Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 386
Filtrar
1.
Traffic ; 25(9): e12955, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39313313

RESUMO

Signaling pathways activated by secreted Wnt ligands play an essential role in tissue development and the progression of diseases, like cancer. Secretion of the lipid-modified Wnt proteins is tightly regulated by a repertoire of intracellular factors. For instance, a membrane protein, Evi, interacts with the Wnt ligand in the ER, and it is essential for its further trafficking and release in the extracellular space. After dissociating from the Wnt, the Wnt-unbound Evi is recycled back to the ER via Golgi. However, where in this trafficking path Wnt proteins dissociate from Evi remains unclear. Here, we have used the Drosophila wing epithelium to trace the route of the Evi-Wg (Wnt homolog) complex leading up to their separation. In these polarized cells, Wg is first trafficked to the apical surface; however, the secretion of Wg is believed to occurs post-internalization via recycling. Our results show that the Evi-Wg complex is internalized from the apical surface and transported to the retromer-positive endosomes. Furthermore, using antibodies that specifically label the Wnt-unbound Evi, we show that Evi and Wg separation occurs post-internalization in the acidic endosomes. These results refine our understanding of the polarized trafficking of Wg and highlight the importance of Wg endocytosis in its secondary secretion.


Assuntos
Proteínas de Drosophila , Endossomos , Transporte Proteico , Proteína Wnt1 , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Endossomos/metabolismo , Proteína Wnt1/metabolismo , Proteína Wnt1/genética , Endocitose/fisiologia , Asas de Animais/metabolismo , Drosophila melanogaster/metabolismo , Drosophila/metabolismo , Proteínas de Membrana/metabolismo
2.
PLoS Genet ; 20(9): e1011387, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39226333

RESUMO

A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogaster wing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.


Assuntos
Dano ao DNA , Drosophila melanogaster , Asas de Animais , Animais , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/genética , Proliferação de Células , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Apoptose , Discos Imaginais/crescimento & desenvolvimento , Discos Imaginais/metabolismo , Cicatrização/genética , Senescência Celular , Sistema de Sinalização das MAP Quinases , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Ciclo Celular
3.
Development ; 151(18)2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39177163

RESUMO

One of the key tissue movements driving closure of a wound is re-epithelialisation. Earlier wound healing studies describe the dynamic cell behaviours that contribute to wound re-epithelialisation, including cell division, cell shape changes and cell migration, as well as the signals that might regulate these cell behaviours. Here, we have used a series of deep learning tools to quantify the contributions of each of these cell behaviours from movies of repairing wounds in the Drosophila pupal wing epithelium. We test how each is altered after knockdown of the conserved wound repair signals Ca2+ and JNK, as well as after ablation of macrophages that supply growth factor signals believed to orchestrate aspects of the repair process. Our genetic perturbation experiments provide quantifiable insights regarding how these wound signals impact cell behaviours. We find that Ca2+ signalling is a master regulator required for all contributing cell behaviours; JNK signalling primarily drives cell shape changes and divisions, whereas signals from macrophages largely regulate cell migration and proliferation. Our studies show deep learning to be a valuable tool for unravelling complex signalling hierarchies underlying tissue repair.


Assuntos
Movimento Celular , Aprendizado Profundo , Transdução de Sinais , Asas de Animais , Cicatrização , Animais , Movimento Celular/genética , Cicatrização/fisiologia , Cicatrização/genética , Asas de Animais/metabolismo , Reepitelização , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Pupa/metabolismo , Macrófagos/metabolismo , Proliferação de Células , Sinalização do Cálcio , Forma Celular , Epitélio/metabolismo
4.
PLoS Biol ; 22(8): e3002780, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39186808

RESUMO

In animals, mitosis involves the breakdown of the nuclear envelope and the sorting of individualized, condensed chromosomes. During mitotic exit, emerging nuclei reassemble a nuclear envelope around a single mass of interconnecting chromosomes. The molecular mechanisms of nuclear reassembly are incompletely understood. Moreover, the cellular and physiological consequences of defects in this process are largely unexplored. Here, we have characterized a mechanism essential for nuclear reassembly in Drosophila. We show that Ankle2 promotes the PP2A-dependent recruitment of BAF and Lamin at reassembling nuclei, and that failures in this mechanism result in severe nuclear defects after mitosis. We then took advantage of perturbations in this mechanism to investigate the physiological responses to nuclear reassembly defects during tissue development in vivo. Partial depletion of Ankle2, BAF, or Lamin in imaginal wing discs results in wing development defects accompanied by apoptosis. We found that blocking apoptosis strongly enhances developmental defects. Blocking p53 does not prevent apoptosis but enhances defects due to the loss of a cell cycle checkpoint. Our results suggest that apoptotic and p53-dependent responses play a crucial role in safeguarding tissue development in response to sporadic nuclear reassembly defects.


Assuntos
Apoptose , Núcleo Celular , Proteínas de Drosophila , Drosophila melanogaster , Mitose , Proteína Supressora de Tumor p53 , Asas de Animais , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Apoptose/genética , Núcleo Celular/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Membrana Nuclear/metabolismo , Laminas/metabolismo , Laminas/genética , Proteínas Nucleares
5.
PLoS Biol ; 22(7): e3002547, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39047051

RESUMO

Despite the deep conservation of the DNA damage response (DDR) pathway, cells in different contexts vary widely in their susceptibility to DNA damage and their propensity to undergo apoptosis as a result of genomic lesions. One of the cell signaling pathways implicated in modulating the DDR is the highly conserved Wnt pathway, which is known to promote resistance to DNA damage caused by ionizing radiation in a variety of human cancers. However, the mechanisms linking Wnt signal transduction to the DDR remain unclear. Here, we use a genetically encoded system in Drosophila to reliably induce consistent levels of DNA damage in vivo, and demonstrate that canonical Wnt signaling in the wing imaginal disc buffers cells against apoptosis in the face of DNA double-strand breaks. We show that Wg, the primary Wnt ligand in Drosophila, activates epidermal growth factor receptor (EGFR) signaling via the ligand-processing protease Rhomboid, which, in turn, modulates the DDR in a Chk2-, p53-, and E2F1-dependent manner. These studies provide mechanistic insight into the modulation of the DDR by the Wnt and EGFR pathways in vivo in a highly proliferative tissue. Furthermore, they reveal how the growth and patterning functions of Wnt signaling are coupled with prosurvival, antiapoptotic activities, thereby facilitating developmental robustness in the face of genomic damage.


Assuntos
Apoptose , Dano ao DNA , Proteínas de Drosophila , Receptores ErbB , Discos Imaginais , Asas de Animais , Via de Sinalização Wnt , Proteína Wnt1 , Animais , Receptores ErbB/metabolismo , Receptores ErbB/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Discos Imaginais/metabolismo , Discos Imaginais/crescimento & desenvolvimento , Proteína Wnt1/metabolismo , Proteína Wnt1/genética , Apoptose/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Quinase do Ponto de Checagem 2/metabolismo , Quinase do Ponto de Checagem 2/genética , Transdução de Sinais , Quebras de DNA de Cadeia Dupla , Receptores de Peptídeos de Invertebrados/metabolismo , Receptores de Peptídeos de Invertebrados/genética , Drosophila/metabolismo , Drosophila/genética , Fatores de Transcrição
6.
Cell Rep ; 43(7): 114398, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38935502

RESUMO

Mechanosensitive Piezo channels regulate cell division, cell extrusion, and cell death. However, systems-level functions of Piezo in regulating organogenesis remain poorly understood. Here, we demonstrate that Piezo controls epithelial cell topology to ensure precise organ growth by integrating live-imaging experiments with pharmacological and genetic perturbations and computational modeling. Notably, the knockout or knockdown of Piezo increases bilateral asymmetry in wing size. Piezo's multifaceted functions can be deconstructed as either autonomous or non-autonomous based on a comparison between tissue-compartment-level perturbations or between genetic perturbation populations at the whole-tissue level. A computational model that posits cell proliferation and apoptosis regulation through modulation of the cutoff tension required for Piezo channel activation explains key cell and tissue phenotypes arising from perturbations of Piezo expression levels. Our findings demonstrate that Piezo promotes robustness in regulating epithelial topology and is necessary for precise organ size control.


