Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 132024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38905123

RESUMO

The brain is consisted of diverse neurons arising from a limited number of neural stem cells. Drosophila neural stem cells called neuroblasts (NBs) produces specific neural lineages of various lineage sizes depending on their location in the brain. In the Drosophila visual processing centre - the optic lobes (OLs), medulla NBs derived from the neuroepithelium (NE) give rise to neurons and glia cells of the medulla cortex. The timing and the mechanisms responsible for the cessation of medulla NBs are so far not known. In this study, we show that the termination of medulla NBs during early pupal development is determined by the exhaustion of the NE stem cell pool. Hence, altering NE-NB transition during larval neurogenesis disrupts the timely termination of medulla NBs. Medulla NBs terminate neurogenesis via a combination of apoptosis, terminal symmetric division via Prospero, and a switch to gliogenesis via Glial Cell Missing (Gcm); however, these processes occur independently of each other. We also show that temporal progression of the medulla NBs is mostly not required for their termination. As the Drosophila OL shares a similar mode of division with mammalian neurogenesis, understanding when and how these progenitors cease proliferation during development can have important implications for mammalian brain size determination and regulation of its overall function.


Every cell in the body can be traced back to a stem cell. For instance, most cells in the adult brains of fruit flies come from a type of stem cell known as a neuroblast. This includes neurons and glial cells (which support and protect neurons) in the optic lobe, the part of the brain that processes visual information. The numbers of neurons and glia in the optic lobe are tightly regulated such that when the right numbers are reached, the neuroblasts stop making more and are terminated. But how and when this occurs is poorly understood. To investigate, Nguyen and Cheng studied when neuroblasts disappear in the optic lobe over the course of development. This revealed that the number of neuroblasts dropped drastically 12 to 18 hours after the fruit fly larvae developed in to pupae, and were completely gone by 30 hours in to pupae life. Further experiments revealed that the timing of this decrease is influenced by neuroepithelium cells, the pool of stem cells that generate neuroblasts during the early stages of development. Nguyen and Cheng found that speeding up this transition so that neuroblasts arise from the neuroepithelium earlier, led neuroblasts to disappear faster from the optic lobe; whereas delaying the transition caused neuroblasts to persist for much longer. Thus, the time at which neuroblasts are born determines when they are terminated. Furthermore, Nguyen and Cheng showed that the neuroblasts were lost through a combination of means. This includes dying via a process called apoptosis, dividing to form two mature neurons, or switching to a glial cell fate. These findings provide a deeper understanding of the mechanisms regulating stem cell pools and their conversion to different cell types, a process that is crucial to the proper development of the brain. How cells divide to form the optic lobe of fruit flies is similar to how new neurons arise in the mammalian brain. Understanding how and when stem cells in the fruit fly brain stop proliferating could therefore provide new insights in to the development of the human brain.


Assuntos
Apoptose , Diferenciação Celular , Proteínas de Drosophila , Células-Tronco Neurais , Células Neuroepiteliais , Neurogênese , Animais , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/citologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Neurogênese/fisiologia , Células Neuroepiteliais/fisiologia , Células Neuroepiteliais/citologia , Neuroglia/fisiologia , Neuroglia/citologia , Drosophila/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/fisiologia , Drosophila melanogaster/citologia , Lobo Óptico de Animais não Mamíferos/citologia , Lobo Óptico de Animais não Mamíferos/crescimento & desenvolvimento , Pupa/crescimento & desenvolvimento , Proteínas de Ligação a DNA , Fatores de Transcrição
2.
EMBO Rep ; 25(4): 1835-1858, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38429578

RESUMO

Cancer cachexia is a tumour-induced wasting syndrome, characterised by extreme loss of skeletal muscle. Defective mitochondria can contribute to muscle wasting; however, the underlying mechanisms remain unclear. Using a Drosophila larval model of cancer cachexia, we observed enlarged and dysfunctional muscle mitochondria. Morphological changes were accompanied by upregulation of beta-oxidation proteins and depletion of muscle glycogen and lipid stores. Muscle lipid stores were also decreased in Colon-26 adenocarcinoma mouse muscle samples, and expression of the beta-oxidation gene CPT1A was negatively associated with muscle quality in cachectic patients. Mechanistically, mitochondrial defects result from reduced muscle insulin signalling, downstream of tumour-secreted insulin growth factor binding protein (IGFBP) homologue ImpL2. Strikingly, muscle-specific inhibition of Forkhead box O (FOXO), mitochondrial fusion, or beta-oxidation in tumour-bearing animals preserved muscle integrity. Finally, dietary supplementation with nicotinamide or lipids, improved muscle health in tumour-bearing animals. Overall, our work demonstrates that muscle FOXO, mitochondria dynamics/beta-oxidation and lipid utilisation are key regulators of muscle wasting in cancer cachexia.


Assuntos
Neoplasias do Colo , Proteínas de Drosophila , Insulinas , Camundongos , Animais , Humanos , Caquexia/etiologia , Caquexia/metabolismo , Drosophila/metabolismo , Dinâmica Mitocondrial , Atrofia Muscular/patologia , Músculo Esquelético/metabolismo , Neoplasias do Colo/metabolismo , Insulinas/metabolismo , Lipídeos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
3.
PLoS Genet ; 19(11): e1011010, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37930995

RESUMO

Damage to light-sensing photoreceptors (PRs) occurs in highly prevalent retinal diseases. As humans cannot regenerate new PRs, these diseases often lead to irreversible blindness. Intriguingly, animals, such as the zebrafish, can regenerate PRs efficiently and restore functional vision. Upon injury, mature Müller glia (MG) undergo reprogramming to adopt a stem cell-like state. This process is similar to cellular dedifferentiation, and results in the generation of progenitor cells, which, in turn, proliferate and differentiate to replace lost retinal neurons. In this study, we tested whether factors involved in dedifferentiation of Drosophila CNS are implicated in the regenerative response in the zebrafish retina. We found that hairy-related 6 (her6) negatively regulates of PR production by regulating the rate of cell divisions in the MG-derived progenitors. prospero homeobox 1a (prox1a) is expressed in differentiated PRs and may promote PR differentiation through phase separation. Interestingly, upon Her6 downregulation, Prox1a is precociously upregulated in the PRs, to promote PR differentiation; conversely, loss of Prox1a also induces a downregulation of Her6. Together, we identified two novel candidates of PR regeneration that cross regulate each other; these may be exploited to promote human retinal regeneration and vision recovery.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Homeodomínio , Retina , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Regeneração Nervosa/fisiologia , Neuroglia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Homeodomínio/genética
4.
EMBO Rep ; 24(12): e57695, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38014610

RESUMO

In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-ß signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-ß signalling to regulate ECM accumulation in the fat body. TGF-ß signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-ß signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.


Assuntos
Caquexia , Neoplasias , Animais , Caquexia/etiologia , Caquexia/metabolismo , Drosophila , Insulina , Corpo Adiposo/metabolismo , Tecido Adiposo/metabolismo , Fator de Crescimento Transformador beta , Neoplasias/complicações
5.
EMBO Rep ; 24(6): e55837, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37039033

RESUMO

Dedifferentiation is the reversion of mature cells to a stem cell-like fate, whereby gene expression programs are altered and genes associated with multipotency are (re)expressed. Misexpression of multipotency factors and pathways causes the formation of ectopic neural stem cells (NSCs). Whether dedifferentiated NSCs faithfully produce the correct number and types of progeny, or undergo timely terminal differentiation, has not been assessed. Here, we show that ectopic NSCs induced via bHLH transcription factor Deadpan (Dpn) expression fail to undergo appropriate temporal progression by constantly expressing mid-temporal transcription factor(tTF), Sloppy-paired 1/2 (Slp). Consequently, this resulted in impaired terminal differenation and generated an excess of Twin of eyeless (Toy)-positive neurons at the expense of Reversed polarity (Repo)-positive glial cells. Preference for a mid-temporal fate in these ectopic NSCs is concordant with an enriched binding of Dpn at mid-tTF loci and a depletion of Dpn binding at early- and late-tTF loci. Retriggering the temporal series via manipulation of the temporal series or cell cycle is sufficient to reinstate neuronal diversity and timely termination.


Assuntos
Proteínas de Drosophila , Células-Tronco Neurais , Proteínas de Drosophila/genética , Células-Tronco Neurais/metabolismo , Fatores de Transcrição/metabolismo , Neurônios/metabolismo , Neuroglia , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento
6.
STAR Protoc ; 3(1): 101230, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35284841

RESUMO

Drosophila has become a popular model for examining the metabolic wasting syndrome, cachexia, characterized by degradation of muscles and fat. Here we present a protocol for quick and consistent scoring of muscle detachment, fat body lipid droplet size, and extracellular matrix (ECM) quantifications in Drosophila larvae. We detail the procedures for dissecting, staining, and imaging third instar Drosophila larval muscle fillets and fat body, and how to utilize FIJI macros for robust quantification of cachectic phenotypes in these dissected tissues. For complete details on the use and execution of this protocol, please refer to Lodge et al. (2021).


Assuntos
Caquexia , Drosophila , Animais , Caquexia/metabolismo , Corpo Adiposo/metabolismo , Larva/metabolismo , Músculos/metabolismo , Fenótipo
7.
Oxf Open Neurosci ; 1: kvac004, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-38596708

RESUMO

The formation of a functional circuitry in the central nervous system (CNS) requires the correct number and subtypes of neural cells. In the developing brain, neural stem cells (NSCs) self-renew while giving rise to progenitors that in turn generate differentiated progeny. As such, the size and the diversity of cells that make up the functional CNS depend on the proliferative properties of NSCs. In the fruit fly Drosophila, where the process of neurogenesis has been extensively investigated, extrinsic factors such as the microenvironment of NSCs, nutrients, oxygen levels and systemic signals have been identified as regulators of NSC proliferation. Here, we review decades of work that explores how extrinsic signals non-autonomously regulate key NSC characteristics such as quiescence, proliferation and termination in the fly.

8.
Dev Cell ; 56(18): 2664-2680.e6, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34473940

RESUMO

Cachexia, the wasting syndrome commonly observed in advanced cancer patients, accounts for up to one-third of cancer-related mortalities. We have established a Drosophila larval model of organ wasting whereby epithelial overgrowth in eye-antennal discs leads to wasting of the adipose tissue and muscles. The wasting is associated with fat-body remodeling and muscle detachment and is dependent on tumor-secreted matrix metalloproteinase 1 (Mmp1). Mmp1 can both modulate TGFß signaling in the fat body and disrupt basement membrane (BM)/extracellular matrix (ECM) protein localization in both the fat body and the muscle. Inhibition of TGFß signaling or Mmps in the fat body/muscle using a QF2-QUAS binary expression system rescues muscle wasting in the presence of tumor. Altogether, our study proposes that tumor-derived Mmps are central mediators of organ wasting in cancer cachexia.


Assuntos
Tecido Adiposo/metabolismo , Metaloproteinases da Matriz/metabolismo , Músculo Esquelético/metabolismo , Neoplasias/metabolismo , Animais , Membrana Basal/metabolismo , Drosophila/metabolismo , Matriz Extracelular/metabolismo , Atrofia Muscular/metabolismo
9.
EMBO Rep ; 22(5): e52130, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33751817

RESUMO

The final size and function of the adult central nervous system (CNS) are determined by neuronal lineages generated by neural stem cells (NSCs) in the developing brain. In Drosophila, NSCs called neuroblasts (NBs) reside within a specialised microenvironment called the glial niche. Here, we explore non-autonomous glial regulation of NB proliferation. We show that lipid droplets (LDs) which reside within the glial niche are closely associated with the signalling molecule Hedgehog (Hh). Under physiological conditions, cortex glial Hh is autonomously required to sustain niche chamber formation. Upon FGF-mediated cortex glial overgrowth, glial Hh non-autonomously activates Hh signalling in the NBs, which in turn disrupts NB cell cycle progression and its ability to produce neurons. Glial Hh's ability to signal to NB is further modulated by lipid storage regulator lipid storage droplet-2 (Lsd-2) and de novo lipogenesis gene fatty acid synthase 1 (Fasn1). Together, our data suggest that glial-derived Hh modified by lipid metabolism mechanisms can affect the neighbouring NB's ability to proliferate and produce neurons.


Assuntos
Proteínas de Drosophila , Células-Tronco Neurais , Animais , Proliferação de Células , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas Hedgehog/genética , Metabolismo dos Lipídeos , Células-Tronco Neurais/metabolismo
10.
Wiley Interdiscip Rev Dev Biol ; 10(4): e394, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32852143

RESUMO

Studies in model organisms have demonstrated that extensive communication occurs between distant organs both during development and in diseases such as cancer. Organs communicate with each other to coordinate growth and reach the correct size, while the fate of tumor cells depend on the outcome of their interaction with the immune system and peripheral tissues. In this review, we outline recent studies in Drosophila, which have enabled an improved understanding of the complex crosstalk between organs in the context of both organismal and tumor growth. We argue that Drosophila is a powerful model organism for studying these interactions, and these studies have the potential for improving our understanding of signaling pathways and candidate factors that mediate this conserved interorgan crosstalk. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Early Embryonic Development > Development to the Basic Body Plan Invertebrate Organogenesis > Flies.


Assuntos
Comunicação , Proteínas de Drosophila/metabolismo , Drosophila/crescimento & desenvolvimento , Desenvolvimento Embrionário , Neoplasias/patologia , Organogênese , Animais , Transdução de Sinais
11.
EMBO J ; 39(21): e106564, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33021741

RESUMO

Glycolysis is a major metabolic process which ensures the break down of glucose into pyruvate via multiple enzymatic steps, but if and how this catabolism can impact on developmental patterning is unclear. In this issue, Spannl et al (2020) demonstrate a novel link between energy metabolism and tissue formation in the fly imaginal discs. They show that ATPs generated via glycolysis maintain active transport of a smoothened inhibitor, which keeps Hh signalling in check to preserve the correct shape and proportion of the developing wing.


Assuntos
Proteínas de Drosophila , Proteínas Hedgehog , Trifosfato de Adenosina/metabolismo , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Glicólise , Proteínas Hedgehog/metabolismo , Potenciais da Membrana , Plasma/metabolismo , Asas de Animais/metabolismo
12.
EMBO J ; 38(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30804004

RESUMO

Rewired metabolism of glutamine in cancer has been well documented, but less is known about other amino acids such as histidine. Here, we use Drosophila cancer models to show that decreasing the concentration of histidine in the diet strongly inhibits the growth of mutant clones induced by loss of Nerfin-1 or gain of Notch activity. In contrast, changes in dietary histidine have much less effect on the growth of wildtype neural stem cells and Prospero neural tumours. The reliance of tumours on dietary histidine and also on histidine decarboxylase (Hdc) depends upon their growth requirement for Myc. We demonstrate that Myc overexpression in nerfin-1 tumours is sufficient to switch their mode of growth from histidine/Hdc sensitive to resistant. This study suggests that perturbations in histidine metabolism selectively target neural tumours that grow via a dedifferentiation process involving large cell size increases driven by Myc.


Assuntos
Desdiferenciação Celular , Neoplasias do Sistema Nervoso Central/patologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histidina/administração & dosagem , Células-Tronco Neurais/patologia , Fatores de Transcrição/metabolismo , Animais , Neoplasias do Sistema Nervoso Central/genética , Neoplasias do Sistema Nervoso Central/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Feminino , Histidina Descarboxilase/genética , Histidina Descarboxilase/metabolismo , Masculino , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fatores de Transcrição/genética
13.
Cell Rep ; 25(6): 1561-1576.e7, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30404010

RESUMO

The ability of cells to stably maintain their fate is governed by specific transcription regulators. Here, we show that the Scalloped (Sd) and Nervous fingers-1 (Nerfin-1) transcription factors physically and functionally interact to maintain medulla neuron fate in the Drosophila melanogaster CNS. Using Targeted DamID, we find that Sd and Nerfin-1 occupy a highly overlapping set of target genes, including regulators of neural stem cell and neuron fate, and signaling pathways that regulate CNS development such as Notch and Hippo. Modulation of either Sd or Nerfin-1 activity causes medulla neurons to dedifferentiate to a stem cell-like state, and this is mediated at least in part by Notch pathway deregulation. Intriguingly, orthologs of Sd and Nerfin-1 have also been implicated in control of neuronal cell fate decisions in both worms and mammals. Our data indicate that this transcription factor pair exhibits remarkable biochemical and functional conservation across metazoans.


Assuntos
Linhagem da Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Desdiferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lobo Óptico de Animais não Mamíferos/citologia , Lobo Óptico de Animais não Mamíferos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
14.
Curr Biol ; 26(8): 1034-42, 2016 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-26996505

RESUMO

A key question in developmental neurobiology is how neural stem cells regulate their proliferative potential and cellular diversity and thus specify the overall size of the brain. Drosophila melanogaster neural stem cells (neuroblasts) are known to regulate their ability to self-renew by asymmetric cell division and produce different types of neurons and glia through sequential expression of temporal transcription factors [1]. Here, we show that the conserved Hippo pathway, a key regulator of epithelial organ size [2-4], restricts neuroblast proliferative potential and neuronal cell number to regulate brain size. The inhibition of Hippo pathway activity via depletion of the core kinases Tao-1, Hippo, or Warts regulates several key characteristics of neuroblasts during postembryonic neurogenesis. The Hippo pathway is required to maintain timely entry and exit from neurogenesis by regulating both neuroblast reactivation from quiescence and the time at which neuroblasts undergo terminal differentiation. Further, it restricts neuroblast cell-cycle speed, specifies cell size, and alters the proportion of neuron types generated during postembryonic neurogenesis. Collectively, deregulation of Hippo signaling in neuroblasts causes a substantial increase in overall brain size. We show that these effects are mediated via the key downstream transcription co-activator Yorkie and that, indeed, Yorkie overexpression in neuroblasts is sufficient to cause brain overgrowth. These studies reveal a novel mechanism that controls stem cell proliferative potential during postembryonic neurogenesis to regulate brain size.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Transdução de Sinais , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proliferação de Células , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células-Tronco Neurais/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Tamanho do Órgão , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteínas de Sinalização YAP
15.
Cell Mol Life Sci ; 73(8): 1641-4, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26817462

RESUMO

The ability to maintain cells in a differentiated state and to prevent them from reprogramming into a multipotent state has recently emerged as a central theme in neural development as well as in oncogenesis. In the developing central nervous system (CNS) of the fruit fly Drosophila, several transcription factors were recently identified to be required in postmitotic cells to maintain differentiation, and in their absence, mature neurons undergo dedifferentiation, giving rise to proliferative neural stem cells and ultimately to tumor growth. In this review, we will highlight the current understanding of dedifferentiation and cell plasticity in the Drosophila CNS.


Assuntos
Encéfalo/citologia , Desdiferenciação Celular/fisiologia , Plasticidade Celular/fisiologia , Drosophila/citologia , Células-Tronco Neurais/citologia , Animais , Diferenciação Celular/fisiologia , Drosophila/metabolismo , Neurônios/citologia
16.
Genes Dev ; 29(2): 129-43, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25593306

RESUMO

Cellular dedifferentiation is the regression of a cell from a specialized state to a more multipotent state and is implicated in cancer. However, the transcriptional network that prevents differentiated cells from reacquiring stem cell fate is so far unclear. Neuroblasts (NBs), the Drosophila neural stem cells, are a model for the regulation of stem cell self-renewal and differentiation. Here we show that the Drosophila zinc finger transcription factor Nervous fingers 1 (Nerfin-1) locks neurons into differentiation, preventing their reversion into NBs. Following Prospero-dependent neuronal specification in the ganglion mother cell (GMC), a Nerfin-1-specific transcriptional program maintains differentiation in the post-mitotic neurons. The loss of Nerfin-1 causes reversion to multipotency and results in tumors in several neural lineages. Both the onset and rate of neuronal dedifferentiation in nerfin-1 mutant lineages are dependent on Myc- and target of rapamycin (Tor)-mediated cellular growth. In addition, Nerfin-1 is required for NB differentiation at the end of neurogenesis. RNA sequencing (RNA-seq) and chromatin immunoprecipitation (ChIP) analysis show that Nerfin-1 administers its function by repression of self-renewing-specific and activation of differentiation-specific genes. Our findings support the model of bidirectional interconvertibility between neural stem cells and their post-mitotic progeny and highlight the importance of the Nerfin-1-regulated transcriptional program in neuronal maintenance.


Assuntos
Desdiferenciação Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Mutação , Neurogênese/genética , Neurônios/citologia , Fatores de Transcrição/genética
17.
Cell ; 146(3): 435-47, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816278

RESUMO

Developing animals survive periods of starvation by protecting the growth of critical organs at the expense of other tissues. Here, we use Drosophila to explore the as yet unknown mechanisms regulating this privileged tissue growth. As in mammals, we observe in Drosophila that the CNS is more highly spared than other tissues during nutrient restriction (NR). We demonstrate that anaplastic lymphoma kinase (Alk) efficiently protects neural progenitor (neuroblast) growth against reductions in amino acids and insulin-like peptides during NR via two mechanisms. First, Alk suppresses the growth requirement for amino acid sensing via Slimfast/Rheb/TOR complex 1. And second, Alk, rather than insulin-like receptor, primarily activates PI3-kinase. Alk maintains PI3-kinase signaling during NR as its ligand, Jelly belly (Jeb), is constitutively expressed from a glial cell niche surrounding neuroblasts. Together, these findings identify a brain-sparing mechanism that shares some regulatory features with the starvation-resistant growth programs of mammalian tumors.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Quinase do Linfoma Anaplásico , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Privação de Alimentos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Larva/crescimento & desenvolvimento , Larva/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Poliploidia
18.
Curr Opin Neurobiol ; 20(1): 50-7, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20079625

RESUMO

Neural stem and progenitor cells generate the central nervous system (CNS) in organisms as diverse as insects and mammals. In Drosophila, multipotent asymmetrically dividing progenitors called neuroblasts produce neurons and glia throughout the developing CNS. Nevertheless, the time-windows of mitotic activity, the division modes, the termination mechanisms and the lineage sizes of individual neuroblasts all vary considerably from region-to-region. Recent studies shed light on some of the mechanisms underlying this neuroblast diversity and, in particular, how proliferation is boosted in two brain regions. In the central brain, some specialised neuroblasts generate intermediate neural progenitors that can each divide multiple times, thus increasing overall lineage size. In the optic lobe, an alternative expansion strategy involves symmetrically dividing neuroepithelial cells generating large numbers of asymmetrically dividing neuroblasts. Evidence is also emerging for a cell-intrinsic timer that alters the properties of each neuroblast with increasing developmental age. The core mechanism corresponds to a series of transcription factors that coordinates temporal changes in neuronal/glial identity with transitions in neuroblast cell-cycle speed, entry into quiescence and, ultimately, with termination.


Assuntos
Proliferação de Células , Sistema Nervoso Central/citologia , Drosophila/citologia , Neurônios/citologia , Células-Tronco/fisiologia , Animais , Divisão Celular/fisiologia , Sistema Nervoso Central/fisiologia , Drosophila/fisiologia , Neurônios/fisiologia , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...