Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 84(4): 727-743.e8, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38325378

RESUMEN

Lysosomes are central to metabolic homeostasis. The microphthalmia bHLH-LZ transcription factors (MiT/TFEs) family members MITF, TFEB, and TFE3 promote the transcription of lysosomal and autophagic genes and are often deregulated in cancer. Here, we show that the GATOR2 complex, an activator of the metabolic regulator TORC1, maintains lysosomal function by protecting MiT/TFEs from proteasomal degradation independent of TORC1, GATOR1, and the RAG GTPase. We determine that in GATOR2 knockout HeLa cells, members of the MiT/TFEs family are ubiquitylated by a trio of E3 ligases and are degraded, resulting in lysosome dysfunction. Additionally, we demonstrate that GATOR2 protects MiT/TFE proteins in pancreatic ductal adenocarcinoma and Xp11 translocation renal cell carcinoma, two cancers that are driven by MiT/TFE hyperactivation. In summary, we find that the GATOR2 complex has independent roles in TORC1 regulation and MiT/TFE protein protection and thus is central to coordinating cellular metabolism with control of the lysosomal-autophagic system.


Asunto(s)
Neoplasias Renales , Factor de Transcripción Asociado a Microftalmía , Humanos , Células HeLa , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteolisis , Autofagia/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas/metabolismo , Neoplasias Renales/metabolismo , Lisosomas/genética , Lisosomas/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo
2.
Dev Cell ; 58(21): 2249-2260.e9, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37647895

RESUMEN

In multicellular lives, the differentiation of stem cells and progenitor cells is often accompanied by a transition from glycolysis to mitochondrial oxidative phosphorylation (OXPHOS). However, the underlying mechanism of this metabolic transition remains largely unknown. In this study, we investigate the role of mechanical stress in activating OXPHOS during differentiation of the female germline cyst in Drosophila. We demonstrate that the surrounding somatic cells flatten the 16-cell differentiating cyst, resulting in an increase of the membrane tension of germ cells inside the cyst. This mechanical stress is necessary to maintain cytosolic Ca2+ concentration in germ cells through a mechanically activated channel, transmembrane channel-like. The sustained cytosolic Ca2+ triggers a CaMKI-Fray-JNK signaling relay, leading to the transcriptional activation of OXPHOS in differentiating cysts. Our findings demonstrate a molecular link between cell mechanics and mitochondrial energy metabolism, with implications for other developmentally orchestrated metabolic transitions in mammals.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Células Germinativas/metabolismo , Metabolismo Energético , Diferenciación Celular , Proteínas de Drosophila/metabolismo , Mamíferos/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(1): e2212330120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36577058

RESUMEN

Target of Rapamycin Complex I (TORC1) is a central regulator of metabolism in eukaryotes that responds to a wide array of negative and positive inputs. The GTPase-activating protein toward Rags (GATOR) signaling pathway acts upstream of TORC1 and is comprised of two subcomplexes. The trimeric GATOR1 complex inhibits TORC1 activity in response to amino acid limitation by serving as a GTPase-activating protein (GAP) for the TORC1 activator RagA/B, a component of the lysosomally located Rag GTPase. The multi-protein GATOR2 complex inhibits the activity of GATOR1 and thus promotes TORC1 activation. Here we report that Wdr59, originally assigned to the GATOR2 complex based on studies performed in tissue culture cells, unexpectedly has a dual function in TORC1 regulation in Drosophila. We find that in the ovary and the eye imaginal disc brain complex, Wdr59 inhibits TORC1 activity by opposing the GATOR2-dependent inhibition of GATOR1. Conversely, in the Drosophila fat body, Wdr59 promotes the accumulation of the GATOR2 component Mio and is required for TORC1 activation. Similarly, in mammalian HeLa cells, Wdr59 prevents the proteolytic destruction of GATOR2 proteins Mio and Wdr24. Consistent with the reduced levels of the TORC1-activating GATOR2 complex, Wdr59KOs HeLa cells have reduced TORC1 activity which is restored along with GATOR2 protein levels upon proteasome inhibition. Taken together, our data support the model that the Wdr59 component of the GATOR2 complex functions to promote or inhibit TORC1 activity depending on cellular context.


Asunto(s)
Proteínas de Drosophila , Drosophila , Complejos Multiproteicos , Proteína Fosfatasa 2 , Animales , Femenino , Humanos , Anticuerpos , Drosophila/metabolismo , Proteínas Activadoras de GTPasa , Células HeLa , Mamíferos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas de Drosophila/metabolismo
4.
Cell Death Dis ; 12(6): 571, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078879

RESUMEN

Target of Rapamycin Complex 1 (TORC1) is a master regulator that coordinates nutrient status with cell metabolism. The GTPase-activating protein towards Rags complex 1 (GATOR1) inhibits TORC1 activity and protects cells from damage during periods of stress. Here we characterize multiple pathways that regulate the expression of the GATOR1 component Nprl3 in Drosophila. We determine that the stability of Nprl3 is impacted by the Unassembled Soluble Complex Proteins Degradation (USPD) pathway. In addition, we find that FK506 binding protein 39 (FKBP39)-dependent proteolytic destruction maintains Nprl3 at low levels in nutrient replete conditions. Nutrient starvation abrogates the degradation of the Nprl3 protein and rapidly promotes Nprl3 accumulation. Consistent with a role in promoting the stability of a TORC1 inhibitor, mutations in fkbp39 decrease TORC1 activity and increase autophagy. Finally, we show that the 5'UTR of nprl3 transcripts contain a functional upstream open reading frame (uORF) that inhibits main ORF translation. In summary, our work has uncovered novel mechanisms of Nprl3 regulation and identifies an important role for FKBP39 in the control of cellular metabolism.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Transcripción/metabolismo , Animales , Autofagia , Drosophila , Transfección
5.
Dev Cell ; 55(3): 272-288.e5, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32898476

RESUMEN

The dysregulation of the metabolic regulator TOR complex I (TORC1) contributes to a wide array of human pathologies. Tuberous sclerosis complex (TSC) is a potent inhibitor of TORC1. Here, we demonstrate that the Rag GTPase acts in both the amino-acid-sensing and growth factor signaling pathways to control TORC1 activity through the regulation of TSC dynamics in HeLa cells and Drosophila. We find that TSC lysosomal-cytosolic exchange increases in response to both amino acid and growth factor restriction. Moreover, the rate of exchange mirrors TSC function, with depletions of the Rag GTPase blocking TSC lysosomal mobility and rescuing TORC1 activity. Finally, we show that the GATOR2 complex controls the phosphorylation of TSC2, which is essential for TSC exchange. Our data support the model that the amino acid and growth factor signaling pathways converge on the Rag GTPase to inhibit TORC1 activity through the regulation of TSC dynamics.


Asunto(s)
Aminoácidos/deficiencia , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Proteínas de Unión al GTP Monoméricas/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Drosophila , Femenino , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Lisosomas/metabolismo , Mutación/genética , Ovario/metabolismo , Fosforilación , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
6.
Aging (Albany NY) ; 11(21): 9811-9828, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31712450

RESUMEN

Aging and age-related diseases occur in almost all organisms. Recently, it was discovered that the inhibition of target of rapamycin complex 1 (TORC1), a conserved complex that mediates nutrient status and cell metabolism, can extend an individual's lifespan and inhibit age-related diseases in many model organisms. However, the mechanism whereby TORC1 affects aging remains elusive. Here, we use a loss-of-function mutation in nprl2, a component of GATOR1 that mediates amino acid levels and inhibits TORC1 activity, to investigate the effect of increased TORC1 activity on the occurrence of age-related digestive dysfunction in Drosophila. We found that the nprl2 mutation decreased Drosophila lifespan. Furthermore, the nprl2 mutant had a distended crop, with food accumulation at an early age. Interestingly, the inappropriate food distribution and digestion along with decreased crop contraction in nprl2 mutant can be rescued by decreasing TORC1 activity. In addition, nprl2-mutant flies exhibited age-related phenotypes in the midgut, including short gut length, a high rate of intestinal stem cell proliferation, and metabolic dysfunction, which could be rescued by inhibiting TORC1 activity. Our findings showed that the gastrointestinal tract aging process is accelerated in nprl2-mutant flies, owing to high TORC1 activity, which suggested that TORC1 promotes digestive tract senescence.


Asunto(s)
Envejecimiento/fisiología , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Motilidad Gastrointestinal , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas Portadoras/genética , Digestión , Proteínas de Drosophila/genética , Masculino , Proteínas Supresoras de Tumor/genética
7.
Elife ; 82019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31650955

RESUMEN

The TORC1 regulator GATOR1/SEACIT controls meiotic entry and early meiotic events in yeast. However, how metabolic pathways influence meiotic progression in metazoans remains poorly understood. Here we examine the role of the TORC1 regulators GATOR1 and GATOR2 in the response to meiotic double-stranded breaks (DSB) during Drosophila oogenesis. We find that in mutants of the GATOR2 component mio, meiotic DSBs trigger the constitutive downregulation of TORC1 activity and a permanent arrest in oocyte growth. Conversely, in GATOR1 mutants, high TORC1 activity results in the delayed repair of meiotic DSBs and the hyperactivation of p53. Unexpectedly, we found that GATOR1 inhibits retrotransposon expression in the presence of meiotic DSBs in a pathway that functions in parallel to p53. Thus, our studies have revealed a link between oocyte metabolism, the repair of meiotic DSBs and retrotransposon expression.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Meiosis , Complejos Multiproteicos/metabolismo , Oogénesis/fisiología , Animales , Regulación de la Expresión Génica , Mapas de Interacción de Proteínas
8.
PLoS Genet ; 15(7): e1008253, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31291240

RESUMEN

Endoreplication is a cell cycle variant that entails cell growth and periodic genome duplication without cell division, and results in large, polyploid cells. Cells switch from mitotic cycles to endoreplication cycles during development, and also in response to conditional stimuli during wound healing, regeneration, aging, and cancer. In this study, we use integrated approaches in Drosophila to determine how mitotic cycles are remodeled into endoreplication cycles, and how similar this remodeling is between induced and developmental endoreplicating cells (iECs and devECs). Our evidence suggests that Cyclin A / CDK directly activates the Myb-MuvB (MMB) complex to induce transcription of a battery of genes required for mitosis, and that repression of CDK activity dampens this MMB mitotic transcriptome to promote endoreplication in both iECs and devECs. iECs and devECs differed, however, in that devECs had reduced expression of E2F1-dependent genes that function in S phase, whereas repression of the MMB transcriptome in iECs was sufficient to induce endoreplication without a reduction in S phase gene expression. Among the MMB regulated genes, knockdown of AurB protein and other subunits of the chromosomal passenger complex (CPC) induced endoreplication, as did knockdown of CPC-regulated cytokinetic, but not kinetochore, proteins. Together, our results indicate that the status of a CycA-Myb-MuvB-AurB network determines the decision to commit to mitosis or switch to endoreplication in both iECs and devECs, and suggest that regulation of different steps of this network may explain the known diversity of polyploid cycle types in development and disease.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Endorreduplicación , Animales , Aurora Quinasa B/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina A/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Femenino , Perfilación de la Expresión Génica , Mitosis , Poliploidía , Proteínas Proto-Oncogénicas c-myb/metabolismo
9.
G3 (Bethesda) ; 6(12): 3859-3867, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27672113

RESUMEN

TORC1 regulates metabolism and growth in response to a large array of upstream inputs. The evolutionarily conserved trimeric GATOR1 complex inhibits TORC1 activity in response to amino acid limitation. In humans, the GATOR1 complex has been implicated in a wide array of pathologies including cancer and hereditary forms of epilepsy. However, the precise role of GATOR1 in animal physiology remains largely undefined. Here, we characterize null mutants of the GATOR1 components nprl2, nprl3, and iml1 in Drosophila melanogaster We demonstrate that all three mutants have inappropriately high baseline levels of TORC1 activity and decreased adult viability. Consistent with increased TORC1 activity, GATOR1 mutants exhibit a cell autonomous increase in cell growth. Notably, escaper nprl2 and nprl3 mutant adults have a profound locomotion defect. In line with a nonautonomous role in the regulation of systemic metabolism, expressing the Nprl3 protein in the fat body, a nutrient storage organ, and hemocytes but not muscles and neurons rescues the motility of nprl3 mutants. Finally, we show that nprl2 and nprl3 mutants fail to activate autophagy in response to amino acid limitation and are extremely sensitive to both amino acid and complete starvation. Thus, in Drosophila, in addition to maintaining baseline levels of TORC1 activity, the GATOR1 complex has retained a critical role in the response to nutrient stress. In summary, the TORC1 inhibitor GATOR1 contributes to multiple aspects of the development and physiology of Drosophila.


Asunto(s)
Drosophila melanogaster/fisiología , Metabolismo Energético , Homeostasis , Complejos Multiproteicos/metabolismo , Estrés Fisiológico , Animales , Animales Modificados Genéticamente , Autofagia , Sistemas CRISPR-Cas , Proliferación Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Genes Letales , Locomoción , Diana Mecanicista del Complejo 1 de la Rapamicina , Unión Proteica , ARN Guía de Kinetoplastida , Eliminación de Secuencia , Serina-Treonina Quinasas TOR/metabolismo
10.
PLoS Genet ; 12(5): e1006036, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27166823

RESUMEN

TORC1 is a master regulator of metabolism in eukaryotes that responds to multiple upstream signaling pathways. The GATOR complex is a newly defined upstream regulator of TORC1 that contains two sub-complexes, GATOR1, which inhibits TORC1 activity in response to amino acid starvation and GATOR2, which opposes the activity of GATOR1. While the GATOR1 complex has been implicated in a wide array of human pathologies including cancer and hereditary forms of epilepsy, the in vivo relevance of the GATOR2 complex remains poorly understood in metazoans. Here we define the in vivo role of the GATOR2 component Wdr24 in Drosophila. Using a combination of genetic, biochemical, and cell biological techniques we demonstrate that Wdr24 has both TORC1 dependent and independent functions in the regulation of cellular metabolism. Through the characterization of a null allele, we show that Wdr24 is a critical effector of the GATOR2 complex that promotes the robust activation of TORC1 and cellular growth in a broad array of Drosophila tissues. Additionally, epistasis analysis between wdr24 and genes that encode components of the GATOR1 complex revealed that Wdr24 has a second critical function, the TORC1 independent regulation of lysosome dynamics and autophagic flux. Notably, we find that two additional members of the GATOR2 complex, Mio and Seh1, also have a TORC1 independent role in the regulation of lysosome function. These findings represent a surprising and previously unrecognized function of GATOR2 complex components in the regulation of lysosomes. Consistent with our findings in Drosophila, through the characterization of a wdr24-/- knockout HeLa cell line we determined that Wdr24 promotes lysosome acidification and autophagic flux in mammalian cells. Taken together our data support the model that Wdr24 is a key effector of the GATOR2 complex, required for both TORC1 activation and the TORC1 independent regulation of lysosomes.


Asunto(s)
Proteínas de Drosophila/genética , Lisosomas/genética , Complejos Multiproteicos/genética , Proteínas Nucleares/genética , Proteínas/genética , Serina-Treonina Quinasas TOR/genética , Animales , Proteínas de Ciclo Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Epistasis Genética , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(52): E5670-7, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512509

RESUMEN

In single-cell eukaryotes the pathways that monitor nutrient availability are central to initiating the meiotic program and gametogenesis. In Saccharomyces cerevisiae an essential step in the transition to the meiotic cycle is the down-regulation of the nutrient-sensitive target of rapamycin complex 1 (TORC1) by the increased minichromosome loss 1/ GTPase-activating proteins toward Rags 1 (Iml1/GATOR1) complex in response to amino acid starvation. How metabolic inputs influence early meiotic progression and gametogenesis remains poorly understood in metazoans. Here we define opposing functions for the TORC1 regulatory complexes Iml1/GATOR1 and GATOR2 during Drosophila oogenesis. We demonstrate that, as is observed in yeast, the Iml1/GATOR1 complex inhibits TORC1 activity to slow cellular metabolism and drive the mitotic/meiotic transition in developing ovarian cysts. In iml1 germline depletions, ovarian cysts undergo an extra mitotic division before meiotic entry. The TORC1 inhibitor rapamycin can suppress this extra mitotic division. Thus, high TORC1 activity delays the mitotic/meiotic transition. Conversely, mutations in Tor, which encodes the catalytic subunit of the TORC1 complex, result in premature meiotic entry. Later in oogenesis, the GATOR2 components Mio and Seh1 are required to oppose Iml1/GATOR1 activity to prevent the constitutive inhibition of TORC1 and a block to oocyte growth and development. To our knowledge, these studies represent the first examination of the regulatory relationship between the Iml1/GATOR1 and GATOR2 complexes within the context of a multicellular organism. Our data imply that the central role of the Iml1/GATOR1 complex in the regulation of TORC1 activity in the early meiotic cycle has been conserved from single cell to multicellular organisms.


Asunto(s)
Proteínas de Drosophila/metabolismo , Meiosis/fisiología , Oocitos/metabolismo , Oogénesis/fisiología , Factores de Transcripción/metabolismo , Animales , Antibacterianos/farmacología , Proteínas de Ciclo Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Meiosis/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/citología , Oogénesis/efectos de los fármacos , Sirolimus/farmacología , Factores de Transcripción/genética
12.
Dev Cell ; 26(4): 329-30, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23987509

RESUMEN

Polycomb-group proteins silence gene expression through epigenetic modification of chromatin. In this issue of Developmental Cell, Iovino et al. (2013) demonstrate that Polycomb repressive complex 2 (PRC2) is required for maintenance of oocyte fate by repressing expression of two critical targets, Cyclin E and dacapo, during the early meiotic cycle.


Asunto(s)
Linaje de la Célula/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Genes cdc , N-Metiltransferasa de Histona-Lisina/metabolismo , Oocitos/citología , Proteínas Represoras/metabolismo , Animales , Femenino
13.
Development ; 138(10): 2133-42, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21521741

RESUMEN

The nuclear pore complex (NPC) mediates the transport of macromolecules between the nucleus and cytoplasm. Recent evidence indicates that structural nucleoporins, the building blocks of the NPC, have a variety of unanticipated cellular functions. Here, we report an unexpected tissue-specific requirement for the structural nucleoporin Seh1 during Drosophila oogenesis. Seh1 is a component of the Nup107-160 complex, the major structural subcomplex of the NPC. We demonstrate that Seh1 associates with the product of the missing oocyte (mio) gene. In Drosophila, mio regulates nuclear architecture and meiotic progression in early ovarian cysts. Like mio, seh1 has a crucial germline function during oogenesis. In both mio and seh1 mutant ovaries, a fraction of oocytes fail to maintain the meiotic cycle and develop as pseudo-nurse cells. Moreover, the accumulation of Mio protein is greatly diminished in the seh1 mutant background. Surprisingly, our characterization of a seh1 null allele indicates that, although required in the female germline, seh1 is dispensable for the development of somatic tissues. Our work represents the first examination of seh1 function within the context of a multicellular organism. In summary, our studies demonstrate that Mio is a novel interacting partner of the conserved nucleoporin Seh1 and add to the growing body of evidence that structural nucleoporins can have novel tissue-specific roles.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Oogénesis/fisiología , Animales , Secuencia de Bases , Proteínas de Ciclo Celular , Cartilla de ADN/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Genes de Insecto , Complejos Multiproteicos , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Oogénesis/genética , Interferencia de ARN , Serina-Treonina Quinasas TOR/metabolismo , Distribución Tisular
14.
Cell Cycle ; 8(10): 1501-9, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19372757

RESUMEN

Development of a fertilized egg into an adult human requires trillions of cell divisions, the vast majority of which duplicate their genome once and only once. Nevertheless, trophoblast giant cells and megakaryocytes in mammals circumvent this rule by duplicating their genome multiple times without undergoing cell division, a process generally referred to as 'endoreduplication'. In contrast, arthropods such as Drosophila endoreduplicate their genome in most larval tissues, as well as in many adult tissues. Endoreduplication requires that cells prevent entrance into or completion of mitosis and cytokinesis under conditions that permit assembly of prereplication complexes. In addition, cells must prevent induction of apoptosis in response to incomplete DNA replication or DNA damage that may occur during the ensuing sequence of 'endocycles'. Thus, developmentally regulated endoreduplication results in terminal cell differentiation. Recent progress has revealed both differences and similarities in the mechanisms employed by flies and mammals to change from mitotic cell cycles to 'endocycles'. The critical step, however, appears to be switching from a CDK-dependent form of the anaphase promoting complex (APC) to one that functions only in the absence of CDK activity.


Asunto(s)
Ciclo Celular , Replicación del ADN , Poliploidía , Animales , Apoptosis , Humanos , Mitosis
15.
Development ; 135(8): 1451-61, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18321983

RESUMEN

The endocycle is a commonly observed variant cell cycle in which cells undergo repeated rounds of DNA replication with no intervening mitosis. How the cell cycle machinery is modified to transform a mitotic cycle into endocycle has long been a matter of interest. In both plants and animals, the transition from the mitotic cycle to the endocycle requires Fzr/Cdh1, a positive regulator of the Anaphase-Promoting Complex/Cyclosome (APC/C). However, because many of its targets are transcriptionally downregulated upon entry into the endocycle, it remains unclear whether the APC/C functions beyond the mitotic/endocycle boundary. Here, we report that APC/C Fzr/Cdh1 activity is required to promote the G/S oscillation of the Drosophila endocycle. We demonstrate that compromising APC/C activity, after cells have entered the endocycle, inhibits DNA replication and results in the accumulation of multiple APC/C targets, including the mitotic cyclins and Geminin. Notably, our data suggest that the activity of APC/C Fzr/Cdh1 during the endocycle is not continuous but is cyclic, as demonstrated by the APC/C-dependent oscillation of the pre-replication complex component Orc1. Taken together, our data suggest a model in which the cyclic activity of APC/C Fzr/Cdh1 during the Drosophila endocycle is driven by the periodic inhibition of Fzr/Cdh1 by Cyclin E/Cdk2. We propose that, as is observed in mitotic cycles, during endocycles, APC/C Fzr/Cdh1 functions to reduce the levels of the mitotic cyclins and Geminin in order to facilitate the relicensing of DNA replication origins and cell cycle progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/citología , Drosophila/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Animales Modificados Genéticamente , Proteínas Cdh1 , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Replicación del ADN , Regulación hacia Abajo , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Genes de Insecto , Mitosis , Modelos Biológicos , Complejo de Reconocimiento del Origen/genética , Complejo de Reconocimiento del Origen/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/genética
16.
EMBO J ; 26(8): 2071-82, 2007 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-17380129

RESUMEN

The endocycle is a developmentally programmed variant cell cycle in which cells undergo repeated rounds of DNA replication with no intervening mitosis. In Drosophila, the endocycle is driven by the oscillations of Cyclin E/Cdk2 activity. How the periodicity of Cyclin E/Cdk2 activity is achieved during endocycles is poorly understood. Here, we demonstrate that the p21(cip1)/p27(kip1)/p57(kip2)-like cyclin-dependent kinase inhibitor (CKI), Dacapo (Dap), promotes replication licensing during Drosophila endocycles by reinforcing low Cdk activity during the endocycle Gap-phase. In dap mutants, cells in the endocycle have reduced levels of the licensing factor Double Parked/Cdt1 (Dup/Cdt1), as well as decreased levels of chromatin-bound minichromosome maintenance (MCM2-7) complex. Moreover, mutations in dup/cdt1 dominantly enhance the dap phenotype in several polyploid cell types. Consistent with a reduced ability to complete genomic replication, dap mutants accumulate increased levels of DNA damage during the endocycle S-phase. Finally, genetic interaction studies suggest that dap functions to promote replication licensing in a subset of Drosophila mitotic cycles.


Asunto(s)
Ciclo Celular/fisiología , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Replicación del ADN/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Animales , Bromodesoxiuridina , Proteínas de Drosophila/genética , Citometría de Flujo , Inmunohistoquímica , Microscopía Electrónica de Rastreo , Mutación/genética , Proteínas Nucleares/genética , Células Fotorreceptoras de Invertebrados/ultraestructura
17.
Dev Cell ; 10(1): 127-35, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16399084

RESUMEN

Animal oocytes undergo a highly conserved developmental arrest in prophase of meiosis I. The maintenance of the prophase I arrest requires the silencing of Cdk1 activity. Drosophila oocytes inhibit the accumulation of the mitotic cyclins, the activating subunits of Cdk1, via a poorly defined posttranscriptional mechanism. Here, we demonstrate that the translational repressor Bruno binds the 3' UTR and inhibits the translation of the mitotic cyclin Cyclin A during prophase of meiosis I. In the absence of Bruno, ovarian cysts enter meiosis but rapidly accumulate high levels of mitotic cyclins and return to the mitotic cycle. Based on our results, we propose a model in which Bruno and the anaphase-promoting complex/cyclosome act together to restrict the accumulation of the mitotic cyclins, and thus Cdk1 activity, during the prophase I arrest of the Drosophila oocyte.


Asunto(s)
Ciclinas/metabolismo , Proteínas de Drosophila/fisiología , Profase Meiótica I/fisiología , Oocitos/fisiología , Proteínas de Unión al ARN/fisiología , Regiones no Traducidas 3'/fisiología , Animales , Animales Modificados Genéticamente , Northern Blotting/métodos , Western Blotting/métodos , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina A/metabolismo , Ciclina B/metabolismo , Ciclinas/clasificación , Drosophila , Proteínas de Drosophila/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Indoles , Ovario/fisiología , Unión Proteica/fisiología , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Tubulina (Proteína)/metabolismo
18.
Oncogene ; 24(17): 2765-75, 2005 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-15838513

RESUMEN

During metazoan development, the organization of the cell cycle is often modified in response to developmental signals. The endocycle provides a dramatic example of this phenomenon. In the endocycle, also referred to as the endoreplicative cycle, cells undergo successive rounds of DNA replication without an intervening mitosis. Often the endocycle is used to expand the genome of a group of specialized cells that are highly biosynthetically active. In these circumstances, large polyploid cells are produced in organisms that are primarily comprised of diploid cells. However, many organisms achieve growth by increasing cell size, rather than cell number. This strategy is more generally exploited in insects and plants. For instance, in the insect Drosophila melanogaster, the majority of the larval tissues, as well as many adult tissues, enter the endocycle and become polyploid. Therefore, Drosophila has been a rich source for studies on endocycle regulation. Recent work from Drosophila is beginning to reveal how developmental signals promote the transition from the mitotic cycle to the endocycle, as well as what drives endocycle progression. In addition, studies on the endocycle have provided insight into the regulatory principles underlying the once per cell cycle replication of the genome, as well as the relationship between S phase and mitosis.


Asunto(s)
Ciclo Celular , Drosophila/citología , Animales , Cromatina/química , Cromatina/fisiología , Ciclinas/fisiología , Replicación del ADN , Drosophila/genética , Proteínas de Drosophila , Proteínas de la Membrana/metabolismo , Receptores Notch , Origen de Réplica , Transducción de Señal
19.
Proc Natl Acad Sci U S A ; 102(12): 4482-7, 2005 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-15767569

RESUMEN

The synaptonemal complex (SC) is intimately involved in the process of meiotic recombination in most organisms, but its exact role remains enigmatic. One reason for this uncertainty is that the overall structure of the SC is evolutionarily conserved, but many SC proteins are not. Two putative SC proteins have been identified in Drosophila: C(3)G and C(2)M. Mutations in either gene cause defects in SC structure and meiotic recombination. Although neither gene is well conserved at the amino acid level, the predicted secondary structure of C(3)G is similar to that of transversefilament proteins, and C(2)M is a distantly related member of the alpha-kleisin family that includes Rec8, a meiosis-specific cohesin protein. Here, we use immunogold labeling of SCs in Drosophila ovaries to localize C(3)G and C(2)M at the EM level. We show that both C(3)G and C(2)M are components of the SC, that the orientation of C(3)G within the SC is similar to other transverse-filament proteins, and that the N terminus of C(2)M is located in the central region adjacent to the lateral elements (LEs). Based on our data and the known phenotypes of C(2)M and C(3)G mutants, we propose a model of SC structure in which C(2)M links C(3)G to the LEs.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Complejo Sinaptonémico/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Ciclo Celular/química , Drosophila/ultraestructura , Proteínas de Drosophila/química , Femenino , Genes de Insecto , Microscopía Inmunoelectrónica , Modelos Biológicos , Mutación , Fenotipo , Complejo Sinaptonémico/química , Complejo Sinaptonémico/ultraestructura
20.
Development ; 132(6): 1165-74, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15703281

RESUMEN

Cyclins regulate progression through the cell cycle. Control of cyclin levels is essential in Drosophila oogenesis for the four synchronous divisions that generate the 16 cell germ line cyst and for ensuring that one cell in each cyst, the oocyte, is arrested in meiosis, while the remaining fifteen cells become polyploid nurse cells. Changes in cyclin levels could be achieved by regulating transcription, translation or protein stability. The proteasome limits cyclin protein levels in the Drosophila ovary, but the mechanisms regulating RNA turnover or translation remain largely unclear. Here, we report the identification of twin, a homolog of the yeast CCR4 deadenylase. We show that twin is important for the number and synchrony of cyst divisions and oocyte fate. Consistent with the deadenylase activity of CCR4 in yeast, our data suggest that Twin controls germ line cyst development by regulating poly(A) tail lengths of several targets including Cyclin A (CycA) RNA. twin mutants exhibit very low expression of Bag-of-marbles (Bam), a regulator of cyst division, indicating that Twin/Ccr4 activity is necessary for wild-type Bam expression. Lowering the levels of CycA or increasing the levels of Bam suppresses the defects we observe in twin ovaries, implicating CycA and Bam as downstream effectors of Twin. We propose that Twin/Ccr4 functions during early oogenesis to coordinate cyst division, oocyte fate specification and egg chamber maturation.


Asunto(s)
Adenosina/metabolismo , Ciclinas/biosíntesis , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Oogénesis/fisiología , Polímeros/metabolismo , Ribonucleasas/metabolismo , Secuencia de Aminoácidos , Animales , Ciclinas/genética , Drosophila/genética , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Femenino , Regulación de la Expresión Génica , Datos de Secuencia Molecular , Oogénesis/genética , ARN Mensajero/metabolismo , Ribonucleasas/genética , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...