Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(11): e2318760121, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38442150

RESUMO

The animal foregut is the first tissue to encounter ingested food, bacteria, and viruses. We characterized the adult Drosophila foregut using transcriptomics to better understand how it triages consumed items for digestion or immune response and manages resources. Cell types were assigned and validated using GFP-tagged and Gal4 reporter lines. Foregut-associated neuroendocrine cells play a major integrative role by coordinating gut activity with nutrition, the microbiome, and circadian cycles; some express clock genes. Multiple epithelial cell types comprise the proventriculus, the central foregut organ that secretes the peritrophic matrix (PM) lining the gut. Analyzing cell types synthesizing individual PM layers revealed abundant mucin production close to enterocytes, similar to the mammalian intestinal mucosa. The esophagus and salivary gland express secreted proteins likely to line the esophageal surface, some of which may generate a foregut commensal niche housing specific gut microbiome species. Overall, our results imply that the foregut coordinates dietary sensing, hormonal regulation, and immunity in a manner that has been conserved during animal evolution.


Assuntos
Líquidos Corporais , Drosophila , Animais , Células Epiteliais , Contagem de Células , Estado Nutricional , Mamíferos
2.
Elife ; 122023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37831064

RESUMO

Highly potent animal stem cells either self renew or launch complex differentiation programs, using mechanisms that are only partly understood. Drosophila female germline stem cells (GSCs) perpetuate without change over evolutionary time and generate cystoblast daughters that develop into nurse cells and oocytes. Cystoblasts initiate differentiation by generating a transient syncytial state, the germline cyst, and by increasing pericentromeric H3K9me3 modification, actions likely to suppress transposable element activity. Relatively open GSC chromatin is further restricted by Polycomb repression of testis or somatic cell-expressed genes briefly active in early female germ cells. Subsequently, Neijre/CBP and Myc help upregulate growth and reprogram GSC metabolism by altering mitochondrial transmembrane transport, gluconeogenesis, and other processes. In all these respects GSC differentiation resembles development of the totipotent zygote. We propose that the totipotent stem cell state was shaped by the need to resist transposon activity over evolutionary timescales.


Most animals are made up of two cell types: germline stem cells, which give rise to reproductive cells (egg and sperm) and pass their DNA to the next generation, and somatic cells, which make up the rest of the body. Transposable elements ­ fragments of DNA that can copy themselves and integrate into different parts of the genome ­ can greatly disrupt the integrity of the germ cell genome. Systems involving small RNAs and DNA methylation, which respectively modify the sequence and structure of the genome, can protect germ cells from the activity of transposable elements. While these systems have been studied extensively in late germ cells, less is known about how they work in germ cells generated early on in development. To investigate, Pang et al. studied the germline stem cells that give rise to eggs in female fruit flies. Techniques that measure DNA modifications showed that these germline stem cells and the cells they give rise to early on are better protected against transposable elements. This is likely due to the unusual cell cycle of early germ cells, which display a very short initial growth phase and special DNA replication timing during the synthesis phase. Until now, the purpose of these long-known cell cycle differences between early and late germ cells was not understood. Experiments also showed known transposable element defences are upregulated before the cell division that produces reproductive cells. DNA becomes more densely packed and germ cells connect with one another, forming germline 'cysts' that allow them to share small RNAs that can suppress transposable elements. Pang et al. propose that these changes compensate for the loss of enhanced repression that occurs in the earlier stem cell stage. Very similar changes also take place in the cells generated from fertilized eggs and in mammalian reproductive cells. Further experiments investigated how these changes impact the transition from stem cell to egg cell, revealing that germline stem cells express a wide diversity of genes, including most genes whose transcripts will be stored in the mature egg later on. Another type of cell produced by germline stem cells known as nurse cells, which synthesize most of the contents of the egg, dramatically upregulate genes supporting growth. Meanwhile, 25% of genes initially expressed in germline stem cells are switched off during the transition, partly due to a mechanism called Polycomb-mediated repression. The findings advance fundamental knowledge of how germline stem cells become egg cells, and could lead to important findings in developmental biology. Furthermore, understanding that for practical applications germline stem cells do not need to retain transposable element controls designed for evolutionary time scales means that removing them may make it easier to obtain and manipulate new stem cell lines and to develop new medical therapies.


Assuntos
Proteínas de Drosophila , Células-Tronco de Oogônios , Animais , Masculino , Drosophila/genética , Cromatina/metabolismo , Células-Tronco de Oogônios/metabolismo , Proteínas de Drosophila/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular/genética , Células Germinativas/metabolismo , Expressão Gênica , Biologia , Drosophila melanogaster/metabolismo
3.
Elife ; 112022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36445738

RESUMO

Recent studies show that pre-follicular mouse oogenesis takes place in germline cysts, highly conserved groups of oogonial cells connected by intercellular bridges that develop as nurse cells as well as an oocyte. Long studied in Drosophila and insect gametogenesis, female germline cysts acquire cytoskeletal polarity and traffic centrosomes and organelles between nurse cells and the oocyte to form the Balbiani body, a conserved marker of polarity. Mouse oocyte development and nurse cell dumping are supported by dynamic, cell-specific programs of germline gene expression. High levels of perinatal germ cell death in this species primarily result from programmed nurse cell turnover after transfer rather than defective oocyte production. The striking evolutionary conservation of early oogenesis mechanisms between distant animal groups strongly suggests that gametogenesis and early embryonic development in vertebrates and invertebrates share even more in common than currently believed.


Assuntos
Drosophila , Oogênese , Feminino , Gravidez , Camundongos , Animais , Células Germinativas , Oócitos , Gametogênese
4.
Cell ; 185(14): 2576-2590.e12, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35623357

RESUMO

Mouse germline cysts, on average, develop into six oocytes supported by 24 nurse cells that transfer cytoplasm and organelles to generate a Balbiani body. We showed that between E14.5 and P5, cysts periodically activate some nurse cells to begin cytoplasmic transfer, which causes them to shrink and turnover within 2 days. Nurse cells die by a programmed cell death (PCD) pathway involving acidification, similar to Drosophila nurse cells, and only infrequently by apoptosis. Prior to initiating transfer, nurse cells co-cluster by scRNA-seq with their pro-oocyte sisters, but during their final 2 days, they cluster separately. The genes promoting oocyte development and nurse cell PCD are upregulated, whereas the genes that repress transfer, such as Tex14, and oocyte factors, such as Nobox and Lhx8, are under-expressed. The transferred nurse cell centrosomes build a cytocentrum that establishes a large microtubule aster in the primordial oocyte that organizes the Balbiani body, defining the earliest oocyte polarity.


Assuntos
Linhagem da Célula , Cistos , Oócitos , Animais , Apoptose , Crescimento Celular , Cistos/genética , Cistos/metabolismo , Citoplasma/metabolismo , Drosophila melanogaster , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Oócitos/citologia , Oócitos/metabolismo , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo
5.
Biol Open ; 10(6)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34156079

RESUMO

During oocyte differentiation in mouse fetal ovaries, sister germ cells are connected by intercellular bridges, forming germline cysts. Within the cyst, primary oocytes form via gaining cytoplasm and organelles from sister germ cells through germ cell connectivity. To uncover the role of intercellular bridges in oocyte differentiation, we analyzed mutant female mice lacking testis-expressed 14 (TEX14), a protein involved in intercellular bridge formation and stabilization. In Tex14 homozygous mutant fetal ovaries, germ cells divide to form a reduced number of cysts in which germ cells remained connected via syncytia or fragmented cell membranes, rather than normal intercellular bridges. Compared with wild-type cysts, homozygous mutant cysts fragmented at a higher frequency and produced a greatly reduced number of primary oocytes with precocious cytoplasmic enrichment and enlarged volume. By contrast, Tex14 heterozygous mutant germline cysts were less fragmented and generate primary oocytes at a reduced size. Moreover, enlarged primary oocytes in homozygous mutants were used more efficiently to sustain folliculogenesis than undersized heterozygous mutant primary oocytes. Our observations directly link the nature of fetal germline cysts to oocyte differentiation and development.


Assuntos
Cistos/embriologia , Cistos/genética , Células Germinativas/citologia , Células Germinativas/metabolismo , Mutação , Oogênese/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Homozigoto , Camundongos , Oócitos/citologia , Oócitos/metabolismo , Fatores de Transcrição/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(33): 20015-20026, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32759216

RESUMO

We sequenced more than 52,500 single cells from embryonic day 11.5 (E11.5) postembryonic day 5 (P5) gonads and performed lineage tracing to analyze primordial follicles and wave 1 medullar follicles during mouse fetal and perinatal oogenesis. Germ cells clustered into six meiotic substages, as well as dying/nurse cells. Wnt-expressing bipotential precursors already present at E11.5 are followed at each developmental stage by two groups of ovarian pregranulosa (PG) cells. One PG group, bipotential pregranulosa (BPG) cells, derives directly from bipotential precursors, expresses Foxl2 early, and associates with cysts throughout the ovary by E12.5. A second PG group, epithelial pregranulosa (EPG) cells, arises in the ovarian surface epithelium, ingresses cortically by E12.5 or earlier, expresses Lgr5, but delays robust Foxl2 expression until after birth. By E19.5, EPG cells predominate in the cortex and differentiate into granulosa cells of quiescent primordial follicles. In contrast, medullar BPG cells differentiate along a distinct pathway to become wave 1 granulosa cells. Reflecting their separate somatic cellular lineages, second wave follicles were ablated by diptheria toxin treatment of Lgr5-DTR-EGFP mice at E16.5 while first wave follicles developed normally and supported fertility. These studies provide insights into ovarian somatic cells and a resource to study the development, physiology, and evolutionary conservation of mammalian ovarian follicles.


Assuntos
Células da Granulosa/citologia , Camundongos/embriologia , Folículo Ovariano/embriologia , Animais , Diferenciação Celular , Linhagem da Célula , Feminino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Células da Granulosa/metabolismo , Camundongos/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Gravidez , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
7.
Elife ; 92020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32175841

RESUMO

Adult Drosophila Malpighian tubules have low rates of cell turnover but are vulnerable to damage caused by stones, like their mammalian counterparts, kidneys. We show that Drosophilarenal stem cells (RSCs) in the ureter and lower tubules comprise a unique, unipotent regenerative compartment. RSCs respond only to loss of nearby principal cells (PCs), cells critical for maintaining ionic balance. Large polyploid PCs are outnumbered by RSCs, which replace each lost cell with multiple PCs of lower ploidy. Notably, RSCs do not replenish principal cells or stellate cells in the upper tubules. RSCs generate daughters by asymmetric Notch signaling, yet RSCs remain quiescent (cell cycle-arrested) without damage. Nevertheless, the capacity for RSC-mediated repair extends the lifespan of flies carrying kidney stones. We propose that abundant, RSC-like stem cells exist in other tissues with low rates of turnover where they may have been mistaken for differentiated tissue cells.


Assuntos
Drosophila melanogaster/fisiologia , Túbulos de Malpighi/citologia , Células-Tronco/fisiologia , Animais , Pontos de Checagem do Ciclo Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Genótipo , Cálculos Renais , Transdução de Sinais , Ureter/fisiologia
8.
Dev Cell ; 47(1): 98-111.e5, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30220569

RESUMO

Tissue homeostasis involves a complex balance of developmental signals and environmental cues that dictate stem cell function. We found that dietary lipids control enteroendocrine cell production from Drosophila posterior midgut stem cells. Dietary cholesterol influences new intestinal cell differentiation in an Hr96-dependent manner by altering the level and duration of Notch signaling. Exogenous lipids modulate Delta ligand and Notch extracellular domain stability and alter their trafficking in endosomal vesicles. Lipid-modulated Notch signaling occurs in other nutrient-dependent tissues, suggesting that Delta trafficking in many cells is sensitive to cellular sterol levels. These diet-mediated alterations in young animals contribute to a metabolic program that persists after the diet changes. A low-sterol diet also slows the proliferation of enteroendocrine tumors initiated by Notch pathway disruption. Thus, a specific dietary nutrient can modify a key intercellular signaling pathway to shift stem cell differentiation and cause lasting changes in tissue structure and physiology.


Assuntos
Colesterol na Dieta/efeitos adversos , Lipídeos/fisiologia , Receptores Notch/efeitos dos fármacos , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Colesterol/metabolismo , Colesterol na Dieta/metabolismo , Proteínas de Drosophila/efeitos dos fármacos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Enterócitos/metabolismo , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/fisiologia , Intestinos/citologia , Peptídeos e Proteínas de Sinalização Intracelular , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Proteínas de Membrana , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Esteróis/metabolismo
9.
PLoS One ; 11(3): e0151251, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26958853

RESUMO

Tissue integrity and homeostasis often rely on the proliferation of stem cells or differentiated cells to replace lost, aged, or damaged cells. Recently, we described an alternative source of cell replacement- the expansion of resident, non-dividing diploid cells by wound-induced polyploidization (WIP). Here we show that the magnitude of WIP is proportional to the extent of cell loss using a new semi-automated assay with single cell resolution. Hippo and JNK signaling regulate WIP; unexpectedly however, JNK signaling through AP-1 limits rather than stimulates the level of Yki activation and polyploidization in the Drosophila epidermis. We found that polyploidization also quantitatively compensates for cell loss in a mammalian tissue, mouse corneal endothelium, where increased cell death occurs with age in a mouse model of Fuchs Endothelial Corneal Dystrophy (FECD). Our results suggest that WIP is an evolutionarily conserved homeostatic mechanism that maintains the size and synthetic capacity of adult tissues.


Assuntos
Proteínas de Drosophila/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Distrofias Hereditárias da Córnea/metabolismo , Modelos Animais de Doenças , Drosophila , Endotélio Corneano/metabolismo , Feminino , Mamíferos , Camundongos , Camundongos Mutantes , Poliploidia , Fator de Transcrição AP-1/metabolismo
10.
Science ; 352(6281): 95-9, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26917595

RESUMO

Oocytes differentiate in diverse species by receiving organelles and cytoplasm from sister germ cells while joined in germline cysts or syncytia. Mouse primordial germ cells form germline cysts, but the role of cysts in oogenesis is unknown. We find that mouse germ cells receive organelles from neighboring cyst cells and build a Balbiani body to become oocytes, whereas nurselike germ cells die. Organelle movement, Balbiani body formation, and oocyte fate determination are selectively blocked by low levels of microtubule-dependent transport inhibitors. Membrane breakdown within the cyst and an apoptosis-like process are associated with organelle transfer into the oocyte, events reminiscent of nurse cell dumping in Drosophila We propose that cytoplasmic and organelle transport plays an evolutionarily conserved and functionally important role in mammalian oocyte differentiation.


Assuntos
Células Gigantes/citologia , Oócitos/citologia , Oogênese , Organelas/fisiologia , Animais , Apoptose , Evolução Biológica , Citoplasma/fisiologia , Citoplasma/ultraestrutura , Feminino , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/fisiologia
11.
Cell ; 164(3): 420-32, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26824655

RESUMO

Reproduction is heavily influenced by nutrition and metabolic state. Many common reproductive disorders in humans are associated with diabetes and metabolic syndrome. We characterized the metabolic mechanisms that support oogenesis and found that mitochondria in mature Drosophila oocytes enter a low-activity state of respiratory quiescence by remodeling the electron transport chain (ETC). This shift in mitochondrial function leads to extensive glycogen accumulation late in oogenesis and is required for the developmental competence of the oocyte. Decreased insulin signaling initiates ETC remodeling and mitochondrial respiratory quiescence through glycogen synthase kinase 3 (GSK3). Intriguingly, we observed similar ETC remodeling and glycogen uptake in maturing Xenopus oocytes, suggesting that these processes are evolutionarily conserved aspects of oocyte development. Our studies reveal an important link between metabolism and oocyte maturation.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio/metabolismo , Oogênese , Xenopus laevis/embriologia , Animais , Drosophila melanogaster/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Feminino , Fatores de Transcrição Forkhead/metabolismo , Mitocôndrias/metabolismo , Proteína Oncogênica v-akt/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Xenopus laevis/metabolismo
12.
Elife ; 42015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25824290

RESUMO

Here, we document a collection of ∼7434 MiMIC (Minos Mediated Integration Cassette) insertions of which 2854 are inserted in coding introns. They allowed us to create a library of 400 GFP-tagged genes. We show that 72% of internally tagged proteins are functional, and that more than 90% can be imaged in unfixed tissues. Moreover, the tagged mRNAs can be knocked down by RNAi against GFP (iGFPi), and the tagged proteins can be efficiently knocked down by deGradFP technology. The phenotypes associated with RNA and protein knockdown typically correspond to severe loss of function or null mutant phenotypes. Finally, we demonstrate reversible, spatial, and temporal knockdown of tagged proteins in larvae and adult flies. This new strategy and collection of strains allows unprecedented in vivo manipulations in flies for many genes. These strategies will likely extend to vertebrates.


Assuntos
Elementos de DNA Transponíveis/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Biblioteca Gênica , Mutagênese Insercional , Interferência de RNA , Animais , Animais Geneticamente Modificados , Western Blotting , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Larva/genética , Larva/metabolismo , Aprendizagem/fisiologia , Microscopia Confocal , Fatores de Tempo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , alfa Catenina/genética , alfa Catenina/metabolismo
13.
PLoS Genet ; 11(2): e1004989, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25695427

RESUMO

Ovulation is critical for successful reproduction and correlates with ovarian cancer risk, yet genetic studies of ovulation have been limited. It has long been thought that the mechanism controlling ovulation is highly divergent due to speciation and fast evolution. Using genetic tools available in Drosophila, we now report that ovulation in Drosophila strongly resembles mammalian ovulation at both the cellular and molecular levels. Just one of up to 32 mature follicles per ovary pair loses posterior follicle cells ("trimming") and protrudes into the oviduct, showing that a selection process prefigures ovulation. Follicle cells that remain after egg release form a "corpus luteum (CL)" at the end of the ovariole, develop yellowish pigmentation, and express genes encoding steroid hormone biosynthetic enzymes that are required for full fertility. Finally, matrix metalloproteinase 2 (Mmp2), a type of protease thought to facilitate mammalian ovulation, is expressed in mature follicle and CL cells. Mmp2 activity is genetically required for trimming, ovulation and CL formation. Our studies provide new insights into the regulation of Drosophila ovulation and establish Drosophila as a model for genetically investigating ovulation in diverse organisms, including mammals.


Assuntos
Corpo Lúteo/crescimento & desenvolvimento , Drosophila melanogaster/crescimento & desenvolvimento , Metaloproteinase 2 da Matriz/genética , Ovulação/genética , Animais , Drosophila melanogaster/genética , Feminino , Fertilidade/genética , Mamíferos/genética , Mamíferos/crescimento & desenvolvimento , Oócitos/crescimento & desenvolvimento , Folículo Ovariano/crescimento & desenvolvimento , Útero/crescimento & desenvolvimento
14.
Genes Dev ; 28(16): 1840-55, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25128500

RESUMO

DNA replication remains unfinished in many Drosophila polyploid cells, which harbor disproportionately fewer copies of late-replicating chromosomal regions. By analyzing paired-end high-throughput sequence data from polytene larval salivary gland cells, we define 112 underreplicated (UR) euchromatic regions 60-480 kb in size. To determine the effects of underreplication on genome integrity, we analyzed anomalous read pairs and breakpoint reads throughout the euchromatic genome. Each UR euchromatic region contains many different deletions 10-500 kb in size, while very few deletions are present in fully replicated chromosome regions or UR zones from embryo DNA. Thus, during endocycles, stalled forks within UR regions break and undergo local repair instead of remaining stable and generating nested forks. As a result, each salivary gland cell contains hundreds of unique deletions that account for their copy number reductions. Similar UR regions and deletions were observed in ovarian DNA, suggesting that incomplete replication, fork breakage, and repair occur widely in polytene cells. UR regions are enriched in genes encoding immunoglobulin superfamily proteins and contain many neurally expressed and homeotic genes. We suggest that the extensive somatic DNA instability described here underlies position effect variegation, molds the structure of polytene chromosomes, and should be investigated for possible functions.


Assuntos
Replicação do DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Cromossomos Politênicos/genética , Glândulas Salivares , Animais , DNA/genética , Quebras de DNA , Reparo do DNA , Feminino , Instabilidade Genômica , Imunoglobulinas/genética , Larva , Ovário , Deleção de Sequência/genética
15.
Curr Biol ; 23(22): 2224-2232, 2013 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-24184101

RESUMO

BACKGROUND: Reestablishing epithelial integrity and biosynthetic capacity is critically important following tissue damage. The adult Drosophila abdominal epithelium provides an attractive new system to address how postmitotic diploid cells contribute to repair. RESULTS: Puncture wounds to the adult Drosophila epidermis close initially by forming a melanized scab. We found that epithelial cells near the wound site fuse to form a giant syncytium, which sends lamellae under the scab to re-epithelialize the damaged site. Other large cells arise more peripherally by initiating endocycles and becoming polyploid, or by cell fusion. Rac GTPase activity is needed for syncytium formation, while the Hippo signaling effector Yorkie modulates both polyploidization and cell fusion. Large cell formation is functionally important because when both polyploidization and fusion are blocked, wounds do not re-epithelialize. CONCLUSIONS: Our observations indicate that cell mass lost upon wounding can be replaced by polyploidization instead of mitotic proliferation. We propose that large cells generated by polyploidization or cell fusion are essential because they are better able than diploid cells to mechanically stabilize wounds, especially those containing permanent acellular structures, such as scar tissue.


Assuntos
Fusão Celular , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Proteínas Nucleares/metabolismo , Poliploidia , Transativadores/metabolismo , Cicatrização/fisiologia , Traumatismos Abdominais/patologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Ciclo Celular , Ciclina E/genética , Ciclina E/metabolismo , Drosophila/citologia , Proteínas de Drosophila/genética , Células Epiteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/genética , Proteínas de Sinalização YAP , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo
16.
Elife ; 2: e00886, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23991285

RESUMO

The Drosophila midgut is maintained throughout its length by superficially similar, multipotent intestinal stem cells that generate new enterocytes and enteroendocrine cells in response to tissue requirements. We found that the midgut shows striking regional differentiation along its anterior-posterior axis. At least ten distinct subregions differ in cell morphology, physiology and the expression of hundreds of genes with likely tissue functions. Stem cells also vary regionally in behavior and gene expression, suggesting that they contribute to midgut sub-specialization. Clonal analyses showed that stem cells generate progeny located outside their own subregion at only one of six borders tested, suggesting that midgut subregions resemble cellular compartments involved in tissue development. Tumors generated by disrupting Notch signaling arose preferentially in three subregions and tumor cells also appeared to respect regional borders. Thus, apparently similar intestinal stem cells differ regionally in cell production, gene expression and in the ability to spawn tumors. DOI:http://dx.doi.org/10.7554/eLife.00886.001.


Assuntos
Compartimento Celular , Drosophila/fisiologia , Células-Tronco/citologia , Animais , Drosophila/citologia
17.
Development ; 140(10): 2075-81, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23578925

RESUMO

Mammalian germ cells divide mitotically and form nests of associated cells just prior to entering meiosis. At least some nests contain germline cysts that arise by synchronous, incomplete mitotic divisions, but others may form by aggregation. To systematically investigate early murine germ cell development, we lineage marked the progeny of individual, newly arrived primordial germ cells in the E10.5 gonad. All the marked germ cells initially develop into clones containing two, four or eight cells, indicating cyst formation. Surprisingly, growing cysts in both sexes partially fragment into smaller cysts prior to completion and associate with cysts from unrelated progenitors. At the time divisions cease, female clones comprise five cysts on average that eventually give rise to about six primordial follicles. Male cyst cells break apart and probably become spermatogonial stem cells. Thus, cysts are invariant units of mouse germ cell development and cyst fragmentation provides insight into the amplification of spermatogonial stem cells and the origin of primordial follicles.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Meiose , Animais , Apoptose , Linhagem da Célula , Proliferação de Células , Cruzamentos Genéticos , Biologia do Desenvolvimento/métodos , Feminino , Masculino , Camundongos , Oócitos/metabolismo , Espermatogênese , Fatores de Tempo
18.
PLoS One ; 7(10): e46109, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056242

RESUMO

Drosophila adult females but not males contain high levels of the steroid hormone ecdysone, however, the roles played by steroid signaling during Drosophila gametogenesis remain poorly understood. Drosophila germ cells in both sexes initially follow a similar pathway. After germline stem cells are established, their daughters form interconnected cysts surrounded by somatic escort (female) or cyst (male) cells and enter meiosis. Subsequently, female cysts acquire a new covering of somatic cells to form follicles. Knocking down expression of the heterodimeric ecdysteroid receptor (EcR/Usp) or the E75 early response gene in escort cells disrupts 16-cell cyst production, meiotic entry and follicle formation. Escort cells lose their squamous morphology and unsheath germ cells. By contrast, disrupting ecdysone signaling in males does not perturb cyst development or ensheathment. Thus, sex-specific steroid signaling is essential for female germ cell development at the time male and female pathways diverge. Our results suggest that steroid signaling plays an important sex-specific role in early germ cell development in Drosophila, a strategy that may be conserved in mammals.


Assuntos
Drosophila/metabolismo , Ecdisona/metabolismo , Células Germinativas/crescimento & desenvolvimento , Meiose , Transdução de Sinais , Animais , Animais Geneticamente Modificados , Apoptose/genética , Forma Celular/genética , Drosophila/genética , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Feminino , Células Germinativas/metabolismo , Células Germinativas/ultraestrutura , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Microscopia Confocal , Microscopia Eletrônica , Mutação , Oogênese/genética , Ovário/citologia , Ovário/metabolismo , Interferência de RNA , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo
19.
J Pathol ; 225(2): 161-2, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21792940

RESUMO

Advances in stem cell research highlight the importance of analysing multicellular interactions in vivo before modelling them in cell culture systems. Gain-of-function assays such as transplantation are useful, but are not equivalent to studying cells in their natural, undisturbed microenvironment.


Assuntos
Células-Tronco/citologia , Urotélio/citologia , Humanos
20.
Development ; 138(11): 2207-15, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21558370

RESUMO

The Drosophila ovariole tip produces new ovarian follicles on a 12-hour cycle by controlling niche-based germline and follicle stem cell divisions and nurturing their developing daughters. Static images provide a thumbnail view of folliculogenesis but imperfectly capture the dynamic cellular interactions that underlie follicle production. We describe a live-imaging culture system that supports normal ovarian stem cell activity, cyst movement and intercellular interaction over 14 hours, which is long enough to visualize all the steps of follicle generation. Our results show that live imaging has unique potential to address diverse aspects of stem cell biology and gametogenesis. Stem cells in cultured tissue respond to insulin and orient their mitotic spindles. Somatic escort cells, the glial-like partners of early germ cells, do not adhere to and migrate along with germline stem cell daughters as previously proposed. Instead, dynamic, microtubule-rich cell membranes pass cysts from one escort cell to the next. Additionally, escort cells are not replenished by the regular division of escort stem cells as previously suggested. Rather, escort cells remain quiescent and divide only to maintain a constant germ cell:escort cell ratio.


Assuntos
Drosophila/embriologia , Oócitos/fisiologia , Células-Tronco/fisiologia , Imagem com Lapso de Tempo/métodos , Animais , Membrana Celular/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Drosophila/citologia , Drosophila/fisiologia , Feminino , Insulina/farmacologia , Microscopia de Fluorescência , Oócitos/citologia , Folículo Ovariano/embriologia , Ovário/embriologia , Ovário/fisiologia , Fuso Acromático/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA