Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
1.
J Agric Food Chem ; 72(25): 14141-14151, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38864686

RESUMO

The cotton aphid, Aphis gossypii, is a polyphagous pest that stunts host plant growth via direct feeding or transmitting plant virus. Due to the long-term application of insecticides, A. gossypii has developed different levels of resistance to numerous insecticides. We found that five field populations had evolved multiple resistances to neonicotinoids. To explore the resistance mechanism mediated by uridine diphosphate glycosyltransferases (UGTs), two upregulated UGT genes in these five strains, UGT350C3 and UGT344L7, were selected for functional analysis of their roles in neonicotinoid detoxification. Transgenic Drosophila bioassay results indicated that compared with the control lines, the UGT350C3 and UGT344L7 overexpression lines were more tolerant to thiamethoxam, imidacloprid, and dinotefuran. Knockdown of UGT350C3 and UGT344L7 significantly increased A. gossypii sensitivity to thiamethoxam, imidacloprid, and dinotefuran. Molecular docking analysis demonstrated that these neonicotinoids could bind to the active pockets of UGT350C3 and UGT344L7. This study provides functional evidence of neonicotinoid detoxification mediated by UGTs and will facilitate further work to identify strategies for preventing the development of neonicotinoid resistance in insects.


Assuntos
Afídeos , Glicosiltransferases , Resistência a Inseticidas , Inseticidas , Neonicotinoides , Nitrocompostos , Animais , Afídeos/genética , Afídeos/enzimologia , Afídeos/efeitos dos fármacos , Neonicotinoides/farmacologia , Neonicotinoides/metabolismo , Neonicotinoides/química , Inseticidas/farmacologia , Inseticidas/química , Inseticidas/metabolismo , Resistência a Inseticidas/genética , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Glicosiltransferases/química , Nitrocompostos/farmacologia , Nitrocompostos/metabolismo , Simulação de Acoplamento Molecular , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Proteínas de Insetos/química , Tiametoxam , Drosophila/genética , Drosophila/enzimologia , Drosophila/efeitos dos fármacos , Drosophila/metabolismo , Guanidinas
2.
J Biol Chem ; 300(1): 105490, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38000659

RESUMO

The C-terminal binding protein (CtBP) is a transcriptional corepressor that plays critical roles in development, tumorigenesis, and cell fate. CtBP proteins are structurally similar to alpha hydroxyacid dehydrogenases and feature a prominent intrinsically disordered region in the C terminus. In the mammalian system, CtBP proteins lacking the C-terminal domain (CTD) are able to function as transcriptional regulators and oligomerize, putting into question the significance of this unstructured domain for gene regulation. Yet, the presence of an unstructured CTD of ∼100 residues, including some short motifs, is conserved across Bilateria, indicating the importance of maintaining this domain over evolutionary time. To uncover the significance of the CtBP CTD, we functionally tested naturally occurring Drosophila isoforms of CtBP that possess or lack the CTD, namely CtBP(L) and CtBP(S). We used the CRISPRi system to recruit dCas9-CtBP(L) and dCas9-CtBP(S) to endogenous promoters to directly compare their transcriptional impacts in vivo. Interestingly, CtBP(S) was able to significantly repress transcription of the Mpp6 promoter, while CtBP(L) was much weaker, suggesting that the long CTD may modulate CtBP's repression activity. In contrast, in cell culture, the isoforms behaved similarly on a transfected Mpp6 reporter gene. The context-specific differences in activity of these two developmentally regulated isoforms suggests that the CTD may help provide a spectrum of repression activity suitable for developmental programs.


Assuntos
Oxirredutases do Álcool , Proteínas de Drosophila , Regulação da Expressão Gênica , Domínios Proteicos , Proteínas Repressoras , Animais , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Drosophila/enzimologia , Drosophila/genética , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/metabolismo , Domínios Proteicos/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Linhagem Celular , Regulação da Expressão Gênica/genética
3.
Arch Insect Biochem Physiol ; 109(1): e21849, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34779010

RESUMO

Matrix metalloproteinase (MMP), a protease enzyme, participates in proteolytic cleavage of extracellular matrix proteins from Drosophila and mammals. But, recent studies have revealed other physiologically important roles of MMP in Drosophila. MMP contributes to cardioblast movement and distribution of collagen proteins during cardiogenesis in developing Drosophila. Tissue remodeling, especially tracheal development is also maintained by MMP. MMP regulates certain immunological functions in Drosophila such as wound repairing, plasmatocyte assemblage at the injured site of the basement membrane and glial response to axon degeneration in Drosophila nervous system. But, the contribution of MMP to tumor formation and metastasis in Drosophila has made it an interesting topic among researchers. Ovulation and egg laying are also found to be affected positively by MMP in Drosophila.


Assuntos
Drosophila/enzimologia , Metaloproteinases da Matriz , Animais , Carcinogênese , Drosophila/crescimento & desenvolvimento , Drosophila/imunologia , Drosophila/fisiologia , Feminino , Metástase Neoplásica , Oviposição , Ovulação/fisiologia
4.
Small GTPases ; 12(5-6): 416-428, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33985411

RESUMO

Epithelial folding is a common means to execute morphogenetic movements. The gastrulating Drosophila embryo offers many examples of epithelial folding events, including the ventral, cephalic, and dorsal furrows. Each of these folding events is associated with changes in intracellular contractility and/or cytoskeleton structures that autonomously promote epithelial folding. Here, we review accumulating evidence that suggests the progression and final form of ventral, cephalic, and dorsal furrows are also influenced by the behaviour of cells neighbouring these folds. We further discuss the prevalence and importance of junctional rearrangements during epithelial folding events, suggesting adherens junction components are prime candidates to modulate the transmission of the intercellular forces that influence folding events. Finally, we discuss how recently developed methods that enable precise spatial and/or temporal control of protein activity allow direct testing of molecular models of morphogenesis in vivo.


Assuntos
Citoesqueleto/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Embrião não Mamífero/fisiologia , Células Epiteliais/fisiologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Morfogênese , Animais , Citoesqueleto/enzimologia , Drosophila/enzimologia , Embrião não Mamífero/citologia , Embrião não Mamífero/enzimologia , Células Epiteliais/enzimologia , Microtúbulos/enzimologia , Microtúbulos/fisiologia
5.
Biochim Biophys Acta Gene Regul Mech ; 1864(2): 194630, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32911111

RESUMO

The Spt-Ada-Gcn5 Acetyltransferase (SAGA) chromatin modifying complex is a critical regulator of gene expression and is highly conserved across species. Subunits of SAGA arrange into discrete modules with lysine aceyltransferase and deubiquitinase activities housed separately. Mutation of the SAGA deubiquitinase module can lead to substantial biological misfunction and diseases such as cancer, neurodegeneration, and blindness. Here, we review the structure and functions of the SAGA deubiquitinase module and regulatory mechanisms acting to control these.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Complexos Multienzimáticos/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Fatores de Transcrição de p300-CBP/metabolismo , Animais , Arabidopsis/enzimologia , Aspergillus nidulans/enzimologia , Ataxina-7/genética , Cegueira/genética , Enzimas Desubiquitinantes/genética , Drosophila/enzimologia , Histonas/metabolismo , Humanos , Camundongos , Complexos Multienzimáticos/genética , Mutação , Neoplasias/genética , Doenças Neurodegenerativas/genética , Peptídeos/genética , Processamento de Proteína Pós-Traducional , RNA Polimerase II/metabolismo , Saccharomyces cerevisiae/enzimologia , Transativadores/genética , Fatores de Transcrição de p300-CBP/genética
6.
J Med Chem ; 63(17): 9876-9887, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787106

RESUMO

Emerging viruses like dengue, West Nile, chikungunya, and Zika can cause widespread viral epidemics. Developing novel drugs or vaccines against specific targets for each virus is a difficult task. As obligate parasites, all viruses exploit common cellular pathways, providing the possibility to develop broad-spectrum antiviral agents targeting host factors. The human DEAD-box RNA helicase DDX3X is an essential cofactor for viral replication but dispensable for cell viability. Herein, we exploited the presence of a unique structural motif of DDX3X not shared by other cellular enzymes to develop a theoretical model to aid in the design of a novel class of highly selective inhibitors acting against such specific targets, thus limiting off-targeting effects. High-throughput virtual screening led us to identify hit compound 5, endowed with promising antienzymatic activity. To improve its aqueous solubility, 5 and its two enantiomers were synthesized and converted into their corresponding acetate salts (compounds 11, 12, and 13). In vitro mutagenesis and biochemical and cellular assays further confirmed that the developed molecules were selective for DDX3X and were able to suppress replication of West Nile and dengue viruses in infected cells in the micromolar range while showing no toxicity for uninfected cells. These results provide proof of principle for a novel strategy in developing highly selective and broad-spectrum antiviral molecules active against emerging and dangerous viral pathogens. This study paves the way for the development of larger focused libraries targeting such domain to expand SAR studies and fully characterize their mode of interaction.


Assuntos
Antivirais/farmacologia , RNA Helicases DEAD-box/antagonistas & inibidores , Vírus da Dengue/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Vírus do Nilo Ocidental/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/toxicidade , Arabidopsis/enzimologia , Linhagem Celular Tumoral , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Drosophila/enzimologia , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/toxicidade , Hepacivirus/enzimologia , Humanos , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Mutação , Estudo de Prova de Conceito , Domínios Proteicos , Replicação Viral/efeitos dos fármacos
7.
PLoS One ; 15(6): e0234744, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32542037

RESUMO

Developmental processes require strict regulation of proliferation, differentiation and patterning for the generation of final organ size. Aberrations in these fundamental events are critically important in tumorigenesis and cancer progression. Salt inducible kinases (Siks) are evolutionarily conserved genes involved in diverse biological processes, including salt sensing, metabolism, muscle, cartilage and bone formation, but their role in development remains largely unknown. Recent findings implicate Siks in mitotic control, and in both tumor suppression and progression. Using a tumor model in the Drosophila eye, we show that perturbation of Sik function exacerbates tumor-like tissue overgrowth and metastasis. Furthermore, we show that both Drosophila Sik genes, Sik2 and Sik3, function in eye development processes. We propose that an important target of Siks may be the Notch signaling pathway, as we demonstrate genetic interaction between Siks and Notch pathway members. Finally, we investigate Sik expression in the developing retina and show that Sik2 is expressed in all photoreceptors, basal to cell junctions, while Sik3 appears to be expressed specifically in R3/R4 cells in the developing eye. Combined, our data suggest that Sik genes are important for eye tissue specification and growth, and that their dysregulation may contribute to tumor formation.


Assuntos
Drosophila/enzimologia , Drosophila/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Notch/metabolismo , Retina/crescimento & desenvolvimento , Animais , Drosophila/metabolismo , Ligação Proteica , Retina/metabolismo
8.
Elife ; 92020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32432549

RESUMO

Histone acetylation and deposition of H2A.Z variant are integral aspects of active transcription. In Drosophila, the single DOMINO chromatin regulator complex is thought to combine both activities via an unknown mechanism. Here we show that alternative isoforms of the DOMINO nucleosome remodeling ATPase, DOM-A and DOM-B, directly specify two distinct multi-subunit complexes. Both complexes are necessary for transcriptional regulation but through different mechanisms. The DOM-B complex incorporates H2A.V (the fly ortholog of H2A.Z) genome-wide in an ATP-dependent manner, like the yeast SWR1 complex. The DOM-A complex, instead, functions as an ATP-independent histone acetyltransferase complex similar to the yeast NuA4, targeting lysine 12 of histone H4. Our work provides an instructive example of how different evolutionary strategies lead to similar functional separation. In yeast and humans, nucleosome remodeling and histone acetyltransferase complexes originate from gene duplication and paralog specification. Drosophila generates the same diversity by alternative splicing of a single gene.


Cells contain a large number of proteins that control the activity of genes in response to various signals and changes in their environment. Often these proteins work together in groups called complexes. In the fruit fly Drosophila melanogaster, one of these complexes is called DOMINO. The DOMINO complex alters gene activity by interacting with other proteins called histones which influence how the genes are packaged and accessed within the cell. DOMINO works in two separate ways. First, it can replace certain histones with other variants that regulate genes differently. Second, it can modify histones by adding a chemical marker to them, which alters how they interact with genes. It was not clear how DOMINO can do both of these things and how that is controlled; but it was known that cells can make two different forms of the central component of the complex, called DOM-A and DOM-B, which are both encoded by the same gene. Scacchetti et al. have now studied fruit flies to understand the activities of these forms. This revealed that they do have different roles and that gene activity in cells changes if either one is lost. The two forms operate as part complexes with different compositions and only DOM-A includes the TIP60 enzyme that is needed to modify histones. As such, it seems that DOM-B primarily replaces histones with variant forms, while DOM-A modifies existing histones. This means that each form has a unique role associated with each of the two known behaviors of this complex. The presence of two different DOMINO complexes is common to flies and, probably, other insects. Yet, in other living things, such as mammals and yeast, their two roles are carried out by protein complexes originating from two distinct genes. This illustrates a concept called convergent evolution, where different organisms find different solutions for the same problem. As such, these findings provide an insight into the challenges encountered through evolution and the diverse solutions that have developed. They will also help us to understand the ways in which protein activities can adapt to different needs over evolutionary time.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/enzimologia , Histona Acetiltransferases/metabolismo , Complexos Multiproteicos/metabolismo , Fatores de Transcrição/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Montagem e Desmontagem da Cromatina , Drosophila/genética , Proteínas de Drosophila/genética , Histona Acetiltransferases/genética , Histonas/genética , Histonas/metabolismo , Complexos Multiproteicos/genética , Nucleossomos/genética , Nucleossomos/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/genética
9.
J Chem Phys ; 150(15): 155101, 2019 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-31005085

RESUMO

ERK2 is a kinase protein that belongs to a Ras/Raf/MEK/ERK signaling pathway, which is activated in response to a range of extracellular signals. Malfunctioning of this cascade leads to a variety of serious diseases, including cancers. This is often caused by mutations in proteins belonging to the cascade, frequently leading to abnormally high activity of the cascade even in the absence of an external signal. One such "gain-of-function" mutation in the ERK2 protein, called a "sevenmaker" mutation (D319N), was discovered in 1994 in Drosophila. The mutation leads to disruption of interactions of other proteins with the D-site of ERK2 and results, contrary to expectations, in an increase of its activity in vivo. However, no molecular mechanism to explain this effect has been presented so far. The difficulty is that this mutation should equally negatively affect interactions of ERK2 with all substrates, activators, and deactivators. In this paper, we present a semiquantitative kinetic network model that gives a possible explanation of the increased activity of mutant ERK2 species. A simplified biochemical network for ERK2, viewed as a system of coupled Michaelis-Menten processes, is presented. Its dynamic properties are calculated explicitly using the method of first-passage processes. The effect of mutation is associated with changes in the strength of interaction energy between the enzyme and the substrates. It is found that the dependence of kinetic properties of the protein on the interaction energy is nonmonotonic, suggesting that some mutations might lead to more efficient catalytic properties, despite weakening intermolecular interactions. Our theoretical predictions agree with experimental observations for the sevenmaker mutation in ERK2. It is also argued that the effect of mutations might depend on the concentrations of substrates.


Assuntos
Mutação com Ganho de Função , Proteína Quinase 1 Ativada por Mitógeno/química , Proteína Quinase 1 Ativada por Mitógeno/genética , Animais , Biocatálise , Drosophila/enzimologia , Cinética , Modelos Químicos , Especificidade por Substrato
10.
J Biol Chem ; 293(43): 16697-16708, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30213861

RESUMO

The Nedd4 family E3 ligases Itch and WWP1/2 play crucial roles in the regulation of cell cycle progression and apoptosis and are closely correlated with cancer development and metastasis. It has been recently shown that the ligase activities of Itch and WWP1/2 are tightly regulated, with the HECT domain sequestered intramolecularly by a linker region connecting WW2 and WW3. Here, we show that a similar autoinhibitory mechanism is utilized by the Drosophila ortholog of Itch and WWP1/2, Suppressor of Deltex (Su(dx)). We show that Su(dx) adopts an inactive steady state with the WW domain region interacting with the HECT domain. We demonstrate that both the linker and preceding WW2 are required for the efficient binding and regulation of Su(dx) HECT. Recruiting the multiple-PY motif-containing adaptor dNdfip via WW domains relieves the inhibitory state of Su(dx) and leads to substrate (e.g. Notch) ubiquitination. Our study demonstrates an evolutionarily conservative mechanism governing the regulation and activation of some Nedd4 family E3 ligases. Our results also suggest a dual regulatory mechanism for specific Notch down-regulation via dNdfip-Su(dx)-mediated Notch ubiquitination.


Assuntos
Proteínas de Drosophila/química , Drosophila/enzimologia , Ubiquitina-Proteína Ligases/química , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Drosophila/química , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Ligação Proteica , Domínios Proteicos , Estrutura Terciária de Proteína , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Domínios WW
11.
J Neurosci ; 38(35): 7701-7712, 2018 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-30030398

RESUMO

In addition to mechanisms promoting protein-synthesis-dependent long-term memory (PSD-LTM), the process appears to also be specifically constrained. We present evidence that the highly conserved receptor tyrosine kinase dAlk is a novel PSD-LTM attenuator in Drosophila Reduction of dAlk levels in adult α/ß mushroom body (MB) neurons during conditioning elevates LTM, whereas its overexpression impairs it. Unlike other memory suppressor proteins and miRNAs, dAlk within the MBs constrains PSD-LTM specifically but constrains learning outside the MBs as previously shown. Dendritic dAlk levels rise rapidly in MB neurons upon conditioning, a process apparently controlled by the 3'UTR of its mRNA, and interruption of the 3'UTR leads to enhanced LTM. Because its activating ligand Jeb is dispensable for LTM attenuation, we propose that postconditioning elevation of dAlk within α/ß dendrites results in its autoactivation and constrains formation of the energy costly PSD-LTM, acting as a novel memory filter.SIGNIFICANCE STATEMENT In addition to the widely studied molecular mechanisms promoting protein-synthesis-dependent long-term memory (PSD-LTM), recent discoveries indicate that the process is also specifically constrained. We describe a role in PSD-LTM constraint for the first receptor tyrosine kinase (RTK) involved in olfactory memory in Drosophila Unlike other memory suppressor proteins and miRNAs, dAlk limits specifically PSD-LTM formation as it does not affect 3 h, or anesthesia-resistant memory. Significantly, we show conditioning-dependent dAlk elevation within the mushroom body dendrites and propose that its local abundance may activate its kinase activity, to mediate imposition of PSD-LTM constraints through yet unknown mechanisms.


Assuntos
Quinase do Linfoma Anaplásico/fisiologia , Aprendizagem da Esquiva/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/fisiologia , Memória de Longo Prazo/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Regiões 3' não Traduzidas , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/genética , Animais , Dendritos/enzimologia , Dendritos/fisiologia , Drosophila/enzimologia , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Indução Enzimática , Larva , Consolidação da Memória , Corpos Pedunculados/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Neurônios/fisiologia , Odorantes , Pirimidinas/farmacologia , Interferência de RNA
12.
Nat Commun ; 8(1): 1649, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29158494

RESUMO

Ash1 is a Trithorax group protein that possesses H3K36-specific histone methyltransferase activity, which antagonizes Polycomb silencing. Here we report the identification of two Ash1 complex subunits, Mrg15 and Nurf55. In vitro, Mrg15 stimulates the enzymatic activity of Ash1. In vivo, Mrg15 is recruited by Ash1 to their common targets, and Mrg15 reinforces Ash1 chromatin association and facilitates the proper deposition of H3K36me2. To dissect the functional role of Mrg15 in the context of the Ash1 complex, we identify an Ash1 point mutation (Ash1-R1288A) that displays a greatly attenuated interaction with Mrg15. Knock-in flies bearing this mutation display multiple homeotic transformation phenotypes, and these phenotypes are partially rescued by overexpressing the Mrg15-Nurf55 fusion protein, which stabilizes the association of Mrg15 with Ash1. In summary, Mrg15 is a subunit of the Ash1 complex, a stimulator of Ash1 enzymatic activity and a critical regulator of the TrxG protein function of Ash1 in Drosophila.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/enzimologia , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Feminino , Histonas/química , Histonas/metabolismo , Humanos , Masculino , Metilação , Mutação , Ligação Proteica , Proteína 4 de Ligação ao Retinoblastoma/genética , Proteína 4 de Ligação ao Retinoblastoma/metabolismo , Fatores de Transcrição/genética
13.
PLoS Genet ; 13(7): e1006910, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28715417

RESUMO

Hox transcription factors specify distinct cell types along the anterior-posterior axis of metazoans by regulating target genes that modulate signaling pathways. A well-established example is the induction of Epidermal Growth Factor (EGF) signaling by an Abdominal-A (Abd-A) Hox complex during the specification of Drosophila hepatocyte-like cells (oenocytes). Previous studies revealed that Abd-A is non-cell autonomously required to promote oenocyte fate by directly activating a gene (rhomboid) that triggers EGF secretion from sensory organ precursor (SOP) cells. Neighboring cells that receive the EGF signal initiate a largely unknown pathway to promote oenocyte fate. Here, we show that Abd-A also plays a cell autonomous role in inducing oenocyte fate by activating the expression of the Pointed-P1 (PntP1) ETS transcription factor downstream of EGF signaling. Genetic studies demonstrate that both PntP1 and PntP2 are required for oenocyte specification. Moreover, we found that PntP1 contains a conserved enhancer (PntP1OE) that is activated in oenocyte precursor cells by EGF signaling via direct regulation by the Pnt transcription factors as well as a transcription factor complex consisting of Abd-A, Extradenticle, and Homothorax. Our findings demonstrate that the same Abd-A Hox complex required for sending the EGF signal from SOP cells, enhances the competency of receiving cells to select oenocyte cell fate by up-regulating PntP1. Since PntP1 is a downstream effector of EGF signaling, these findings provide insight into how a Hox factor can both trigger and potentiate the EGF signal to promote an essential cell fate along the body plan.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Fator de Crescimento Epidérmico/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Drosophila/enzimologia , Proteínas de Drosophila/genética , Elementos Facilitadores Genéticos , Fator de Crescimento Epidérmico/genética , Regulação da Expressão Gênica no Desenvolvimento , Hepatócitos/citologia , Hepatócitos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas/genética , Órgãos dos Sentidos/crescimento & desenvolvimento , Fatores de Transcrição/genética
14.
Mol Cells ; 40(4): 280-290, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28359145

RESUMO

Several lines of evidence suggest that endoplasmic reticulum (ER) stress plays a critical role in the pathogenesis of many neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Protein tyrosine phosphatase 1B (PTP1B) is known to regulate the ER stress signaling pathway, but its role in neuronal systems in terms of ER stress remains largely unknown. Here, we showed that rotenone-induced toxicity in human neuroblastoma cell lines and mouse primary cortical neurons was ameliorated by PTP1B inhibition. Moreover, the increase in the level of ER stress markers (eIF2α phosphorylation and PERK phosphorylation) induced by rotenone treatment was obviously suppressed by concomitant PTP1B inhibition. However, the rotenone-induced production of reactive oxygen species (ROS) was not affected by PTP1B inhibition, suggesting that the neuroprotective effect of the PTP1B inhibitor is not associated with ROS production. Moreover, we found that MG132-induced toxicity involving proteasome inhibition was also ameliorated by PTP1B inhibition in a human neuroblastoma cell line and mouse primary cortical neurons. Consistently, downregulation of the PTP1B homologue gene in Drosophila mitigated rotenone- and MG132-induced toxicity. Taken together, these findings indicate that PTP1B inhibition may represent a novel therapeutic approach for ER stress-mediated neurodegenerative diseases.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Doenças Neurodegenerativas/enzimologia , Neurônios/efeitos dos fármacos , Neuroproteção , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Animais , Morte Celular , Córtex Cerebral/enzimologia , Regulação para Baixo , Drosophila/enzimologia , Fator de Iniciação 2 em Eucariotos/efeitos dos fármacos , Humanos , Leupeptinas/farmacologia , Camundongos , Neurônios/enzimologia , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Rotenona/farmacologia , Células Tumorais Cultivadas , Desacopladores/farmacologia , eIF-2 Quinase/efeitos dos fármacos
15.
RNA ; 22(10): 1550-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27484313

RESUMO

MicroRNAs (miRNAs) recruit the CCR4-NOT complex, which contains two deadenylases, CCR4 and CAF1, to promote shortening of the poly(A) tail. Although both CCR4 and CAF1 generally have a strong preference for poly(A) RNA substrates, it has been reported from yeast to humans that they can also remove non-A residues in vitro to various degrees. However, it remains unknown how CCR4 and CAF1 remove non-A sequences. Herein we show that Drosophila miRNAs can promote the removal of 3'-terminal non-A residues in an exonucleolytic manner, but only if an upstream poly(A) sequence exists. This non-A removing reaction is directly catalyzed by both CCR4 and CAF1 and depends on the balance between the length of the internal poly(A) sequence and that of the downstream non-A sequence. These results suggest that the CCR4-NOT complex has an intrinsic activity to remove the 3'-terminal non-A modifications downstream from the poly(A) tail.


Assuntos
Proteínas de Drosophila/metabolismo , Proteína 4 de Ligação ao Retinoblastoma/metabolismo , Ribonucleases/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Drosophila/enzimologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Poliadenilação , Proteínas de Ligação a RNA , Proteína 4 de Ligação ao Retinoblastoma/genética , Ribonucleases/genética
16.
Elife ; 52016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27268300

RESUMO

A broadly known method to stimulate the growth potential of axons is to elevate intracellular levels of cAMP, however the cellular pathway(s) that mediate this are not known. Here we identify the Dual Leucine-zipper Kinase (DLK, Wnd in Drosophila) as a critical target and effector of cAMP in injured axons. DLK/Wnd is thought to function as an injury 'sensor', as it becomes activated after axonal damage. Our findings in both Drosophila and mammalian neurons indicate that the cAMP effector kinase PKA is a conserved and direct upstream activator of Wnd/DLK. PKA is required for the induction of Wnd signaling in injured axons, and DLK is essential for the regenerative effects of cAMP in mammalian DRG neurons. These findings link two important mediators of responses to axonal injury, DLK/Wnd and cAMP/PKA, into a unified and evolutionarily conserved molecular pathway for stimulating the regenerative potential of injured axons.


Assuntos
AMP Cíclico/metabolismo , Drosophila/enzimologia , Drosophila/fisiologia , MAP Quinase Quinase Quinases/metabolismo , Regeneração Nervosa , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Camundongos
17.
J Econ Entomol ; 109(2): 973-81, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26921228

RESUMO

Spotted wing drosophila, Drosophila suzukii Matsumura, is an invasive and economically damaging pest in Europe and North America. The females have a serrated ovipositor that enables them to infest almost all ripening small fruits. To understand the physiological and metabolic basis of spotted wing drosophila food preferences for healthy ripening fruits, we investigated the biological and biochemical characteristics of spotted wing drosophila and compared them with those of Drosophila melanogaster Meigen. We found that the susceptibility to oxidative stressors was significantly increased in spotted wing drosophila compared with those of D. melanogaster. In addition, we found that spotted wing drosophila had significantly reduced glutathione-S transferase (GST) activity and gene numbers. Furthermore, fructose concentrations found in spotted wing drosophila were significantly lower than those of D. melanogaster. Our data strongly suggest that the altered food preferences of spotted wing drosophila may stem from evolutionary adaptations to fresh foods accompanied by alterations in carbohydrate metabolism and GST activities.


Assuntos
Adaptação Biológica , Drosophila/enzimologia , Frutose/metabolismo , Glutationa Transferase/metabolismo , Espécies Introduzidas , Animais , Drosophila/genética , Proteínas de Drosophila/genética , Estresse Oxidativo , Sinapsinas/genética , Proteínas Supressoras de Tumor/genética
18.
Nat Commun ; 7: 10512, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26813350

RESUMO

It has been postulated that synaptic potentiation during waking is offset by a homoeostatic reduction in net synaptic strength during sleep. However, molecular mechanisms to support such a process are lacking. Here we demonstrate that deficiencies in the RNA-editing gene Adar increase sleep due to synaptic dysfunction in glutamatergic neurons in Drosophila. Specifically, the vesicular glutamate transporter is upregulated, leading to over-activation of NMDA receptors, and the reserve pool of glutamatergic synaptic vesicles is selectively expanded in Adar mutants. Collectively these changes lead to sustained neurotransmitter release under conditions that would otherwise result in synaptic depression. We propose that a shift in the balance from synaptic depression towards synaptic potentiation in sleep-promoting neurons underlies the increased sleep pressure of Adar-deficient animals. Our findings provide a plausible molecular mechanism linking sleep and synaptic plasticity.


Assuntos
Adenosina Desaminase/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/enzimologia , Drosophila/fisiologia , Ácido Glutâmico/metabolismo , Plasticidade Neuronal , Adenosina Desaminase/genética , Animais , Drosophila/genética , Proteínas de Drosophila/genética , Feminino , Masculino , Neurônios/metabolismo , Neurotransmissores/metabolismo , Edição de RNA , Sono , Vesículas Sinápticas/metabolismo
19.
Genes Dev ; 30(24): 2696-2709, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087714

RESUMO

Disruption of apical-basal polarity is implicated in developmental disorders and cancer; however, the mechanisms connecting cell polarity proteins with intracellular signaling pathways are largely unknown. We determined previously that membrane-associated guanylate kinase (MAGUK) protein discs large homolog 5 (DLG5) functions in cell polarity and regulates cellular proliferation and differentiation via undefined mechanisms. We report here that DLG5 functions as an evolutionarily conserved scaffold and negative regulator of Hippo signaling, which controls organ size through the modulation of cell proliferation and differentiation. Affinity purification/mass spectrometry revealed a critical role of DLG5 in the formation of protein assemblies containing core Hippo kinases mammalian ste20 homologs 1/2 (MST1/2) and Par-1 polarity proteins microtubule affinity-regulating kinases 1/2/3 (MARK1/2/3). Consistent with this finding, Hippo signaling is markedly hyperactive in mammalian Dlg5-/- tissues and cells in vivo and ex vivo and in Drosophila upon dlg5 knockdown. Conditional deletion of Mst1/2 fully rescued the phenotypes of brain-specific Dlg5 knockout mice. Dlg5 also interacts genetically with Hippo effectors Yap1/Taz Mechanistically, we show that DLG5 inhibits the association between MST1/2 and large tumor suppressor homologs 1/2 (LATS1/2), uses its scaffolding function to link MST1/2 with MARK3, and inhibits MST1/2 kinase activity. These data reveal a direct connection between cell polarity proteins and Hippo, which is essential for proper development of multicellular organisms.


Assuntos
Polaridade Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Células Cultivadas , Drosophila/embriologia , Drosophila/enzimologia , Drosophila/genética , Deleção de Genes , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Domínios Proteicos , Proteínas Serina-Treonina Quinases/genética , Proteômica , Interferência de RNA , Proteínas Supressoras de Tumor/genética
20.
Genetics ; 201(4): 1511-23, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26482795

RESUMO

CTP synthase (CTPsyn) plays an essential role in DNA, RNA, and lipid synthesis. Recent studies in bacteria, yeast, and Drosophila all reveal a polymeric CTPsyn structure, which dynamically regulates its enzymatic activity. However, the molecular mechanism underlying the formation of CTPsyn polymers is not completely understood. In this study, we found that reversible ubiquitination regulates the dynamic assembly of the filamentous structures of Drosophila CTPsyn. We further determined that the proto-oncogene Cbl, an E3 ubiquitin ligase, controls CTPsyn filament formation in endocycles. While the E3 ligase activity of Cbl is required for CTPsyn filament formation, Cbl does not affect the protein levels of CTPsyn. It remains unclear whether the regulation of CTPsyn filaments by Cbl is through direct ubiquitination of CTPsyn. In the absence of Cbl or with knockdown of CTPsyn, the progression of the endocycle-associated S phase was impaired. Furthermore, overexpression of wild-type, but not enzymatically inactive CTPsyn, rescued the endocycle defect in Cbl mutant cells. Together, these results suggest that Cbl influences the nucleotide pool balance and controls CTPsyn filament formation in endocycles. This study links Cbl-mediated ubiquitination to the polymerization of a metabolic enzyme and reveals a role for Cbl in endocycles during Drosophila development.


Assuntos
Carbono-Nitrogênio Ligases/metabolismo , Citoesqueleto/metabolismo , Replicação do DNA , Drosophila/genética , Animais , DNA , Drosophila/enzimologia , Feminino , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA