Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2626: 69-87, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36715900

RESUMO

The ability to conduct spatially controlled RNA interference (RNAi) for gene knockdown using the UAS/Gal4 system is among the most appealing techniques available for analysis of gene function in the Drosophila ovary. While gene knockdown experiments in somatic cells in the developing organism (i.e., embryos and larvae) are effectively and commonly performed, the use of RNAi in adult ovarian cells can be hampered by the unintended deleterious effects of Gal4 expression in "off-target" developing tissues. Mosaic analysis overcomes these problems by imparting temporal and spatial control over gene manipulation, providing a useful tool to compare manipulated cells with wild-type cells in the same tissue. Here, we provide a method to utilize the UAS/Gal4 system in combination with the Flippase (FLP)-Flippase Recognition Target (FRT) system to generate positively labeled "FLP-Out" clones expressing a chosen RNAi in both the germline and the soma in the Drosophila ovary. This protocol outlines each step of the generation of clones and the selection of appropriate fly stocks and reagents, providing a guide to this powerful tool in the Drosophila genetic toolbox. These techniques allow for RNAi analysis within a specific cell type, providing an opportunity to study a variety of unique aspects of cell function that would not be possible in more traditional RNAi-based experiments.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Feminino , Drosophila/genética , Drosophila/metabolismo , Interferência de RNA , Ovário/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Técnicas Genéticas , Drosophila melanogaster/genética
2.
Methods Mol Biol ; 2626: 135-150, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36715903

RESUMO

In many species, oocytes are initially formed by the mitotic divisions of germline stem cells and their differentiating daughters. These progenitor cells are frequently interconnected in structures called cysts, which may function to safeguard oocyte quality. In Drosophila, an essential germline-specific organelle called the fusome helps maintain and coordinate the mitotic divisions of both germline stem cells and cyst cells. The fusome also serves as a useful experimental marker to identify germ cells during their mitotic divisions. Fusomes are cytoplasmic organelles composed of microtubules, endoplasmic reticulum-derived vesicles, and a meshwork of membrane skeleton proteins. The fusome branches as mitotic divisions progress, traversing the intercellular bridges of germline stem cell/cystoblast pairs and cysts. Here, we provide a protocol to visualize fusome morphology in fixed tissue by stabilizing microtubules and immunostaining for α-Tubulin and other protein constituents of the fusome. We identify a variety of fluorophore-tagged proteins that are useful for visualizing the fusome and describe how these might be combined experimentally. Taken together, these tools provide a valuable resource to interrogate the genetic control of germline stem cell function, oocyte selection, and asymmetric division.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Tubulina (Proteína) , Oócitos/metabolismo , Células Germinativas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Membrana , Imunofluorescência , Oogênese
3.
Dev Dyn ; 252(3): 415-428, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36308715

RESUMO

BACKGROUND: Reproductive capacity in many organisms is maintained by germline stem cells (GSCs). A complex regulatory network influences stem cell fate, including intrinsic factors, local signals, and hormonal and nutritional cues. Posttranscriptional regulatory mechanisms ensure proper cell fate transitions, promoting germ cell differentiation to oocytes. As essential RNA binding proteins with constitutive functions in RNA metabolism, heterogeneous nuclear ribonucleoproteins (hnRNPs) have been implicated in GSC function and axis specification during oocyte development. HnRNPs support biogenesis, localization, maturation, and translation of nascent transcripts. Whether and individual hnRNPs specifically regulate GSC function has yet to be explored. RESULTS: We demonstrate that hnRNPs are expressed in distinct patterns in the Drosophila germarium. We show that three hnRNPs, squid, hephaestus, and Hrb27C are cell-autonomously required in GSCs for their maintenance. Although these hnRNPs do not impact adhesion of GSCs to adjacent cap cells, squid and hephaestus (but not Hrb27C) are necessary for proper bone morphogenetic protein signaling in GSCs. Moreover, Hrb27C promotes proper GSC proliferation, whereas hephaestus promotes cyst division. CONCLUSIONS: We find that hnRNPs are independently and intrinsically required in GSCs for their maintenance in adults. Our results support the model that hnRNPs play unique roles in stem cells essential for their self-renewal and proliferation.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Diferenciação Celular , Drosophila/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Células Germinativas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Oócitos , Oogênese/fisiologia , Proteínas de Ligação a RNA/metabolismo
4.
Dev Biol ; 494: 1-12, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36450333

RESUMO

Germ cell development requires interplay between factors that balance cell fate and division. Early in their development, germ cells in many organisms divide mitotically with incomplete cytokinesis. Key regulatory events then lead to the specification of mature gametes, marked by the switch to a meiotic cell cycle program. Though the regulation of germ cell proliferation and meiosis are well understood, how these events are coordinated during development remains incompletely described. Originally characterized in their role as nucleo-cytoplasmic shuttling proteins, ß-importins exhibit diverse functions during male and female gametogenesis. Here, we describe novel, distinct roles for the ß-importin, Transportin-Serine/Arginine rich (Tnpo-SR), as a regulator of the mitosis to meiosis transition in the Drosophila ovary. We find that Tnpo-SR is necessary for germline stem cell (GSC) establishment and self-renewal, likely by controlling the response of GSCs to bone morphogenetic proteins. Depletion of Tnpo-SR results in germ cell counting defects and loss of oocyte identity. We show that in the absence of Tnpo-SR, proteins typically suppressed in germ cells when they exit mitosis fail to be down-regulated, and oocyte-specific factors fail to accumulate. Together, these findings provide new insight into the balance between germ cell division and differentiation and identify novel roles for ß-importins in germ cell development.


Assuntos
Drosophila , Carioferinas , Animais , Feminino , Masculino , Arginina , beta Carioferinas , Diferenciação Celular , Células Germinativas , Meiose , Mitose , Oócitos , Células-Tronco
6.
Vitam Horm ; 116: 327-362, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33752824

RESUMO

Maternal nutrition and physiology are intimately associated with reproductive success in diverse organisms. Despite decades of study, the molecular mechanisms linking maternal diet to the production and quality of oocytes remain poorly defined. Nuclear receptors (NRs) link nutritional signals to cellular responses and are essential for oocyte development. The fruit fly, Drosophila melanogaster, is an excellent genetically tractable model to study the relationship between NR signaling and oocyte production. In this review, we explore how NRs in Drosophila regulate the earliest stages of oocyte development. Long-recognized as an essential mediator of developmental transitions, we focus on the intrinsic roles of the Ecdysone Receptor and its ligand, ecdysone, in oogenesis. We also review recent studies suggesting broader roles for NRs as regulators of maternal physiology and their impact specifically on oocyte production. We propose that NRs form the molecular basis of a broad physiological surveillance network linking maternal diet with oocyte production. Given the functional conservation between Drosophila and humans, continued experimental investigation into the molecular mechanisms by which NRs promote oogenesis will likely aid our understanding of human fertility.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Oogênese , Receptores de Esteroides , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Células Germinativas/fisiologia , Oogênese/genética , Oogênese/fisiologia , Receptores de Esteroides/genética , Receptores de Esteroides/fisiologia , Células-Tronco/fisiologia
7.
G3 (Bethesda) ; 11(2)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33693603

RESUMO

Gamete production in mammals and insects is controlled by cell signaling pathways that facilitate communication between germ cells and somatic cells. Nuclear receptor signaling is a key mediator of many aspects of reproduction, including gametogenesis. For example, the NR5A subfamily of nuclear receptors is essential for gonad development and sex steroid production in mammals. Despite the original identification of the NR5A subfamily in the model insect Drosophila melanogaster, it has been unclear whether Drosophila NR5A receptors directly control oocyte production. Ftz-f1 is expressed throughout the ovary, including in germline stem cells, germline cysts, and several populations of somatic cells. We show that ftz-f1 is required in follicle cells prior to stage 10 to promote egg chamber survival at the mid-oogenesis checkpoint. Our data suggest that egg chamber death in the absence of ftz-f1 is due, at least in part, to failure of follicle cells to exit the mitotic cell cycle or failure to accumulate oocyte-specific factors in the germline. Taken together, these results show that, as in mammals, the NR5A subfamily promotes maximal reproductive output in Drosophila. Our data underscore the importance of nuclear receptors in the control of reproduction and highlight the utility of Drosophila oogenesis as a key model for unraveling the complexity of nuclear receptor signaling in gametogenesis.


Assuntos
Proteínas de Drosophila , Drosophila , Receptores Nucleares Órfãos , Animais , Drosophila melanogaster , Feminino , Oogênese , Ovário
8.
Front Cell Dev Biol ; 8: 19, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117961

RESUMO

Gametes are highly specialized cell types produced by a complex differentiation process. Production of viable oocytes requires a series of precise and coordinated molecular events. Early in their development, germ cells are an interconnected group of mitotically dividing cells. Key regulatory events lead to the specification of mature oocytes and initiate a switch to the meiotic cell cycle program. Though the chromosomal events of meiosis have been extensively studied, it is unclear how other aspects of oocyte specification are temporally coordinated. The fruit fly, Drosophila melanogaster, has long been at the forefront as a model system for genetics and cell biology research. The adult Drosophila ovary continuously produces germ cells throughout the organism's lifetime, and many of the cellular processes that occur to establish oocyte fate are conserved with mammalian gamete development. Here, we review recent discoveries from Drosophila that advance our understanding of how early germ cells balance mitotic exit with meiotic initiation. We discuss cell cycle control and establishment of cell polarity as major themes in oocyte specification. We also highlight a germline-specific organelle, the fusome, as integral to the coordination of cell division, cell polarity, and cell fate in ovarian germ cells. Finally, we discuss how the molecular controls of the cell cycle might be integrated with cell polarity and cell fate to maintain oocyte production.

9.
Gene Expr Patterns ; 34: 119074, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31563631

RESUMO

The insect steroid hormone ecdysone is a key regulator of oogenesis in Drosophila melanogaster and many other species. Despite the diversity of cellular functions of ecdysone in oogenesis, the molecular regulation of most ecdysone-responsive genes in ovarian cells remains largely unexplored. We performed a functional screen using the UAS/Gal4 system to identify non-coding cis-regulatory elements within well-characterized ecdysone-response genes capable of driving transcription of an indelible reporter in ovarian cells. Using two publicly available transgenic collections (the FlyLight and Vienna Tiles resources), we tested 62 Gal4 drivers corresponding to ecdysone-response genes EcR, usp, E75, br, ftz-f1 and Hr3. We observed 31 lines that were sufficient to drive a UAS-lacZ reporter in discrete cell populations in the ovary. Reporter expression was reproducibly observed in both somatic and germ cells at distinct stages of oogenesis, including those previously characterized as critical points of ecdysone regulation. Our studies identified several useful new reagents, adding to the UAS/Gal4 toolkit available for genetic analysis of oogenesis in Drosophila. Further, our study provides novel insight into the molecular regulation of ecdysone signaling in oogenesis.


Assuntos
Ecdisona/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Oogênese/genética , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Expressão Gênica/genética , Células Germinativas/metabolismo , Ovário/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Transdução de Sinais , Fatores de Transcrição/genética
10.
Dev Biol ; 454(2): 97-107, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31251895

RESUMO

Dietary proteins are crucial for oogenesis. The Target of Rapamycin (TOR) is a major nutrient sensor controlling organismal growth and fertility, but the downstream effectors of TOR signaling remain largely uncharacterized. We previously identified Drosophila Spargel/dPGC-1 as a terminal effector of the TOR-TSC pathway, and now report that Spargel connects nutrition to oogenesis. We found that Spargel is expressed predominantly in the ovaries of adult flies, and germline spargel knockdown inhibits cyst growth, ultimately leading to egg chamber degeneration and female sterility. In situ staining demonstrated nuclear localization of Spargel in the nurse cells and follicle cells of the ovariole. Furthermore, Spargel/dPGC-1 expression is influenced by dietary yeast concentration and TOR signaling, suggesting Spargel/dPGC-1 might transmit nutrient-mediated signals into ovarian growth. We propose that potentiating Spargel/dPGC-1 expression in the ovary is instrumental in nutrient-mediated regulation of oogenesis.


Assuntos
Proteínas de Drosophila/metabolismo , Oogênese/fisiologia , Ovário/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Animais , Proteínas Alimentares/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/metabolismo , Feminino , Células Germinativas/metabolismo , Nutrientes , Folículo Ovariano/metabolismo , Ovário/crescimento & desenvolvimento , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fator B de Elongação Transcricional Positiva/fisiologia , Transdução de Sinais , Sirolimo/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/fisiologia
11.
Dev Biol ; 429(1): 118-131, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28711427

RESUMO

Development of multicellular organisms relies upon the coordinated regulation of cellular differentiation and proliferation. Growing evidence suggests that some molecular regulatory pathways associated with the cell cycle machinery also dictate cell fate; however, it remains largely unclear how the cell cycle is remodeled in concert with cell differentiation. During Drosophila oogenesis, mature oocytes are created through a series of precisely controlled division and differentiation steps, originating from a single tissue-specific stem cell. Further, germline stem cells (GSCs) and their differentiating progeny remain in a predominantly linear arrangement as oogenesis proceeds. The ability to visualize the stepwise events of differentiation within the context of a single tissue make the Drosophila ovary an exceptional model for study of cell cycle remodeling. To describe how the cell cycle is remodeled in germ cells as they differentiate in situ, we used the Drosophila Fluorescence Ubiquitin-based Cell Cycle Indicator (Fly-FUCCI) system, in which degradable versions of GFP::E2f1 and RFP::CycB fluorescently label cells in each phase of the cell cycle. We found that the lengths of the G1, S, and G2 phases of the cell cycle change dramatically over the course of differentiation, and identified the 4/8-cell cyst as a key developmental transition state in which cells prepare for specialized cell cycles. Our data suggest that the transcriptional activator E2f1, which controls the transition from G1 to S phase, is a key regulator of mitotic divisions in the early germline. Our data support the model that E2f1 is necessary for proper GSC proliferation, self-renewal, and daughter cell development. In contrast, while E2f1 degradation by the Cullin 4 (Cul4)-containing ubiquitin E3 ligase (CRL4) is essential for developmental transitions in the early germline, our data do not support a role for E2f1 degradation as a mechanism to limit GSC proliferation or self-renewal. Taken together, these findings provide further insight into the regulation of cell proliferation and the acquisition of differentiated cell fate, with broad implications across developing tissues.


Assuntos
Ciclo Celular , Diferenciação Celular , Drosophila melanogaster/citologia , Células Germinativas/citologia , Animais , Biomarcadores/metabolismo , Ciclo Celular/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Feminino , Fluorescência , Genes Reporter , Mitose/genética , Morfogênese/genética , Ovário/citologia , Proteólise , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo , Ubiquitina/metabolismo , Regulação para Cima/genética
12.
Methods Mol Biol ; 1622: 161-184, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28674809

RESUMO

Elucidating the full repertoire of molecular mechanisms that promote stem cell maintenance requires sophisticated techniques for identifying and characterizing gene function in stem cells in their native environment. Ovarian germline stem cells in the fruit fly, Drosophila melanogaster, are an ideal model to study the complex molecular mechanisms driving stem cell function in vivo. A variety of new genetic tools make RNAi a useful complement to traditional genetic mutants for the investigation of the molecular mechanisms guiding ovarian germline stem cell function. Here, we provide a detailed guide for using targeted RNAi knockdown for the discovery of gene function in ovarian germline stem cells and their progeny.


Assuntos
Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Interferência de RNA , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Células Germinativas/citologia , Microscopia Confocal , Oogênese/genética , Ovário/metabolismo , Fenótipo
13.
Curr Stem Cell Rep ; 3(1): 9-18, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28458991

RESUMO

PURPOSE OF REVIEW: Stem cells respond to local paracrine signals; more recently, however, systemic hormones have also emerged as key regulators of stem cells. This review explores the role of steroid hormones in stem cells, using the Drosophila germline stem cell as a centerpiece for discussion. RECENT FINDINGS: Stem cells sense and respond directly and indirectly to steroid hormones, which regulate diverse sets of target genes via interactions with nuclear hormone receptors. Hormone-regulated networks likely integrate the actions of multiple systemic signals to adjust the activity of stem cell lineages in response to changes in physiological status. SUMMARY: Hormones are inextricably linked to animal physiology, and can control stem cells and their local niches. Elucidating the molecular mechanisms of hormone signaling in stem cells is essential for our understanding of the fundamental underpinnings of stem cell biology, and for informing new therapeutic interventions against cancers or for regenerative medicine.

14.
Cytometry A ; 91(3): 270-280, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27984679

RESUMO

Application of fluid shear stress to adherent cells dramatically influences their cytoskeletal makeup and differentially regulates their migratory phenotype. Because cytoskeletal rearrangements are necessary for cell motility and migration, preserving these adaptations under in vitro conditions and in the presence of fluid flow are physiologically essential. With this in mind, parallel plate flow chambers and microchannels are often used to conduct in vitro perfusion experiments. However, both of these systems currently lack capacity to accurately study cell migration in the same location where cells were perfused. The most common perfusion/migration assays involve cell perfusion followed by trypsinization which can compromise adaptive cytoskeletal geometry and lead to misleading phenotypic conclusions. The purpose of this study was to quantitatively highlight some limitations commonly found with currently used cell migration approaches and to introduce two new advances which use additive manufacturing (3D printing) or laser capture microdissection (LCM) technology. The residue-free 3D printed insert allows accurate cell seeding within defined areas, increases cell yield for downstream analyses, and more closely resembles the reported levels of fluid shear stress calculated with computational fluid dynamics as compared to other residue-free cell seeding techniques. The LCM approach uses an ultraviolet laser for "touchless technology" to rapidly and accurately introduce a custom-sized wound area in otherwise inaccessible perfusion microchannels. The wound area introduced by LCM elicits comparable migration characteristics compared to traditional pipette tip-induced injuries. When used in perfusion experiments, both of these newly characterized tools were effective in yielding similar results yet without the limitations of the traditional modalities. These innovative methods provide valuable tools for exploring mechanisms of clinically important aspects of cell migration fundamental to the pathogenesis of many flow-mediated disorders and are applicable to other perfusion-based models where migration is of central importance. © 2016 International Society for Advancement of Cytometry.


Assuntos
Técnicas de Cultura de Células/métodos , Movimento Celular/efeitos dos fármacos , Estresse Mecânico , Citoesqueleto/ultraestrutura , Humanos , Perfusão , Tripsina/farmacologia
15.
G3 (Bethesda) ; 6(8): 2629-42, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27226164

RESUMO

Multiple aspects of Drosophila oogenesis, including germline stem cell activity, germ cell differentiation, and follicle survival, are regulated by the steroid hormone ecdysone. While the transcriptional targets of ecdysone signaling during development have been studied extensively, targets in the ovary remain largely unknown. Early studies of salivary gland polytene chromosomes led to a model in which ecdysone stimulates a hierarchical transcriptional cascade, wherein a core group of ecdysone-sensitive transcription factors induce tissue-specific responses by activating secondary branches of transcriptional targets. More recently, genome-wide approaches have identified hundreds of putative ecdysone-responsive targets. Determining whether these putative targets represent bona fide targets in vivo, however, requires that they be tested via traditional mutant analysis in a cell-type specific fashion. To investigate the molecular mechanisms whereby ecdysone signaling regulates oogenesis, we used genetic mosaic analysis to screen putative ecdysone-responsive genes for novel roles in the control of the earliest steps of oogenesis. We identified a cohort of genes required for stem cell maintenance, stem and progenitor cell proliferation, and follicle encapsulation, growth, and survival. These genes encode transcription factors, chromatin modulators, and factors required for RNA transport, stability, and ribosome biogenesis, suggesting that ecdysone might control a wide range of molecular processes during oogenesis. Our results suggest that, although ecdysone target genes are known to have cell type-specific roles, many ecdysone response genes that control larval or pupal cell types at developmental transitions are used reiteratively in the adult ovary. These results provide novel insights into the molecular mechanisms by which ecdysone signaling controls oogenesis, laying new ground for future studies.


Assuntos
Drosophila melanogaster/genética , Ecdisona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Oogênese/genética , Ovário/fisiologia , Animais , Animais Geneticamente Modificados , Linhagem da Célula , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Ecdisona/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas Genéticas , Ribonucleoproteínas Nucleares Heterogêneas/genética , Larva/efeitos dos fármacos , Larva/genética , Mosaicismo , Mutação , Oogênese/efeitos dos fármacos , Ovário/citologia , Pupa/genética , Receptores de Esteroides/genética , Células-Tronco/fisiologia
16.
Mech Dev ; 138 Pt 3: 291-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26369283

RESUMO

Efficient replication of the genome and the establishment of endogenous chromatin states are processes that are essential to eukaryotic life. It is well documented that Mcm10 is intimately linked to both of these important biological processes; therefore, it is not surprising that Mcm10 is commonly misregulated in many human cancers. Most of the research regarding the biological roles of Mcm10 has been performed in single-cell or cell-free in-vitro systems. Though these systems are informative, they are unable to provide information on the cell-specific function of Mcm10 in the context of the tissue and organ systems that comprise multicellular eukaryotes. We therefore sought to identify the potential biological functions of Mcm10 in the context of a complex multicellular organism by continuing our analysis in Drosophila using three novel hypomorphic alleles. Observation of embryonic nuclear morphology and quantification of embryo hatch rates reveal that maternal loading of Mcm10 is required for embryonic nuclear stability, and suggest a role for Mcm10 post zygotic transition. Contrary to the essential nature of Mcm10 depicted in the literature, it does not appear to be required for adult viability in Drosophila if embryonic requirements are met. Although not required for adult somatic viability, analysis of fecundity and ovarian morphology in mutant females suggest that Mcm10 plays a role in maintenance of the female germline. Taken together, our results demonstrate critical roles for Mcm10 during early embryogenesis, and mark the first data linking Mcm10 to female specific reproduction in multicellular eukaryotes.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/fisiologia , Oogênese/genética , Oogênese/fisiologia , Animais , Animais Geneticamente Modificados , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Fertilidade/genética , Fertilidade/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Instabilidade Genômica , Humanos , Masculino , Mutação
17.
Genetics ; 199(1): 17-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25573011

RESUMO

Achieving a thorough understanding of the events and ramifications of meiosis is a common learning objective for undergraduate introductory biology, genetics, and cell biology courses. Meiosis is also one of the most challenging cellular processes for students to conceptualize. Connecting textbook descriptions of meiosis to current research in the field of genetics in a problem-based learning format may aid students' understanding of this important biological concept. This primer seeks to assist students and instructors by providing an introductory framework upon which to integrate discussions of current meiosis research into traditional genetics or cell biology curriculum.


Assuntos
Drosophila/genética , Genética/educação , Meiose , Complexo Sinaptonêmico/metabolismo , Animais , Drosophila/metabolismo , Drosophila/fisiologia , Oogênese , Complexo Sinaptonêmico/genética
18.
Dev Biol ; 400(1): 33-42, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25624267

RESUMO

Nuclear hormone receptors have emerged as important regulators of mammalian and Drosophila adult physiology, affecting such seemingly diverse processes as adipogenesis, carbohydrate metabolism, circadian rhythm, stem cell function, and gamete production. Although nuclear hormone receptors Ecdysone Receptor (EcR) and Ultraspiracle (Usp) have multiple known roles in Drosophila development and regulate key processes during oogenesis, the adult function of the majority of nuclear hormone receptors remains largely undescribed. Ecdysone-induced protein 78C (E78), a nuclear hormone receptor closely related to Drosophila E75 and to mammalian Rev-Erb and Peroxisome Proliferator Activated Receptors, was originally identified as an early ecdysone target; however, it has remained unclear whether E78 significantly contributes to adult physiology or reproductive function. To further explore the biological function of E78 in oogenesis, we used available E78 reporters and created a new E78 loss-of-function allele. We found that E78 is expressed throughout the germline during oogenesis, and is important for proper egg production and for the maternal control of early embryogenesis. We showed that E78 is required during development to establish the somatic germline stem cell (GSC) niche, and that E78 function in the germline promotes the survival of developing follicles. Consistent with its initial discovery as an ecdysone-induced target, we also found significant genetic interactions between E78 and components of the ecdysone-signaling pathway. Taken together with the previously described roles of EcR, Usp, and E75, our results suggest that nuclear hormone receptors are critical for the broad transcriptional control of a wide variety of cellular processes during oogenesis.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Germinativas/fisiologia , Oogênese/fisiologia , Folículo Ovariano/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Primers do DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Galactosídeos , Indóis , Microscopia de Fluorescência , Interferência de RNA , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
19.
Development ; 140(3): 530-40, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23293285

RESUMO

Stem cells must proliferate while maintaining 'stemness'; however, much remains to be learned about how factors that control the division of stem cells influence their identity. Multiple stem cell types display cell cycles with short G1 phases, thought to minimize susceptibility to differentiation factors. Drosophila female germline stem cells (GSCs) have short G1 and long G2 phases, and diet-dependent systemic factors often modulate G2. We previously observed that Cyclin E (CycE), a known G1/S regulator, is atypically expressed in GSCs during G2/M; however, it remained unclear whether CycE has cell cycle-independent roles in GSCs or whether it acts exclusively by modulating the cell cycle. In this study, we detected CycE activity during G2/M, reflecting its altered expression pattern, and showed that CycE and its canonical partner, Cyclin-dependent kinase 2 (Cdk2), are required not only for GSC proliferation, but also for GSC maintenance. In genetic mosaics, CycE- and Cdk2-deficient GSCs are rapidly lost from the niche, remain arrested in a G1-like state, and undergo excessive growth and incomplete differentiation. However, we found that CycE controls GSC maintenance independently of its role in the cell cycle; GSCs harboring specific hypomorphic CycE mutations are not efficiently maintained despite normal proliferation rates. Finally, CycE-deficient GSCs have an impaired response to niche bone morphogenetic protein signals that are required for GSC self-renewal, suggesting that CycE modulates niche-GSC communication. Taken together, these results show unequivocally that the roles of CycE/Cdk2 in GSC division cycle regulation and GSC maintenance are separable, and thus potentially involve distinct sets of phosphorylation targets.


Assuntos
Proliferação de Células , Ciclina E/metabolismo , Drosophila/citologia , Nicho de Células-Tronco , Células-Tronco/citologia , Alelos , Animais , Apoptose , Divisão Celular , Ciclina B/genética , Ciclina B/metabolismo , Ciclina E/genética , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Fase G2 , Células Germinativas/citologia , Células Germinativas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Junções Intercelulares/genética , Junções Intercelulares/metabolismo , Mutação , Fenótipo , Transdução de Sinais , Células-Tronco/metabolismo
20.
Wiley Interdiscip Rev Dev Biol ; 1(5): 657-74, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23799567

RESUMO

Adult stem cells are inextricably linked to whole-body physiology and nutrient availability through complex systemic signaling networks. A full understanding of how stem cells sense and respond to dietary fluctuations will require identifying key systemic mediators, as well as elucidating how they are regulated and integrated with local and intrinsic factors across multiple tissues. Studies focused on the Drosophila germline have generated valuable insights into how stem cells are controlled by diet-dependent pathways, and increasing evidence suggests that diverse adult stem cell populations respond to nutrients through similar mechanisms. Systemic signals, including nutrients themselves and diet-regulated hormones such as Insulin/Insulin-like growth factor or steroid hormones, can directly or indirectly affect stem cell behavior by modifying local cell-cell communication or intrinsic factors. The physiological regulation of stem cells in response to nutritional status not only is a fascinating biological problem, but also has clinical implications, as research in this field holds the key to noninvasive approaches for manipulating stem cells in vivo. In addition, given the known associations between diet, stem cells, and cancer risk, this research may inspire novel anticancer therapies.


Assuntos
Células-Tronco Adultas , Dieta , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Ecdisona/metabolismo , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Humanos , Redes e Vias Metabólicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...