Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
PLoS Biol ; 21(5): e3001665, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37252939

RESUMEN

Epithelial repair relies on the activation of stress signaling pathways to coordinate tissue repair. Their deregulation is implicated in chronic wound and cancer pathologies. Using TNF-α/Eiger-mediated inflammatory damage to Drosophila imaginal discs, we investigate how spatial patterns of signaling pathways and repair behaviors arise. We find that Eiger expression, which drives JNK/AP-1 signaling, transiently arrests proliferation of cells in the wound center and is associated with activation of a senescence program. This includes production of the mitogenic ligands of the Upd family, which allows JNK/AP-1-signaling cells to act as paracrine organizers of regeneration. Surprisingly, JNK/AP-1 cell-autonomously suppress activation of Upd signaling via Ptp61F and Socs36E, both negative regulators of JAK/STAT signaling. As mitogenic JAK/STAT signaling is suppressed in JNK/AP-1-signaling cells at the center of tissue damage, compensatory proliferation occurs by paracrine activation of JAK/STAT in the wound periphery. Mathematical modelling suggests that cell-autonomous mutual repression between JNK/AP-1 and JAK/STAT is at the core of a regulatory network essential to spatially separate JNK/AP-1 and JAK/STAT signaling into bistable spatial domains associated with distinct cellular tasks. Such spatial stratification is essential for proper tissue repair, as coactivation of JNK/AP-1 and JAK/STAT in the same cells creates conflicting signals for cell cycle progression, leading to excess apoptosis of senescently stalled JNK/AP-1-signaling cells that organize the spatial field. Finally, we demonstrate that bistable separation of JNK/AP-1 and JAK/STAT drives bistable separation of senescent signaling and proliferative behaviors not only upon tissue damage, but also in RasV12, scrib tumors. Revealing this previously uncharacterized regulatory network between JNK/AP-1, JAK/STAT, and associated cell behaviors has important implications for our conceptual understanding of tissue repair, chronic wound pathologies, and tumor microenvironments.


Asunto(s)
Proteínas de Drosophila , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción STAT/metabolismo , Drosophila/metabolismo , Proliferación Celular , Quinasas Janus/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
2.
Nature ; 629(8012): 534-535, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38658714
3.
PLoS Genet ; 18(12): e1010516, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36520882

RESUMEN

Regeneration relies on cell proliferation to restore damaged tissues. Multiple signaling pathways activated by local or paracrine cues have been identified to promote regenerative proliferation. How different types of tissue damage may activate distinct signaling pathways and how these differences converge on regenerative proliferation is less well defined. To better understand how tissue damage and proliferative signals are integrated during regeneration, we investigate models of compensatory proliferation in Drosophila imaginal discs. We find that compensatory proliferation is associated with a unique cell cycle profile, which is characterized by short G1 and G2 phases and, surprisingly, by acceleration of the S-phase. S-phase acceleration can be induced by two distinct signaling signatures, aligning with inflammatory and non-inflammatory tissue damage. Specifically, non-autonomous activation of JAK/STAT and Myc in response to inflammatory damage, or local activation of Ras/ERK and Hippo/Yki in response to elevated cell death, promote accelerated nucleotide incorporation during S-phase. This previously unappreciated convergence of different damaging insults on the same regenerative cell cycle program reconciles previous conflicting observations on proliferative signaling in different tissue regeneration and tumor models.


Asunto(s)
Proteínas de Drosophila , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Transducción de Señal/fisiología , Drosophila/metabolismo , Proliferación Celular/genética , División Celular , Drosophila melanogaster/metabolismo
4.
Development ; 147(24)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33355242

RESUMEN

One of the central questions in developmental biology concerns how cells become organized into tissues of the correct size, shape and polarity. This organization depends on the implementation of a cell's genetic information to give rise to specific and coordinated cell behaviors, including cell division and cell shape change. The execution of these cell behaviors requires the active generation of mechanical forces. However, understanding how force generation is controlled and, importantly, coordinated among many cells in a tissue was little explored until the early 2000s. Suzanne Eaton was one of the pioneers in this emerging field of developmental tissue mechanics. As we briefly review here, she connected the quantitative analysis of cell behaviors with genetic assays, and integrated physical modeling with measurements of mechanical forces to reveal fundamental insights into epithelial morphogenesis at cell- and tissue-level scales.


Asunto(s)
Forma de la Célula/genética , Desarrollo Embrionario/genética , Mecanotransducción Celular/genética , Morfogénesis/genética , Animales , Fenómenos Biomecánicos , División Celular/genética , Drosophila/genética , Drosophila/crecimiento & desarrollo , Embrión no Mamífero
5.
Development ; 146(17)2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31399470

RESUMEN

How actomyosin generates forces at epithelial adherens junctions has been extensively studied. However, less is known about how a balance between internal and external forces establishes epithelial cell, tissue and organ shape. We used the Drosophila egg chamber to investigate how contractility at adherens junctions in the follicle epithelium is modulated to accommodate and resist forces arising from the growing germ line. We found that between stages 6 and 9, adherens junction tension in the post-mitotic epithelium decreases, suggesting that the junctional network relaxes to accommodate germline growth. At that time, a prominent medial Myosin II network coupled to corrugating adherens junctions develops. Local enrichment of medial Myosin II in main body follicle cells resists germline-derived forces, thus constraining apical areas and, consequently, cuboidal cell shapes at stage 9. At the tissue and organ level, local reinforcement of medial junction architecture ensures the timely contact of main body cells with the expanding oocyte and imposes circumferential constraints on the germ line guiding egg elongation. Our study provides insight into how adherens junction tension promotes cell and tissue shape transitions while integrating the growth and shape of an internally enclosed structure in vivo.


Asunto(s)
Forma de la Célula/fisiología , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Actomiosina/metabolismo , Uniones Adherentes/metabolismo , Animales , Proteínas del Dominio Armadillo/metabolismo , Cadherinas/metabolismo , Proteínas de Drosophila/metabolismo , Células Epiteliales/citología , Femenino , Miosina Tipo II/metabolismo , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Ovario/citología , Factores de Transcripción/metabolismo
6.
Development ; 143(16): 2907-19, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27385008

RESUMEN

Tissue homeostasis relies on the ability of tissues to respond to stress. Tissue regeneration and tumour models in Drosophila have shown that c-Jun amino-terminal kinase (JNK) acts as a prominent stress-response pathway promoting injury-induced apoptosis and compensatory proliferation. A central question remaining unanswered is how both responses are balanced by activation of a single pathway. Signalling through the Janus kinase/Signal transducers and activators of transcription (JAK/STAT) pathway, which is a potential JNK target, is implicated in promoting compensatory proliferation. While we observe JAK/STAT activation in imaginal discs upon damage, our data demonstrate that JAK/STAT and its downstream effector Zfh2 promote the survival of JNK signalling cells. The JNK component fos and the pro-apoptotic gene hid are regulated in a JAK/STAT-dependent manner. This molecular pathway restrains JNK-induced apoptosis and spatial propagation of JNK signalling, thereby limiting the extent of tissue damage, as well as facilitating systemic and proliferative responses to injury. We find that the pro-survival function of JAK/STAT also drives tumour growth under conditions of chronic stress. Our study defines the function of JAK/STAT in tissue stress and illustrates how crosstalk between conserved signalling pathways establishes an intricate equilibrium between proliferation, apoptosis and survival to restore tissue homeostasis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores de Transcripción STAT/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila/fisiología , Proteínas de Drosophila/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fosforilación/genética , Fosforilación/fisiología , Factores de Transcripción STAT/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
7.
Cell Tissue Res ; 356(3): 477-93, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24728925

RESUMEN

Cells respond to extra- and intra-cellular signals by dynamically changing their gene expression patterns. After termination of the original signal, new expression patterns are maintained by epigenetic DNA and histone modifications. This represents a powerful mechanism that enables long-term phenotypic adaptation to transient signals. Adaptation of epigenetic landscapes is important for mediating cellular differentiation during development and allows adjustment to altered environmental conditions throughout life. Work over the last decade has begun to elucidate the way that extra- and intra-cellular signals lead to changes in gene expression patterns by directly modulating the function of chromatin-associated proteins. Here, we review key signaling-to-chromatin pathways that are specifically thought to target Polycomb and Trithorax group complexes, a classic example of epigenetically acting gene silencers and activators important in development, stem cell differentiation and cancer. We discuss the influence that signals triggered by kinase cascades, metabolic fluctuations and cell-cycle dynamics have on the function of these protein complexes. Further investigation into these pathways will be important for understanding the mechanisms that maintain epigenetic stability and those that promote epigenetic plasticity.


Asunto(s)
Cromatina/metabolismo , Epigénesis Genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Transducción de Señal , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Cromatina/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Proteína de la Leucemia Mieloide-Linfoide/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas del Grupo Polycomb/genética , Células Madre/metabolismo , Células Madre/patología
8.
Curr Biol ; 34(5): 980-996.e6, 2024 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-38350446

RESUMEN

Tissue-intrinsic error correction enables epithelial cells to detect abnormal neighboring cells and facilitate their removal from the tissue. One of these pathways, "interface surveillance," is triggered by cells with aberrant developmental and cell-fate-patterning pathways. It remains unknown which molecular mechanisms provide cells with the ability to compare fate between neighboring cells. We demonstrate that Drosophila imaginal discs express an array of cell surface molecules previously implicated in neuronal axon guidance processes. They include members of the Robo, Teneurin, Ephrin, Toll-like, or atypical cadherin families. Importantly, a mismatch in expression levels of these cell surface molecules between adjacent cells is sufficient to induce interface surveillance, indicating that differences in expression levels between neighboring cells, rather than their absolute expression levels, are crucial. Specifically, a mismatch in Robo2 and Robo3, but not Robo1, induces enrichment of actin, myosin II, and Ena/Vasp, as well as activation of JNK and apoptosis at clonal interfaces. Moreover, Robo2 can induce interface surveillance independently of its cytosolic domain and without the need for the Robo-ligand Slit. The expression of Robo2 and other cell surface molecules, such as Teneurins or the Ephrin receptor is regulated by fate-patterning pathways intrinsic and extrinsic to the wing disc, as well as by expression of oncogenic RasV12. Combined, we demonstrate that neighboring cells respond to a mismatch in surface code patterns mediated by specific transmembrane proteins and reveal a novel function for these cell surface proteins in cell fate recognition and removal of aberrant cells during development and homeostasis of epithelial tissues.


Asunto(s)
Proteínas de Drosophila , Receptores Inmunológicos , Humanos , Animales , Receptores Inmunológicos/metabolismo , Proteínas Roundabout , Drosophila/fisiología , Axones/fisiología , Proteínas de Drosophila/metabolismo , Efrinas/metabolismo
9.
Elife ; 122024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38189792

RESUMEN

Environmental factors, infection, or injury can cause oxidative stress in diverse tissues and loss of tissue homeostasis. Effective stress response cascades, conserved from invertebrates to mammals, ensure reestablishment of homeostasis and tissue repair. Hemocytes, the Drosophila blood-like cells, rapidly respond to oxidative stress by immune activation. However, the precise signals how they sense oxidative stress and integrate these signals to modulate and balance the response to oxidative stress in the adult fly are ill-defined. Furthermore, hemocyte diversification was not explored yet on oxidative stress. Here, we employed high-throughput single nuclei RNA-sequencing to explore hemocytes and other cell types, such as fat body, during oxidative stress in the adult fly. We identified distinct cellular responder states in plasmatocytes, the Drosophila macrophages, associated with immune response and metabolic activation upon oxidative stress. We further define oxidative stress-induced DNA damage signaling as a key sensor and a rate-limiting step in immune-activated plasmatocytes controlling JNK-mediated release of the pro-inflammatory cytokine unpaired-3. We subsequently tested the role of this specific immune activated cell stage during oxidative stress and found that inhibition of DNA damage signaling in plasmatocytes, as well as JNK or upd3 overactivation, result in a higher susceptibility to oxidative stress. Our findings uncover that a balanced composition and response of hemocyte subclusters is essential for the survival of adult Drosophila on oxidative stress by regulating systemic cytokine levels and cross-talk to other organs, such as the fat body, to control energy mobilization.


Asunto(s)
Artrópodos , Drosophila , Animales , Estrés Oxidativo , Macrófagos , Citocinas , Daño del ADN , Mamíferos
10.
Development ; 137(14): 2353-64, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20534670

RESUMEN

In addition to apicobasal polarization, some epithelia also display polarity within the plane of the epithelium. To what extent polarized endocytosis plays a role in the establishment and maintenance of planar cell polarity (PCP) is at present unclear. Here, we investigated the role of Rabenosyn-5 (Rbsn-5), an evolutionarily conserved effector of the small GTPase Rab5, in the development of Drosophila wing epithelium. We found that Rbsn-5 regulates endocytosis at the apical side of the wing epithelium and, surprisingly, further uncovered a novel function of this protein in PCP. At early stages of pupal wing development, the PCP protein Fmi redistributes between the cortex and Rab5- and Rbsn-5-positive early endosomes. During planar polarization, Rbsn-5 is recruited at the apical cell boundaries and redistributes along the proximodistal axis in an Fmi-dependent manner. At pre-hair formation, Rbsn-5 accumulates at the bottom of emerging hairs. Loss of Rbsn-5 causes intracellular accumulation of Fmi and typical PCP alterations such as defects in cell packing, in the polarized distribution of PCP proteins, and in hair orientation and formation. Our results suggest that establishment of planar polarity requires the activity of Rbsn-5 in regulating both the endocytic trafficking of Fmi at the apical cell boundaries and hair morphology.


Asunto(s)
Polaridad Celular/fisiología , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo , Animales , Polaridad Celular/genética , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiología , Endocitosis/genética , Endocitosis/fisiología , Morfogénesis/genética , Morfogénesis/fisiología , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Alas de Animales/fisiología
11.
Elife ; 122023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36744859

RESUMEN

Tissue-intrinsic defense mechanisms eliminate aberrant cells from epithelia and thereby maintain the health of developing tissues or adult organisms. 'Interface surveillance' comprises one such distinct mechanism that specifically guards against aberrant cells which undergo inappropriate cell fate and differentiation programs. The cellular mechanisms which facilitate detection and elimination of these aberrant cells are currently unknown. We find that in Drosophila imaginal discs, clones of cells with inappropriate activation of cell fate programs induce bilateral JNK activation at clonal interfaces, where wild type and aberrant cells make contact. JNK activation is required to drive apoptotic elimination of interface cells. Importantly, JNK activity and apoptosis are highest in interface cells within small aberrant clones, which likely supports the successful elimination of aberrant cells when they arise. Our findings are consistent with a model where clone size affects the topology of interface contacts and thereby the strength of JNK activation in wild type and aberrant interface cells. Bilateral JNK activation is unique to 'interface surveillance' and is not observed in other tissue-intrinsic defense mechanisms, such as classical 'cell-cell competition'. Thus, bilateral JNK interface signaling provides an independent tissue-level mechanism to eliminate cells with inappropriate developmental fate but normal cellular fitness. Finally, oncogenic Ras-expressing clones activate 'interface surveillance' but evade elimination by bilateral JNK activation. Combined, our work establishes bilateral JNK interface signaling and interface apoptosis as a new hallmark of interface surveillance and highlights how oncogenic mutations evade tumor suppressor function encoded by this tissue-intrinsic surveillance system.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Células Epiteliales , Proteínas Quinasas JNK Activadas por Mitógenos , Animales , Apoptosis , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Epitelio/metabolismo , Genes Supresores de Tumor , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Células Epiteliales/citología , Células Epiteliales/metabolismo
12.
Elife ; 122023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37133250

RESUMEN

Wound response programs are often activated during neoplastic growth in tumors. In both wound repair and tumor growth, cells respond to acute stress and balance the activation of multiple programs, including apoptosis, proliferation, and cell migration. Central to those responses are the activation of the JNK/MAPK and JAK/STAT signaling pathways. Yet, to what extent these signaling cascades interact at the cis-regulatory level and how they orchestrate different regulatory and phenotypic responses is still unclear. Here, we aim to characterize the regulatory states that emerge and cooperate in the wound response, using the Drosophila melanogaster wing disc as a model system, and compare these with cancer cell states induced by rasV12scrib-/- in the eye disc. We used single-cell multiome profiling to derive enhancer gene regulatory networks (eGRNs) by integrating chromatin accessibility and gene expression signals. We identify a 'proliferative' eGRN, active in the majority of wounded cells and controlled by AP-1 and STAT. In a smaller, but distinct population of wound cells, a 'senescent' eGRN is activated and driven by C/EBP-like transcription factors (Irbp18, Xrp1, Slow border, and Vrille) and Scalloped. These two eGRN signatures are found to be active in tumor cells at both gene expression and chromatin accessibility levels. Our single-cell multiome and eGRNs resource offers an in-depth characterization of the senescence markers, together with a new perspective on the shared gene regulatory programs acting during wound response and oncogenesis.


Asunto(s)
Proteínas de Drosophila , Neoplasias , Animales , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Redes Reguladoras de Genes , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias/patología , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo
13.
Nat Commun ; 13(1): 6377, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36289235

RESUMEN

Cooperative morphogenesis of cell lineages underlies the development of functional units and organs. To study mechanisms driving the coordination of lineages, we investigated soma-germline interactions during oogenesis. From invertebrates to vertebrates, oocytes develop as part of a germline cyst that consists of the oocyte itself and so-called nurse cells, which feed the oocyte and are eventually removed. The enveloping somatic cells specialize to facilitate either oocyte maturation or nurse cell removal, which makes it essential to establish the right match between germline and somatic cells. We uncover that the transcriptional regulator Eya, expressed in the somatic lineage, controls bilateral cell-cell affinity between germline and somatic cells in Drosophila oogenesis. Employing functional studies and mathematical modelling, we show that differential affinity and the resulting forces drive somatic cell redistribution over the germline surface and control oocyte growth to match oocyte and nurse cells with their respective somatic cells. Thus, our data demonstrate that differential affinity between cell lineages is sufficient to drive the complex assembly of inter-lineage functional units and underlies tissue self-organization during Drosophila oogenesis.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Linaje de la Célula/genética , Oogénesis/genética , Oocitos/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
14.
Dev Cell ; 9(6): 805-17, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16326392

RESUMEN

The mechanisms that order cellular packing geometry are critical for the functioning of many tissues, but they are poorly understood. Here, we investigate this problem in the developing wing of Drosophila. The surface of the wing is decorated by hexagonally packed hairs that are uniformly oriented by the planar cell polarity pathway. They are constructed by a hexagonal array of wing epithelial cells. Wing epithelial cells are irregularly arranged throughout most of development, but they become hexagonally packed shortly before hair formation. During the process, individual cell boundaries grow and shrink, resulting in local neighbor exchanges, and Cadherin is actively endocytosed and recycled through Rab11 endosomes. Hexagonal packing depends on the activity of the planar cell polarity proteins. We propose that these proteins polarize trafficking of Cadherin-containing exocyst vesicles during junction remodeling. This may be a common mechanism for the action of planar cell polarity proteins in diverse systems.


Asunto(s)
Polaridad Celular , Drosophila melanogaster/crecimiento & desarrollo , Células Epiteliales/citología , Genes de Insecto , Transducción de Señal , Alas de Animales/citología , Animales , Cadherinas/genética , Cadherinas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Endocitosis , Endosomas , Células Epiteliales/metabolismo , Exocitosis , Receptores Frizzled , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Transporte de Proteínas , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Alas de Animales/metabolismo , Proteínas de Unión al GTP rab/metabolismo
15.
Elife ; 82019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30735120

RESUMEN

The restoration of homeostasis after tissue damage relies on proper spatial-temporal control of damage-induced apoptosis and compensatory proliferation. In Drosophila imaginal discs these processes are coordinated by the stress response pathway JNK. We demonstrate that JNK signaling induces a dose-dependent extension of G2 in tissue damage and tumors, resulting in either transient stalling or a prolonged but reversible cell cycle arrest. G2-stalling is mediated by downregulation of the G2/M-specific phosphatase String(Stg)/Cdc25. Ectopic expression of stg is sufficient to suppress G2-stalling and reveals roles for stalling in survival, proliferation and paracrine signaling. G2-stalling protects cells from JNK-induced apoptosis, but under chronic conditions, reduces proliferative potential of JNK-signaling cells while promoting non-autonomous proliferation. Thus, transient cell cycle stalling in G2 has key roles in wound healing but becomes detrimental upon chronic JNK overstimulation, with important implications for chronic wound healing pathologies or tumorigenic transformation.


Asunto(s)
Senescencia Celular/genética , Discos Imaginales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Estrés Fisiológico/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Puntos de Control del Ciclo Celular/genética , División Celular/genética , Proliferación Celular/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Humanos , Discos Imaginales/crecimiento & desarrollo , Discos Imaginales/lesiones , Sistema de Señalización de MAP Quinasas/genética , Cicatrización de Heridas/genética
16.
Methods Mol Biol ; 420: 265-75, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18641953

RESUMEN

Drosophila pupal (P) wing development entails a series of dynamic developmental events, such as epithelial and glial morphogenesis, that are of outstanding interest to cell biologists. Here, we first describe how to prepare P and prepupal (PP) wings for immunofluorescence microscopy. This protocol has been optimized to visualize wing epithelial architecture, such as polarized cortical domains of planar cell polarity proteins. We then provide a protocol to prepare pupae for whole mount live imaging of P wings. This procedure has allowed us to live-image glial cell migration and proliferation along wing sensory nerves.


Asunto(s)
Biología Evolutiva/métodos , Drosophila melanogaster/fisiología , Microscopía Fluorescente/métodos , Animales , Tipificación del Cuerpo , Movimiento Celular , Polaridad Celular , Células Epiteliales/citología , Genes de Insecto , Metamorfosis Biológica , Microscopía Fluorescente/instrumentación , Morfogénesis , Neuroglía/citología , Neuronas Aferentes/metabolismo , Pupa/metabolismo , Alas de Animales/embriología
17.
Epigenetics Chromatin ; 11(1): 38, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29970137

RESUMEN

Unfortunately, the original version of this article contained a typographical error in one of the author names. The name of the author Alexey Pindyurin was incorrectly spelt as Alexey Pinduyrin. The correct spelling is included here and has been updated in the original article.

18.
Epigenetics Chromatin ; 11(1): 27, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29871666

RESUMEN

BACKGROUND: Tracking dynamic protein-chromatin interactions in vivo is key to unravel transcriptional and epigenetic transitions in development and disease. However, limited availability and heterogeneous tissue composition of in vivo source material impose challenges on many experimental approaches. RESULTS: Here we adapt cell-type-specific DamID-seq profiling for use in Drosophila imaginal discs and make FLP/FRT-based induction accessible to GAL driver-mediated targeting of specific cell lineages. In a proof-of-principle approach, we utilize ubiquitous DamID expression to describe dynamic transitions of Polycomb-binding sites during wing imaginal disc development and in a scrib tumorigenesis model. We identify Atf3 and Ets21C as novel Polycomb target genes involved in scrib tumorigenesis and suggest that target gene regulation by Atf3 and AP-1 transcription factors, as well as modulation of insulator function, plays crucial roles in dynamic Polycomb-binding at target sites. We establish these findings by DamID-seq analysis of wing imaginal disc samples derived from 10 larvae. CONCLUSIONS: Our study opens avenues for robust profiling of small cell population in imaginal discs in vivo and provides insights into epigenetic changes underlying transcriptional responses to tumorigenic transformation.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Drosophila/genética , Drosophila/genética , Discos Imaginales/crecimiento & desarrollo , Animales , Sitios de Unión , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Drosophila/embriología , Drosophila/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Discos Imaginales/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Unión Proteica , Análisis de Secuencia de ADN/métodos
19.
Sci Rep ; 7: 42786, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28218282

RESUMEN

While calcium signaling in excitable cells, such as muscle or neurons, is extensively characterized, calcium signaling in epithelial tissues is little understood. Specifically, the range of intercellular calcium signaling patterns elicited by tightly coupled epithelial cells and their function in the regulation of epithelial characteristics are little explored. We found that in Drosophila imaginal discs, a widely studied epithelial model organ, complex spatiotemporal calcium dynamics occur. We describe patterns that include intercellular waves traversing large tissue domains in striking oscillatory patterns as well as spikes confined to local domains of neighboring cells. The spatiotemporal characteristics of intercellular waves and oscillations arise as emergent properties of calcium mobilization within a sheet of gap-junction coupled cells and are influenced by cell size and environmental history. While the in vivo function of spikes, waves and oscillations requires further characterization, our genetic experiments suggest that core calcium signaling components guide actomyosin organization. Our study thus suggests a possible role for calcium signaling in epithelia but importantly, introduces a model epithelium enabling the dissection of cellular mechanisms supporting the initiation, transmission and regeneration of long-range intercellular calcium waves and the emergence of oscillations in a highly coupled multicellular sheet.


Asunto(s)
Calcio/metabolismo , Drosophila melanogaster/metabolismo , Epitelio/fisiología , Discos Imaginales/citología , Animales , Señalización del Calcio , Tamaño de la Célula , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Discos Imaginales/metabolismo , Modelos Biológicos
20.
PLoS One ; 12(5): e0177408, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28510597

RESUMEN

The nuclear acetyltransferase MOF (KAT8 in mammals) is a subunit of at least two multi-component complexes involved in transcription regulation. In the context of complexes of the 'Non-Specific-Lethal' (NSL) type it controls transcription initiation of many nuclear housekeeping genes and of mitochondrial genes. While this function is conserved in metazoans, MOF has an additional, specific function in Drosophila in the context of dosage compensation. As a subunit of the male-specific-lethal dosage compensation complex (MSL-DCC) it contributes to the doubling of transcription output from the single male X chromosome by acetylating histone H4. Proper dosage compensation requires finely tuned levels of MSL-DCC and an appropriate distribution of MOF between the regulatory complexes. The amounts of DCC formed depends directly on the levels of the male-specific MSL2, which orchestrates the assembly of the DCC, including MOF recruitment. We found earlier that MSL2 is an E3 ligase that ubiquitylates most MSL proteins, including MOF, suggesting that ubiquitylation may contribute to a quality control of MOF's overall levels and folding state as well as its partitioning between the complex entities. We now used mass spectrometry to map the lysines in MOF that are ubiquitylated by MSL2 in vitro and identified in vivo ubiquitylation sites of MOF in male and female cells. MSL2-specific ubiquitylation in vivo could not be traced due to the dominance of other, sex-independent ubiquitylation events and conceivably may be rare or transient. Expressing appropriately mutated MOF derivatives we assessed the importance of the ubiquitylated lysines for dosage compensation by monitoring DCC formation and X chromosome targeting in cultured cells, and by genetic complementation of the male-specific-lethal mof2 allele in flies. Our study provides a comprehensive analysis of MOF ubiquitylation as a reference for future studies.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/metabolismo , Regulación Alostérica , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Activación Enzimática , Histona Acetiltransferasas/genética , Mutación , Proteínas Nucleares/genética , Unión Proteica , Factores de Transcripción/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA