Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 17(10): e1009834, 2021 10.
Article in English | MEDLINE | ID: mdl-34644293

ABSTRACT

Stem cells have the potential to maintain undifferentiated state and differentiate into specialized cell types. Despite numerous progress has been achieved in understanding stem cell self-renewal and differentiation, many fundamental questions remain unanswered. In this study, we identify dRTEL1, the Drosophila homolog of Regulator of Telomere Elongation Helicase 1, as a novel regulator of male germline stem cells (GSCs). Our genome-wide transcriptome analysis and ChIP-Seq results suggest that dRTEL1 affects a set of candidate genes required for GSC maintenance, likely independent of its role in DNA repair. Furthermore, dRTEL1 prevents DNA damage-induced checkpoint activation in GSCs. Finally, dRTEL1 functions to sustain Stat92E protein levels, the key player in GSC maintenance. Together, our findings reveal an intrinsic role of the DNA helicase dRTEL1 in maintaining male GSC and provide insight into the function of dRTEL1.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Germ Cells/physiology , Stem Cells/physiology , Animals , Cell Self Renewal/genetics , DNA Helicases/metabolism , DNA Repair/genetics , Female , Male , Signal Transduction/genetics , Transcriptome/genetics
2.
Development ; 146(13)2019 06 28.
Article in English | MEDLINE | ID: mdl-31164352

ABSTRACT

One fundamental property of a stem cell niche is the exchange of molecular signals between its component cells. Niche models, such as the Drosophila melanogaster testis, have been instrumental in identifying and studying the conserved genetic factors that contribute to niche molecular signalling. Here, we identify jam packed (jam), an allele of Striatin interacting protein (Strip), which is a core member of the highly conserved Striatin-interacting phosphatase and kinase (STRIPAK) complex. In the developing Drosophila testis, Strip cell-autonomously regulates the differentiation and morphology of the somatic lineage, and non-cell-autonomously regulates the proliferation and differentiation of the germline lineage. Mechanistically, Strip acts in the somatic lineage with its STRIPAK partner, Connector of kinase to AP-1 (Cka), where they negatively regulate the Jun N-terminal kinase (JNK) signalling pathway. Our study reveals a novel role for Strip/Cka in JNK pathway regulation during spermatogenesis within the developing Drosophila testis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drosophila Proteins/physiology , Drosophila melanogaster , MAP Kinase Signaling System/genetics , Phosphate-Binding Proteins/physiology , Spermatogenesis/genetics , Animals , Animals, Genetically Modified , Cell Differentiation , Down-Regulation/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Male , Testis/cytology , Testis/growth & development , Testis/metabolism
3.
Behav Brain Res ; 359: 918-926, 2019 02 01.
Article in English | MEDLINE | ID: mdl-29935278

ABSTRACT

Autism spectrum disorder (ASD) is a heterogeneous and highly heritable condition with multiple aetiologies. Although the biological mechanisms underlying ASD are not fully understood, evidence suggests that dysregulation of serotonergic systems play an important role in ASD psychopathology. Preclinical models using mice with altered serotonergic neurotransmission may provide insight into the role of serotonin in behaviours relevant to clinical features of ASD. For example, BALB/c mice carry a loss-of-function single nucleotide polymorphism (SNP; C1473 G) in tryptophan hydroxylase 2 (Tph2), which encodes the brain-specific isoform of the rate-limiting enzyme for serotonin synthesis, and these mice frequently have been used to model symptoms of ASD. In this study, juvenile male BALB/c (G/G; loss-of-function variant) and C57BL/6 J (C/C; wild type variant) mice, were exposed to the three-chamber sociability test, and one week later to the elevated plus-maze (EPM). Tryptophan hydroxylase 2 (TPH2) activity was measured following injection of the aromatic amino acid decarboxylase (AADC)-inhibitor, NSD-1015, and subsequent HPLC detection of 5-hydroxytryptophan (5-HTP) within subregions of the dorsal raphe nucleus (DR) and median raphe nucleus (MnR). The BALB/c mice showed reduced social behaviour and increased anxious behaviour, as well as decreased 5-HTP accumulation in the rostral and mid-rostrocaudal DR. In the full cohort of mice, TPH2 activity in the mid-rostrocaudal DR was correlated with anxious behaviour in the EPM, however these correlations were not statistically significant within each strain, suggesting that TPH2 activity was not directly associated with either anxiety or sociability. Further research is therefore required to more fully understand how serotonergic systems are involved in mouse behaviours that resemble some of the clinical features of ASD.


Subject(s)
Anxiety/metabolism , Raphe Nuclei/metabolism , Social Behavior , Tryptophan Hydroxylase/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Anxiety/drug therapy , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Genotype , Humans , Hydrazines/therapeutic use , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , Tryptophan Hydroxylase/genetics
4.
Stem Cell Reports ; 7(6): 1152-1163, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27974223

ABSTRACT

The Drosophila testis has been fundamental to understanding how stem cells interact with their endogenous microenvironment, or niche, to control organ growth in vivo. Here, we report the identification of two independent alleles for the highly conserved tumor suppressor gene, Retinoblastoma-family protein (Rbf), in a screen for testis phenotypes in X chromosome third-instar lethal alleles. Rbf mutant alleles exhibit overproliferation of spermatogonial cells, which is phenocopied by the molecularly characterized Rbf11 null allele. We demonstrate that Rbf promotes cell-cycle exit and differentiation of the somatic and germline stem cells of the testes. Intriguingly, depletion of Rbf specifically in the germline does not disrupt stem cell differentiation, rather Rbf loss of function in the somatic lineage drives overproliferation and differentiation defects in both lineages. Together our observations suggest that Rbf in the somatic lineage controls germline stem cell renewal and differentiation non-autonomously via essential roles in the microenvironment of the germline lineage.


Subject(s)
Cell Lineage , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Retinoblastoma Protein/metabolism , Spermatogenesis , Stem Cells/cytology , Testis/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Proliferation , Germ Cells/cytology , Germ Cells/metabolism , Larva , Male , Mutation/genetics , Stem Cell Niche , Stem Cells/metabolism
5.
Development ; 143(1): 35-44, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26586222

ABSTRACT

The mechanism for the basal targeting of the Miranda (Mira) complex during the asymmetric division of Drosophila neuroblasts (NBs) is yet to be fully understood. We have identified conserved Phosphotyrosyl phosphatase activator (PTPA) as a novel mediator for the basal localization of the Mira complex in larval brain NBs. In mutant Ptpa NBs, Mira remains cytoplasmic during early mitosis and its basal localization is delayed until anaphase. Detailed analyses indicate that PTPA acts independent of and before aPKC to localize Mira. Mechanistically, our data show that the phosphorylation status of the T591 residue determines the subcellular localization of Mira and that PTPA facilitates the dephosphorylation of T591. Furthermore, PTPA associates with the Protein phosphatase 4 complex to mediate localization of Mira. On the basis of these results, a two-step process for the basal localization of Mira during NB division is revealed: cortical association of Mira mediated by the PTPA-PP4 complex is followed by apical aPKC-mediated basal restriction.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/embryology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Protein Kinase C/metabolism , Animals , Asymmetric Cell Division/physiology , Cell Line , Phosphoprotein Phosphatases/metabolism , Phosphorylation
6.
Adv Exp Med Biol ; 786: 79-102, 2013.
Article in English | MEDLINE | ID: mdl-23696353

ABSTRACT

The Drosophila central nervous system develops from polarised asymmetric divisions of precursor cells, called neuroblasts. Decades of research on neuroblasts have resulted in a substantial understanding of the factors and molecular events responsible for fate decisions of neuroblasts and their progeny. Furthermore, the cell-cycle dependent mechanisms responsible for asymmetric cortical protein localisation, resulting in the unequal partitioning between daughters, are beginning to be exposed. Disruption to the appropriate partitioning of proteins between neuroblasts and differentiation-committed daughters can lead to supernumerary neuroblast-like cells and the formation of tumours. Many of the factors responsible for regulating asymmetric division of Drosophila neuroblasts are evolutionarily conserved and, in many cases, have been shown to play a functionally conserved role in mammalian neurogenesis. Recent genome-wide studies coupled with advancements in live-imaging technologies have opened further avenues of research into neuroblast biology. We review our current understanding of the molecular mechanisms regulating neuroblast divisions, a powerful system to model mammalian neurogenesis and tumourigenesis.


Subject(s)
Central Nervous System/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Neurons/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Division/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Central Nervous System/cytology , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Signal Transduction
7.
Cell Adh Migr ; 3(4): 425-32, 2009.
Article in English | MEDLINE | ID: mdl-19829062

ABSTRACT

Metazoan growth and development is maintained by populations of undifferentiated cells, commonly known as stem cells. Stem cells possess several characteristic properties, including dividing through self-renewing divisions and generating progeny that differentiate to have specialized cell fates. Multiple signaling pathways have been identified which coordinate stem cell proliferation with maintenance and differentiation. Relatively recently, the small, non-protein coding microRNAs (miRNAs) have been identified to function as important regulators in stem cell development. Individual miRNAs are capable of directing the translational repression of many mRNAs targets, generating widespread changes in gene expression. In addition, dysfunction of miRNA expression is commonly associated with cancer development. Cancer stem cells, which are likely responsible for initiating and maintaining tumorigenesis, share many similarities with stem cells and some mechanisms of miRNA function may be in common between these two cell types.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , MicroRNAs/physiology , Stem Cells/cytology , Stem Cells/physiology , Animals , Humans
8.
J Cell Biol ; 180(2): 267-72, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18209103

ABSTRACT

Over the past decade, many of the key components of the genetic machinery that regulate the asymmetric division of Drosophila melanogaster neural progenitors, neuroblasts, have been identified and their functions elucidated. Studies over the past two years have shown that many of these identified components act to regulate the self-renewal versus differentiation decision and appear to function as tumor suppressors during larval nervous system development. In this paper, we highlight the growing number of molecules that are normally considered to be key regulators of cell cycle events/progression that have recently been shown to impinge on the neuroblast asymmetric division machinery to control asymmetric protein localization and/or the decision to self-renew or differentiate.


Subject(s)
Drosophila Proteins/analysis , Drosophila melanogaster/chemistry , Drosophila melanogaster/cytology , Neoplasms/metabolism , Animals , Cell Cycle , Cell Division , Drosophila melanogaster/metabolism , Humans , Larva , Neurons/cytology
9.
Nature ; 449(7158): 96-100, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17805297

ABSTRACT

Self-renewal and differentiation are cardinal features of stem cells. Asymmetric cell division provides one fundamental mechanism by which stem cell self-renewal and differentiation are balanced. A failure of this balance could lead to diseases such as cancer. During asymmetric division of stem cells, factors controlling their self-renewal and differentiation are unequally segregated between daughter cells. Numb is one such factor that is segregated to the differentiating daughter cell during the stem-cell-like neuroblast divisions in Drosophila melanogaster, where it inhibits self-renewal. The localization and function of Numb is cell-cycle-dependent. Here we show that Polo (ref. 13), a key cell cycle regulator, the mammalian counterparts of which have been implicated as oncogenes as well as tumour suppressors, acts as a tumour suppressor in the larval brain. Supernumerary neuroblasts are produced at the expense of neurons in polo mutants. Polo directly phosphorylates Partner of Numb (Pon, ref. 16), an adaptor protein for Numb, and this phosphorylation event is important for Pon to localize Numb. In polo mutants, the asymmetric localization of Pon, Numb and atypical protein kinase C are disrupted, whereas other polarity markers are largely unaffected. Overexpression of Numb suppresses neuroblast overproliferation caused by polo mutations, suggesting that Numb has a major role in mediating this effect of Polo. Our results reveal a biochemical link between the cell cycle and the asymmetric protein localization machinery, and indicate that Polo can inhibit progenitor self-renewal by regulating the localization and function of Numb.


Subject(s)
Carrier Proteins/metabolism , Cell Polarity , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Juvenile Hormones/metabolism , Neurons/cytology , Protein Serine-Threonine Kinases/metabolism , Stem Cells/cytology , Animals , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Differentiation , Cell Division , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Gene Expression , Larva/cytology , Larva/metabolism , Neurons/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Transport , Spindle Apparatus/metabolism , Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
BMC Genet ; 7: 33, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16749923

ABSTRACT

BACKGROUND: The asymmetric segregation of determinants during cell division is a fundamental mechanism for generating cell fate diversity during development. In Drosophila, neural precursors (neuroblasts) divide in a stem cell-like manner generating a larger apical neuroblast and a smaller basal ganglion mother cell. The cell fate determinant Prospero and its adapter protein Miranda are asymmetrically localized to the basal cortex of the dividing neuroblast and segregated into the GMC upon cytokinesis. Previous screens to identify components of the asymmetric division machinery have concentrated on embryonic phenotypes. However, such screens are reaching saturation and are limited in that the maternal contribution of many genes can mask the effects of zygotic loss of function, and other approaches will be necessary to identify further genes involved in neuroblast asymmetric division. RESULTS: We have performed a genetic screen in the third instar larval brain using the basal localization of Miranda as a marker for neuroblast asymmetry. In addition to the examination of pupal lethal mutations, we have employed the MARCM (Mosaic Analysis with a Repressible Cell Marker) system to generate postembryonic clones of mutations with an early lethal phase. We have screened a total of 2,300 mutagenized chromosomes and isolated alleles affecting cell fate, the localization of basal determinants or the orientation of the mitotic spindle. We have also identified a number of complementation groups exhibiting defects in cell cycle progression and cytokinesis, including both novel genes and new alleles of known components of these processes. CONCLUSION: We have identified four mutations which affect the process of neuroblast asymmetric division. One of these, mapping to the imaginal discs arrested locus, suggests a novel role for the anaphase promoting complex/cyclosome (APC/C) in the targeting of determinants to the basal cortex. The identification and analysis of the remaining mutations will further advance our understanding of the process of asymmetric cell division. We have also isolated a number of mutations affecting cell division which will complement the functional genomics approaches to this process being employed by other laboratories. Taken together, these results demonstrate the value of mosaic screens in the identification of genes involved in neuroblast division.


Subject(s)
Drosophila/genetics , Ganglia, Invertebrate/embryology , Germ-Line Mutation , Mosaicism , Animals , Brain/cytology , Brain/embryology , Cell Cycle Proteins/genetics , Drosophila/embryology , Drosophila Proteins/genetics , Female , Ganglia, Invertebrate/cytology , Genes, Lethal , Genetic Testing , Larva/genetics , Larva/growth & development , Male , Phenotype
11.
Dev Cell ; 9(3): 312-3, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16139221

ABSTRACT

Three current papers in Cell and in this issue of Developmental Cell highlight the role of the exocyst in recycling of membrane proteins from endosomes to the plasma membrane in asymmetric cell division and polarized epithelial cells.


Subject(s)
Cell Polarity/physiology , Animals , Cell Division/physiology , Cell Membrane/physiology , Drosophila/metabolism , Endosomes/metabolism , Endosomes/physiology , Epithelial Cells/physiology , Oocytes/physiology , Sense Organs/physiology
12.
J Cell Sci ; 116(Pt 21): 4277-81, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14514883

ABSTRACT

Despite a century of research into the nature of animal cell division, a molecular explanation for the positioning of the actomyosin contractile ring has remained elusive. The discovery of a novel interaction between regulators of Rho family small GTPases has revealed a link between the mitotic microtubules and the contractile ring during the later stages of mitosis. The properties of the interacting Rho regulators suggest a molecular model for the positioning and initiation of contractile ring furrowing in animal cells. In this 'double ring' model, centralspindlin complexes, localized by the action of their kinesin-like protein component, position and activate a cortical equatorial ring of Rho GTPase exchange factors. The resulting ring of activated Rho would then trigger a cascade of events leading to formation and constriction of the contractile ring.


Subject(s)
Actomyosin/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism , Animals , Cell Division , Contractile Proteins/metabolism , Drosophila Proteins/metabolism
13.
Dev Cell ; 4(1): 29-39, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12530961

ABSTRACT

The mechanism that positions the cytokinetic contractile ring is unknown, but derives from the spindle midzone. We show that an interaction between the Rho GTP exchange factor, Pebble, and the Rho family GTPase-activating protein, RacGAP50C, connects the contractile ring to cortical microtubules at the site of furrowing in D. melanogaster cells. Pebble regulates actomyosin organization, while RacGAP50C and its binding partner, the Pavarotti kinesin-like protein, regulate microtubule bundling. All three factors are required for cytokinesis. As furrowing begins, these proteins colocalize to a cortical equatorial ring. We propose that RacGAP50C-Pavarotti complexes travel on cortical microtubules to the cell equator, where they associate with the Pebble RhoGEF to position contractile ring formation and coordinate F-actin and microtubule remodeling during cytokinesis.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubules/metabolism , Animals , Cell Division , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Macromolecular Substances , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rho Guanine Nucleotide Exchange Factors , Spindle Apparatus , Two-Hybrid System Techniques , Wings, Animal/cytology , Wings, Animal/embryology , Wings, Animal/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...