Assuntos
Células Epiteliais , Canais Iônicos , Canais Iônicos/metabolismo , Canais Iônicos/genética , Animais , Tamanho do Órgão , Células Epiteliais/metabolismo , Camundongos , Proliferação de Células , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Apoptose , Humanos , Epitélio/metabolismo
7.
Commun Biol ; 7(1): 533, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710747

RESUMO

Insect wing development is a fascinating and intricate process that involves the regulation of wing size through cell proliferation and apoptosis. In this study, we find that Ter94, an AAA-ATPase, is essential for proper wing size dependently on its ATPase activity. Loss of Ter94 enables the suppression of Hippo target genes. When Ter94 is depleted, it results in reduced wing size and increased apoptosis, which can be rescued by inhibiting the Hippo pathway. Biochemical experiments reveal that Ter94 reciprocally binds to Mer, a critical upstream component of the Hippo pathway, and disrupts its interaction with Ex and Kib. This disruption prevents the formation of the Ex-Mer-Kib complex, ultimately leading to the inactivation of the Hippo pathway and promoting proper wing development. Finally, we show that hVCP, the human homolog of Ter94, is able to substitute for Ter94 in modulating Drosophila wing size, underscoring their functional conservation. In conclusion, Ter94 plays a positive role in regulating wing size by interfering with the Ex-Mer-Kib complex, which results in the suppression of the Hippo pathway.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Proteínas de Membrana , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Proteínas Supressoras de Tumor , Asas de Animais , Animais , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/genética , Apoptose , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo
8.
Biomolecules ; 14(5)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38785929

RESUMO

Suppressor of deltex (Su(dx)) is a Drosophila melanogaster member of the NEDD4 family of the HECT domain E3 ubiquitin ligases. Su(dx) acts as a regulator of Notch endocytic trafficking, promoting Notch lysosomal degradation and the down-regulation of both ligand-dependent and ligand-independent signalling, the latter involving trafficking through the endocytic pathway and activation of the endo/lysosomal membrane. Mutations of Su(dx) result in developmental phenotypes in the Drosophila wing that reflect increased Notch signalling, leading to gaps in the specification of the wing veins, and Su(dx) functions to provide the developmental robustness of Notch activity to environmental temperature shifts. The full developmental functions of Su(dx) are unclear; however, this is due to a lack of a clearly defined null allele. Here we report the first defined null mutation of Su(dx), generated by P-element excision, which removes the complete open reading frame. We show that the mutation is recessive-viable, with the Notch gain of function phenotypes affecting wing vein and leg development. We further uncover new roles for Su(dx) in Drosophila oogenesis, where it regulates interfollicular stalk formation, egg chamber separation and germline cyst enwrapment by the follicle stem cells. Interestingly, while the null allele exhibited a gain in Notch activity during oogenesis, the previously described Su(dx)SP allele, which carries a seven amino acid in-frame deletion, displayed a Notch loss of function phenotypes and an increase in follicle stem cell turnover. This is despite both alleles displaying similar Notch gain of function in wing development. We attribute this unexpected context-dependent outcome of Su(dx)sp being due to the partial retention of function by the intact C2 and WW domain regions of the protein. Our results extend our understanding of the developmental role of Su(dx) in the tissue renewal and homeostasis of the Drosophila ovary and illustrate the importance of examining an allelic series of mutations to fully understand developmental functions.


Assuntos
Alelos , Proteínas de Drosophila , Drosophila melanogaster , Oogênese , Receptores Notch , Animais , Oogênese/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , Feminino , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo , Mutação , Transdução de Sinais , Fenótipo , Proteínas de Membrana
9.
Cell Rep ; 43(5): 114147, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38662541

RESUMO

Butterfly wings display a diversity of cell types, including large polyploid scale cells, yet the molecular basis of such diversity is poorly understood. To explore scale cell diversity at a transcriptomic level, we employ single-cell RNA sequencing of ∼5,200 large cells (>6 µm) from 22.5- to 25-h male pupal forewings of the butterfly Bicyclus anynana. Using unsupervised clustering, followed by in situ hybridization, immunofluorescence, and CRISPR-Cas9 editing of candidate genes, we annotate various cell types on the wing. We identify genes marking non-innervated scale cells, pheromone-producing glandular cells, and innervated sensory cell types. We show that senseless, a zinc-finger transcription factor, and HR38, a hormone receptor, determine the identity, size, and color of different scale cell types and are important regulators of scale cell differentiation. This dataset and the identification of various wing cell-type markers provide a foundation to compare and explore scale cell-type diversification across arthropod species.


Assuntos
Borboletas , Pupa , Análise de Célula Única , Asas de Animais , Animais , Borboletas/genética , Asas de Animais/metabolismo , Asas de Animais/citologia , Pupa/metabolismo , Análise de Célula Única/métodos , Masculino , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Transcriptoma/genética
10.
Cell Mol Life Sci ; 81(1): 195, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38653877

RESUMO

The Notch pathway is an evolutionarily conserved signaling system that is intricately regulated at multiple levels and it influences different aspects of development. In an effort to identify novel components involved in Notch signaling and its regulation, we carried out protein interaction screens which identified non-muscle myosin II Zipper (Zip) as an interacting partner of Notch. Physical interaction between Notch and Zip was further validated by co-immunoprecipitation studies. Immunocytochemical analyses revealed that Notch and Zip co-localize within same cytoplasmic compartment. Different alleles of zip also showed strong genetic interactions with Notch pathway components. Downregulation of Zip resulted in wing phenotypes that were reminiscent of Notch loss-of-function phenotypes and a perturbed expression of Notch downstream targets, Cut and Deadpan. Further, synergistic interaction between Notch and Zip resulted in highly ectopic expression of these Notch targets. Activated Notch-induced tumorous phenotype of larval tissues was enhanced by over-expression of Zip. Notch-Zip synergy resulted in the activation of JNK pathway that consequently lead to MMP activation and proliferation. Taken together, our results suggest that Zip may play an important role in regulation of Notch signaling.


Assuntos
Proteínas de Drosophila , Proteínas de Membrana , Cadeias Pesadas de Miosina , Receptores Notch , Transdução de Sinais , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores Notch/metabolismo , Receptores Notch/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Drosophila/metabolismo , Drosophila/genética , Fenótipo , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz/genética , Proliferação de Células , Miosina Tipo II/metabolismo , Miosina Tipo II/genética
11.
Nat Commun ; 13(1): 5670, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36167844

RESUMO

Insect wing polyphenism is characterized by its ability to produce two or more distinct wing morphs from a single genotype in response to changing environments. However, the molecular basis of this phenomenon remains poorly understood. Here, we identified a zinc finger homeodomain transcription factor Zfh1 that acts as an upstream regulator for the development of long-winged (LW) or shorted-winged (SW) morphs in planthoppers. Knockdown of Zfh1 directs SW-destined nymphs to develop into LW morphs by down-regulating the transcriptional level of FoxO, a prominent downstream effector of the insulin/IGF signaling (IIS) pathway. The balance between transcriptional regulation via the Zfh1-FoxO cascade and post-translational regulation via the IIS-FoxO cascade provides a flexible regulatory mechanism for the development of alternative wing morphs. These findings help us understand how phenotypic diversity is generated by altering the activity of conserved proteins, and provide an extended framework for the evolution of wing morphological diversity in insects.


