética , Proteína Wnt1/genética , Animais , Morte Celular , Instabilidade Cromossômica , Cromossomos de Insetos/química , Cromossomos de Insetos/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Larva/citologia , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Masculino , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Núcleosídeo-Difosfato Quinase/antagonistas & inibidores , Núcleosídeo-Difosfato Quinase/metabolismo , Fenótipo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Asas de Animais/citologia , Asas de Animais/crescimento & desenvolvimento , Proteína Wnt1/metabolismo
2.
Front Physiol ; 10: 983, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31427986

RESUMO

The Awd (abnormal wing discs) gene is the Drosophila homolog of human NME1 and NME2 metastasis suppressor genes. These genes play a key role in tumor progression. Extensive studies revealed that intracellular NME1/2 protein levels could be related to either favorable or poor prognosis depending on tissue context. More recently, extracellular activities of NME1/2 proteins have also been reported, including a tumor- promoting function. We used Drosophila as a genetic model to investigate the mechanism controlling intra- and extracellular levels of NME1/2. We examined the role of several components of the ESCRT (endosomal sorting complex required for transport) complex in controlling Awd trafficking. We show that the Vps28 component of the ESCRT-I complex is required for maintenance of normal intracellular level of Awd in larval adipocytes. We already showed that blocking of Shibire (Shi)/Dynamin function strongly- lowers Awd intracellular level. To further investigate this down regulative effect, we analyzed the distribution of endosomal markers in wild type and Shi-defective adipocytes. Our results suggest that Awd does not enter CD63-positive endosomes. Interestingly, we found that in fat body cells, Awd partly- colocalizes with the ESCRT accessory component ALiX, the ALG-2 (apoptosis-linked gene 2)-interacting protein X. Moreover, we show that the intracellular levels of both proteins are downregulated by blocking the function of the Dynamin encoded by the shibire gene.

4.
Lab Invest ; 98(2): 248-257, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29035383

RESUMO

The Nm23/NME gene family has been under intensive study since Nm23H1/NME1 was identified as the first metastasis suppressor. Inverse correlation between the expression levels of NME1/2 and prognosis has indeed been demonstrated in different tumor cohorts. Interestingly, the presence of NME proteins in the extracellular environment in normal and tumoral conditions has also been noted. In many reported cases, however, these extracellular NME proteins exhibit anti-differentiation or oncogenic functions, contradicting their canonical anti-metastatic action. This emerging field thus warrants further investigation. In this review, we summarize the current understanding of extracellular NME proteins. A role in promoting stem cell pluripotency and inducing development of central nervous system as well as a neuroprotective function of extracellular NME have been suggested. Moreover, a tumor-promoting function of extracellular NME also emerged at least in some tumor cohorts. In this complex scenario, the secretory mechanism through which NME proteins exit cells is far from being understood. Recently, some evidence obtained in the Drosophila and cancer cell line models points to the involvement of Dynamin in controlling the balance between intra- and extracellular levels of NME. Further analyses on extracellular NME will lead to a better understanding of its physiological function and in turn will allow understanding of how its deregulation contributes to carcinogenesis.


Assuntos
Espaço Extracelular/enzimologia , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neoplasias/enzimologia , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Nucleosídeo NM23 Difosfato Quinases/genética , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia
5.
Sci Rep ; 7(1): 16820, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203880

RESUMO

The abnormal wing discs (awd) gene encodes the Drosophila homolog of NME1/NME2 metastasis suppressor genes. Awd acts in multiple tissues where its function is critical in establishing and maintaining epithelial integrity. Here, we analysed awd gene function in Drosophila epithelial cells using transgene-mediated RNA interference and genetic mosaic analysis. We show that awd knockdown in larval wing disc epithelium leads to chromosomal instability (CIN) and induces apoptosis mediated by activation of c-Jun N-terminal kinase. Forced maintenance of Awd depleted cells, by expressing the cell death inhibitor p35, downregulates atypical protein kinase C and DE-Cadherin. Consistent with their loss of cell polarity and enhanced level of matrix metalloproteinase 1, cells delaminate from wing disc epithelium. Furthermore, the DNA content profile of these cells indicates that they are aneuploid. Overall, our data demonstrate a novel function for awd in maintenance of genomic stability. Our results are consistent with other studies reporting that NME1 down-regulation induces CIN in human cell lines and suggest that Drosophila model could be successfully used to study in vivo the impact of NME/Awd - induced genomic instability on tumour development and metastasis formation.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Instabilidade Genômica , Núcleosídeo-Difosfato Quinase/genética , Aneuploidia , Animais , Caderinas/metabolismo , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Larva/crescimento & desenvolvimento , Larva/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Microscopia de Fluorescência , Núcleosídeo-Difosfato Quinase/antagonistas & inibidores , Núcleosídeo-Difosfato Quinase/metabolismo , Proteína Quinase C/metabolismo , Interferência de RNA , Asas de Animais/metabolismo , Asas de Animais/patologia
6.
Naunyn Schmiedebergs Arch Pharmacol ; 389(11): 1171-1182, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27449069

RESUMO

Dynamin GTPase (Dyn) plays a critical role in membrane-remodelling events underlying endocytosis. Studies in Drosophila identified a functional interaction between the Dyn homologue, encoded by the shibire (shi) gene, and Abnormal wing discs (Awd), a nucleoside diphosphate kinase (NDPK) that is the homologue of group I Nme human genes. These Drosophila studies showed that awd mutations enhance mutant shi phenotype and thus indicated the existence of a highly specific interaction between these genes. Furthermore, in human cells, it has been shown that Nme proteins promote Dyn activity in different membrane compartments through spatially controlled supply of GTP. Interestingly, Awd and Nme proteins have been detected in the extracellular environment. While no role has been inferred to extracellular Awd, presence of Nme1 in cancer patient serum is an unfavourable prognostic marker. In the present work, we used Drosophila and human cell line models to investigate the shuttling Awd/Nme1 proteins between intracellular and extracellular spaces. By using classic and reverse genetic approaches, we show that downregulation of Shi/Dyn1 activity enhances extracellular Awd/Nme1 in both Drosophila and human colon cell lines. We extended our analyses to colon cancer cell lines and found that knocking down Dyn1, besides to raise Nme1 extracellular amount, downregulates expression of molecular components that play key roles in tumour invasion. Interestingly, in vivo analyses of Drosophila larval adipocytes show that the conditional block of Shi activity greatly reduces intracellular amount of Awd confirming that Shi plays a key role in controlling the balance between intracellular and extracellular Awd.


Assuntos
Neoplasias do Colo/enzimologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Dinamina I/metabolismo , Dinaminas/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Núcleosídeo-Difosfato Quinase/metabolismo , Adipócitos/enzimologia , Animais , Animais Geneticamente Modificados , Neoplasias do Colo/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Dinamina I/genética , Dinaminas/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genótipo , Células HT29 , Humanos , Larva/enzimologia , Mutação , Nucleosídeo NM23 Difosfato Quinases/genética , Núcleosídeo-Difosfato Quinase/genética , Fenótipo , Interferência de RNA , Transfecção
7.
BMC Biol ; 12: 12, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24528630

RESUMO

BACKGROUND: The Drosophila abnormal wing discs (awd) belongs to a highly conserved family of genes implicated in metastasis suppression, metabolic homeostasis and epithelial morphogenesis. The cellular function of the mammalian members of this family, the Nm23 proteins, has not yet been clearly defined. Previous awd genetic analyses unraveled its endocytic role that is required for proper internalization of receptors controlling different signaling pathways. In this study, we analyzed the role of Awd in controlling Notch signaling during development. RESULTS: To study the awd gene function we used genetic mosaic approaches to obtain cells homozygous for a loss of function allele. In awd mutant follicle cells and wing disc cells, Notch accumulates in enlarged early endosomes, resulting in defective Notch signaling. Our results demonstrate that awd function is required before γ-secretase mediated cleavage since over-expression of the constitutively active form of the Notch receptor in awd mutant follicle cells allows rescue of the signaling. By using markers of different endosomal compartments we show that Notch receptor accumulates in early endosomes in awd mutant follicle cells. A trafficking assay in living wing discs also shows that Notch accumulates in early endosomes. Importantly, constitutively active Rab5 cannot rescue the awd phenotype, suggesting that awd is required for Rab5 function in early endosome maturation. CONCLUSIONS: In this report we demonstrate that awd is essential for Notch signaling via its endocytic role. In addition, we identify the endocytic step at which Awd function is required for Notch signaling and we obtain evidence indicating that Awd is necessary for Rab5 function. These findings provide new insights into the developmental and pathophysiological function of this important gene family.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Nucleosídeo NM23 Difosfato Quinases/genética , Núcleosídeo-Difosfato Quinase/metabolismo , Receptores Notch/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Animais , Proliferação de Células , Células Clonais , Vesículas Citoplasmáticas , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Endocitose , Endossomos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Discos Imaginais/citologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Mutação/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Metástase Neoplásica , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Transporte Proteico , Asas de Animais/citologia , Asas de Animais/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo
8.
Development ; 137(9): 1493-503, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20388653

RESUMO

Mutations in the human von Hippel-Lindau (VHL) genes are the cause of VHL disease, which displays multiple benign and malignant tumors. The VHL gene has been shown to regulate angiogenic potential and glycolic metabolism via its E3 ubiquitin ligase function against the alpha subunit of hypoxia-inducible factor (HIF). However, many other HIF-independent functions of VHL have been identified and recent evidence indicates that the canonical function cannot fully explain the VHL mutant cell phenotypes. Many of these functions have not been verified in genetically tractable systems. Using an established follicular epithelial model in Drosophila, we show that the Drosophila VHL gene is involved in epithelial morphogenesis via stabilizing microtubule bundles and aPKC. Microtubule defects in VHL mutants lead to mislocalization of aPKC and subsequent loss of epithelial integrity. Destabilizing microtubules in ex vivo culture of wild-type egg chambers can also result in aPKC mislocalization and epithelial defects. Importantly, paclitaxel-induced stabilization of microtubules can rescue the aPKC localization phenotype in Drosophila VHL mutant follicle cells. The results establish a developmental function of the VHL gene that is relevant to its tumor-suppressor activity.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Drosophila/genética , Epitélio/embriologia , Microtúbulos/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Animais , Proteínas de Drosophila/genética , Estabilidade Enzimática , Feminino , Imuno-Histoquímica , Imunoprecipitação , Técnicas In Vitro , Masculino , Microscopia Confocal , Folículo Ovariano/citologia , Folículo Ovariano/embriologia , Proteína Supressora de Tumor Von Hippel-Lindau/genética
9.
Dev Genes Evol ; 217(7): 529-40, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17569083

RESUMO

Drosophila vitelline membrane gene VM32E is expressed in the follicle cells of the stage 10 egg chamber and shows a peculiar temporal and spatial expression pattern compared to the other members of the same gene family. Previous work has led us to demonstrate that Decapentaplegic (Dpp) signaling represses the expression of the VM32E gene in the centripetal follicle cells. In this paper, we describe another level of complexity of the VM32E gene expression regulation. Through clonal analyses, we show that the expression of the VM32E gene in the main body follicle cells is modulated by the epidermal growth factor receptor (Egfr) activity. In follicle cell clones expressing a constitutively active form of the Egfr, the VM32E gene is downregulated, while the loss of the Egfr activity upregulates VM32E expression. In addition, we show that the ectopic expression of the Egfr-induced ETS transcription factor PointedP2 (PntP2) affects the expression of the VM32E gene. From these results and our previously published data, it appears that the proper patterning of follicle cells, defined by Dpp and Egfr signaling pathways, controls the VM32E gene expression pattern. This may suggest that a fine tuning of the expression of specific eggshell structural genes could be part of the complex process that leads to a proper eggshell assembly.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas do Ovo/genética , Receptores ErbB/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Oogênese/genética , Proteínas Quinases/metabolismo , Receptores de Peptídeos de Invertebrados/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Proteínas do Ovo/metabolismo , Imunofluorescência , Hibridização In Situ , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Dev Dyn ; 232(3): 845-54, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15712201

RESUMO

The Drosophila Akt (dAkt) serine/threonine kinase is a component of the insulin receptor/PI3K signaling pathway that regulates cell growth. Here, we show that this kinase is expressed during Drosophila oogenesis and is required for egg chamber development. Loss of dAkt function in follicle cells causes a cell-autonomous reduction of cell size while expression of the constitutively active myristylated form of this kinase (dAkt(myr)) causes increased cell size. Accordingly, expression of the antagonist dPTEN in the same follicular domains causes reduced follicle cell size. Perturbations of dAkt function do not affect follicle cell proliferation or cell death. Of interest, expression of dAkt(myr) in the posterior domain of the follicular epithelium causes a delay in the posterior movement of follicular epithelium and dumpless-like egg chambers. It appears that dAkt is required for maintaining the continuity of cell size within the follicular epithelium, which in turn is necessary for its proper morphogenesis.


Assuntos
Tamanho Celular , Proteínas de Drosophila , Drosophila/embriologia , Oogênese/fisiologia , Folículo Ovariano/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Embrião não Mamífero , Feminino , Folículo Ovariano/metabolismo , Ovário/citologia , Proteínas Proto-Oncogênicas c-akt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA