Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Annu Rev Cell Dev Biol ; 29: 107-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23937350

RESUMO

Adult animals rely on populations of stem cells to ensure organ function throughout their lifetime. Stem cells are governed by signals from stem cell niches, and much is known about how single niches promote stemness and direct stem cell behavior. However, most organs contain a multitude of stem cell-niche units, which are often distributed across the entire expanse of the tissue. Beyond the biology of individual stem cell-niche interactions, the next challenge is to uncover the tissue-level processes that orchestrate spatial control of stem-based renewal, repair, and remodeling throughout a whole organ. Here we examine what is known about higher order mechanisms for interniche coordination in epithelial organs, whose simple geometry offers a promising entry point for understanding the regulation of niche number, distribution, and activity. We also consider the potential existence of stem cell territories and how tissue architecture may influence niche coordination.


Assuntos
Células Epiteliais/fisiologia , Epitélio/fisiologia , Células-Tronco/fisiologia , Animais , Epitélio/embriologia , Organogênese , Regeneração , Nicho de Células-Tronco , Células-Tronco/citologia
2.
Cell ; 147(3): 603-14, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-22036568

RESUMO

Throughout life, adult organs continually adapt to variable environmental factors. Adaptive mechanisms must fundamentally differ from homeostatic maintenance, but little is known about how physiological factors elicit tissue remodeling. Here, we show that specialized stem cell responses underlie the adaptive resizing of a mature organ. In the adult Drosophila midgut, intestinal stem cells interpret a nutrient cue to "break homeostasis" and drive growth when food is abundant. Activated in part by niche production of insulin, stem cells direct a growth program through two altered modes of behavior: accelerated division rates and predominance of symmetric division fates. Together, these altered modes produce a net increase in total intestinal cells, which is reversed upon withdrawal of food. Thus, tissue renewal programs are not committed to maintain cellular equilibrium; stem cells can remodel organs in response to physiological triggers.


Assuntos
Divisão Celular , Drosophila/citologia , Drosophila/fisiologia , Células-Tronco/citologia , Animais , Drosophila/crescimento & desenvolvimento , Jejum , Feminino , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citologia , Células-Tronco/metabolismo
3.
Genes Dev ; 32(2): 156-164, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29440263

RESUMO

Insulin resistance, the failure to activate insulin signaling in the presence of ligand, leads to metabolic diseases, including type 2 diabetes. Physical activity and mechanical stress have been shown to protect against insulin resistance, but the molecular mechanisms remain unclear. Here, we address this relationship in the Drosophila larval fat body, an insulin-sensitive organ analogous to vertebrate adipose tissue and livers. We found that insulin signaling in Drosophila fat body cells is abolished in the absence of physical activity and mechanical stress even when excess insulin is present. Physical movement is required for insulin sensitivity in both intact larvae and fat bodies cultured ex vivo. Interestingly, the insulin receptor and other downstream components are recruited to the plasma membrane in response to mechanical stress, and this membrane localization is rapidly lost upon disruption of larval or tissue movement. Sensing of mechanical stimuli is mediated in part by integrins, whose activation is necessary and sufficient for mechanical stress-dependent insulin signaling. Insulin resistance develops naturally during the transition from the active larval stage to the immotile pupal stage, suggesting that regulation of insulin sensitivity by mechanical stress may help coordinate developmental programming with metabolism.


Assuntos
Proteínas de Drosophila/metabolismo , Insulina/fisiologia , Integrinas/metabolismo , Receptor de Insulina/metabolismo , Estresse Mecânico , Animais , Membrana Celular , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Matriz Extracelular/metabolismo , Cadeias beta de Integrinas/metabolismo , Larva/metabolismo , Movimento , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Talina/metabolismo
4.
Nature ; 541(7637): 417-420, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28077876

RESUMO

As malignant tumours develop, they interact intimately with their microenvironment and can activate autophagy, a catabolic process which provides nutrients during starvation. How tumours regulate autophagy in vivo and whether autophagy affects tumour growth is controversial. Here we demonstrate, using a well characterized Drosophila melanogaster malignant tumour model, that non-cell-autonomous autophagy is induced both in the tumour microenvironment and systemically in distant tissues. Tumour growth can be pharmacologically restrained using autophagy inhibitors, and early-stage tumour growth and invasion are genetically dependent on autophagy within the local tumour microenvironment. Induction of autophagy is mediated by Drosophila tumour necrosis factor and interleukin-6-like signalling from metabolically stressed tumour cells, whereas tumour growth depends on active amino acid transport. We show that dormant growth-impaired tumours from autophagy-deficient animals reactivate tumorous growth when transplanted into autophagy-proficient hosts. We conclude that transformed cells engage surrounding normal cells as active and essential microenvironmental contributors to early tumour growth through nutrient-generating autophagy.


Assuntos
Autofagia , Drosophila melanogaster/citologia , Modelos Biológicos , Neoplasias/patologia , Microambiente Tumoral , Aminoácidos/metabolismo , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Transporte Biológico , Proliferação de Células , Modelos Animais de Doenças , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Feminino , Interleucina-6/metabolismo , Proteínas de Membrana , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
5.
Proc Natl Acad Sci U S A ; 117(21): 11531-11540, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32414916

RESUMO

A polarized architecture is central to both epithelial structure and function. In many cells, polarity involves mutual antagonism between the Par complex and the Scribble (Scrib) module. While molecular mechanisms underlying Par-mediated apical determination are well-understood, how Scrib module proteins specify the basolateral domain remains unknown. Here, we demonstrate dependent and independent activities of Scrib, Discs-large (Dlg), and Lethal giant larvae (Lgl) using the Drosophila follicle epithelium. Our data support a linear hierarchy for localization, but rule out previously proposed protein-protein interactions as essential for polarization. Cortical recruitment of Scrib does not require palmitoylation or polar phospholipid binding but instead an independent cortically stabilizing activity of Dlg. Scrib and Dlg do not directly antagonize atypical protein kinase C (aPKC), but may instead restrict aPKC localization by enabling the aPKC-inhibiting activity of Lgl. Importantly, while Scrib, Dlg, and Lgl are each required, all three together are not sufficient to antagonize the Par complex. Our data demonstrate previously unappreciated diversity of function within the Scrib module and begin to define the elusive molecular functions of Scrib and Dlg.


Assuntos
Polaridade Celular/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila , Células Epiteliais , Proteínas de Membrana/fisiologia , Animais , Drosophila/citologia , Drosophila/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Epitélio/fisiologia , Feminino , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia , Proteína Quinase C , Proteínas Supressoras de Tumor
6.
Development ; 141(15): 2978-83, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25053431

RESUMO

The Par-3/Par-6/aPKC complex is the primary determinant of apical polarity in epithelia across animal species, but how the activity of this complex is restricted to allow polarization of the basolateral domain is less well understood. In Drosophila, several multiprotein modules antagonize the Par complex through a variety of means. Here we identify a new mechanism involving regulated protein degradation. Strong mutations in supernumerary limbs (slmb), which encodes the substrate adaptor of an SCF-class E3 ubiquitin ligase, cause dramatic loss of polarity in imaginal discs accompanied by tumorous proliferation defects. Slmb function is required to restrain apical aPKC activity in a manner that is independent of endolysosomal trafficking and parallel to the Scribble module of junctional scaffolding proteins. The involvement of the Slmb E3 ligase in epithelial polarity, specifically limiting Par complex activity to distinguish the basolateral domain, points to parallels with polarization of the C. elegans zygote.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas de Drosophila/fisiologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteína Quinase C/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Alelos , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células , Transformação Celular Neoplásica , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Endossomos/metabolismo , Proteínas F-Box/fisiologia , Feminino , Lisossomos/metabolismo , Mutação , Fenótipo , Transporte Proteico , Ubiquitina-Proteína Ligases/genética
7.
Development ; 141(14): 2796-802, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25005475

RESUMO

Scribble (Scrib) module proteins are major regulators of cell polarity, but how they influence membrane traffic is not known. Endocytosis is also a key regulator of polarity through roles that remain unclear. Here we link Scrib to a specific arm of the endocytic trafficking system. Drosophila mutants that block AP-2-dependent endocytosis share many phenotypes with Scrib module mutants, but Scrib module mutants show intact internalization and endolysosomal transport. However, defective traffic of retromer pathway cargo is seen, and retromer components show strong genetic interactions with the Scrib module. The Scrib module is required for proper retromer localization to endosomes and promotes appropriate cargo sorting into the retromer pathway via both aPKC-dependent and -independent mechanisms. We propose that the Scrib module regulates epithelial polarity by influencing endocytic itineraries of Crumbs and other retromer-dependent cargo.


Assuntos
Polaridade Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Endocitose , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas de Membrana/metabolismo , Complexo 2 de Proteínas Adaptadoras/metabolismo , Animais , Proliferação de Células , Drosophila melanogaster/enzimologia , Olho/citologia , Olho/metabolismo , Feminino , Lisossomos/metabolismo , Mutação/genética , Folículo Ovariano/citologia , Folículo Ovariano/enzimologia , Fenótipo , Proteína Quinase C/metabolismo , Transporte Proteico
8.
bioRxiv ; 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-39026720

RESUMO

While paradigms for patterning of cell fates in development are well-established, paradigms for patterning morphogenesis, particularly when organ shape is influenced by the extracellular matrix (ECM), are less so. Morphogenesis of the Drosophila egg chamber (follicle) depends on anterior-posterior distribution of basement membrane (BM) components such as Collagen IV (Col4), whose symmetric gradient creates tissue mechanical properties that specify the degree of elongation. Here we show that the gradient is not regulated by Col4 transcription but instead relies on post-transcriptional mechanisms. The metalloprotease ADAMTS-A, expressed in a gradient inverse to that of Col4, limits Col4 deposition in the follicle center and manipulation of its levels can cause either organ hyper- or hypo-elongation. We present evidence that ADAMTS-A acts within the secretory pathway, rather than extracellularly, to limit Col4 incorporation into the BM. High levels of ADAMTS-A in follicle termini are normally dispensable but suppress Col4 incorporation when transcription is elevated. Our data show how an organ can employ patterned expression of ECM proteases with intracellular as well as extracellular activity to specify BM properties that control shape.

9.
bioRxiv ; 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38585959

RESUMO

Tumors can induce systemic disturbances in distant organs, leading to physiological changes that enhance host morbidity. In Drosophila cancer models, tumors have been known for decades to cause hypervolemic 'bloating' of the abdominal cavity. Here we use allograft and transgenic tumors to show that hosts display fluid retention associated with autonomously defective secretory capacity of fly renal tubules, which function analogous to those of the human kidney. Excretion from these organs is blocked by abnormal cells that originate from inappropriate activation of normally quiescent renal stem cells (RSCs). Blockage is initiated by IL-6-like oncokines that perturb renal water-transporting cells, and trigger a damage response in RSCs that proceeds pathologically. Thus, a chronic inflammatory state produced by the tumor causes paraneoplastic fluid dysregulation by altering cellular homeostasis of host renal units.

10.
Development ; 137(11): 1825-32, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20460366

RESUMO

Evidence indicates that endosomal entry promotes signaling by the Notch receptor, but the mechanisms involved are not clear. In a search for factors that regulate Notch activation in endosomes, we isolated mutants in Drosophila genes that encode subunits of the vacuolar ATPase (V-ATPase) proton pump. Cells lacking V-ATPase function display impaired acidification of the endosomal compartment and a correlated failure to degrade endocytic cargoes. V-ATPase mutant cells internalize Notch and accumulate it in the lysosome, but surprisingly also show a substantial loss of both physiological and ectopic Notch activation in endosomes. V-ATPase activity is required in signal-receiving cells for Notch signaling downstream of ligand activation but upstream of gamma-secretase-dependent S3 cleavage. These data indicate that V-ATPase, probably via acidification of early endosomes, promotes not only the degradation of Notch in the lysosome but also the activation of Notch signaling in endosomes. The results also suggest that the ionic properties of the endosomal lumen might regulate Notch cleavage, providing a rationale for physiological as well as pathological endocytic control of Notch activity.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Receptores Notch/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/genética , Endossomos/metabolismo , Olho/crescimento & desenvolvimento , Olho/metabolismo , Olho/ultraestrutura , Genes de Insetos , Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , Microscopia Eletrônica de Transmissão , Mutação , Receptores Notch/genética , Transdução de Sinais , ATPases Vacuolares Próton-Translocadoras/genética
11.
Dev Cell ; 58(3): 211-223.e5, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36708706

RESUMO

Shaping of developing organs requires dynamic regulation of force and resistance to achieve precise outcomes, but how organs monitor tissue mechanical properties is poorly understood. We show that in developing Drosophila follicles (egg chambers), a single pair of cells performs such monitoring to drive organ shaping. These anterior polar cells secrete a matrix metalloproteinase (MMP) that specifies the appropriate degree of tissue elongation, rather than hyper- or hypo-elongated organs. MMP production is negatively regulated by basement membrane (BM) mechanical properties, which are sensed through focal adhesion signaling and autonomous contractile activity; MMP then reciprocally regulates BM remodeling, particularly at the anterior region. Changing BM properties at remote locations alone is sufficient to induce a remodeling response in polar cells. We propose that this small group of cells senses both local and distant stiffness cues to produce factors that pattern the organ's BM mechanics, ensuring proper tissue shape and reproductive success.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Membrana Basal , Morfogênese/fisiologia , Matriz Extracelular
12.
Curr Biol ; 33(14): 3002-3010.e6, 2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37354901

RESUMO

Malignant tumors trigger a complex network of inflammatory and wound repair responses, prompting Dvorak's characterization of tumors as "wounds that never heal."1 Some of these responses lead to profound defects in blood clotting, such as disseminated intravascular coagulopathy (DIC), which correlate with poor prognoses.2,3,4 Here, we demonstrate that a new tumor model in Drosophila provokes phenotypes that resemble coagulopathies observed in patients. Fly ovarian tumors overproduce multiple secreted components of the clotting cascade and trigger hypercoagulation of fly blood (hemolymph). Hypercoagulation occurs shortly after tumor induction and is transient; it is followed by a hypocoagulative state that is defective in wound healing. Cellular clotting regulators accumulate on the tumor over time and are depleted from the body, suggesting that hypocoagulation is caused by exhaustion of host clotting components. We show that rescuing coagulopathy by depleting a tumor-produced clotting factor improves survival of tumor-bearing flies, despite the fact that flies have an open (non-vascular) circulatory system. As clinical studies suggest that lethality in patients with high serum levels of clotting components can be independent of thrombotic events,5,6 our work establishes a platform for identifying alternative mechanisms by which tumor-driven coagulopathy triggers early mortality. Moreover, it opens up exploration of other conserved mechanisms of host responses to chronic wounds.


Assuntos
Modelos Animais de Doenças , Animais , Transtornos da Coagulação Sanguínea/etiologia , Neoplasias Ovarianas/complicações , Transcriptoma
13.
Nat Cell Biol ; 7(12): 1232-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16258546

RESUMO

Intracellular protein transport is a key factor in epithelial cell polarity. Here we report that mutations in two core components of the vesicle trafficking machinery - a syntaxin and a Rab protein - cause an expansion of the apical membrane domain of Drosophila melanogaster epithelia; this polarity defect is coupled with overproliferation to form neoplastic tumours. Surprisingly, these proteins are associated with the endocytic, and not the exocytic, pathway. The syntaxin Avalanche (Avl) localizes to early endosomes, and loss of avl results in the cellular accumulation of specific membrane proteins, including the Notch signalling receptor and the polarity determinant Crumbs (Crb). Protein accumulation results from a failure in endosomal entry and progression towards lysosomal degradation; these and other avl phenotypes are also detected in Rab5 null mutant cells. Overexpression of Crb alone is sufficient to induce overproliferation of wild-type imaginal tissue, suggesting that polarity alterations in avl and Rab5 mutants directly contribute to tumour formation. Our findings reveal a critical and specific role for endocytic traffic in the control of both apico-basal polarity and cell proliferation.


Assuntos
Drosophila melanogaster/citologia , Endocitose/fisiologia , Células Epiteliais/citologia , Animais , Membrana Celular/fisiologia , Polaridade Celular , Proliferação de Células , Células Epiteliais/ultraestrutura , Proteínas de Membrana/metabolismo , Mutação , Transporte Proteico , Proteínas Qa-SNARE/genética , Proteínas rab5 de Ligação ao GTP/genética
14.
Biol Open ; 11(7)2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35722710

RESUMO

The compartmentalized domains of polarized epithelial cells arise from mutually antagonistic actions between the apical Par complex and the basolateral Scrib module. In Drosophila, the Scrib module proteins Scribble (Scrib) and Discs-large (Dlg) are required to limit Lgl phosphorylation at the basolateral cortex, but how Scrib and Dlg could carry out such a 'protection' activity is not clear. We tested Protein Phosphatase 1α (PP1) as a potential mediator of this activity, but demonstrate that a significant component of Scrib and Dlg regulation of Lgl is PP1 independent, and found no evidence for a Scrib-Dlg-PP1 protein complex. However, the Dlg SH3 domain plays a role in Lgl protection and, in combination with the N-terminal region of the Dlg HOOK domain, in recruitment of Scrib to the membrane. We identify a 'minimal Dlg' comprised of the SH3 and HOOK domains that is both necessary and sufficient for Scrib localization and epithelial polarity function in vivo. This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas de Drosophila , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Epiteliais/metabolismo , Humanos
15.
Science ; 376(6590): 297-301, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35420935

RESUMO

Animals have evolved mechanisms, such as cell competition, to remove dangerous or nonfunctional cells from a tissue. Tumor necrosis factor signaling can eliminate clonal malignancies from Drosophila imaginal epithelia, but why this pathway is activated in tumor cells but not normal tissue is unknown. We show that the ligand that drives elimination is present in basolateral circulation but remains latent because it is spatially segregated from its apically localized receptor. Polarity defects associated with malignant transformation cause receptor mislocalization, allowing ligand binding and subsequent apoptotic signaling. This process occurs irrespective of the neighboring cells' genotype and is thus distinct from cell competition. Related phenomena at epithelial wound sites are required for efficient repair. This mechanism of polarized compartmentalization of ligand and receptor can generally monitor epithelial integrity to promote tissue homeostasis.


Assuntos
Competição entre as Células , Transformação Celular Neoplásica , Proteínas de Drosophila , Drosophila melanogaster , Células Epiteliais , Animais , Polaridade Celular/fisiologia , Transformação Celular Neoplásica/patologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/fisiologia , Células Epiteliais/fisiologia , Discos Imaginais/citologia , Ligantes , Transdução de Sinais
16.
Biol Open ; 11(11)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36355597

RESUMO

Animal organs maintain tissue integrity and ensure removal of aberrant cells through several types of surveillance mechanisms. One prominent example is the elimination of polarity-deficient mutant cells within developing Drosophila imaginal discs. This has been proposed to require heterotypic cell competition dependent on the receptor tyrosine phosphatase PTP10D within the mutant cells. We report here experiments to test this requirement in various contexts and find that PTP10D is not obligately required for the removal of scribble (scrib) mutant and similar polarity-deficient cells. Our experiments used identical stocks with which another group can detect the PTP10D requirement, and our results do not vary under several husbandry conditions including high and low protein food diets. Although we are unable to identify the source of the discrepant results, we suggest that the role of PTP10D in polarity-deficient cell elimination may not be absolute.


Assuntos
Proteínas de Drosophila , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Competição entre as Células , Drosophila/genética , Drosophila/metabolismo , Células Clonais/metabolismo
17.
Nat Cell Biol ; 5(1): 53-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12510194

RESUMO

Polarized cells contain numerous membrane domains, but it is unclear how the formation of these domains is coordinated to create a single integrated cell architecture. Genetic screens of Drosophila melanogaster embryos have identified three complexes, each containing one of the PDZ domain proteins--Stardust (Sdt), Bazooka (Baz) and Scribble (Scrib)--that control epithelial polarity and formation of zonula adherens. We find that these complexes can be ordered into a single regulatory hierarchy that is initiated by cell adhesion-dependent recruitment of the Baz complex to the zonula adherens. The Scrib complex represses apical identity along basolateral surfaces by antagonizing Baz-initiated apical polarity. The Sdt-containing Crb complex is recruited apically by the Baz complex to counter antagonistic Scrib activity. Thus, a finely tuned balance between Scrib and Crb complex activity sets the limits of the apical and basolateral membrane domains and positions cell junctions. Our data suggest a model in which the maturation of epithelial cell polarity is driven by integration of the sequential activities of PDZ-based protein complexes.


Assuntos
Proteínas de Transporte/metabolismo , Polaridade Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Células Epiteliais/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Núcleosídeo-Fosfato Quinase/metabolismo , Alelos , Animais , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Células Epiteliais/citologia , Guanilato Quinases , Proteínas de Membrana/genética , Núcleosídeo-Fosfato Quinase/genética
18.
PLoS Genet ; 4(1): e8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18208331

RESUMO

Intense investigation has identified an elaborate protein network controlling epithelial polarity. Although precise subcellular targeting of apical and basolateral determinants is required for epithelial architecture, little is known about how the individual determinant proteins become localized within the cell. Through a genetic screen for epithelial defects in the Drosophila follicle cells, we have found that the cytoplasmic Dynein motor is an essential regulator of apico-basal polarity. Our data suggest that Dynein acts through the cytoplasmic scaffolding protein Stardust (Sdt) to localize the transmembrane protein Crumbs, in part through the apical targeting of specific sdt mRNA isoforms. We have mapped the sdt mRNA localization signal to an alternatively spliced coding exon. Intriguingly, the presence or absence of this exon corresponds to a developmental switch in sdt mRNA localization in which apical transcripts are only found during early stages of epithelial development, while unlocalized transcripts predominate in mature epithelia. This work represents the first demonstration that Dynein is required for epithelial polarity and suggests that mRNA localization may have a functional role in the regulation of apico-basal organization. Moreover, we introduce a unique mechanism in which alternative splicing of a coding exon is used to control mRNA localization during development.


Assuntos
Polaridade Celular , Proteínas de Drosophila/metabolismo , Dineínas/fisiologia , Células Epiteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana Transportadoras/metabolismo , Núcleosídeo-Fosfato Quinase/metabolismo , RNA Mensageiro/metabolismo , Alelos , Animais , Animais Geneticamente Modificados , Drosophila/citologia , Drosophila/embriologia , Drosophila/metabolismo , Proteínas de Drosophila/genética , Dineínas/genética , Embrião não Mamífero , Feminino , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Guanilato Quinases , Imuno-Histoquímica , Hibridização In Situ , Proteínas de Membrana Transportadoras/genética , Mutação , Núcleosídeo-Fosfato Quinase/genética , Faloidina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Rodaminas/metabolismo , Transgenes
19.
Nat Rev Cancer ; 21(11): 687-700, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34389815

RESUMO

There is a large gap between the deep understanding of mechanisms driving tumour growth and the reasons why patients ultimately die of cancer. It is now appreciated that interactions between the tumour and surrounding non-tumour (sometimes referred to as host) cells play critical roles in mortality as well as tumour progression, but much remains unknown about the underlying molecular mechanisms, especially those that act beyond the tumour microenvironment. Drosophila has a track record of high-impact discoveries about cell-autonomous growth regulation, and is well suited to now probe mysteries of tumour - host interactions. Here, we review current knowledge about how fly tumours interact with microenvironmental stroma, circulating innate immune cells and distant organs to influence disease progression. We also discuss reciprocal regulation between tumours and host physiology, with a particular focus on paraneoplasias. The fly's simplicity along with the ability to study lethality directly provide an opportunity to shed new light on how cancer actually kills.


Assuntos
Modelos Animais de Doenças , Progressão da Doença , Drosophila melanogaster , Neoplasias/patologia , Microambiente Tumoral , Animais , Drosophila melanogaster/citologia , Drosophila melanogaster/imunologia , Humanos , Imunidade Inata , Neoplasias/imunologia , Neoplasias/mortalidade , Microambiente Tumoral/imunologia
20.
Mol Biol Cell ; 32(21): ar23, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34495684

RESUMO

Scribble (Scrib), Discs-large (Dlg), and Lethal giant larvae (Lgl) are basolateral regulators of epithelial polarity and tumor suppressors whose molecular mechanisms of action remain unclear. We used proximity biotinylation to identify proteins localized near Dlg in the Drosophila wing imaginal disc epithelium. In addition to expected membrane- and cytoskeleton-associated protein classes, nuclear proteins were prevalent in the resulting mass spectrometry dataset, including all four members of the nucleosome remodeling factor (NURF) chromatin remodeling complex. Subcellular fractionation demonstrated a nuclear pool of Dlg and proximity ligation confirmed its position near the NURF complex. Genetic analysis showed that NURF activity is also required for the overgrowth of dlg tumors, and this growth suppression correlated with a reduction in Hippo pathway gene expression. Together, these data suggest a nuclear role for Dlg in regulating chromatin and transcription through a more direct mechanism than previously thought.


Assuntos
Proteínas de Drosophila/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Polaridade Celular/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Células Epiteliais/metabolismo , Epitélio , Discos Imaginais/metabolismo , Proteínas de Membrana/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Proteínas Nucleares/metabolismo , Proteína Quinase C/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA