Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
EMBO J ; 40(9): e106388, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33634906

RESUMO

Transposable elements (TEs) play a significant role in evolution, contributing to genetic variation. However, TE mobilization in somatic cells is not well understood. Here, we address the prevalence of transposition in a somatic tissue, exploiting the Drosophila midgut as a model. Using whole-genome sequencing of in vivo clonally expanded gut tissue, we have mapped hundreds of high-confidence somatic TE integration sites genome-wide. We show that somatic retrotransposon insertions are associated with inactivation of the tumor suppressor Notch, likely contributing to neoplasia formation. Moreover, applying Oxford Nanopore long-read sequencing technology we provide evidence for tissue-specific differences in retrotransposition. Comparing somatic TE insertional activity with transcriptomic and small RNA sequencing data, we demonstrate that transposon mobility cannot be simply predicted by whole tissue TE expression levels or by small RNA pathway activity. Finally, we reveal that somatic TE insertions in the adult fly intestine are enriched in genic regions and in transcriptionally active chromatin. Together, our findings provide clear evidence of ongoing somatic transposition in Drosophila and delineate previously unknown features underlying somatic TE mobility in vivo.


Assuntos
Elementos de DNA Transponíveis , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Neoplasias Intestinais/genética , Receptores Notch/genética , Animais , Evolução Clonal , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Masculino , Especificidade de Órgãos , Recombinação Genética , Análise de Sequência de RNA/métodos , Sequenciamento Completo do Genoma
2.
Development ; 149(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36399062

RESUMO

While testing for genome instability in Drosophila as reported by unscheduled upregulation of UAS-GFP in cells that co-express GAL80 and GAL4, we noticed that, as expected, background levels were low in most developing tissues. However, GFP-positive clones were frequent in the larval brain. Most of these clones originated from central brain neural stem cells. Using imaging-based approaches and genome sequencing, we show that these unscheduled clones do not result from chromosome loss or mutations in GAL80. We have named this phenomenon 'Illuminati'. Illuminati is strongly enhanced in brat tumors and is also sensitive to environmental conditions such as food content and temperature. Illuminati is suppressed by Su(var)2-10, but it is not significantly affected by several modifiers of position effect variegation or Gal4::UAS variegation. We conclude that Illuminati identifies a previously unknown type of functional instability that may have important implications in development and disease.


Assuntos
Proteínas de Drosophila , Células-Tronco Neurais , Animais , Drosophila/genética , Drosophila melanogaster/genética , Mutação/genética , Expressão Gênica , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética
3.
Genome Res ; 31(8): 1419-1432, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34168010

RESUMO

Spontaneous mutations can alter tissue dynamics and lead to cancer initiation. Although large-scale sequencing projects have illuminated processes that influence somatic mutation and subsequent tumor evolution, the mutational dynamics operating in the very early stages of cancer development are currently not well understood. To explore mutational processes in the early stages of cancer evolution, we exploited neoplasia arising spontaneously in the Drosophila intestine. Analysing whole-genome sequencing data with a dedicated bioinformatic pipeline, we found neoplasia formation to be driven largely through the inactivation of Notch by structural variants, many of which involve highly complex genomic rearrangements. The genome-wide mutational burden in neoplasia was found to be similar to that of several human cancers. Finally, we identified genomic features associated with spontaneous mutation, and defined the evolutionary dynamics and mutational landscape operating within intestinal neoplasia over the short lifespan of the adult fly. Our findings provide unique insight into mutational dynamics operating over a short timescale in the genetic model system, Drosophila melanogaster.


Assuntos
Drosophila melanogaster , Drosophila , Animais , Drosophila/genética , Drosophila melanogaster/genética , Genômica , Intestinos , Mutação , Células-Tronco
4.
EMBO J ; 36(13): 1928-1945, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28533229

RESUMO

How terminal cell fates are specified in dynamically renewing adult tissues is not well understood. Here we explore terminal cell fate establishment during homeostasis using the enteroendocrine cells (EEs) of the adult Drosophila midgut as a paradigm. Our data argue against the existence of local feedback signals, and we identify Numb as an intrinsic regulator of EE fate. Our data further indicate that Numb, with alpha-adaptin, acts upstream or in parallel of known regulators of EE fate to limit Notch signaling, thereby facilitating EE fate acquisition. We find that Numb is regulated in part through its asymmetric and symmetric distribution during stem cell divisions; however, its de novo synthesis is also required during the differentiation of the EE cell. Thus, this work identifies Numb as a crucial factor for cell fate choice in the adult Drosophila intestine. Furthermore, our findings demonstrate that cell-intrinsic control mechanisms of terminal cell fate acquisition can result in a balanced tissue-wide production of terminally differentiated cell types.


Assuntos
Diferenciação Celular , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Células Enteroendócrinas/fisiologia , Regulação da Expressão Gênica , Hormônios Juvenis/metabolismo , Animais , Intestinos/fisiologia , Transdução de Sinais
5.
PLoS Genet ; 14(11): e1007773, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30452449

RESUMO

Precise regulation of stem cell self-renewal and differentiation properties is essential for tissue homeostasis. Using the adult Drosophila intestine to study molecular mechanisms controlling stem cell properties, we identify the gene split-ends (spen) in a genetic screen as a novel regulator of intestinal stem cell fate (ISC). Spen family genes encode conserved RNA recognition motif-containing proteins that are reported to have roles in RNA splicing and transcriptional regulation. We demonstrate that spen acts at multiple points in the ISC lineage with an ISC-intrinsic function in controlling early commitment events of the stem cells and functions in terminally differentiated cells to further limit the proliferation of ISCs. Using two-color cell sorting of stem cells and their daughters, we characterize spen-dependent changes in RNA abundance and exon usage and find potential key regulators downstream of spen. Our work identifies spen as an important regulator of adult stem cells in the Drosophila intestine, provides new insight to Spen-family protein functions, and may also shed light on Spen's mode of action in other developmental contexts.


Assuntos
Células-Tronco Adultas/citologia , Autorrenovação Celular/genética , Autorrenovação Celular/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/citologia , Drosophila melanogaster/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Células-Tronco Adultas/metabolismo , Animais , Animais Geneticamente Modificados , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Proteínas de Homeodomínio/antagonistas & inibidores , Intestinos/citologia , Masculino , Modelos Biológicos , Mutação , Proteínas Nucleares/antagonistas & inibidores , Interferência de RNA , Proteínas de Ligação a RNA , Receptores Notch/metabolismo , Transdução de Sinais
6.
EMBO J ; 31(11): 2473-85, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22522699

RESUMO

The Drosophila adult posterior midgut has been identified as a powerful system in which to study mechanisms that control intestinal maintenance, in normal conditions as well as during injury or infection. Early work on this system has established a model of tissue turnover based on the asymmetric division of intestinal stem cells. From the quantitative analysis of clonal fate data, we show that tissue turnover involves the neutral competition of symmetrically dividing stem cells. This competition leads to stem-cell loss and replacement, resulting in neutral drift dynamics of the clonal population. As well as providing new insight into the mechanisms regulating tissue self-renewal, these findings establish intriguing parallels with the mammalian system, and confirm Drosophila as a useful model for studying adult intestinal maintenance.


Assuntos
Divisão Celular , Drosophila melanogaster/fisiologia , Homeostase/fisiologia , Intestinos/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Proliferação de Células , Drosophila melanogaster/citologia , Feminino , Intestinos/citologia , Células-Tronco/citologia
7.
Biochim Biophys Acta ; 1830(2): 2307-22, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22917651

RESUMO

BACKGROUND: Notch signaling plays a critical role in multiple developmental programs and not surprisingly, the Notch pathway has also been implicated in the regulation of many adult stem cells, such as those in the intestine, skin, lungs, hematopoietic system, and muscle. SCOPE OF REVIEW: In this review, we will first describe molecular mechanisms of Notch component modulation including recent advances in this field and introduce the fundamental principles of Notch signaling controlling cell fate decisions. We will then illustrate its important and varied functions in major stem cell model systems including: Drosophila and mammalian intestinal stem cells and mammalian skin, lung, hematopoietic and muscle stem cells. MAJOR CONCLUSIONS: The Notch receptor and its ligands are controlled by endocytic processes that regulate activation, turnover, and recycling. Glycosylation of the Notch extracellular domain has important modulatory functions on interactions with ligands and on proper receptor activity. Notch can mediate cell fate decisions including proliferation, lineage commitment, and terminal differentiation in many adult stem cell types. Certain cell fate decisions can have precise requirements for levels of Notch signaling controlled through modulatory regulation. GENERAL SIGNIFICANCE: We describe the current state of knowledge of how the Notch receptor is controlled through its interaction with ligands and how this is regulated by associated factors. The functional consequences of Notch receptor activation on cell fate decisions are discussed. We illustrate the importance of Notch's role in cell fate decisions in adult stem cells using examples from the intestine, skin, lung, blood, and muscle. This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Assuntos
Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Humanos , Transcrição Gênica
8.
Development ; 138(21): 4585-95, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21965616

RESUMO

Tight regulation of self-renewal and differentiation of adult stem cells ensures that tissues are properly maintained. In the Drosophila intestine, both commitment, i.e. exit from self-renewal, and terminal differentiation are controlled by Notch signaling. Here, we show that distinct requirements for Notch activity exist: commitment requires high Notch activity, whereas terminal differentiation can occur with lower Notch activity. We identified the gene GDP-mannose 4,6-dehydratase (Gmd), a modulator of Notch signaling, as being required for commitment but dispensable for terminal differentiation. Gmd loss resulted in aberrant, self-renewing stem cell divisions that generated extra ISC-like cells defective in Notch reporter activation, as well as wild-type-like cell divisions that produced properly terminally differentiated cells. Lowering Notch signaling using additional genetic means, we provided further evidence that commitment has a higher Notch signaling requirement than terminal differentiation. Our work suggests that a commitment requirement for high-level Notch activity safeguards the stem cells from loss through differentiation, revealing a novel role for the importance of Notch signaling levels in this system.


Assuntos
Diferenciação Celular/fisiologia , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/fisiologia , Intestinos/citologia , Receptores Notch/metabolismo , Células-Tronco/fisiologia , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Genes Reporter , Hidroliases/genética , Hidroliases/metabolismo , Intestinos/fisiologia , Mutação , Receptores Notch/genética , Transdução de Sinais/fisiologia , Células-Tronco/citologia
9.
Curr Biol ; 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38823381

RESUMO

Host-microbe interactions influence intestinal stem cell (ISC) activity to modulate epithelial turnover and composition. Here, we investigated the functional impacts of viral infection on intestinal homeostasis and the mechanisms by which viral infection alters ISC activity. We report that Drosophila A virus (DAV) infection disrupts intestinal homeostasis in Drosophila by inducing sustained ISC proliferation, resulting in intestinal dysplasia, loss of gut barrier function, and reduced lifespan. We found that additional viruses common in laboratory-reared Drosophila also promote ISC proliferation. The mechanism of DAV-induced ISC proliferation involves progenitor-autonomous epidermal growth factor receptor (EGFR) signaling, c-Jun N-terminal kinase (JNK) activity in enterocytes, and requires Sting-dependent nuclear factor κB (NF-κB) (Relish) activity. We further demonstrate that activating Sting-Relish signaling is sufficient to induce ISC proliferation, promote intestinal dysplasia, and reduce lifespan in the absence of infection. Our results reveal that viral infection can significantly disrupt intestinal physiology, highlight a novel role for Sting-Relish signaling, and support a role for viral infection in aging.

10.
STAR Protoc ; 5(1): 102851, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38354083

RESUMO

Applying mechanical forces to tissues helps to understand morphogenesis and homeostasis. Additionally, recording the dynamics of living tissues under mechanical constraints is needed to explore tissue biomechanics. Here, we present a protocol to 3D-print a StretchCo device and use it to apply uniaxial mechanical stress on the Drosophila pupal dorsal thorax epithelium. We describe steps for 3D printing, polydimethylsiloxane (PDMS) strip cutting, and glue preparation. We detail procedures for PDMS strip mounting, tissue compaction, and live imaging upon force application. For additional details on the use and execution of this protocol, please refer to Cachoux et al. (2023)1 from which the StretchCo machine has been derived.


Assuntos
Dimetilpolisiloxanos , Drosophila , Animais , Epitélio , Morfogênese , Fenômenos Biomecânicos , Estresse Mecânico
11.
Development ; 137(5): 705-14, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20147375

RESUMO

Adult stem cells maintain tissue homeostasis by controlling the proper balance of stem cell self-renewal and differentiation. The adult midgut of Drosophila contains multipotent intestinal stem cells (ISCs) that self-renew and produce differentiated progeny. Control of ISC identity and maintenance is poorly understood. Here we find that transcriptional repression of Notch target genes by a Hairless-Suppressor of Hairless complex is required for ISC maintenance, and identify genes of the Enhancer of split complex [E(spl)-C] as the major targets of this repression. In addition, we find that the bHLH transcription factor Daughterless is essential to maintain ISC identity and that bHLH binding sites promote ISC-specific enhancer activity. We propose that Daughterless-dependent bHLH activity is important for the ISC fate and that E(spl)-C factors inhibit this activity to promote differentiation.


Assuntos
Proliferação de Células , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Mucosa Intestinal/metabolismo , Células-Tronco/fisiologia , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Diferenciação Celular/genética , Drosophila/metabolismo , Drosophila/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/fisiologia , Feminino , Intestinos/fisiologia , Modelos Biológicos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia
12.
Med Sci (Paris) ; 29(1): 75-81, 2013 Jan.
Artigo em Francês | MEDLINE | ID: mdl-23351697

RESUMO

Constant renewal of cells occurs in most tissues throughout the adult lifetime and is insured by the activity of resident stem cells. Recent work has demonstrated the presence of adult stem cells in the Drosophila intestine and consequently, the Drosophila intestine has become a powerful model to understand adult stem cells in vivo. In this review, we summarize our current understanding of the mechanisms controlling cell fate decisions of the intestinal stem cells with a particular focus on the role of the Notch pathway in this process. We also summarize what is known about proliferation control of the intestinal stem cells, which is crucial to maintain tissue homeostasis during normal and environmentally stressful conditions.


Assuntos
Células-Tronco Adultas , Drosophila melanogaster/citologia , Animais , Proliferação de Células , Homeostase , Intestinos/citologia , Modelos Animais , Receptores Notch
13.
Dev Cell ; 58(24): 3048-3063.e6, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38056452

RESUMO

Tissue homeostasis relies on rewiring of stem cell transcriptional programs into those of differentiated cells. Here, we investigate changes in chromatin occurring in a bipotent adult stem cells. Combining mapping of chromatin-associated factors with statistical modeling, we identify genome-wide transitions during differentiation in the adult Drosophila intestinal stem cell (ISC) lineage. Active, stem-cell-enriched genes transition to a repressive heterochromatin protein-1-enriched state more prominently in enteroendocrine cells (EEs) than in enterocytes (ECs), in which the histone H1-enriched Black state is preeminent. In contrast, terminal differentiation genes associated with metabolic functions follow a common path from a repressive, primed, histone H1-enriched Black state in ISCs to active chromatin states in EE and EC cells. Furthermore, we find that lineage priming has an important function in adult ISCs, and we identify histone H1 as a mediator of this process. These data define underlying principles of chromatin changes during adult multipotent stem cell differentiation.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Histonas/metabolismo , Cromatina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Linhagem da Célula , Intestinos , Diferenciação Celular/genética
14.
Cell Rep ; 42(12): 113485, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38032794

RESUMO

During development and aging, genome mutation leading to loss of heterozygosity (LOH) can uncover recessive phenotypes within tissue compartments. This phenomenon occurs in normal human tissues and is prevalent in pathological genetic conditions and cancers. While studies in yeast have defined DNA repair mechanisms that can promote LOH, the predominant pathways and environmental triggers in somatic tissues of multicellular organisms are not well understood. Here, we investigate mechanisms underlying LOH in intestinal stem cells in Drosophila. Infection with the pathogenic bacteria, Erwinia carotovora carotovora 15, but not Pseudomonas entomophila, increases LOH frequency. Using whole genome sequencing of somatic LOH events, we demonstrate that they arise primarily via mitotic recombination. Molecular features and genetic evidence argue against a break-induced replication mechanism and instead support cross-over via double Holliday junction-based repair. This study provides a mechanistic understanding of mitotic recombination, an important mediator of LOH, and its effects on stem cells in vivo.


Assuntos
Drosophila , Recombinação Genética , Animais , Humanos , Drosophila/genética , Recombinação Genética/genética , Reparo do DNA , Perda de Heterozigosidade , Saccharomyces cerevisiae/genética , Células-Tronco
15.
Curr Biol ; 32(17): R931-R934, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36099901

RESUMO

Epidermal growth factor receptor signaling is central to cell proliferation, growth, and survival and is often deregulated in cancers. A new study links downstream effectors of this receptor to stem cell growth via mitochondrial biogenesis and metabolic reprogramming.


Assuntos
Biogênese de Organelas , Células-Tronco , Proliferação de Células , Receptores ErbB , Transdução de Sinais
16.
Dev Cell ; 10(2): 245-55, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16459303

RESUMO

Endocytosis of Notch receptor ligands in signaling cells is essential for Notch receptor activation. In Drosophila, the E3 ubiquitin ligase Neuralized (Neur) promotes the endocytosis and signaling activity of the ligand Delta (Dl). In this study, we identify proteins of the Bearded (Brd) family as interactors of Neur. We show that Tom, a prototypic Brd family member, inhibits Neur-dependent Notch signaling. Overexpression of Tom inhibits the endocytosis of Dl and interferes with the interaction of Dl with Neur. Deletion of the Brd gene complex results in ectopic endocytosis of Dl in dorsal cells of stage 5 embryos. This defect in Dl trafficking is associated with ectopic expression of the single-minded gene, a direct Notch target gene that specifies the mesectoderm. We propose that inhibition of Neur by Brd proteins is important for precise spatial regulation of Dl signaling.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/genética , Proteínas de Ligação a DNA/genética , Drosophila/embriologia , Drosophila/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Endocitose , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Ligantes , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Fosfoproteínas/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética
17.
Curr Opin Cell Biol ; 73: 58-68, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34217969

RESUMO

Stem cells have essential functions in the development and maintenance of our organs. Improper regulation of adult stem cells and tissue homeostasis can result in cancers and age-dependent decline. Therefore, understanding how tissue-specific stem cells can accurately renew tissues is an important aim of regenerative medicine. The Drosophila midgut harbors multipotent adult stem cells that are essential to renew the gut in homeostatic conditions and upon stress-induced regeneration. It is now a widely used model system to decipher regulatory mechanisms of stem cell biology. Here, we review recent findings on how adult intestinal stem cells differentiate, interact with their environment, and change during aging.


Assuntos
Células-Tronco Adultas , Drosophila , Animais , Drosophila melanogaster , Homeostase , Intestinos , Modelos Biológicos , Células-Tronco
18.
Artigo em Inglês | MEDLINE | ID: mdl-31932318

RESUMO

Adult stem cells fuel tissue homeostasis and regeneration through their unique ability to self-renew and differentiate into specialized cells. Thus, their DNA provides instructions that impact the tissue as a whole. Since DNA is not an inert molecule, but rather dynamic, interacting with a myriad of chemical and physical factors, it encounters damage from both endogenous and exogenous sources. Damage to DNA introduces deviations from its normal intact structure and, if left unrepaired, may result in a genetic mutation. In turn, mutant genomes of stem and progenitor cells are inherited in cells of the lineage, thus eroding the genetic information that maintains homeostasis of the somatic cell population. Errors arising in stem and progenitor cells will have a substantially larger impact on the tissue in which they reside than errors occurring in postmitotic differentiated cells. Therefore, maintaining the integrity of genomic DNA within our stem cells is essential to protect the instructions necessary for rebuilding healthy tissues during homeostatic renewal. In this review, we will first discuss DNA damage arising in stem cells and cell- and tissue-intrinsic mechanisms that protect against harmful effects of this damage. Secondly, we will examine how erroneous DNA repair and persistent DNA damage in stem and progenitor cells impact stem cells and tissues in the context of cancer initiation and aging. Finally, we will discuss the use of invertebrate and vertebrate model systems to address unanswered questions on the role that DNA damage and mutation may play in aging and precancerous conditions.


Assuntos
Envelhecimento/genética , Dano ao DNA , Genoma , Neoplasias/patologia , Células-Tronco , Animais , Humanos , Mutação
19.
Dev Cell ; 49(4): 556-573.e6, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31112698

RESUMO

Chromatin remodeling accompanies differentiation, however, its role in self-renewal is less well understood. We report that in Drosophila, the chromatin remodeler Kismet/CHD7/CHD8 limits intestinal stem cell (ISC) number and proliferation without affecting differentiation. Stem-cell-specific whole-genome profiling of Kismet revealed its enrichment at transcriptionally active regions bound by RNA polymerase II and Brahma, its recruitment to the transcription start site of activated genes and developmental enhancers and its depletion from regions bound by Polycomb, Histone H1, and heterochromatin Protein 1. We demonstrate that the Trithorax-related/MLL3/4 chromatin modifier regulates ISC proliferation, colocalizes extensively with Kismet throughout the ISC genome, and co-regulates genes in ISCs, including Cbl, a negative regulator of Epidermal Growth Factor Receptor (EGFR). Loss of kismet or trr leads to elevated levels of EGFR protein and signaling, thereby promoting ISC self-renewal. We propose that Kismet with Trr establishes a chromatin state that limits EGFR proliferative signaling, preventing tumor-like stem cell overgrowths.


Assuntos
Cromatina/metabolismo , DNA Helicases/metabolismo , Proteínas de Drosophila/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Montagem e Desmontagem da Cromatina/fisiologia , DNA Helicases/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/metabolismo , Receptores ErbB/metabolismo , Histona-Lisina N-Metiltransferase/fisiologia , Histonas/metabolismo , Proteínas de Homeodomínio/fisiologia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Receptores de Peptídeos de Invertebrados/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
20.
Curr Biol ; 12(24): 2098-110, 2002 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-12498684

RESUMO

BACKGROUND: The putative guanine nucleotide exchange factor Lte1 plays an essential role in promoting exit from mitosis at low temperatures. Lte1 is thought to activate a Ras-like signaling cascade, the mitotic exit network (MEN). MEN promotes the release of the protein phosphatase Cdc14 from the nucleolus during anaphase, and this release is a prerequisite for exit from mitosis. Lte1 is present throughout the cell during G1 but is sequestered in the bud during S phase and mitosis by an unknown mechanism. RESULTS: We show that anchorage of Lte1 in the bud requires septins, the cell polarity determinants Cdc42 and Cla4, and Kel1. Lte1 physically associates with Kel1 and requires Kel1 for its localization in the bud, suggesting a role for Kel1 in anchoring Lte1 at the bud cortex. Our data further implicate the PAK-like protein kinase Cla4 in controlling Lte1 phosphorylation and localization. CLA4 is required for Lte1 phosphorylation and bud localization. Furthermore, when overexpressed, CLA4 induces Lte1 phosphorylation and localization to regions of polarized growth. Finally, we show that Cdc14, directly or indirectly, controls Lte1 dephosphorylation and delocalization from the bud during exit from mitosis. CONCLUSION: Restriction of Lte1 to the bud cortex depends on the cortical proteins Cdc42 and Kel1 and the septin ring. Cla4 and Cdc14 promote and demote Lte1 localization at and from the bud cortex, respectively, suggesting not only that the phosphorylation status of Lte1 controls its localization but also indicating that Cla4 and Cdc14 are key regulators of the spatial asymmetry of Lte1.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas de Ciclo Celular/metabolismo , Polaridade Celular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citologia , Actinas/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Polaridade Celular/efeitos dos fármacos , Citoplasma/metabolismo , Proteínas do Citoesqueleto/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/genética , Microtúbulos/metabolismo , Mitose , Mutação , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Proteínas Tirosina Fosfatases/genética , Proteínas/genética , Proteínas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/genética , Tiazóis/farmacologia , Tiazolidinas , Proteína cdc42 de Saccharomyces cerevisiae de Ligação ao GTP/genética , Proteína cdc42 de Saccharomyces cerevisiae de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA