Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Biol ; 223(5)2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38470362

RESUMO

The eukaryotic p24 family, consisting of α-, ß-, γ- and δ-p24 subfamilies, has long been known to be involved in regulating secretion. Despite increasing interest in these proteins, fundamental questions remain about their role. Here, we systematically investigated Drosophila p24 proteins. We discovered that members of all four p24 subfamilies are required for general secretion and that their localizations between ER exit site (ERES) and Golgi are interdependent in an α→ßδ→γ sequence. We also found that localization of p24 proteins and ERES determinant Tango1 requires interaction through their respective GOLD and SH3 lumenal domains, with Tango1 loss sending p24 proteins to the plasma membrane and vice versa. Finally, we show that p24 loss expands the COPII zone at ERES and increases the number of ER-Golgi vesicles, supporting a restrictive role of p24 proteins on vesicle budding for efficient transport. Our results reveal Tango1-p24 interplay as central to the generation of a stable ER-Golgi interface.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto , Proteínas de Drosophila , Retículo Endoplasmático , Complexo de Golgi , Proteínas de Membrana Transportadoras , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Membrana Celular , Drosophila melanogaster , Proteínas de Drosophila/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Domínios de Homologia de src , Proteínas de Membrana Transportadoras/metabolismo
2.
PLoS Biol ; 21(3): e3002050, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36947563

RESUMO

Knowledge of adipogenetic mechanisms is essential to understand and treat conditions affecting organismal metabolism and adipose tissue health. In Drosophila, mature adipose tissue (fat body) exists in larvae and adults. In contrast to the well-known development of the larval fat body from the embryonic mesoderm, adult adipogenesis has remained mysterious. Furthermore, conclusive proof of its physiological significance is lacking. Here, we show that the adult fat body originates from a pool of undifferentiated mesodermal precursors that migrate from the thorax into the abdomen during metamorphosis. Through in vivo imaging, we found that these precursors spread from the ventral midline and cover the inner surface of the abdomen in a process strikingly reminiscent of embryonic mesoderm migration, requiring fibroblast growth factor (FGF) signaling as well. FGF signaling guides migration dorsally and regulates adhesion to the substrate. After spreading is complete, precursor differentiation involves fat accumulation and cell fusion that produces mature binucleate and tetranucleate adipocytes. Finally, we show that flies where adult adipogenesis is impaired by knock down of FGF receptor Heartless or transcription factor Serpent display ectopic fat accumulation in oenocytes and decreased resistance to starvation. Our results reveal that adult adipogenesis occurs de novo during metamorphosis and demonstrate its crucial physiological role.


Assuntos
Adipogenia , Drosophila , Animais , Drosophila/metabolismo , Corpo Adiposo/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Tecido Adiposo/metabolismo
3.
Front Cell Dev Biol ; 11: 1034484, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38264353

RESUMO

The fusion of epithelial sheets is an essential and conserved morphogenetic event that requires the maintenance of tissue continuity. This is secured by membrane-bound or diffusible signals that instruct the epithelial cells, in a coordinated fashion, to change shapes and adhesive properties and when, how and where to move. Here we show that during Dorsal Closure (DC) in Drosophila, the Jun kinase (JNK) signaling pathway modulates integrins expression and ensures tissue endurance. An excess of JNK activity, as an outcome of a failure in the negative feedback implemented by the dual-specificity phosphatase Puckered (Puc), promotes the loss of integrins [the ß-subunit Myospheroid (Mys)] and amnioserosa detachment. Likewise, integrins signal back to the pathway to regulate the duration and strength of JNK activity. Mys is necessary for the regulation of JNK activity levels and in its absence, puc expression is downregulated and JNK activity increases.

4.
Dis Model Mech ; 15(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34842272

RESUMO

Development involves tightly paced, reproducible sequences of events, yet it must adjust to conditions external to it, such as resource availability and organismal damage. A major mediator of damage-induced immune responses in vertebrates and insects is JAK/STAT signaling. At the same time, JAK/STAT activation by the Drosophila Upd cytokines is pleiotropically involved in normal development of multiple organs. Whether inflammatory and developmental JAK/STAT roles intersect is unknown. Here, we show that JAK/STAT is active during development of the prothoracic gland (PG), which controls metamorphosis onset through ecdysone production. Reducing JAK/STAT signaling decreased PG size and advanced metamorphosis. Conversely, JAK/STAT hyperactivation by overexpression of pathway components or SUMOylation loss caused PG hypertrophy and metamorphosis delay. Tissue damage and tumors, known to secrete Upd cytokines, also activated JAK/STAT in the PG and delayed metamorphosis, at least in part by inducing expression of the JAK/STAT target Apontic. JAK/STAT damage signaling, therefore, regulates metamorphosis onset by co-opting its developmental role in the PG. Our findings in Drosophila provide insights on how systemic effects of damage and cancer can interfere with hormonally controlled development and developmental transitions.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ecdisona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais/fisiologia
5.
Cell Rep ; 36(11): 109707, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525362

RESUMO

Secretory cargos are collected at endoplasmic reticulum (ER) exit sites (ERES) before transport to the Golgi apparatus. Decades of research have provided many details of the molecular events underlying ER-Golgi exchanges. Essential questions, however, remain about the organization of the ER-Golgi interface in cells and the type of membrane structures mediating traffic from ERES. To investigate these, we use transgenic tagging in Drosophila flies, 3D-structured illumination microscopy (SIM), and focused ion beam scanning electron microscopy (FIB-SEM) to characterize ERES-Golgi units in collagen-producing fat body, imaginal discs, and imaginal discs overexpressing ERES determinant Tango1. Facing ERES, we find a pre-cis-Golgi region, equivalent to the vertebrate ER-Golgi intermediate compartment (ERGIC), involved in both anterograde and retrograde transport. This pre-cis-Golgi is continuous with the rest of the Golgi, not a separate compartment or collection of large carriers, for which we find no evidence. We observe, however, many vesicles, as well as pearled tubules connecting ERES and Golgi.


Assuntos
Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Drosophila/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Fator 1 de Ribosilação do ADP/metabolismo , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Transporte Biológico , Proteínas de Drosophila/metabolismo , Retículo Endoplasmático/química , Complexo de Golgi/química , Proteínas da Matriz do Complexo de Golgi/metabolismo , Microscopia Eletrônica de Varredura , Proteínas Monoméricas de Ligação ao GTP/metabolismo
6.
J Cell Biol ; 220(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33263729

RESUMO

Mechanoreceptor cells develop a specialized cytoskeleton that plays structural and sensory roles at the site of mechanotransduction. However, little is known about how the cytoskeleton is organized and formed. Using electron tomography and live-cell imaging, we resolve the 3D structure and dynamics of the microtubule-based cytoskeleton in fly campaniform mechanosensory cilia. Investigating the formation of the cytoskeleton, we find that katanin p60-like 1 (kat-60L1), a neuronal type of microtubule-severing enzyme, serves two functions. First, it amplifies the mass of microtubules to form the dense microtubule arrays inside the sensory cilia. Second, it generates short microtubules that are required to build the nanoscopic cytoskeleton at the mechanotransduction site. Additional analyses further reveal the functional roles of Patronin and other potential factors in the local regulatory network. In all, our results characterize the specialized cytoskeleton in fly external mechanosensory cilia at near-molecular resolution and provide mechanistic insights into how it is formed.


Assuntos
Proteínas de Drosophila/metabolismo , Katanina/metabolismo , Mecanotransdução Celular , Animais , Polaridade Celular , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestrutura , Extremidades/fisiologia , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Modelos Biológicos , Organelas/metabolismo , Organelas/ultraestrutura , Receptores de Superfície Celular/metabolismo
7.
J Genet Genomics ; 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29935791

RESUMO

Collagens are large secreted trimeric proteins making up most of the animal extracellular matrix. Secretion of collagen has been a focus of interest for cell biologists in recent years because collagen trimers are too large and rigid to fit into the COPII vesicles mediating transport from the endoplasmic reticulum (ER) to the Golgi. Collagen-specific mechanisms to create enlarged ER-to-Golgi transport carriers have been postulated, including cargo loading by conserved ER exit site (ERES) protein Tango1. Here, we report an RNAi screening for genes involved in collagen secretion in Drosophila. In this screening, we examined distribution of GFP-tagged Collagen IV in live animals and found 88 gene hits for which the knockdown produced intracellular accumulation of Collagen IV in the fat body, the main source of matrix proteins in the larva. Among these hits, only two affected collagen secretion specifically: PH4αEFB and Plod, encoding enzymes known to mediate posttranslational modification of collagen in the ER. Every other intracellular accumulation hit affected general secretion, consistent with the notion that secretion of collagen does not use a specific mode of vesicular transport, but the general secretory pathway. Included in our hits are many known players in the eukaryotic secretory machinery, like COPII and COPI components, SNAREs and Rab-GTPase regulators. Our further analysis of the involvement of Rab-GTPases in secretion shows that Rab1, Rab2 and RabX3, are all required at ERES, each of them differentially affecting ERES morphology. Abolishing activity of all three by Rep knockdown, in contrast, led to uncoupling of ERES and Golgi. We additionally present a characterization of a screening hit we named trabuco (tbc), encoding an ERES-localized TBC domain-containing Rab-GAP. Finally, we discuss the success of our screening in identifying secretory pathway genes in comparison to two previous secretion screenings in Drosophila S2 cells.

8.
Development ; 145(13)2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29853618

RESUMO

Although the specific form of an organ is frequently important for its function, the mechanisms underlying organ shape are largely unknown. In Drosophila, the wings and halteres, homologous appendages of the second and third thoracic segments, respectively, bear different forms: wings are flat, whereas halteres are globular, and yet both characteristic shapes are essential for a normal flight. The Hox gene Ultrabithorax (Ubx) governs the difference between wing and haltere development, but how Ubx function in the appendages prevents or allows flat or globular shapes is unknown. Here, we show that Ubx downregulates Matrix metalloproteinase 1 (Mmp1) expression in the haltere pouch at early pupal stage, which in turn prevents the rapid clearance of Collagen IV compared with the wing disc. This difference is instrumental in determining cell shape changes, expansion of the disc and apposition of dorsal and ventral layers, all of these phenotypic traits being characteristic of wing pouch development. Our results suggest that Ubx regulates organ shape by controlling Mmp1 expression, and the extent and timing of extracellular matrix degradation.


Assuntos
Proteínas de Drosophila/biossíntese , Matriz Extracelular/metabolismo , Proteínas de Homeodomínio/biossíntese , Discos Imaginais/embriologia , Metaloproteinase 1 da Matriz/metabolismo , Fatores de Transcrição/biossíntese , Asas de Animais/embriologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Matriz Extracelular/genética , Proteínas de Homeodomínio/genética , Metaloproteinase 1 da Matriz/genética , Fatores de Transcrição/genética
9.
J Cell Biol ; 216(4): 1035-1049, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28280122

RESUMO

Exit of secretory cargo from the endoplasmic reticulum (ER) takes place at specialized domains called ER exit sites (ERESs). In mammals, loss of TANGO1 and other MIA/cTAGE (melanoma inhibitory activity/cutaneous T cell lymphoma-associated antigen) family proteins prevents ER exit of large cargoes such as collagen. Here, we show that Drosophila melanogaster Tango1, the only MIA/cTAGE family member in fruit flies, is a critical organizer of the ERES-Golgi interface. Tango1 rings hold COPII (coat protein II) carriers and Golgi in close proximity at their center. Loss of Tango1, present at ERESs in all tissues, reduces ERES size and causes ERES-Golgi uncoupling, which impairs secretion of not only collagen, but also all other cargoes we examined. Further supporting an organizing role of Tango1, its overexpression creates more and larger ERESs. Our results suggest that spatial coordination of ERES, carrier, and Golgi elements through Tango1's multiple interactions increases secretory capacity in Drosophila and allows secretion of large cargo.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/fisiologia , Animais , Transporte Biológico/fisiologia , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/fisiologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Complexo de Golgi/metabolismo , Complexo de Golgi/fisiologia , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia , Proteínas de Transporte Vesicular/metabolismo
11.
Open Biol ; 5(7): 140171, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26202785

RESUMO

Signalling networks that control the life or death of a cell are of central interest in modern biology. While the defined roles of the c-Jun N-terminal kinase (JNK) pathway in regulating cell death have been well-established, additional factors that modulate JNK-mediated cell death have yet to be fully elucidated. To identify novel regulators of JNK-dependent cell death, we performed a dominant-modifier screen in Drosophila and found that the Toll pathway participates in JNK-mediated cell death. Loss of Toll signalling suppresses ectopically and physiologically activated JNK signalling-induced cell death. Our epistasis analysis suggests that the Toll pathway acts as a downstream modulator for JNK-dependent cell death. In addition, gain of JNK signalling results in Toll pathway activation, revealed by stimulated transcription of Drosomycin (Drs) and increased cytoplasm-to-nucleus translocation of Dorsal. Furthermore, the Spätzle (Spz) family ligands for the Toll receptor are transcriptionally upregulated by activated JNK signalling in a non-cell-autonomous manner, providing a molecular mechanism for JNK-induced Toll pathway activation. Finally, gain of Toll signalling exacerbates JNK-mediated cell death and promotes cell death independent of caspases. Thus, we have identified another important function for the evolutionarily conserved Toll pathway, in addition to its well-studied roles in embryonic dorso-ventral patterning and innate immunity.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Sistema de Sinalização das MAP Quinases , Receptores Toll-Like/metabolismo , Animais , Morte Celular/genética , Olho Composto de Artrópodes/citologia , Olho Composto de Artrópodes/crescimento & desenvolvimento , Drosophila/citologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Receptores Toll-Like/fisiologia , Asas de Animais/citologia , Asas de Animais/crescimento & desenvolvimento
12.
Elife ; 4: e07187, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-26090908

RESUMO

Many chronic diseases are associated with fibrotic deposition of Collagen and other matrix proteins. Little is known about the factors that determine preferential onset of fibrosis in particular tissues. Here we show that plasma membrane (PM) overgrowth causes pericellular Collagen accumulation in Drosophila adipocytes. We found that loss of Dynamin and other endocytic components causes pericellular trapping of outgoing Collagen IV due to dramatic cortex expansion when endocytic removal of PM is prevented. Deposits also form in the absence of negative Toll immune regulator Cactus, excess PM being caused in this case by increased secretion. Finally, we show that trimeric Collagen accumulation, downstream of Toll or endocytic defects, activates a tissue damage response. Our work indicates that traffic imbalances and PM topology may contribute to fibrosis. It also places fibrotic deposits both downstream and upstream of immune signaling, consistent with the chronic character of fibrotic diseases.


Assuntos
Adipócitos/fisiologia , Membrana Celular/metabolismo , Colágeno/metabolismo , Drosophila/fisiologia , Animais
13.
Annu Rev Genet ; 47: 51-74, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23988119

RESUMO

Cancer was seen for a long time as a strictly cell-autonomous process in which oncogenes and tumor-suppressor mutations drive clonal cell expansions. Research in the past decade, however, paints a more integrative picture of communication and interplay between neighboring cells in tissues. It is increasingly clear as well that tumors, far from being homogenous lumps of cells, consist of different cell types that function together as complex tissue-level communities. The repertoire of interactive cell behaviors and the quantity of cellular players involved call for a social cell biology that investigates these interactions. Research into this social cell biology is critical for understanding development of normal and tumoral tissues. Such complex social cell biology interactions can be parsed in Drosophila. Techniques in Drosophila for analysis of gene function and clonal behavior allow us to generate tumors and dissect their complex interactive biology with cellular resolution. Here, we review recent Drosophila research aimed at understanding tissue-level biology and social cell interactions in tumors, highlighting the principles these studies reveal.


Assuntos
Comunicação Celular/genética , Transformação Celular Neoplásica/genética , Drosophila melanogaster/genética , Animais , Animais Geneticamente Modificados , Membrana Basal/fisiologia , Adesão Celular , Morte Celular , Divisão Celular , Microambiente Celular , Células Clonais/citologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Genes de Insetos , Genes Supressores de Tumor , Homeostase , Humanos , Discos Imaginais/citologia , Imunidade Inata , Mitose/efeitos da radiação , Mosaicismo , Neoplasias/genética , Neoplasias/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Células-Tronco Neoplásicas/citologia , Recombinação Genética/efeitos da radiação , Evasão Tumoral
14.
Dev Cell ; 21(2): 245-56, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21839919

RESUMO

Basement membranes (BMs) are resilient polymer structures that surround organs in all animals. Tissues, however, undergo extensive morphological changes during development. It is not known whether the assembly of BM components plays an active morphogenetic role. To study in vivo the biogenesis and assembly of Collagen IV, the main constituent of BMs, we used a GFP-based RNAi method (iGFPi) designed to knock down any GFP-trapped protein in Drosophila. We found with this method that Collagen IV is synthesized by the fat body, secreted to the hemolymph (insect blood), and continuously incorporated into the BMs of the larva. We also show that incorporation of Collagen IV determines organ shape, first by mechanically constricting cells and second through recruitment of Perlecan, which counters constriction by Collagen IV. Our results uncover incorporation of Collagen IV and Perlecan into BMs as a major determinant of organ shape and animal form.


Assuntos
Forma Celular/fisiologia , Colágeno Tipo IV/metabolismo , Corpo Adiposo/citologia , Corpo Adiposo/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Asas de Animais/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados , Membrana Basal/metabolismo , Colágeno Tipo IV/genética , Drosophila , Proteínas de Drosophila/genética , Corpo Adiposo/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Larva , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Interferência de RNA/fisiologia , Asas de Animais/ultraestrutura
15.
Proc Natl Acad Sci U S A ; 108(22): 9232-7, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21576456

RESUMO

Cellular imbalances of cholesterol and fatty acid metabolism result in pathological processes, including atherosclerosis and metabolic syndrome. Recent work from our group and others has shown that the intronic microRNAs hsa-miR-33a and hsa-miR-33b are located within the sterol regulatory element-binding protein-2 and -1 genes, respectively, and regulate cholesterol homeostasis in concert with their host genes. Here, we show that miR-33a and -b also regulate genes involved in fatty acid metabolism and insulin signaling. miR-33a and -b target key enzymes involved in the regulation of fatty acid oxidation, including carnitine O-octaniltransferase, carnitine palmitoyltransferase 1A, hydroxyacyl-CoA-dehydrogenase, Sirtuin 6 (SIRT6), and AMP kinase subunit-α. Moreover, miR-33a and -b also target the insulin receptor substrate 2, an essential component of the insulin-signaling pathway in the liver. Overexpression of miR-33a and -b reduces both fatty acid oxidation and insulin signaling in hepatic cell lines, whereas inhibition of endogenous miR-33a and -b increases these two metabolic pathways. Together, these data establish that miR-33a and -b regulate pathways controlling three of the risk factors of metabolic syndrome, namely levels of HDL, triglycerides, and insulin signaling, and suggest that inhibitors of miR-33a and -b may be useful in the treatment of this growing health concern.


Assuntos
Ácidos Graxos/metabolismo , Insulina/metabolismo , MicroRNAs/biossíntese , Animais , Doenças Cardiovasculares/metabolismo , Colesterol/metabolismo , Citoplasma/metabolismo , Drosophila melanogaster/metabolismo , Homeostase , Humanos , Imuno-Histoquímica/métodos , Lipídeos/química , Fosforilação , Processamento Pós-Transcricional do RNA , Transdução de Sinais
16.
Nature ; 463(7280): 545-8, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-20072127

RESUMO

Human tumours have a large degree of cellular and genetic heterogeneity. Complex cell interactions in the tumour and its microenvironment are thought to have an important role in tumorigenesis and cancer progression. Furthermore, cooperation between oncogenic genetic lesions is required for tumour development; however, it is not known how cell interactions contribute to oncogenic cooperation. The genetic techniques available in the fruitfly Drosophila melanogaster allow analysis of the behaviour of cells with distinct mutations, making this the ideal model organism with which to study cell interactions and oncogenic cooperation. In Drosophila eye-antennal discs, cooperation between the oncogenic protein Ras(V12) (ref. 5) and loss-of-function mutations in the conserved tumour suppressor scribbled (scrib) gives rise to metastatic tumours that display many characteristics observed in human cancers. Here we show that clones of cells bearing different mutations can cooperate to promote tumour growth and invasion in Drosophila. We found that the Ras(V12) and scrib(-) mutations can also cause tumours when they affect different adjacent epithelial cells. We show that this interaction between Ras(V12) and scrib(-) clones involves JNK signalling propagation and JNK-induced upregulation of JAK/STAT-activating cytokines, a compensatory growth mechanism for tissue homeostasis. The development of Ras(V12) tumours can also be triggered by tissue damage, a stress condition that activates JNK signalling. Given the conservation of the pathways examined here, similar cooperative mechanisms could have a role in the development of human cancers.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas ras/metabolismo , Animais , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Janus Quinases/metabolismo , MAP Quinase Quinase 4/metabolismo , Neoplasias/patologia , Fatores de Transcrição STAT/metabolismo
17.
Dev Cell ; 16(3): 458-65, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19289090

RESUMO

Oncogenic alterations in epithelial tissues often trigger apoptosis, suggesting an evolutionary mechanism by which organisms eliminate aberrant cells from epithelia. In Drosophila imaginal epithelia, clones of cells mutant for tumor suppressors, such as scrib or dlg, lose their polarity and are eliminated by cell death. Here, we show that Eiger, the Drosophila tumor necrosis factor (TNF), behaves like a tumor suppressor that eliminates oncogenic cells from epithelia through a local endocytic JNK-activation mechanism. In the absence of Eiger, these polarity-deficient clones are no longer eliminated; instead, they grow aggressively into tumors. We show that in scrib clones endocytosis is elevated, which translocates Eiger to endocytic vesicles and leads to activation of apoptotic JNK signaling. Furthermore, blocking endocytosis prevents both JNK activation and cell elimination. Our data indicate that TNF signaling and the endocytic machinery could be components of an evolutionarily conserved fail-safe mechanism by which animals protect against neoplastic development.


Assuntos
Proteínas de Drosophila/fisiologia , Proteínas de Membrana/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Animais Geneticamente Modificados , Apoptose , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Drosophila/fisiologia , Proteínas de Drosophila/genética , Endocitose , Epitélio/crescimento & desenvolvimento , Genes de Insetos , Genes Supressores de Tumor , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Proteínas de Membrana/genética , Mutação , Neoplasias Experimentais/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética
18.
Dis Model Mech ; 1(2-3): 144-54; discussion 153, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19048077

RESUMO

Studies in mice and humans have demonstrated a role for the immune system in preventing the growth of tumors. Deciphering the mechanisms involved in the immune response to tumors is essential to our understanding of immune recognition and cancer progression. Here we report an innate immune response to tumors in Drosophila melanogaster. We found that circulating blood cells, termed hemocytes, adhere to tumors upon detection of basement membrane disruption, and subsequently counter their growth. Basement membrane components are remarkably conserved throughout the animal kingdom, providing a unique structure for the immune system to sense tissue integrity. Further, we show that tissue damage activates JNK signaling in both tumors and aseptic wounds, causing expression of JAK/STAT-activating cytokines. Cytokine secretion from the injured tissue is amplified into a systemic response through the induction of additional cytokine expression in the hemocytes and the fat body, resulting in hemocyte proliferation. Our findings reveal common mechanisms in the response to tumors and wounds in flies. A similar innate reaction may underlie the response to tumors and tissue damage in vertebrates and humans.


Assuntos
Drosophila melanogaster/imunologia , Imunidade Inata , Neoplasias Experimentais/imunologia , Animais , Adesão Celular , Hemócitos/patologia , Neoplasias Experimentais/patologia
19.
Proc Natl Acad Sci U S A ; 104(8): 2721-6, 2007 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-17301221

RESUMO

Organ and tissue integrity is often maintained in animals by a specialized extracellular matrix structure called the basement membrane (BM). Accumulated evidence indicates that BM remodeling occurs during development and tumor invasion. Although the BM organizes and functions at the organ level, most past studies have explored its biochemical and in vitro properties. In this study, we monitor the BM in vivo during developmental tissue invasion for disc eversion and tumor invasion in Drosophila and modulate BM integrity with genetic alterations affecting either the whole organism or the targeted discs or tumors. We observe that the degradation of BM by the discs or the tumors is an early event during invasion processes and that preventing BM degradation completely blocks both tissue and tumor invasion, indicating that modulation of BM is essential for developmental and tumor invasion. Furthermore, elements of the invasion machinery, including JNK-induced matrix metalloproteinase (MMP) expression, are shared by both disc eversion and tumor invasion processes. Moreover, we show that although expression of MMP inhibitor, TIMP, is sufficient to halt developmental invasion, inhibition of proteases by both TIMP and RECK are required to block tumor invasion. These data suggest that tumor cells have a more robust invasion mechanism and could acquire metastatic behavior by co-opting developmental invasion programs. This type of co-option may be a general feature contributing to the progression of tumors. Finally, although past efforts using MMP inhibitors have not yielded much success, our results strongly argue that BM modulation could be a critical target for cancer therapy.


Assuntos
Membrana Basal/metabolismo , Drosophila melanogaster/embriologia , Neoplasias/patologia , Asas de Animais/embriologia , Animais , Drosophila melanogaster/enzimologia , Ativação Enzimática , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinases da Matriz/metabolismo , Modelos Biológicos , Neoplasias/enzimologia , Pupa/citologia , Transdução de Sinais , Asas de Animais/anormalidades , Asas de Animais/citologia
20.
Dev Cell ; 7(3): 387-99, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15363413

RESUMO

Drosophila imaginal discs are monolayered epithelial invaginations that grow during larval stages and evert at metamorphosis to assemble the adult exoskeleton. They consist of columnar cells, forming the imaginal epithelium, as well as squamous cells, which constitute the peripodial epithelium and stalk (PS). Here, we uncover a new morphogenetic/cellular mechanism for disc eversion. We show that imaginal discs evert by apposing their peripodial side to the larval epidermis and through the invasion of the larval epidermis by PS cells, which undergo a pseudo-epithelial-mesenchymal transition (PEMT). As a consequence, the PS/larval bilayer is perforated and the imaginal epithelia protrude, a process reminiscent of other developmental events, such as epithelial perforation in chordates. When eversion is completed, PS cells localize to the leading front, heading disc expansion. We found that the JNK pathway is necessary for PS/larval cells apposition, the PEMT, and the motile activity of leading front cells.


Assuntos
Drosophila/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Quinases JNK Ativadas por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Transdução de Sinais , Asas de Animais/embriologia , Animais , Padronização Corporal , Divisão Celular , Linhagem Celular , Drosophila melanogaster/metabolismo , Embrião não Mamífero/fisiologia , Epiderme/embriologia , Epiderme/metabolismo , Epitélio/metabolismo , Imuno-Histoquímica , MAP Quinase Quinase 4 , Mesoderma/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Fenótipo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...