Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2677: 113-125, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37464238

RESUMO

Live imaging of adult tissue stem cell niches provides key insights into the dynamic behavior of stem cells, their differentiating progeny, and their neighboring support cells, but few niches are amenable to this approach. Here, we discuss a technique for long-term live imaging of the Drosophila testis stem cell niche. Culturing whole testes ex vivo for up to 18 h allows for tracking of cell-type-specific behaviors under normal and various chemically or genetically modified conditions. Fixing and staining tissues after live imaging allows for the molecular confirmation of cell identity and behavior. By using live imaging in intact niches, we can better uncover the cellular and molecular mechanisms that regulate stem cell function in vivo.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Masculino , Testículo , Nicho de Células-Tronco/fisiologia , Células-Tronco , Drosophila melanogaster
2.
Elife ; 122023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36795469

RESUMO

Proper differentiation of sperm from germline stem cells, essential for production of the next generation, requires dramatic changes in gene expression that drive remodeling of almost all cellular components, from chromatin to organelles to cell shape itself. Here, we provide a single nucleus and single cell RNA-seq resource covering all of spermatogenesis in Drosophila starting from in-depth analysis of adult testis single nucleus RNA-seq (snRNA-seq) data from the Fly Cell Atlas (FCA) study. With over 44,000 nuclei and 6000 cells analyzed, the data provide identification of rare cell types, mapping of intermediate steps in differentiation, and the potential to identify new factors impacting fertility or controlling differentiation of germline and supporting somatic cells. We justify assignment of key germline and somatic cell types using combinations of known markers, in situ hybridization, and analysis of extant protein traps. Comparison of single cell and single nucleus datasets proved particularly revealing of dynamic developmental transitions in germline differentiation. To complement the web-based portals for data analysis hosted by the FCA, we provide datasets compatible with commonly used software such as Seurat and Monocle. The foundation provided here will enable communities studying spermatogenesis to interrogate the datasets to identify candidate genes to test for function in vivo.


Assuntos
Células-Tronco Adultas , Testículo , Animais , Masculino , Testículo/metabolismo , Drosophila , RNA-Seq , Sêmen
3.
Development ; 150(2)2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36503989

RESUMO

The adult Drosophila testis contains a well-defined niche created by a cluster of hub cells, which secrete signals that maintain adjacent germline stem cells and somatic cyst stem cells (CySCs). Hub cells are normally quiescent in adult flies but can exit quiescence, delaminate from the hub and convert into CySCs after ablation of all CySCs. The opposite event, CySC conversion into hub cells, was proposed to occur under physiological conditions, but the frequency of this event is debated. Here, to probe further the question of whether or not hub cells can be regenerated, we developed methods to genetically ablate some or all hub cells. Surprisingly, when flies were allowed to recover from ablation, the missing hub cells were not replaced. Hub cells did not exit quiescence after partial ablation of hub cells, and labeled cells from outside the hub did not enter the hub during or after ablation. Despite its ability to exit quiescence in response to CySC ablation, we conclude that the hub in the adult Drosophila testis does not have a mechanism to replenish missing hub cells.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Masculino , Proteínas de Drosophila/genética , Testículo , Células-Tronco/fisiologia , Nicho de Células-Tronco , Células Germinativas/fisiologia , Drosophila melanogaster , Diferenciação Celular/fisiologia
4.
Elife ; 112022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35468055

RESUMO

Adult stem cells are maintained in niches, specialized microenvironments that regulate their self-renewal and differentiation. In the adult Drosophila testis stem cell niche, somatic hub cells produce signals that regulate adjacent germline stem cells (GSCs) and somatic cyst stem cells (CySCs). Hub cells are normally quiescent, but after complete genetic ablation of CySCs, they can proliferate and transdifferentiate into new CySCs. Here we find that Epidermal growth factor receptor (EGFR) signaling is upregulated in hub cells after CySC ablation and that the ability of testes to recover from ablation is inhibited by reduced EGFR signaling. In addition, activation of the EGFR pathway in hub cells is sufficient to induce their proliferation and transdifferentiation into CySCs. We propose that EGFR signaling, which is normally required in adult cyst cells, is actively inhibited in adult hub cells to maintain their fate but is repurposed to drive stem cell regeneration after CySC ablation.


Assuntos
Cistos , Proteínas de Drosophila , Animais , Transdiferenciação Celular , Cistos/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores ErbB/metabolismo , Masculino , Receptores de Peptídeos de Invertebrados/genética , Receptores de Peptídeos de Invertebrados/metabolismo , Células-Tronco/fisiologia , Testículo/metabolismo , Microambiente Tumoral
5.
Science ; 375(6584): eabk2432, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35239393

RESUMO

For more than 100 years, the fruit fly Drosophila melanogaster has been one of the most studied model organisms. Here, we present a single-cell atlas of the adult fly, Tabula Drosophilae, that includes 580,000 nuclei from 15 individually dissected sexed tissues as well as the entire head and body, annotated to >250 distinct cell types. We provide an in-depth analysis of cell type-related gene signatures and transcription factor markers, as well as sexual dimorphism, across the whole animal. Analysis of common cell types between tissues, such as blood and muscle cells, reveals rare cell types and tissue-specific subtypes. This atlas provides a valuable resource for the Drosophila community and serves as a reference to study genetic perturbations and disease models at single-cell resolution.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Transcriptoma , Animais , Núcleo Celular/metabolismo , Bases de Dados Genéticas , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Genes de Insetos , Masculino , RNA-Seq , Caracteres Sexuais , Análise de Célula Única , Fatores de Transcrição/genética
6.
Cell Rep ; 24(13): 3466-3476.e8, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30257208

RESUMO

Homeostasis in adult tissues depends on the precise regulation of stem cells and their surrounding microenvironments, or niches. Here, we show that the cell cycle inhibitor and tumor suppressor Retinoblastoma (RB) is a critical regulator of niche cells in the Drosophila testis. The testis contains a single niche, composed of somatic hub cells, that signals to adjacent germline and somatic stem cells. Hub cells are normally quiescent, but knockdown of the RB homolog Rbf in these cells causes them to proliferate and convert to somatic stem cells. Over time, mutant hub cell clusters enlarge and split apart, forming ectopic hubs surrounded by active stem cells. Furthermore, we show that Rbf's ability to restrict niche number depends on the transcription factors E2F and Escargot and the adhesion molecule E-cadherin. Together this work reveals how precise modulation of niche cells, not only the stem cells they support, can drive regeneration and disease.


Assuntos
Autorrenovação Celular , Proteínas de Drosophila/metabolismo , Proteína do Retinoblastoma/metabolismo , Nicho de Células-Tronco , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Animais , Caderinas/metabolismo , Proliferação de Células , Proteínas de Drosophila/genética , Drosophila melanogaster , Masculino , Proteína do Retinoblastoma/genética , Testículo/citologia , Fatores de Transcrição/genética
7.
Methods Mol Biol ; 1463: 63-74, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27734347

RESUMO

Live imaging of adult tissue stem cell niches provides key insights into the dynamic behavior of stem cells, their differentiating progeny, and their neighboring support cells, but few niches are amenable to this approach. Here we discuss a technique for long-term live imaging of the Drosophila testis stem cell niche. Culturing whole testes ex vivo for up to 12.5 h allows for tracking of cell-type specific behaviors under normal and various chemically or genetically modified conditions. Fixing and staining tissues after live imaging allows for the molecular confirmation of cell identity and behavior. Utilization of live imaging in intact niches will facilitate further understanding of the cellular and molecular mechanisms that regulate stem cell function in vivo.


Assuntos
Drosophila/citologia , Testículo/ultraestrutura , Imagem com Lapso de Tempo/métodos , Animais , Rastreamento de Células , Masculino , Microscopia Confocal/métodos , Técnicas de Cultura de Órgãos , Coloração e Rotulagem , Nicho de Células-Tronco , Testículo/citologia , Fixação de Tecidos
8.
Development ; 143(5): 754-63, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26811385

RESUMO

Sexual identity is continuously maintained in specific differentiated cell types long after sex determination occurs during development. In the adult Drosophila testis, the putative transcription factor Chronologically inappropriate morphogenesis (Chinmo) acts with the canonical male sex determinant DoublesexM (Dsx(M)) to maintain the male identity of somatic cyst stem cells and their progeny. Here we find that ectopic expression of chinmo is sufficient to induce a male identity in adult ovarian somatic cells, but it acts through a Dsx(M)-independent mechanism. Conversely, the feminization of the testis somatic stem cell lineage caused by loss of chinmo is enhanced by expression of the canonical female sex determinant Dsx(F), indicating that chinmo acts in parallel with the canonical sex determination pathway to maintain the male identity of testis somatic cells. Consistent with this finding, ectopic expression of female sex determinants in the adult testis disrupts tissue morphology. The miRNA let-7 downregulates chinmo in many contexts, and ectopic expression of let-7 in the adult testis is sufficient to recapitulate the chinmo loss-of-function phenotype, but we find no apparent phenotypes upon removal of let-7 in the adult ovary or testis. Our finding that chinmo is necessary and sufficient to promote a male identity in adult gonadal somatic cells suggests that the sexual identity of somatic cells can be reprogrammed in the adult Drosophila ovary as well as in the testis.


Assuntos
Linhagem da Célula , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Proteínas do Tecido Nervoso/genética , Ovário/citologia , Animais , Diferenciação Celular , Proteínas de Drosophila/fisiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Janus Quinases/metabolismo , Masculino , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Oogênese , Fenótipo , Fatores de Transcrição STAT/metabolismo , Processos de Determinação Sexual , Testículo/citologia
9.
Dev Biol ; 404(1): 27-39, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25941003

RESUMO

Tissue-specific stem cells are thought to resist environmental insults better than their differentiating progeny, but this resistance varies from one tissue to another, and the underlying mechanisms are not well-understood. Here, we use the Drosophila testis as a model system to study the regulation of cell death within an intact niche. This niche contains sperm-producing germline stem cells (GSCs) and accompanying somatic cyst stem cells (or CySCs). Although many signals are known to promote stem cell self-renewal in this tissue, including the highly conserved JAK-STAT pathway, the response of these stem cells to potential death-inducing signals, and factors promoting stem cell survival, have not been characterized. Here we find that both GSCs and CySCs resist cell death better than their differentiating progeny, under normal laboratory conditions and in response to potential death-inducing stimuli such as irradiation or starvation. To ask what might be promoting stem cell survival, we characterized the role of the anti-apoptotic gene Drosophila inhibitor of apoptosis 1 (diap1) in testis stem cells. DIAP1 protein is enriched in the GSCs and CySCs and is a JAK-STAT target. diap1 is necessary for survival of both GSCs and CySCs, and ectopic up-regulation of DIAP1 in somatic cyst cells is sufficient to non-autonomously rescue stress-induced cell death in adjacent differentiating germ cells (spermatogonia). Altogether, our results show that niche signals can promote stem cell survival by up-regulation of highly conserved anti-apoptotic proteins, and suggest that this strategy may underlie the ability of stem cells to resist death more generally.


Assuntos
Sobrevivência Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Proteínas Inibidoras de Apoptose/metabolismo , Transdução de Sinais , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Proteínas Inibidoras de Apoptose/genética , Masculino , Espermatogônias/citologia , Espermatogônias/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Testículo/citologia , Testículo/metabolismo , Regulação para Cima
10.
Dev Cell ; 31(4): 474-86, 2014 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-25453558

RESUMO

Local signals maintain adult stem cells in many tissues. Whether the sexual identity of adult stem cells must also be maintained was not known. In the adult Drosophila testis niche, local Jak-STAT signaling promotes somatic cyst stem cell (CySC) renewal through several effectors, including the putative transcription factor Chronologically inappropriate morphogenesis (Chinmo). Here, we find that Chinmo also prevents feminization of CySCs. Chinmo promotes expression of the canonical male sex determination factor DoublesexM (Dsx(M)) within CySCs and their progeny, and ectopic expression of DsxM in the CySC lineage partially rescues the chinmo sex transformation phenotype, placing Chinmo upstream of Dsx(M). The Dsx homolog DMRT1 prevents the male-to-female conversion of differentiated somatic cells in the adult mammalian testis, but its regulation is not well understood. Our work indicates that sex maintenance occurs in adult somatic stem cells and that this highly conserved process is governed by effectors of niche signals. PAPERCLIP:


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Janus Quinases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição STAT/metabolismo , Processos de Determinação Sexual/fisiologia , Células-Tronco/citologia , Testículo/metabolismo , Envelhecimento , Animais , Diferenciação Celular/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Masculino , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo
11.
PLoS Genet ; 10(11): e1004713, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25375180

RESUMO

Stem cells in tissues reside in and receive signals from local microenvironments called niches. Understanding how multiple signals within niches integrate to control stem cell function is challenging. The Drosophila testis stem cell niche consists of somatic hub cells that maintain both germline stem cells and somatic cyst stem cells (CySCs). Here, we show a role for the axon guidance pathway Slit-Roundabout (Robo) in the testis niche. The ligand Slit is expressed specifically in hub cells while its receptor, Roundabout 2 (Robo2), is required in CySCs in order for them to compete for occupancy in the niche. CySCs also require the Slit-Robo effector Abelson tyrosine kinase (Abl) to prevent over-adhesion of CySCs to the niche, and CySCs mutant for Abl outcompete wild type CySCs for niche occupancy. Both Robo2 and Abl phenotypes can be rescued through modulation of adherens junction components, suggesting that the two work together to balance CySC adhesion levels. Interestingly, expression of Robo2 requires JAK-STAT signaling, an important maintenance pathway for both germline and cyst stem cells in the testis. Our work indicates that Slit-Robo signaling affects stem cell function downstream of the JAK-STAT pathway by controlling the ability of stem cells to compete for occupancy in their niche.


Assuntos
Janus Quinases/genética , Proteínas do Tecido Nervoso/biossíntese , Receptores Imunológicos/biossíntese , Fatores de Transcrição STAT/genética , Células-Tronco/metabolismo , Testículo/metabolismo , Animais , Diferenciação Celular/genética , Drosophila melanogaster , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Humanos , Janus Quinases/biossíntese , Masculino , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Fatores de Transcrição STAT/biossíntese , Transdução de Sinais , Nicho de Células-Tronco/genética , Células-Tronco/citologia , Testículo/crescimento & desenvolvimento , Proteínas Roundabout
12.
Dev Biol ; 394(1): 129-41, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25093968

RESUMO

Stem cell regulation by local signals is intensely studied, but less is known about the effects of hormonal signals on stem cells. In Drosophila, the primary steroid twenty-hydroxyecdysone (20E) regulates ovarian germline stem cells (GSCs) but was considered dispensable for testis GSC maintenance. Male GSCs reside in a microenvironment (niche) generated by somatic hub cells and adjacent cyst stem cells (CySCs). Here, we show that depletion of 20E from adult males by overexpressing a dominant negative form of the Ecdysone receptor (EcR) or its heterodimeric partner ultraspiracle (usp) causes GSC and CySC loss that is rescued by 20E feeding, uncovering a requirement for 20E in stem cell maintenance. EcR and USP are expressed, activated and autonomously required in the CySC lineage to promote CySC maintenance, as are downstream genes ftz-f1 and E75. In contrast, GSCs non-autonomously require ecdysone signaling. Global inactivation of EcR increases cell death in the testis that is rescued by expression of EcR-B2 in the CySC lineage, indicating that ecdysone signaling supports stem cell viability primarily through a specific receptor isoform. Finally, EcR genetically interacts with the NURF chromatin-remodeling complex, which we previously showed maintains CySCs. Thus, although 20E levels are lower in males than females, ecdysone signaling acts through distinct cell types and effectors to ensure both ovarian and testis stem cell maintenance.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Ecdisterona/metabolismo , Células-Tronco Embrionárias/fisiologia , Receptores de Esteroides/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Apoptose/genética , Diferenciação Celular/fisiologia , Sobrevivência Celular/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/biossíntese , Drosophila , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/metabolismo , Ecdisterona/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas , Masculino , Isoformas de Proteínas , Receptores de Esteroides/biossíntese , Transdução de Sinais , Testículo , Fatores de Transcrição/biossíntese
13.
Adv Exp Med Biol ; 786: 247-67, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696361

RESUMO

Adult stem cells are essential for the regeneration and repair of tissues in an organism. Signals from many different pathways converge to regulate stem cell maintenance and differentiation while preventing overproliferation. Although each population of adult stem cells is unique, common themes arise by comparing the regulation of various stem cell types in an organism or by comparing similar stem cell types across species. The JAK-STAT signaling pathway, identified nearly two decades ago, is now known to be involved in many biological processes including the regulation of stem cells. Studies in Drosophila first implicated JAK-STAT signaling in the control of stem cell maintenance in the male germline stem cell microenvironment, or niche; subsequently it has been shown play a role in other niches in both Drosophila and mammals. In this chapter, we will address the role of JAK-STAT signaling in stem cells in the germline, intestinal, hematopoietic and neuronal niches in Drosophila as well as the hematopoietic and neuronal niches in mammals. We will comment on how the study of JAK-STAT signaling in invertebrate systems has helped to advance our understanding of signaling in vertebrates. In addition to the role of JAK- STAT signaling in stem cell niche homeostasis, we will also discuss the diseases, including cancers, that can arise when this pathway is misregulated.


Assuntos
Células-Tronco Adultas/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Janus Quinases/genética , Fatores de Transcrição STAT/genética , Células-Tronco Adultas/citologia , Animais , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Células Germinativas/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Janus Quinases/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Nicho de Células-Tronco
14.
Trends Cell Biol ; 23(8): 357-64, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23597843

RESUMO

Adult stem cells reside in local microenvironments (niches) that produce signals regulating the outcome of stem cell divisions and stem cell-niche interactions. Limited space and signals in the niche often force stem cells to compete with one another. Although previous studies have uncovered several examples of genetically distinct stem cells competing for niche access, recent studies demonstrate that genetically equivalent stem cells compete under normal conditions, resulting in dynamic stem cell behavior in the niche. New work in multiple vertebrate and invertebrate tissues shows that stem cell competition occurs continuously and mutations disrupting the balance between competing stem cells can cause diseases and defects in the niche. This review discusses recent insights into stem cell competition in mammals and Drosophila.


Assuntos
Nicho de Células-Tronco , Células-Tronco/citologia , Animais , Drosophila melanogaster/citologia , Hematopoese , Mamíferos/metabolismo , Especificidade de Órgãos
15.
Spermatogenesis ; 2(3): 137-144, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23087833

RESUMO

The ability of stem cells to divide asymmetrically to produce both self-renewing and differentiating daughter cells sustains many adult tissues, but germline stem cells (GSCs) are unique among stem cells as they perpetuate the genome of the species. The cellular and molecular mechanisms regulating most mammalian stem cells in their endogenous local microenvironments, or niches, are quite challenging to study. However, studies of stem cell niches such as those found in the Drosophila gonads have proven very useful. In these tissues, GSCs are housed in a readily identifiable niche, and the ability to genetically manipulate these cells and their neighbors has uncovered several fundamental mechanisms that are relevant to stem cells more generally. Here, we summarize recent work on the regulation of GSCs in the Drosophila testis niche by intercellular signals, and on the intracellular mechanisms that cooperate with these signals to ensure the survival of the germline. This review focuses on GSCs within the adult Drosophila testis; somatic stem cells in this tissue are reviewed by Zoller and Schulz in this issue.(1) For a review of the testis niche as a whole, see de Cuevas and Matunis,(2) and for more comprehensive reviews of the Drosophila testis, refer to Fuller(3) and Davies and Fuller.(4).

16.
Development ; 138(14): 2861-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21693509

RESUMO

In metazoans, tissue maintenance and regeneration depend on adult stem cells, which are characterized by their ability to self-renew and generate differentiating progeny in response to the needs of the tissues in which they reside. In the Drosophila testis, germline and somatic stem cells are housed together in a common niche, where they are regulated by local signals, epigenetic mechanisms and systemic factors. These stem cell populations in the Drosophila testis have the unique advantage of being easy to identify and manipulate, and hence much progress has been made in understanding how this niche operates. Here, we summarize recent work on stem cells in the adult Drosophila testis and discuss the remarkable ability of these stem cells to respond to change within the niche.


Assuntos
Drosophila/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Homeostase/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Testículo/citologia , Animais , Moléculas de Adesão Celular/metabolismo , Polaridade Celular/fisiologia , Drosophila/fisiologia , Epigênese Genética/fisiologia , Masculino , MicroRNAs/metabolismo
17.
Cell Stem Cell ; 6(6): 557-67, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20569693

RESUMO

Regulation of stem cells depends on both tissue-specific transcriptional regulators and changes in chromatin organization, yet the coordination of these events in endogenous niches is poorly understood. In the Drosophila testis, local JAK-STAT signaling maintains germline and somatic stem cells (GSCs and cyst progenitor cells, or CPCs) in a single niche. Here we show that epigenetic regulation via the nucleosome-remodeling factor (NURF) complex ensures GSC and CPC maintenance by positively regulating JAK-STAT signaling, thereby preventing premature differentiation. Conversely, NURF is not required in early differentiating daughter cells of either lineage. Because three additional ATP-dependent chromatin remodelers (ACF, CHRAC, and dMi-2/NuRD) are dispensable for stem cell maintenance in the testis, epigenetic regulation of stem cells within this niche may rely primarily on NURF. Thus, local signals cooperate with specific chromatin-remodeling complexes in intact niches to coordinately regulate a common set of target genes to prevent premature stem cell differentiation.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Fatores de Transcrição STAT/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Adenosina Trifosfatases/genética , Animais , Montagem e Desmontagem da Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Drosophila/genética , Proteínas de Drosophila/genética , Células-Tronco Embrionárias/patologia , Regulação da Expressão Gênica no Desenvolvimento , Janus Quinases/genética , Janus Quinases/metabolismo , Masculino , Mutação/genética , RNA Interferente Pequeno/genética , Fatores de Transcrição STAT/genética , Transdução de Sinais/genética , Espermatogênese/genética , Nicho de Células-Tronco/patologia , Proteínas Supressoras da Sinalização de Citocina/genética , Testículo/embriologia , Testículo/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
Fly (Austin) ; 3(4): 283-5, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19923919

RESUMO

The reversal of cellular differentiation, or dedifferentiation, has fascinated biologists for many decades. While cells can be re-programmed extensively in culture, examples of in vivo dedifferentiation have recently emerged in both vertebrate and invertebrate systems, allowing for analysis of this intriguing process under more physiologically relevant conditions. Studies suggest that dedifferentiation occurs not only during large-scale cellular regeneration, but also at low levels to replenish stem cells lost due to normal turnover. Our recent paper demonstrates a novel method to induce the dedifferentiation of lineage-committed stem cell daughters back into germline stem cells (GSCs) in the Drosophila testis. We also show a requirement for activation of Janus kinase-signal transducer and activator of transcription (Jak-STAT) signaling in this process, and suggest that normally non-motile germline cells gain mobility and out-compete resident somatic cells for occupancy in the stem cell-maintaining microenvironment (niche). Here, we discuss what our findings reveal about stem cell competition and the capacity of various cell types to dedifferentiate.


Assuntos
Desdiferenciação Celular/fisiologia , Movimento Celular/fisiologia , Drosophila/fisiologia , Modelos Biológicos , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Animais , Proteínas de Drosophila/metabolismo , Masculino , Espermatogônias/citologia
19.
Cell Stem Cell ; 5(2): 191-203, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19664993

RESUMO

Differentiating cells can dedifferentiate to replace stem cells in aged or damaged tissues, but the underlying mechanisms are unknown. In the Drosophila testis, a cluster of stromal cells called the hub creates a niche by locally activating Janus kinase-signal transducer and activator of transcription (Jak-STAT) signaling in adjacent germline and somatic stem cells. Here, we establish a system to study spermatogonial dedifferentiation. Ectopically expressing the differentiation factor bag-of-marbles (Bam) removes germline stem cells from the niche. However, withdrawing ectopic Bam causes interconnected spermatogonia to fragment, move into the niche, exchange positions with resident somatic stem cells, and establish contact with the hub. Concomitantly, actin-based protrusions appear on subsets of spermatogonia, suggesting acquired motility. Furthermore, global downregulation of Jak-STAT signaling inhibits dedifferentiation, indicating that normal levels of pathway activation are required to promote movement of spermatogonia into the niche during dedifferentiation, where they outcompete somatic stem cells for niche occupancy.


Assuntos
Proteínas de Drosophila/metabolismo , Espermatogênese , Espermatogônias/citologia , Nicho de Células-Tronco/citologia , Testículo/citologia , Animais , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Janus Quinases/metabolismo , Masculino , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/fisiologia , Espermatogônias/metabolismo , Nicho de Células-Tronco/metabolismo , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA