Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2677: 139-149, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37464240

RESUMO

Stem cell pools are dynamic and capable of reacting to insults like injury and starvation. Recent work has highlighted the key role of dedifferentiation as a conserved mechanism for replenishing stem cell pools after their loss, thereby maintaining tissue homeostasis. The testis of the fruit fly Drosophila melanogaster offers a simple but powerful system to study dedifferentiation, the process by which differentiating spermatogonia can revert their fate to become fully functional germline stem cells (GSCs). Dedifferentiated GSCs show interesting characteristics, such as being more proliferative than their wild-type sibling GSCs. To facilitate the study of the cellular and molecular mechanisms underlying the process of germline dedifferentiation in the Drosophila testis, here we describe techniques for inducing high rates of dedifferentiation and for unambiguously labeling dedifferentiated GSCs.


Assuntos
Proteínas de Drosophila , Testículo , Animais , Masculino , Espermatogônias , Drosophila , Drosophila melanogaster , Proteínas de Drosophila/genética , Células-Tronco , Diferenciação Celular , Células Germinativas
2.
Dev Cell ; 57(1): 80-94.e7, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34942115

RESUMO

Niches maintain a finite pool of stem cells via restricted space and short-range signals. Stem cells compete for limited niche resources, but the mechanisms regulating competition are poorly understood. Using the Drosophila testis model, we show that germline stem cells (GSCs) lacking the transcription factor Chinmo gain a competitive advantage for niche access. Surprisingly, chinmo-/- GSCs rely on a new mechanism of competition in which they secrete the extracellular matrix protein Perlecan to selectively evict non-mutant GSCs and then upregulate Perlecan-binding proteins to remain in the altered niche. Over time, the GSC pool can be entirely replaced with chinmo-/- cells. As a consequence, the mutant chinmo allele acts as a gene drive element; the majority of offspring inherit the allele despite the heterozygous genotype of the parent. Our results suggest that the influence of GSC competition may extend beyond individual stem cell niche dynamics to population-level allelic drift and evolution.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Animais , Diferenciação Celular/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Matriz Extracelular/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Germinativas/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Masculino , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/fisiologia , Testículo/metabolismo , Fatores de Transcrição/metabolismo
3.
Dev Cell ; 56(16): 2284-2294.e6, 2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34363758

RESUMO

Aging causes stem cell dysfunction as a result of extrinsic and intrinsic changes. Decreased function of the stem cell niche is an important contributor to this dysfunction. We use the Drosophila testis to investigate what factors maintain niche cells. The testis niche comprises quiescent "hub" cells and supports two mitotic stem cell pools: germline stem cells and somatic cyst stem cells (CySCs). We identify the cell-cycle-responsive Dp/E2f1 transcription factor as a crucial non-autonomous regulator required in CySCs to maintain hub cell quiescence. Dp/E2f1 inhibits local Activin ligands through production of the Activin antagonist Follistatin (Fs). Inactivation of Dp/E2f1 or Fs in CySCs or promoting Activin receptor signaling in hub cells causes transdifferentiation of hub cells into fully functional CySCs. This Activin-dependent communication between CySCs and hub regulates the physiological decay of the niche with age and demonstrates that hub cell quiescence results from signals from surrounding stem cells.


Assuntos
Proteínas de Drosophila/metabolismo , Folistatina/metabolismo , Nicho de Células-Tronco , Fatores de Transcrição/metabolismo , Ativinas/metabolismo , Animais , Proliferação de Células , Transdiferenciação Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Masculino , Espermatozoides/citologia , Espermatozoides/metabolismo , Espermatozoides/fisiologia , Testículo/citologia , Fatores de Transcrição/genética
4.
Int J Mol Sci ; 22(11)2021 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-34073743

RESUMO

The Jun N-terminal kinase (JNK) pathway is an evolutionary conserved kinase cascade best known for its roles during stress-induced apoptosis and tumor progression. Recent findings, however, have identified new roles for this pleiotropic pathway in stem cells during regenerative responses and in cellular plasticity. Here, we provide an overview of recent findings about the new roles of JNK signaling in stem cell biology using two well-established Drosophila models: the testis and the intestine. We highlight the pathway's roles in processes such as proliferation, death, self-renewal and reprogramming, and discuss the known parallels between flies and mammals.


Assuntos
Drosophila/metabolismo , Homeostase , Sistema de Sinalização das MAP Quinases , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Drosophila/fisiologia , Feminino , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Células-Tronco/fisiologia
5.
G3 (Bethesda) ; 11(4)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33751104

RESUMO

One of the best examples of sexual dimorphism is the development and function of the gonads, ovaries and testes, which produce sex-specific gametes, oocytes, and spermatids, respectively. The development of these specialized germ cells requires sex-matched somatic support cells. The sexual identity of somatic gonadal cells is specified during development and must be actively maintained during adulthood. We previously showed that the transcription factor Chinmo is required to ensure the male sexual identity of somatic support cells in the Drosophila melanogaster testis. Loss of chinmo from male somatic gonadal cells results in feminization: they transform from squamous to epithelial-like cells that resemble somatic cells in the female gonad but fail to properly ensheath the male germline, causing infertility. To identify potential target genes of Chinmo, we purified somatic cells deficient for chinmo from the adult Drosophila testis and performed next-generation sequencing to compare their transcriptome to that of control somatic cells. Bioinformatics revealed 304 and 1549 differentially upregulated and downregulated genes, respectively, upon loss of chinmo in early somatic cells. Using a combination of methods, we validated several differentially expressed genes. These data sets will be useful resources to the community.


Assuntos
Células-Tronco Adultas , Proteínas de Drosophila , Células-Tronco Adultas/metabolismo , Animais , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Gônadas/metabolismo , Masculino , Proteínas do Tecido Nervoso/genética , Testículo/metabolismo , Fatores de Transcrição/genética , Transcriptoma
6.
G3 (Bethesda) ; 9(8): 2609-2622, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31227525

RESUMO

Cell competition is the elimination of one viable population of cells (the losers) by a neighboring fitter population (the winners) and was discovered by studies in the Drosophila melanogaster wing imaginal disc. Supercompetition is a process in which cells with elevated JAK/STAT signaling or increased Myc become winners and outcompete wild-type neighbors. To identify the genes that are differentially regulated in STAT supercompetitors, we purified these cells from Drosophila wing imaginal discs and performed next-generation sequencing. Their transcriptome was compared to those of control wing disc cells and Myc supercompetitors. Bioinformatics revealed that STAT and Myc supercompetitors have distinct transcriptomes with only 41 common differentially regulated genes. Furthermore, STAT supercompetitors have elevated reactive oxygen species, an anti-oxidant response and increased ecdysone signaling. Using a combination of methods, we validated 13 differentially expressed genes. These data sets will be useful resources to the community.


Assuntos
Drosophila/genética , Drosophila/metabolismo , Ecdisona/metabolismo , Estresse Oxidativo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Animais , Sítios de Ligação , Biologia Computacional/métodos , Genoma , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Janus Quinases/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo
7.
Dis Model Mech ; 12(5)2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31072879

RESUMO

Myeloproliferative neoplasms (MPNs) are clonal hematopoietic disorders that cause excessive production of myeloid cells. Most MPN patients have a point mutation in JAK2 (JAK2V617F ), which encodes a dominant-active kinase that constitutively triggers JAK/STAT signaling. In Drosophila, this pathway is simplified, with a single JAK, Hopscotch (Hop), and a single STAT transcription factor, Stat92E. The hopTumorous-lethal [hopTum ] allele encodes a dominant-active kinase that induces sustained Stat92E activation. Like MPN patients, hopTum mutants have significantly more myeloid cells, which form invasive tumors. Through an unbiased genetic screen, we found that heterozygosity for Enhancer of Polycomb [E(Pc)], a component of the Tip60 lysine acetyltransferase complex (also known as KAT5 in humans), significantly increased tumor burden in hopTum animals. Hematopoietic depletion of E(Pc) or other Tip60 components in an otherwise wild-type background also induced blood cell tumors. The E(Pc) tumor phenotype was dependent on JAK/STAT activity, as concomitant depletion of hop or Stat92E inhibited tumor formation. Stat92E target genes were significantly upregulated in E(Pc)-mutant myeloid cells, indicating that loss of E(Pc) activates JAK/STAT signaling. Neither the hop nor Stat92E gene was upregulated upon hematopoietic E(Pc) depletion, suggesting that the regulation of the JAK/STAT pathway by E(Pc) is dependent on substrates other than histones. Indeed, E(Pc) depletion significantly increased expression of Hop protein in myeloid cells. This study indicates that E(Pc) works as a tumor suppressor by attenuating Hop protein expression and ultimately JAK/STAT signaling. Since loss-of-function mutations in the human homologs of E(Pc) and Tip60 are frequently observed in cancer, our work could lead to new treatments for MPN patients.This article has an associated First Person interview with the first author of the paper.


Assuntos
Carcinogênese/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neoplasias Hematológicas/metabolismo , Histona Acetiltransferases/metabolismo , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Hematopoese , Heterozigoto , Melanoma/patologia , Modelos Biológicos , Fenótipo , Fatores de Transcrição/metabolismo
8.
Development ; 146(2)2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30696713

RESUMO

The JAK/STAT pathway is a conserved metazoan signaling system that transduces cues from extracellular cytokines into transcriptional changes in the nucleus. JAK/STAT signaling is best known for its roles in immunity. However, recent work has demonstrated that it also regulates critical homeostatic processes in germline and somatic stem cells, as well as regenerative processes in several tissues, including the gonad, intestine and appendages. Here, we provide an overview of JAK/STAT signaling in stem cells and regeneration, focusing on Drosophila and highlighting JAK/STAT pathway functions in proliferation, survival and cell competition that are conserved between Drosophila and vertebrates.


Assuntos
Drosophila/citologia , Drosophila/fisiologia , Janus Quinases/metabolismo , Regeneração/fisiologia , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Vertebrados/fisiologia , Animais , Células-Tronco/metabolismo
9.
Dev Cell ; 46(6): 672-674, 2018 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-30253165

RESUMO

Competitive interactions between neighboring cells require fitness comparison and local killing, but the signals regulating these processes are unknown. In this issue, Alpar et al. (2018) demonstrate that fitter cells secrete serine proteases to create a local burst of active Spätzle, triggering Toll signaling and apoptosis in less fit neighbors.


Assuntos
Proteínas de Drosophila , Receptores Toll-Like , Apoptose , Serina Endopeptidases , Transdução de Sinais
10.
Elife ; 72018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29985130

RESUMO

Exhaustion of stem cells is a hallmark of aging. In the Drosophila testis, dedifferentiated germline stem cells (GSCs) derived from spermatogonia increase during lifespan, leading to the model that dedifferentiation counteracts the decline of GSCs in aged males. To test this, we blocked dedifferentiation by mis-expressing the differentiation factor bag of marbles (bam) in spermatogonia while lineage-labeling these cells. Strikingly, blocking bam-lineage dedifferentiation under normal conditions in virgin males has no impact on the GSC pool. However, in mated males or challenging conditions, inhibiting bam-lineage dedifferentiation markedly reduces the number of GSCs and their ability to proliferate and differentiate. We find that bam-lineage derived GSCs have significantly higher proliferation rates than sibling GSCs in the same testis. We determined that Jun N-terminal kinase (JNK) activity is autonomously required for bam-lineage dedifferentiation. Overall, we show that dedifferentiation provides a mechanism to maintain the germline and ensure fertility under chronically stressful conditions.


Assuntos
Desdiferenciação Celular , Drosophila melanogaster/citologia , Sistema de Sinalização das MAP Quinases , Espermatogônias/enzimologia , Espermatogônias/patologia , Células-Tronco/enzimologia , Células-Tronco/patologia , Estresse Fisiológico , Testículo/patologia , Envelhecimento/metabolismo , Animais , Linhagem da Célula , Ativação Enzimática , Masculino , Modelos Biológicos , Espermatogênese
11.
PLoS Genet ; 14(2): e1007203, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29389999

RESUMO

Reproduction in sexually dimorphic animals relies on successful gamete production, executed by the germline and aided by somatic support cells. Somatic sex identity in Drosophila is instructed by sex-specific isoforms of the DMRT1 ortholog Doublesex (Dsx). Female-specific expression of Sex-lethal (Sxl) causes alternative splicing of transformer (tra) to the female isoform traF. In turn, TraF alternatively splices dsx to the female isoform dsxF. Loss of the transcriptional repressor Chinmo in male somatic stem cells (CySCs) of the testis causes them to "feminize", resembling female somatic stem cells in the ovary. This somatic sex transformation causes a collapse of germline differentiation and male infertility. We demonstrate this feminization occurs by transcriptional and post-transcriptional regulation of traF. We find that chinmo-deficient CySCs upregulate tra mRNA as well as transcripts encoding tra-splice factors Virilizer (Vir) and Female lethal (2)d (Fl(2)d). traF splicing in chinmo-deficient CySCs leads to the production of DsxF at the expense of the male isoform DsxM, and both TraF and DsxF are required for CySC sex transformation. Surprisingly, CySC feminization upon loss of chinmo does not require Sxl but does require Vir and Fl(2)d. Consistent with this, we show that both Vir and Fl(2)d are required for tra alternative splicing in the female somatic gonad. Our work reveals the need for transcriptional regulation of tra in adult male stem cells and highlights a previously unobserved Sxl-independent mechanism of traF production in vivo. In sum, transcriptional control of the sex determination hierarchy by Chinmo is critical for sex maintenance in sexually dimorphic tissues and is vital in the preservation of fertility.


Assuntos
Proteínas de Drosophila/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/genética , Processos de Determinação Sexual/genética , Diferenciação Sexual/genética , Testículo/embriologia , Processamento Alternativo/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Proteínas do Tecido Nervoso/genética , Ovário/embriologia , Ovário/metabolismo , Proteínas de Ligação a RNA/fisiologia , Testículo/metabolismo
12.
J Cell Biol ; 216(12): 4165-4182, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29051265

RESUMO

Isoprenylcysteine carboxyl methyltransferase (ICMT) methylesterifies C-terminal prenylcysteine residues of CaaX proteins and some RAB GTPases. Deficiency of either ICMT or NOTCH1 accelerates pancreatic neoplasia in Pdx1-Cre;LSL-KrasG12D mice, suggesting that ICMT is required for NOTCH signaling. We used Drosophila melanogaster wing vein and scutellar bristle development to screen Rab proteins predicted to be substrates for ICMT (ste14 in flies). We identified Rab7 and Rab8 as ICMT substrates that when silenced phenocopy ste14 deficiency. ICMT, RAB7, and RAB8 were all required for efficient NOTCH1 signaling in mammalian cells. Overexpression of RAB8 rescued NOTCH activation after ICMT knockdown both in U2OS cells expressing NOTCH1 and in fly wing vein development. ICMT deficiency induced mislocalization of GFP-RAB7 and GFP-RAB8 from endomembrane to cytosol, enhanced binding to RABGDI, and decreased GTP loading of RAB7 and RAB8. Deficiency of ICMT, RAB7, or RAB8 led to mislocalization and diminished processing of NOTCH1-GFP. Thus, NOTCH signaling requires ICMT in part because it requires methylated RAB7 and RAB8.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , GTP Fosfo-Hidrolases/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Metiltransferases/genética , Receptor Notch1/genética , Proteínas rab de Ligação ao GTP/genética , Alquil e Aril Transferases/genética , Alquil e Aril Transferases/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Metilação , Camundongos , Osteoblastos/metabolismo , Osteoblastos/patologia , Proteínas Metiltransferases/deficiência , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Asas de Animais , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
13.
G3 (Bethesda) ; 7(8): 2427-2438, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28620086

RESUMO

A gain-of-function mutation in the tyrosine kinase JAK2 (JAK2V617F ) causes human myeloproliferative neoplasms (MPNs). These patients present with high numbers of myeloid lineage cells and have numerous complications. Since current MPN therapies are not curative, there is a need to find new regulators and targets of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling that may represent additional clinical interventions . Drosophila melanogaster offers a low complexity model to study MPNs as JAK/STAT signaling is simplified with only one JAK [Hopscotch (Hop)] and one STAT (Stat92E). hopTumorous-lethal(Tum-l) is a gain-of-function mutation that causes dramatic expansion of myeloid cells, which then form lethal melanotic tumors. Through an F1 deficiency (Df) screen, we identified 11 suppressors and 35 enhancers of melanotic tumors in hopTum-l animals. Dfs that uncover the Hippo (Hpo) pathway genes expanded (ex) and warts (wts) strongly enhanced the hopTum-l tumor burden, as did mutations in ex, wts, and other Hpo pathway genes. Target genes of the Hpo pathway effector Yorkie (Yki) were significantly upregulated in hopTum-l blood cells, indicating that Yki signaling was increased. Ectopic hematopoietic activation of Yki in otherwise wild-type animals increased hemocyte proliferation but did not induce melanotic tumors. However, hematopoietic depletion of Yki significantly reduced the hopTum-l tumor burden, demonstrating that Yki is required for melanotic tumors in this background. These results support a model in which elevated Yki signaling increases the number of hemocytes, which become melanotic tumors as a result of elevated JAK/STAT signaling.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Testes Genéticos , Neoplasias Hematológicas/genética , Janus Quinases/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Alelos , Animais , Proliferação de Células , Hematopoese , Hemócitos/metabolismo , Mutação/genética , Carga Tumoral , Regulação para Cima/genética , Proteínas de Sinalização YAP
14.
Development ; 143(21): 3914-3925, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27633989

RESUMO

Stem cells reside in niches that provide signals to maintain self-renewal, and differentiation is viewed as a passive process that depends on loss of access to these signals. Here, we demonstrate that the differentiation of somatic cyst stem cells (CySCs) in the Drosophila testis is actively promoted by PI3K/Tor signaling, as CySCs lacking PI3K/Tor activity cannot differentiate properly. We find that an insulin peptide produced by somatic cells immediately outside of the stem cell niche acts locally to promote somatic differentiation through Insulin-like receptor (InR) activation. These results indicate that there is a local 'differentiation' niche that upregulates PI3K/Tor signaling in the early daughters of CySCs. Finally, we demonstrate that CySCs secrete the Dilp-binding protein ImpL2, the Drosophila homolog of IGFBP7, into the stem cell niche, which blocks InR activation in CySCs. Thus, we show that somatic cell differentiation is controlled by PI3K/Tor signaling downstream of InR and that the local production of positive and negative InR signals regulates the differentiation niche. These results support a model in which leaving the stem cell niche and initiating differentiation are actively induced by signaling.


Assuntos
Diferenciação Celular , Proteínas de Drosophila/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Testículo/embriologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrião não Mamífero , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/genética , Células-Tronco/metabolismo , Testículo/citologia
15.
PLoS Genet ; 12(1): e1005815, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26807580

RESUMO

The Drosophila testis is a well-established system for studying stem cell self-renewal and competition. In this tissue, the niche supports two stem cell populations, germ line stem cells (GSCs), which give rise to sperm, and somatic stem cells called cyst stem cells (CySCs), which support GSCs and their descendants. It has been established that CySCs compete with each other and with GSCs for niche access, and mutations have been identified that confer increased competitiveness to CySCs, resulting in the mutant stem cell and its descendants outcompeting wild type resident stem cells. Socs36E, which encodes a negative feedback inhibitor of the JAK/STAT pathway, was the first identified regulator of niche competition. The competitive behavior of Socs36E mutant CySCs was attributed to increased JAK/STAT signaling. Here we show that competitive behavior of Socs36E mutant CySCs is due in large part to unbridled Mitogen-Activated Protein Kinase (MAPK) signaling. In Socs36E mutant clones, MAPK activity is elevated. Furthermore, we find that clonal upregulation of MAPK in CySCs leads to their outcompetition of wild type CySCs and of GSCs, recapitulating the Socs36E mutant phenotype. Indeed, when MAPK activity is removed from Socs36E mutant clones, they lose their competitiveness but maintain self-renewal, presumably due to increased JAK/STAT signaling in these cells. Consistently, loss of JAK/STAT activity in Socs36E mutant clones severely impairs their self-renewal. Thus, our results enable the genetic separation of two essential processes that occur in stem cells. While some niche signals specify the intrinsic property of self-renewal, which is absolutely required in all stem cells for niche residence, additional signals control the ability of stem cells to compete with their neighbors. Socs36E is node through which these processes are linked, demonstrating that negative feedback inhibition integrates multiple aspects of stem cell behavior.


Assuntos
Diferenciação Celular/genética , Proteínas de Drosophila/genética , Células Germinativas/crescimento & desenvolvimento , Células-Tronco/citologia , Proteínas Supressoras da Sinalização de Citocina/genética , Testículo/crescimento & desenvolvimento , Animais , Proteínas de Drosophila/biossíntese , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Transdução de Sinais/genética , Espermatozoides/citologia , Espermatozoides/crescimento & desenvolvimento , Nicho de Células-Tronco/genética , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Testículo/metabolismo
16.
Cell Stem Cell ; 17(2): 133-4, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26253198

RESUMO

Niche cells produce secreted factors that promote the self-renewal of stem cells in their immediate proximity, but how signaling is restricted to stem cells is not well understood. Inaba et al. (2015) report that microtubule (MT) structures called MT-nanotubes control activation of the primary self-renewal pathway in Drosophila testes.


Assuntos
Drosophila melanogaster/citologia , Microtúbulos/química , Microtúbulos/metabolismo , Nanotubos , Transdução de Sinais , Nicho de Células-Tronco/fisiologia , Testículo/citologia , Animais , Masculino
17.
EMBO J ; 33(20): 2295-313, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25092766

RESUMO

Neutral competition, an emerging feature of stem cell homeostasis, posits that individual stem cells can be lost and replaced by their neighbors stochastically, resulting in chance dominance of a clone at the niche. A single stem cell with an oncogenic mutation could bias this process and clonally spread the mutation throughout the stem cell pool. The Drosophila testis provides an ideal system for testing this model. The niche supports two stem cell populations that compete for niche occupancy. Here, we show that cyst stem cells (CySCs) conform to the paradigm of neutral competition and that clonal deregulation of either the Hedgehog (Hh) or Hippo (Hpo) pathway allows a single CySC to colonize the niche. We find that the driving force behind such behavior is accelerated proliferation. Our results demonstrate that a single stem cell colonizes its niche through oncogenic mutation by co-opting an underlying homeostatic process.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Superfície Celular/genética , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Clonais , Drosophila , Masculino , Modelos Teóricos , Mutação , Fenótipo , Transdução de Sinais , Nicho de Células-Tronco/genética , Células-Tronco/citologia , Testículo/citologia , Testículo/metabolismo
18.
Semin Cell Dev Biol ; 28: 96-103, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24685611

RESUMO

Sustained activation of the JAK/STAT pathway is causal to human cancers. This pathway is less complex in Drosophila, and its dysregulation has been linked to several tumor models in this organism. Here, we discuss models of metastatic epithelial and hematopoietic tumors that are causally linked to dysregulation of JAK/STAT signaling in Drosophila. First, we focus on cancer models in imaginal discs where ectopic expression of the JAK/STAT pathway ligand Unpaired downstream of distinct tumor suppressors has emerged as an unexpected mediator of neoplastic transformation. We also discuss the collaboration between STAT and oncogenic Ras in epithelial transformation. Second, we examine hematopoietic tumors, where mutations that cause hyperactive JAK/STAT signaling are necessary and sufficient for "fly leukemia". We highlight the important contributions that genetic screens in Drosophila have made to understanding the JAK/STAT pathway, its developmental roles, and how its function is co-opted during tumorigenesis.


Assuntos
Proteínas de Drosophila/metabolismo , Janus Quinases/metabolismo , Neoplasias/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Animais , Transformação Celular Neoplásica , Drosophila , Humanos
19.
Development ; 141(5): 988-1000, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24550108

RESUMO

A conventional view of development is that cells cooperate to build an organism. However, based on studies of Drosophila, it has been known for years that viable cells can be eliminated by their neighbours through a process termed cell competition. New studies in mammals have revealed that this process is universal and that many factors and mechanisms are conserved. During cell competition, cells with lower translation rates or those with lower levels of proteins involved in signal transduction, polarity and cellular growth can survive in a homogenous environment but are killed when surrounded by cells of higher fitness. Here, we discuss recent advances in the field as well as the mechanistic steps involved in this phenomenon, which have shed light on how and why cell competition exists in developing and adult organisms.


Assuntos
Comunicação Celular/fisiologia , Animais , Drosophila , Proteínas de Drosophila/metabolismo , Transdução de Sinais/fisiologia
20.
J Clin Invest ; 123(11): 4681-94, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24216479

RESUMO

RAS is the most frequently mutated oncogene in human cancers. Despite decades of effort, anti-RAS therapies have remained elusive. Isoprenylcysteine carboxylmethyltransferase (ICMT) methylates RAS and other CaaX-containing proteins, but its potential as a target for cancer therapy has not been fully evaluated. We crossed a Pdx1-Cre;LSL-KrasG12D mouse, which is a model of pancreatic ductal adenocarcinoma (PDA), with a mouse harboring a floxed allele of Icmt. Surprisingly, we found that ICMT deficiency dramatically accelerated the development and progression of neoplasia. ICMT-deficient pancreatic ductal epithelial cells had a slight growth advantage and were resistant to premature senescence by a mechanism that involved suppression of cyclin-dependent kinase inhibitor 2A (p16INK4A) expression. ICMT deficiency precisely phenocopied Notch1 deficiency in the Pdx1-Cre;LSL-KrasG12D model by exacerbating pancreatic intraepithelial neoplasias, promoting facial papillomas, and derepressing Wnt signaling. Silencing ICMT in human osteosarcoma cells decreased Notch1 signaling in response to stimulation with cell-surface ligands. Additionally, targeted silencing of Ste14, the Drosophila homolog of Icmt, resulted in defects in wing development, consistent with Notch loss of function. Our data suggest that ICMT behaves like a tumor suppressor in PDA because it is required for Notch1 signaling.


Assuntos
Genes ras , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Metiltransferases/deficiência , Receptor Notch1/metabolismo , Animais , Animais Geneticamente Modificados , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Feminino , Humanos , Masculino , Metaplasia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Mutação , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Proteínas Metiltransferases/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...