Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 890
Filtrar
1.
Mol Biol Cell ; 35(6): ar77, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38598296

RESUMO

In favorable conditions, eukaryotic cells proceed irreversibly through the cell division cycle (G1-S-G2-M) in order to produce two daughter cells with the same number and identity of chromosomes of their progenitor. The integrity of this process is maintained by "checkpoints" that hold a cell at particular transition points of the cycle until all requisite events are completed. The crucial functions of these checkpoints seem to depend on irreversible bistability of the underlying checkpoint control systems. Bistability of cell cycle transitions has been confirmed experimentally in frog egg extracts, budding yeast cells and mammalian cells. For fission yeast cells, a recent paper by Patterson et al. (2021) provides experimental evidence for an abrupt transition from G2 phase into mitosis, and we show that these data are consistent with a stochastic model of a bistable switch governing the G2/M checkpoint. Interestingly, our model suggests that their experimental data could also be explained by a reversible/sigmoidal switch, and stochastic simulations confirm this supposition. We propose a simple modification of their experimental protocol that could provide convincing evidence for (or against) bistability of the G2/M transition in fission yeast.


Assuntos
Mitose , Schizosaccharomyces , Schizosaccharomyces/metabolismo , Mitose/fisiologia , Ciclo Celular/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular , Fase G2/fisiologia , Proteínas de Schizosaccharomyces pombe/metabolismo
2.
Int J Mol Sci ; 23(4)2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35216269

RESUMO

Atherosclerosis is a multicausal disease characterized by the formation of cholesterol-containing plaque in the pronounced intima nearest to the heart's elastic-type arteries that have high levels of blood circulation. Plaques are formed due to arterial pressure-induced damage to the endothelium in areas of turbulent blood flow. It is found in the majority of the Western population, including young people. This denies the monogenic mechanism of atherogenesis. In 1988, Orekhov et al. and Kawai et al. discovered that the presence of atherogenic (modified, including oxidized ones) LDLs is necessary for atherogenesis. On the basis of our discovery, suggesting that the overloading of enterocytes with lipids could lead to the formation of modified LDLs, we proposed a new hypothesis explaining the main factors of atherogenesis. Indeed, when endothelial cells are damaged and then pass through the G2 phase of their cell cycle they secrete proteins into their basement membrane. This leads to thickening of the basement membrane and increases its affinity to LDL especially for modified ones. When the enterocyte transcytosis pathway is overloaded with fat, very large chylomicrons are formed, which have few sialic acids, circulate in the blood for a long time, undergo oxidation, and can induce the production of autoantibodies. It is the sialic acids that shield the short forks of the polysaccharide chains to which autoantibodies are produced. Here, these data are evaluated from the point of view of our new model.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fase G2/fisiologia , Humanos , Lipoproteínas LDL/metabolismo , Oxirredução , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Transcitose/fisiologia
3.
Development ; 148(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34370012

RESUMO

Drosophila female germline stem cells (GSCs) are found inside the cellular niche at the tip of the ovary. They undergo asymmetric divisions to renew the stem cell lineage and to produce sibling cystoblasts that will in turn enter differentiation. GSCs and cystoblasts contain spectrosomes, membranous structures essential for orientation of the mitotic spindle and that, particularly in GSCs, change shape depending on the cell cycle phase. Using live imaging and a fusion protein of GFP and the spectrosome component Par-1, we follow the complete spectrosome cycle throughout GSC division and quantify the relative duration of the different spectrosome shapes. We also determine that the Par-1 kinase shuttles between the spectrosome and the cytoplasm during mitosis and observe the continuous addition of new material to the GSC and cystoblast spectrosomes. Next, we use the Fly-FUCCI tool to define, in live and fixed tissues, that GSCs have a shorter G1 compared with the G2 phase. The observation of centrosomes in dividing GSCs allowed us to determine that centrosomes separate very early in G1, before centriole duplication. Furthermore, we show that the anterior centrosome associates with the spectrosome only during mitosis and that, upon mitotic spindle assembly, it translocates to the cell cortex, where it remains anchored until centrosome separation. Finally, we demonstrate that the asymmetric division of GSCs is not an intrinsic property of these cells, as the spectrosome of GSC-like cells located outside of the niche can divide symmetrically. Thus, GSCs display unique properties during division, a behaviour influenced by the surrounding niche.


Assuntos
Divisão Celular Assimétrica/fisiologia , Centrossomo/fisiologia , Drosophila/fisiologia , Células Germinativas/fisiologia , Ovário/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Centrossomo/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Feminino , Fase G1/fisiologia , Fase G2/fisiologia , Células Germinativas/metabolismo , Mitose/fisiologia , Ovário/metabolismo , Fuso Acromático/fisiologia , Células-Tronco/metabolismo
4.
Mol Biol Cell ; 32(20): br2, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34260268

RESUMO

Heterotrimeric G proteins (αßγ) function at the cytoplasmic surface of a cell's plasma membrane to transduce extracellular signals into cellular responses. However, numerous studies indicate that G proteins also play noncanonical roles at unique intracellular locations. Previous work has established that G protein ßγ subunits (Gßγ) regulate a signaling pathway on the cytoplasmic surface of Golgi membranes that controls the exit of select protein cargo. Now, we demonstrate a novel role for Gßγ in regulating mitotic Golgi fragmentation, a key checkpoint of the cell cycle that occurs in the late G2 phase. We show that small interfering RNA-mediated depletion of Gß1 and Gß2 in synchronized cells causes a decrease in the number of cells with fragmented Golgi in late G2 and a delay of entry into mitosis and progression through G2/M. We also demonstrate that during G2/M Gßγ acts upstream of protein kinase D and regulates the phosphorylation of the Golgi structural protein GRASP55. Expression of Golgi-targeted GRK2ct, a Gßγ-sequestering protein used to inhibit Gßγ signaling, also causes a decrease in Golgi fragmentation and a delay in mitotic progression. These results highlight a novel role for Gßγ in regulation of Golgi structure.


Assuntos
Pontos de Checagem da Fase G2 do Ciclo Celular/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Complexo de Golgi/fisiologia , Ciclo Celular/fisiologia , Membrana Celular/metabolismo , Fase G2/fisiologia , Complexo de Golgi/metabolismo , Proteínas da Matriz do Complexo de Golgi/metabolismo , Células HeLa , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Mitose/fisiologia , Fosforilação , Proteína Quinase C/metabolismo , Transporte Proteico/fisiologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia
5.
J Ethnopharmacol ; 269: 113686, 2021 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-33309918

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: The Menispermaceae plant Tiliacora racemosa is immensely popular in Indian traditional Ayurvedic medicine as "Krishnavetra" for its remarkable anti-cancerous property, and is commonly used by tribal population for the treatment of skin infections, snake bites and filariasis. AIM OF THE STUDY: This present study intends to identify the modus operandi behind the cytotoxic activity of Tiliacora racemosa leaves in cervical cancer cells SiHa. Focus has been instilled in the ability of the plant extract to target multiple signaling pathways leading to cell cycle arrest and cell death in SiHa cells, followed by a pharmacological characterization to identify the bioactive principle. MATERIALS AND METHODS: T. racemosa leaves extracted in methanol, ethyl acetate, hexane and aqueous solvent were screened for cytotoxicity in HeLa, SiHa, C33A (cervical cancer cells) and HEK cells by MTT assay. SiHa cells were treated with the most potent extract (TRM). Cellular morphology, clonogenic and wound healing potential, presence of intracellular ROS and NO, lipid peroxidation, activity of cellular antioxidants (SOD, CAT, GSH), DNA damage detection by comet assay and localisation of γ-H2AX foci, intracellular expression of PARP-1, Bax/Bcl2 and caspase-3, loss in mitochondrial membrane potential by JC1 (flow cytometry) and Rh123 (microscopy), cell cycle analysis, Annexin-FITC assay, AO/EtBr microscopy and apoptotic proteome profiling were undertaken in the treated cells. All the related proteins were studied by immunoblots. Effect of NAC (ROS-scavenger) on cell viability, DNA damage and apoptosis were studied. Phytochemical characterization of all TR extracts was followed by LC-MS analysis of TRM and isolated alkaloid of TR was assessed for cytotoxicity. RESULTS: The methanol extract of T. racemosa (TRM) rich in bisbenzylisoquinoline and other alkaloids impeded the proliferation of cervical cancer cells SiHa in vitro through disruption of cellular redox homeostasis caused by increase in cellular ROS and NO with concomitant decrease in the cellular antioxidants. Double-stranded DNA damage was noted from γH2AX foci accumulation and Parp-1 activation leading to ATM-Chk2-p53 pathway arresting the cells at G2/M-phase through cyclin B1 inhibition. The mitochondrial membrane potential was also disturbed leading to caspase-3 dependent apoptotic induction by both extrinsic and intrinsic pathway. Immunoblots show TRM also inhibited PI3K/Akt and NFκB pathway. NAC pre-treatment rescued the cell viability proving DNA damage and apoptosis to be direct consequences of ROS overproduction. Lastly, the therapeutic potential of T. racemosa is was hypothesized to be possibly derived from its alkaloid content. CONCLUSION: This study proves the age old ethnnopharmacological anticancer role of T. racemosa. The leaf extracts inhibited the anomalous proliferation of SiHa cells by virtue of G2/M-phase cell cycle arrest and apoptotic cell death. Oxidative stress mediated double stranded DNA damage paved the way towards apoptotic cell death through multiple routes, including PI3K/Akt/NFκB pathway. The abundant alkaloid content of T. racemosa was denoted as the probable responsible cytotoxic principle.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Menispermaceae , Estresse Oxidativo/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Divisão Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Dano ao DNA/fisiologia , Relação Dose-Resposta a Droga , Feminino , Fase G2/fisiologia , Células HEK293 , Células HeLa , Humanos , Estresse Oxidativo/fisiologia , Folhas de Planta , Neoplasias do Colo do Útero/tratamento farmacológico
6.
Cell Cycle ; 19(21): 2897-2905, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33043808

RESUMO

The precise control of the cell cycle G2 phase to Mitosis (M phase) transition is central for cell fate determination. The commonly used methods for assessing G2 to M phase progression are based on synchronizing cells and involve perturbation of the natural cell cycle progression. Additionally, these methods are often time-consuming and labor-intensive. Here, we report a flow cytometry-based method that offers a kinetic analysis of G2 to M phase progression in asynchronous cells using nocodazole, 5-Ethynyl-2´-deoxyuridine staining, and histone H3 serine 28 phosphorylation (pH3) staining. Nocodazole is used to collect mitotic cells and prevent their progression into G1, at the same time EdU is added for use as a dump channel during analysis. The remaining cells can then be identified as either G1 or G2/M based on their DNA content. Finally, G2 and M phase cells can be separated based on a mitotic marker, phosphorylation of ser28 on histone H3. While developed to assay G2/M phase progression, this method also resolves G1/S phase progression with no additional steps other than analysis. Compared to double thymidine block, this method does not require extended pre-treatments and is compatible with a greater variety of cell lines, while at the same time offering enhanced consistency and temporal resolution.


Assuntos
Citometria de Fluxo/métodos , Fase G2/fisiologia , Mitose/fisiologia , Ciclo Celular/fisiologia , Linhagem Celular , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Histonas/metabolismo , Humanos , Cinética , Nocodazol/metabolismo , Fosforilação/fisiologia
7.
FEMS Yeast Res ; 20(4)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401321

RESUMO

Pinostrobin, a flavonoid compound known for its diverse pharmacological actions, including anti-leukemic and anti-inflammatory activities, has been repeatedly isolated by various screenings, but its action mechanism is still obscure. Previously, pinostrobin was rediscovered in our laboratory using a yeast-based assay procedure devised specifically for the inhibitory effect on the activated Ca2+ signaling that leads the cells to severe growth retardation in the G2 phase. Here, we attempted to identify target of pinostrobin employing the genetic techniques available in the yeast. Using various genetically engineered yeast strains in which the Ca2+-signaling cascade can be activated by the controlled expression of the various signaling molecules of the cascade, its target was narrowed down to Swe1, the cell-cycle regulatory protein kinase. The Swe1 kinase is situated at the downstream of the Ca2+-signaling cascade and downregulates the Cdc28/Clb complex by phosphorylating the Cdc28 moiety of the complex in the G2 phase. We further demonstrated that pinostrobin inhibits the protein kinase activity of Swe1 in vivo as estimated by the decreased level of Cdc28 phosphorylation at Tyr-19. Since the yeast SWE1 gene is an ortholog for the human WEE1 gene, our finding implied a potentiality of pinostrobin as the G2 checkpoint abrogator in cancer chemotherapy.


Assuntos
Cálcio/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Flavanonas/farmacologia , Fase G2/genética , Regulação Fúngica da Expressão Gênica , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Ciclo Celular/genética , Fase G2/fisiologia , Genes Fúngicos , Proteínas Tirosina Quinases/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais/efeitos dos fármacos
8.
Mol Cell Biol ; 40(8)2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-31964753

RESUMO

Cdk2-dependent TopBP1-treslin interaction is critical for DNA replication initiation. However, it remains unclear how this association is terminated after replication initiation is finished. Here, we demonstrate that phosphorylation of TopBP1 by Akt coincides with cyclin A activation during S and G2 phases and switches the TopBP1-interacting partner from treslin to E2F1, which results in the termination of replication initiation. Premature activation of Akt in G1 phase causes an early switch and inhibits DNA replication. TopBP1 is often overexpressed in cancer and can bypass control by Cdk2 to interact with treslin, leading to enhanced DNA replication. Consistent with this notion, reducing the levels of TopBP1 in cancer cells restores sensitivity to a Cdk2 inhibitor. Together, our study links Cdk2 and Akt pathways to the control of DNA replication through the regulation of TopBP1-treslin interaction. These data also suggest an important role for TopBP1 in driving abnormal DNA replication in cancer.


Assuntos
Proteínas de Transporte/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Pontos de Checagem do Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Linhagem Celular , Quinase 2 Dependente de Ciclina/genética , Ciclinas/genética , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Fase G2/fisiologia , Humanos , Proteínas Nucleares/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Fase S/fisiologia
9.
Mol Biol Cell ; 31(8): 725-740, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31995441

RESUMO

E2F8 is a transcriptional repressor that antagonizes E2F1 at the crossroads of the cell cycle, apoptosis, and cancer. Previously, we discovered that E2F8 is a direct target of the APC/C ubiquitin ligase. Nevertheless, it remains unknown how E2F8 is dynamically controlled throughout the entirety of the cell cycle. Here, using newly developed human cell-free systems that recapitulate distinct inter-mitotic and G1 phases and a continuous transition from prometaphase to G1, we reveal an interlocking dephosphorylation switch coordinating E2F8 degradation with mitotic exit and the activation of APC/CCdh1. Further, we uncover differential proteolysis rates for E2F8 at different points within G1 phase, accounting for its accumulation in late G1 while APC/CCdh1 is still active. Finally, we demonstrate that the F-box protein Cyclin F regulates E2F8 in G2-phase. Altogether, our data define E2F8 regulation throughout the cell cycle, illuminating an extensive coordination between phosphorylation, ubiquitination and transcription in mammalian cell cycle.


Assuntos
Ciclo Celular/fisiologia , Proteínas Repressoras/metabolismo , Motivos de Aminoácidos , Subunidade Apc1 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Sistema Livre de Células , Ciclinas/metabolismo , Fator de Transcrição E2F1/metabolismo , Fase G1/fisiologia , Fase G2/fisiologia , Células HeLa , Humanos , Mitose/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteólise , Proteínas Recombinantes/metabolismo , Ubiquitinação
10.
Nanomedicine (Lond) ; 14(22): 2941-2955, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31755353

RESUMO

Aim: We aimed to characterize the tumor-targeting and radiosensitization properties of the photo-responsive gold nanoparticles (AuNPs) decorated photolabile diazirine group and folic acid for improved radiotherapy and computed tomography imaging of tumors. Methods: Folic acid and photolabile diazirine group were covalently conjugated on the surface of AuNPs to afford the desired photo-responsive dAuNP-FA (AuNPs capped with poly(ethylene) glycol ligands bearing photolabile diazirine group and folic acid). The probes were intravenously injected into tumor-bearing mice followed by photocrosslinking upon 405 nm laser irradiation for radiotherapy and computed tomography imaging of tumors in vivo. Results: Light-triggered crosslinking of AuNPs in vivo remarkably enhanced the accumulation and retention of AuNPs within tumors. Conclusion: We have successfully developed a novel photo-responsive Au particle-based tumor theranostic probe showing remarkably improved tumor targeting ability and radiosensitization effect.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Radiossensibilizantes/química , Tomografia Computadorizada por Raios X/métodos , Animais , Divisão Celular/genética , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Ácido Fólico/química , Fase G2/genética , Fase G2/fisiologia , Humanos , Camundongos , Polietilenoglicóis/química
11.
EMBO J ; 38(20): e101430, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31475738

RESUMO

E2F7 and E2F8 act as tumor suppressors via transcriptional repression of genes involved in S-phase entry and progression. Previously, we demonstrated that these atypical E2Fs are degraded by APC/CCdh1 during G1 phase of the cell cycle. However, the mechanism driving the downregulation of atypical E2Fs during G2 phase is unknown. Here, we show that E2F7 is targeted for degradation by the E3 ubiquitin ligase SCFcyclin F during G2. Cyclin F binds via its cyclin domain to a conserved C-terminal CY motif on E2F7. An E2F7 mutant unable to interact with SCFcyclin F remains stable during G2. Furthermore, SCFcyclin F can also interact and induce degradation of E2F8. However, this does not require the cyclin domain of SCFcyclin F nor the CY motifs in the C-terminus of E2F8, implying a different regulatory mechanism than for E2F7. Importantly, depletion of cyclin F causes an atypical-E2F-dependent delay of the G2/M transition, accompanied by reduced expression of E2F target genes involved in DNA repair. Live cell imaging of DNA damage revealed that cyclin F-dependent regulation of atypical E2Fs is critical for efficient DNA repair and cell cycle progression.


Assuntos
Ciclinas/metabolismo , Reparo do DNA , Fator de Transcrição E2F7/metabolismo , Fase G2/fisiologia , Proteólise , Proteínas Repressoras/metabolismo , Pontos de Checagem do Ciclo Celular , Ciclinas/genética , Dano ao DNA , Replicação do DNA , Fator de Transcrição E2F7/genética , Células HeLa , Humanos , Ligação Proteica , Proteínas Repressoras/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
13.
Dev Growth Differ ; 61(6): 357-364, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31199000

RESUMO

Primordial germ cells (PGCs) are reprogrammed into pluripotent embryonic germ cells (EGCs) under specific culture conditions, but the detailed mechanisms of PGC reprogramming have not yet been fully clarified. Previous studies have demonstrated that AKT, an important intracellular signaling molecule, promotes reprogramming of PGCs into EGCs. Because AKT likely inhibits p53 functions to enhance PGC reprogramming, and p53 negatively regulates cell cycle progression, we analyzed cell cycle changes in PGCs following AKT activation and found that the ratio of PGCs in the G1/G0 phase was decreased while that of PGCs in the G2/M phase was increased after AKT activation. We also showed that the expression of the CDK inhibitor p27kip1, which prevents the G1­S transition and is transcriptionally activated by p53, was significantly downregulated by AKT activation. The results suggested that the characteristic cell cycle changes of PGCs by AKT activation are, at least in part, due to decreased expression of p27kip1 . We also investigated changes in histone H3K27 tri-methylation (H3K27me3) by AKT activation in PGCs, because we previously found that decreased H3K27me3 was involved in PGC reprogramming via upregulation of cyclin D1. We observed that AKT activation in PGCs resulted in H3K27 hypomethylation. In addition, DZNeP, an inhibitor of the H3K27 trimethyl transferase Ezh2, stimulated EGC formation. These results together suggested that AKT activation promotes G1-S transition and downregulates H3K27me3 to enhance PGC reprogramming.


Assuntos
Reprogramação Celular/fisiologia , Ciclina D1/metabolismo , Células Germinativas Embrionárias/citologia , Células Germinativas Embrionárias/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fase G1 , Fase G2 , Histonas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Ativação Enzimática , Fase G1/fisiologia , Fase G2/fisiologia , Masculino , Metilação , Camundongos , Camundongos Transgênicos , Transdução de Sinais
14.
Dev Cell ; 49(2): 293-300.e3, 2019 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-30905769

RESUMO

Quiescent neural stem cells (NSCs) in the adult brain are regenerative cells that could be activated therapeutically to repair damage. It is becoming apparent that quiescent NSCs exhibit heterogeneity in their propensity for activation and in the progeny that they generate. We discovered recently that NSCs undergo quiescence in either G0 or G2 in the Drosophila brain, challenging the notion that all quiescent stem cells are G0 arrested. We found that G2-quiescent NSCs become activated prior to G0 NSCs. Here, we show that the cyclin-dependent kinase inhibitor Dacapo (Dap; ortholog of p57KIP2) determines whether NSCs enter G0 or G2 quiescence during embryogenesis. We demonstrate that the dorsal patterning factor, Muscle segment homeobox (Msh; ortholog of MSX1/2/3) binds directly to the Dap locus and induces Dap expression in dorsal NSCs, resulting in G0 arrest, while more ventral NSCs undergo G2 quiescence. Our results reveal region-specific regulation of stem cell quiescence.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Proteínas de Drosophila/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Fase G2/fisiologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fase de Repouso do Ciclo Celular/fisiologia
15.
J Cell Biol ; 218(3): 871-894, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30674580

RESUMO

Hook proteins are evolutionarily conserved dynein adaptors that promote assembly of highly processive dynein-dynactin motor complexes. Mammals express three Hook paralogs, namely Hook1, Hook2, and Hook3, that have distinct subcellular localizations and expectedly, distinct cellular functions. Here we demonstrate that Hook2 binds to and promotes dynein-dynactin assembly specifically during mitosis. During the late G2 phase, Hook2 mediates dynein-dynactin localization at the nuclear envelope (NE), which is required for centrosome anchoring to the NE. Independent of its binding to dynein, Hook2 regulates microtubule nucleation at the centrosome; accordingly, Hook2-depleted cells have reduced astral microtubules and spindle positioning defects. Besides the centrosome, Hook2 localizes to and recruits dynactin and dynein to the central spindle. Dynactin-dependent targeting of centralspindlin complex to the midzone is abrogated upon Hook2 depletion; accordingly, Hook2 depletion results in cytokinesis failure. We find that the zebrafish Hook2 homologue promotes dynein-dynactin association and was essential for zebrafish early development. Together, these results suggest that Hook2 mediates assembly of the dynein-dynactin complex and regulates mitotic progression and cytokinesis.


Assuntos
Citocinese/fisiologia , Fase G2/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Centrômero/genética , Centrômero/metabolismo , Dineínas/genética , Dineínas/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Membrana Nuclear/genética , Fuso Acromático/genética , Fuso Acromático/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
16.
J Biol Chem ; 294(3): 968-980, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30459232

RESUMO

The centromere is an evolutionarily conserved eukaryotic protein machinery essential for precision segregation of the parental genome into two daughter cells during mitosis. Centromere protein A (CENP-A) organizes the functional centromere via a constitutive centromere-associated network composing the CENP-T complex. However, how CENP-T assembles onto the centromere remains elusive. Here we show that CENP-T binds directly to Holliday junction recognition protein (HJURP), an evolutionarily conserved chaperone involved in loading CENP-A. The binding interface of HJURP was mapped to the C terminus of CENP-T. Depletion of HJURP by CRISPR-elicited knockout minimized recruitment of CENP-T to the centromere, indicating the importance of HJURP in CEPN-T loading. Our immunofluorescence analyses indicate that HJURP recruits CENP-T to the centromere in S/G2 phase during the cell division cycle. Significantly, the HJURP binding-deficient mutant CENP-T6L failed to locate to the centromere. Importantly, CENP-T insufficiency resulted in chromosome misalignment, in particular chromosomes 15 and 18. Taken together, these data define a novel molecular mechanism underlying the assembly of CENP-T onto the centromere by a temporally regulated HJURP-CENP-T interaction.


Assuntos
Proteína Centromérica A/metabolismo , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fase G2/fisiologia , Fase S/fisiologia , Centrômero/genética , Proteína Centromérica A/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Células HEK293 , Células HeLa , Humanos
17.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30487274

RESUMO

Vesicular stomatitis virus (VSV) (a rhabdovirus) and its variant VSV-ΔM51 are widely used model systems to study mechanisms of virus-host interactions. Here, we investigated how the cell cycle affects replication of these viruses using an array of cell lines with different levels of impairment of antiviral signaling and a panel of chemical compounds arresting the cell cycle at different phases. We observed that all compounds inducing cell cycle arrest in G2/M phase strongly enhanced the replication of VSV-ΔM51 in cells with functional antiviral signaling. G2/M arrest strongly inhibited type I and type III interferon (IFN) production as well as expression of IFN-stimulated genes in response to exogenously added IFN. Moreover, G2/M arrest enhanced the replication of Sendai virus (a paramyxovirus), which is also highly sensitive to the type I IFN response but did not stimulate the replication of a wild-type VSV that is more effective at evading antiviral responses. In contrast, the positive effect of G2/M arrest on virus replication was not observed in cells defective in IFN signaling. Altogether, our data show that replication of IFN-sensitive cytoplasmic viruses can be strongly stimulated during G2/M phase as a result of inhibition of antiviral gene expression, likely due to mitotic inhibition of transcription, a global repression of cellular transcription during G2/M phase. The G2/M phase thus could represent an "Achilles' heel" of the infected cell, a phase when the cell is inadequately protected. This model could explain at least one of the reasons why many viruses have been shown to induce G2/M arrest.IMPORTANCE Vesicular stomatitis virus (VSV) (a rhabdovirus) and its variant VSV-ΔM51 are widely used model systems to study mechanisms of virus-host interactions. Here, we investigated how the cell cycle affects replication of VSV and VSV-ΔM51. We show that G2/M cell cycle arrest strongly enhances the replication of VSV-ΔM51 (but not of wild-type VSV) and Sendai virus (a paramyxovirus) via inhibition of antiviral gene expression, likely due to mitotic inhibition of transcription, a global repression of cellular transcription during G2/M phase. Our data suggest that the G2/M phase could represent an "Achilles' heel" of the infected cell, a phase when the cell is inadequately protected. This model could explain at least one of the reasons why many viruses have been shown to induce G2/M arrest, and it has important implications for oncolytic virotherapy, suggesting that frequent cell cycle progression in cancer cells could make them more permissive to viruses.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Vesiculovirus/genética , Replicação Viral/genética , Animais , Antivirais/farmacologia , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Citoplasma , Fase G2/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular/fisiologia , Expressão Gênica/genética , Humanos , Interferon Tipo I/metabolismo , Interferon gama/metabolismo , Interferons , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus de RNA/imunologia , Vírus de RNA/metabolismo , Vírus Sendai/genética , Vírus Sendai/metabolismo , Transdução de Sinais , Vírus da Estomatite Vesicular Indiana/genética , Vesiculovirus/metabolismo , Proteínas da Matriz Viral/genética , Replicação Viral/imunologia , Interferon lambda
18.
Mol Cancer Res ; 16(11): 1785-1797, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30018032

RESUMO

Aurora A kinase (AURKA) is a master cell-cycle regulator that is often dysregulated in human cancers. Its overexpression has been associated with genome instability and oncogenic transformation. The protein kinase D (PKD) family is an emerging therapeutic target of cancer. Aberrant PKD activation has been implicated in tumor growth and survival, yet the underlying mechanisms remain to be elucidated. This study identified, for the first time, a functional crosstalk between PKD2 and Aurora A kinase in cancer cells. The data demonstrate that PKD2 is catalytically active during the G2-M phases of the cell cycle, and inactivation or depletion of PKD2 causes delay in mitotic entry due to downregulation of Aurora A, an effect that can be rescued by overexpression of Aurora A. Moreover, PKD2 localizes in the centrosome with Aurora A by binding to γ-tubulin. Knockdown of PKD2 caused defects in centrosome separation, elongated G2 phase, mitotic catastrophe, and eventually cell death via apoptosis. Mechanistically, PKD2 interferes with Fbxw7 function to protect Aurora A from ubiquitin- and proteasome-dependent degradation. Taken together, these results identify PKD as a cell-cycle checkpoint kinase that positively modulates G2-M transition through Aurora A kinase in mammalian cells.Implications: PKD2 is a novel cell-cycle regulator that promotes G2-M transition by modulating Aurora A kinase stability in cancer cells and suggests the PKD2/Aurora A kinase regulatory axis as new therapeutic targets for cancer treatment. Mol Cancer Res; 16(11); 1785-97. ©2018 AACR.


Assuntos
Aurora Quinase A/metabolismo , Centrossomo/enzimologia , Proteínas Quinases/metabolismo , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Centrossomo/metabolismo , Regulação para Baixo , Proteína 7 com Repetições F-Box-WD/metabolismo , Fase G2/fisiologia , Células HeLa , Humanos , Células PC-3 , Proteína Quinase D2 , Ubiquitinação
19.
J Cell Biol ; 217(9): 3019-3029, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-29930206

RESUMO

Cytoplasmic dynein is involved in diverse cell cycle-dependent functions regulated by several accessory factors, including Nde1 and Ndel1. Little is known about the role of these proteins in dynein cargo binding, and less is known about their cell cycle--dependent dynein regulation. Using Nde1 RNAi, mutant cDNAs, and a phosphorylation site-specific antibody, we found a specific association of phospho-Nde1 with the late G2-M nuclear envelope and prophase to anaphase kinetochores, comparable to the pattern for the Nde1 interactor CENP-F. Phosphomutant-Nde1 associated only with prometaphase kinetochores and showed weaker CENP-F binding in in vitro assays. Nde1 RNAi caused severe delays in mitotic progression, which were substantially rescued by both phosphomimetic and phosphomutant Nde1. Expression of a dynein-binding-deficient Nde1 mutant reduced kinetochore dynein by half, indicating a major role for Nde1 in kinetochore dynein recruitment. These results establish CENP-F as the first well-characterized Nde1 cargo protein, and reveal phosphorylation control of Nde1 cargo binding throughout a substantial fraction of the cell cycle.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Dineínas do Citoplasma/metabolismo , Cinetocoros/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Anáfase/fisiologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Fase G2/fisiologia , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/genética , Mitose/genética , Fosforilação , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética
20.
Plant Physiol Biochem ; 126: 39-46, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29499434

RESUMO

Ostreococcus tauri is the smallest free-living unicellular organism with one copy of each core cell cycle genes in its genome. There is a growing interest in this green algae due to its evolutionary origin. Since O. tauri is diverged early in the green lineage, relatively close to the ancestral eukaryotic cell, it might hold a key phylogenetic position in the eukaryotic tree of life. In this study, we focus on the regulatory network of its cell division cycle. We propose a mathematical modelling framework to integrate the existing knowledge of cell cycle network of O. tauri. We observe that feedback loop regulation of both G1/S and G2/M transitions in O. tauri is conserved, which can make the transition bistable. This is essential to make the transition irreversible as shown in other eukaryotic organisms. By performing sequence analysis, we also predict the presence of the Greatwall/PP2A pathway in the cell cycle of O. tauri. Since O. tauri cell cycle machinery is conserved, the exploration of the dynamical characteristic of the cell division cycle will help in further understanding the regulation of cell cycle in higher eukaryotes.


Assuntos
Clorófitas/metabolismo , Fase G1/fisiologia , Fase G2/fisiologia , Redes Reguladoras de Genes/fisiologia , Clorófitas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...