Assuntos
Hemípteros , Asas de Animais , Animais , Regulação da Expressão Gênica , Hemípteros/genética , Insulina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Asas de Animais/metabolismo
12.
EMBO Rep ; 23(11): e54025, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36134875

RESUMO

Adenosine triphosphate (ATP) production and utilization is critically important for animal development. How these processes are regulated in space and time during tissue growth remains largely unclear. We used a FRET-based sensor to dynamically monitor ATP levels across a growing tissue, using the Drosophila larval wing disc. Although steady-state levels of ATP are spatially uniform across the wing pouch, inhibiting oxidative phosphorylation reveals spatial differences in metabolic behavior, whereby signaling centers at compartment boundaries produce more ATP from glycolysis than the rest of the tissue. Genetic perturbations indicate that the conserved Hedgehog signaling pathway can enhance ATP production by glycolysis. Collectively, our work suggests the existence of a homeostatic feedback loop between Hh signaling and glycolysis, advancing our understanding of the connection between conserved developmental patterning genes and ATP production during animal tissue development.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Asas de Animais/metabolismo , Glicólise , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo
13.
Cells ; 11(16)2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-36010619

RESUMO

The loss-of-function conditions for an l(3)malignant brain tumour (l(3)mbt) in larvae reared at 29 °C results in malignant brain tumours and hyperplastic imaginal discs. Unlike the former that have been extensively characterised, little is known about the latter. Here we report the results of a study of the hyperplastic l(3)mbt mutant wing imaginal discs. We identify the l(3)mbt wing disc tumour transcriptome and find it to include genes involved in reactive oxygen species (ROS) metabolism. Furthermore, we show the presence of oxidative stress in l(3)mbt hyperplastic discs, even in apoptosis-blocked conditions, but not in l(3)mbt brain tumours. We also find that chemically blocking oxidative stress in l(3)mbt wing discs reduces the incidence of wing disc overgrowths. Our results reveal the involvement of oxidative stress in l(3)mbt wing discs hyperplastic growth.


Assuntos
Proteínas de Drosophila , Discos Imaginais , Animais , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Discos Imaginais/metabolismo , Estresse Oxidativo , Asas de Animais/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-35895928

RESUMO

The main environmental problem in urban areas, especially in Brazil, is the discharge of untreated sewage. The in vivo Drosophila melanogaster Somatic Mutation and Recombination Test (SMART) was used to assess the genotoxicity of surface waters from three different sites in the Tocantins River, Brazil. The in silico approach was used to search for known and predicted interactions between environmental chemicals found in our samples and Drosophila and human proteins. The genotoxicity tests were performed in standard (ST) and high bioactivation (HB) crosses with samples collected at two periods, the rainy and dry seasons. Mutant spot frequencies found in treatments with unprocessed water from the test sites were compared with the frequencies observed in negative controls. The collection points were represented as sites A, B and C along Tocantins River. Sites A and B are located in Porto Nacional City, whereas site C is located in Palmas City. Considering the rainy season collection, positive responses in the ST cross were observed for sites A and C (89.47% and 85% of recombination, respectively) and in the HB cross for sites A, B and C (88.24%, 84.21% and 82.35% of recombination, respectively). The positive results in the dry season were restricted to sites A and B (88.89% and 85.71% of recombination, respectively) in the HB cross. In accordance with in vivo and in silico results, we hypothesize that ribosomal proteins (RPs) in fruit fly and humans are depleted in cells exposed to heavy metal causing DNA damage and chromosome instability, increasing homologous recombination.


Assuntos
Drosophila melanogaster , Rios , Animais , Brasil , Cidades , Dano ao DNA , Drosophila , Drosophila melanogaster/genética , Humanos , Água/metabolismo , Asas de Animais/metabolismo
15.
Nat Cell Biol ; 24(4): 424-433, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35301437

RESUMO

During development, morphogen gradients encode positional information to pattern morphological structures during organogenesis1. Some gradients, like that of Dpp in the fly wing, remain proportional to the size of growing organs-that is, they scale. Gradient scaling keeps morphological patterns proportioned in organs of different sizes2,3. Here we show a mechanism of scaling that ensures that, when the gradient is smaller than the organ, cell death trims the developing tissue to match the size of the gradient. Scaling is controlled by molecular associations between Dally and Pentagone, known factors involved in scaling, and a key factor that mediates cell death, Flower4-6. We show that Flower activity in gradient expansion is not dominated by cell death, but by the activity of Dally/Pentagone on scaling. Here we show a potential connection between scaling and cell death that may uncover a molecular toolbox hijacked by tumours.


Assuntos
Proteínas de Drosophila , Animais , Padronização Corporal/fisiologia , Morte Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Asas de Animais/metabolismo
16.
Cell Death Dis ; 13(2): 101, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35110540

RESUMO

Hippo signaling is a conserved mechanism for controlling organ growth. Increasing evidence suggests that Hippo signaling is modulated by various cellular factors for normal development and tumorigenesis. Hence, identification of these factors is pivotal for understanding the mechanism for the regulation of Hippo signaling. Drosophila Mnat9 is a putative N-acetyltransferase that is required for cell survival by affecting JNK signaling. Here we show that Mnat9 is involved in the negative regulation of Hippo signaling. RNAi knockdown of Mnat9 in the eye disc suppresses the rough eye phenotype of overexpressing Crumbs (Crb), an upstream factor of the Hippo pathway. Conversely, Mnat9 RNAi enhances the eye phenotype caused by overexpressing Expanded (Ex) or Warts (Wts) that acts downstream to Crb. Similar genetic interactions between Mnat9 and Hippo pathway genes are found in the wing. The reduced wing phenotype of Mnat9 RNAi is suppressed by overexpression of Yorkie (Yki), while it is suppressed by knockdown of Hippo upstream factors like Ex, Merlin, or Kibra. Mnat9 co-immunoprecipitates with Mer, implying their function in a protein complex. Furthermore, Mnat9 overexpression together with Hpo knockdown causes tumorous overgrowth in the abdomen. Our data suggest that Mnat9 is required for organ growth and can induce tumorous growth by negatively regulating the Hippo signaling pathway.


Assuntos
Carcinogênese/metabolismo , Drosophila melanogaster/metabolismo , Via de Sinalização Hippo , Acetiltransferases N-Terminal/metabolismo , Animais , Carcinogênese/patologia , Olho Composto de Artrópodes/crescimento & desenvolvimento , Olho Composto de Artrópodes/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Acetiltransferases N-Terminal/genética , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno , Asas de Animais/metabolismo
17.
Development ; 149(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35005772

RESUMO

Aggressive neoplastic growth can be initiated by a limited number of genetic alterations, such as the well-established cooperation between loss of cell architecture and hyperactive signaling pathways. However, our understanding of how these different alterations interact and influence each other remains very incomplete. Using Drosophila paradigms of imaginal wing disc epithelial growth, we have monitored the changes in Notch pathway activity according to the polarity status of cells (scrib mutant). We show that the scrib mutation impacts the direct transcriptional output of the Notch pathway, without altering the global distribution of Su(H), the Notch-dedicated transcription factor. The Notch-dependent neoplasms require, however, the action of a group of transcription factors, similar to those previously identified for Ras/scrib neoplasm (namely AP-1, Stat92E, Ftz-F1 and basic leucine zipper factors), further suggesting the importance of this transcription factor network during neoplastic growth. Finally, our work highlights some Notch/scrib specificities, in particular the role of the PAR domain-containing basic leucine zipper transcription factor and Notch direct target Pdp1 for neoplastic growth.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Receptores Notch/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/antagonistas & inibidores , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Carcinogênese , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Larva/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Neoplasias/metabolismo , Neoplasias/patologia , Interferência de RNA , Transdução de Sinais , Asas de Animais/metabolismo
18.
Development ; 149(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35037688

RESUMO

A limited BMP signaling range in the stem cell niche of the ovary protects against germ cell tumors and promotes germ cell homeostasis. The canonical repressor of BMP signaling in both the Drosophila embryo and wing disc is the transcription factor Brinker (Brk), yet the expression and potential role of Brk in the germarium has not previously been described. Here, we find that brk expression requires a promoter-proximal element (PPE) to support long-distance enhancer action as well as to drive expression in the germarium. Furthermore, PPE subdomains have different activities; in particular, the proximal portion acts as a damper to regulate brk levels precisely. Using PPE mutants as well as tissue-specific RNA interference and overexpression, we show that altering brk expression within either the soma or the germline affects germ cell homeostasis. Remarkably, we find that Decapentaplegic (Dpp), the main BMP ligand and canonical antagonist of Brk, is upregulated by Brk in the escort cells of the germarium, demonstrating that Brk can positively regulate this pathway.


Assuntos
Proteínas de Drosophila/metabolismo , Células Germinativas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Diferenciação Celular , Drosophila , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Embrião não Mamífero/metabolismo , Feminino , Células Germinativas/citologia , Ovário/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA de Cadeia Dupla/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transdução de Sinais , Regulação para Cima , Asas de Animais/metabolismo
19.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35086929

RESUMO

In Drosophila melanogaster, loss of regenerative capacity in wing imaginal discs coincides with an increase in systemic levels of the steroid hormone ecdysone, a key coordinator of their developmental progression. Regenerating discs release the relaxin hormone Dilp8 (Drosophila insulin-like peptide 8) to limit ecdysone synthesis and extend the regenerative period. Here, we describe how regenerating tissues produce a biphasic response to ecdysone levels: lower concentrations of ecdysone promote local and systemic regenerative signaling, whereas higher concentrations suppress regeneration through the expression of broad splice isoforms. Ecdysone also promotes the expression of wingless during both regeneration and normal development through a distinct regulatory pathway. This dual role for ecdysone explains how regeneration can still be completed successfully in dilp8- mutant larvae: higher ecdysone levels increase the regenerative activity of tissues, allowing regeneration to reach completion in a shorter time. From these observations, we propose that ecdysone hormone signaling functions to coordinate regeneration with developmental progression.


Assuntos
Ecdisona/metabolismo , Regeneração/fisiologia , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hormônios Esteroides Gonadais/metabolismo , Discos Imaginais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Larva/crescimento & desenvolvimento , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Asas de Animais/metabolismo , Proteína Wnt1/metabolismo
20.
Nat Commun ; 12(1): 2159, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33846330

RESUMO

N6-methyladenosine (m6A), the most abundant internal modification in eukaryotic mRNA, is installed by a multi-component writer complex; however, the exact roles of each component remain poorly understood. Here we show that a potential E3 ubiquitin ligase Hakai colocalizes and interacts with other m6A writer components, and Hakai mutants exhibit typical m6A pathway defects in Drosophila, such as lowered m6A levels in mRNA, aberrant Sxl alternative splicing, wing and behavior defects. Hakai, Vir, Fl(2)d and Flacc form a stable complex, and disruption of either Hakai, Vir or Fl(2)d led to the degradation of the other three components. Furthermore, MeRIP-seq indicates that the effective m6A modification is mostly distributed in 5' UTRs in Drosophila, in contrast to the mammalian system. Interestingly, we demonstrate that m6A modification is deposited onto the Sxl mRNA in a sex-specific fashion, which depends on the m6A writer. Together, our work not only advances the understanding of mechanism and regulation of the m6A writer complex, but also provides insights into how Sxl cooperate with the m6A pathway to control its own splicing.


Assuntos
Adenosina/análogos & derivados , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Regiões 5' não Traduzidas/genética , Adenosina/metabolismo , Processamento Alternativo/genética , Animais , Sequência de Bases , Comportamento Animal , Códon de Iniciação/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Metilação , Mutação/genética , Ligação Proteica , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Caracteres Sexuais , Ubiquitina-Proteína Ligases/genética , Asas de Animais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA