Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.639
Filtrar
1.
Development ; 149(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35588250

RESUMO

Although lengthening of the cell cycle and G1 phase is a generic feature of tissue maturation during development, the underlying mechanism remains poorly understood. Here, we develop a time-lapse imaging strategy to measure the four cell cycle phases in single chick neural progenitor cells in their endogenous environment. We show that neural progenitors are widely heterogeneous with respect to cell cycle length. This variability in duration is distributed over all phases of the cell cycle, with the G1 phase contributing the most. Within one cell cycle, each phase duration appears stochastic and independent except for a correlation between S and M phase duration. Lineage analysis indicates that the majority of daughter cells may have a longer G1 phase than mother cells, suggesting that, at each cell cycle, a mechanism lengthens the G1 phase. We identify that the CDC25B phosphatase known to regulate the G2/M transition indirectly increases the duration of the G1 phase, partly through delaying passage through the restriction point. We propose that CDC25B increases the heterogeneity of G1 phase length, revealing a previously undescribed mechanism of G1 lengthening that is associated with tissue development.


Assuntos
Células-Tronco Neurais , Ciclo Celular/fisiologia , Divisão Celular , Fase G1/fisiologia , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
2.
Dev Cell ; 57(5): 561-563, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35290777

RESUMO

Cellular differentiation can entail changes to the cell cycle. In this issue of Developmental Cell, Han et al. show that the transcription factor MUTE directly activates expression of the cyclin-dependent kinase (CDK) inhibitor SIAMESE RELATED 4 (SMR4), thereby slowing down G1 during the transition to stomatal differentiation.


Assuntos
Quinases Ciclina-Dependentes , Ciclinas , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Divisão Celular , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Desaceleração , Fase G1/fisiologia
3.
FEMS Yeast Res ; 22(1)2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-34905017

RESUMO

Molecular signal transduction networks, which conduct transcription at the G1 to S phase transition of the eukaryotic cell division cycle have been identified in diverse taxa from mammals to baker's yeast with analogous functional organization. However, regarding some network components, such as the transcriptional regulators STB1 and WHI5, only few orthologs exist, which are confined to individual Saccharomycotina species. While Whi5 has been characterized as yeast analog of human Rb protein, in the particular case of Stb1 (Sin three binding protein 1) identification of functional analogs emerges as difficult because to date its exact functionality still remains obscured. By aiming to resolve Stb1's enigmatic role this Perspective article especially surveys works covering relations between Cyclin/CDKs, the heteromeric transcription factor complexes SBF (Swi4/Swi6) and MBF (Mbp1/Swi6), as well as additional coregulators (Whi5, Sin3, Rpd3, Nrm1) which are collectively associated with the orderly transcription at 'Start' of the Saccharomyces cerevisiae cell cycle. In this context, interaction capacities of the Sin3-scaffold protein are widely surveyed because its four PAH domains (Paired Amphiphatic Helix) represent a 'recruitment-code' for gene-specific targeting of repressive histone deacetylase activity (Rpd3) via different transcription factors. Here, Stb1 plays a role in Sin3's action on transcription at the G1/S-boundary. Through bioinformatic analyses a potential Sin3-interaction domain (SID) was detected in Stb1, and beyond that, connections within the G1/S-regulatory network are discussed in structural and evolutionary context thereby providing conceptual perspectives.


Assuntos
Redes Reguladoras de Genes , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Fatores de Transcrição , Fase G1/fisiologia , Regulação Fúngica da Expressão Gênica , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
4.
Science ; 374(6565): 347-351, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34648313

RESUMO

Cell division is thought to be initiated by cyclin-dependent kinases (Cdks) inactivating key transcriptional inhibitors. In budding yeast, the G1 cyclin Cln3-Cdk1 complex is thought to directly phosphorylate the Whi5 protein, thereby releasing the transcription factor SBF and committing cells to division. We report that Whi5 is a poor substrate of Cln3-Cdk1, which instead phosphorylates the RNA polymerase II subunit Rpb1's C-terminal domain on S5 of its heptapeptide repeats. Cln3-Cdk1 binds SBF-regulated promoters and Cln3's function can be performed by the canonical S5 kinase Ccl1-Kin28 when synthetically recruited to SBF. Thus, we propose that Cln3-Cdk1 triggers cell division by phosphorylating Rpb1 at SBF-regulated promoters to promote transcription. Our findings blur the distinction between cell cycle and transcriptional Cdks to highlight the ancient relationship between these two processes.


Assuntos
Proteína Quinase CDC28 de Saccharomyces cerevisiae/metabolismo , Divisão Celular/fisiologia , Ciclinas/metabolismo , RNA Polimerase II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiologia , Proteína Quinase CDC28 de Saccharomyces cerevisiae/genética , Divisão Celular/genética , Ciclinas/genética , Fase G1/genética , Fase G1/fisiologia , Regulação Fúngica da Expressão Gênica , Fosforilação , Regiões Promotoras Genéticas , Domínios Proteicos , RNA Polimerase II/química , Proteínas Repressoras/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Fatores de Transcrição/metabolismo
5.
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
6.
Sci Rep ; 11(1): 14736, 2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282211

RESUMO

During early G1 phase, Rb is exclusively mono-phosphorylated by cyclin D:Cdk4/6, generating 14 different isoforms with specific binding patterns to E2Fs and other cellular protein targets. While mono-phosphorylated Rb is dispensable for early G1 phase progression, interfering with cyclin D:Cdk4/6 kinase activity prevents G1 phase progression, questioning the role of cyclin D:Cdk4/6 in Rb inactivation. To dissect the molecular functions of cyclin D:Cdk4/6 during cell cycle entry, we generated a single cell reporter for Cdk2 activation, RB inactivation and cell cycle entry by CRISPR/Cas9 tagging endogenous p27 with mCherry. Through single cell tracing of Cdk4i cells, we identified a time-sensitive early G1 phase specific Cdk4/6-dependent phosphorylation gradient that regulates cell cycle entry timing and resides between serum-sensing and cyclin E:Cdk2 activation. To reveal the substrate identity of the Cdk4/6 phosphorylation gradient, we performed whole proteomic and phospho-proteomic mass spectrometry, and identified 147 proteins and 82 phospho-peptides that significantly changed due to Cdk4 inhibition in early G1 phase. In summary, we identified novel (non-Rb) cyclin D:Cdk4/6 substrates that connects early G1 phase functions with cyclin E:Cdk2 activation and Rb inactivation by hyper-phosphorylation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Fase G1/fisiologia , Divisão Celular , Células Cultivadas , Ciclina D/metabolismo , Ciclina E/metabolismo , Humanos , Proteínas Oncogênicas/metabolismo , Fosforilação , Proteoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína do Retinoblastoma/metabolismo
7.
Mol Cell ; 81(12): 2596-2610.e7, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33961796

RESUMO

p53-binding protein 1 (53BP1) regulates both the DNA damage response and p53 signaling. Although 53BP1's function is well established in DNA double-strand break repair, how its role in p53 signaling is modulated remains poorly understood. Here, we identify the scaffolding protein AHNAK as a G1 phase-enriched interactor of 53BP1. We demonstrate that AHNAK binds to the 53BP1 oligomerization domain and controls its multimerization potential. Loss of AHNAK results in hyper-accumulation of 53BP1 on chromatin and enhanced phase separation, culminating in an elevated p53 response, compromising cell survival in cancer cells but leading to senescence in non-transformed cells. Cancer transcriptome analyses indicate that AHNAK-53BP1 cooperation contributes to the suppression of p53 target gene networks in tumors and that loss of AHNAK sensitizes cells to combinatorial cancer treatments. These findings highlight AHNAK as a rheostat of 53BP1 function, which surveys cell proliferation by preventing an excessive p53 response.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Fase G1/fisiologia , Histonas/metabolismo , Humanos , Células MCF-7 , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/fisiologia
8.
Int J Mol Sci ; 22(6)2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33802105

RESUMO

Several lines of evidence suggest the existence in the eukaryotic cells of a tight, yet largely unexplored, connection between DNA replication and sister chromatid cohesion. Tethering of newly duplicated chromatids is mediated by cohesin, an evolutionarily conserved hetero-tetrameric protein complex that has a ring-like structure and is believed to encircle DNA. Cohesin is loaded onto chromatin in telophase/G1 and converted into a cohesive state during the subsequent S phase, a process known as cohesion establishment. Many studies have revealed that down-regulation of a number of DNA replication factors gives rise to chromosomal cohesion defects, suggesting that they play critical roles in cohesion establishment. Conversely, loss of cohesin subunits (and/or regulators) has been found to alter DNA replication fork dynamics. A critical step of the cohesion establishment process consists in cohesin acetylation, a modification accomplished by dedicated acetyltransferases that operate at the replication forks. Defects in cohesion establishment give rise to chromosome mis-segregation and aneuploidy, phenotypes frequently observed in pre-cancerous and cancerous cells. Herein, we will review our present knowledge of the molecular mechanisms underlying the functional link between DNA replication and cohesion establishment, a phenomenon that is unique to the eukaryotic organisms.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos/fisiologia , Replicação do DNA/fisiologia , Fase G1/fisiologia , Telófase/fisiologia , Animais , Humanos
9.
Cells ; 10(3)2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33806417

RESUMO

Most Cyclin-dependent kinases (Cdks) are redundant for normal cell division. Here we tested whether these redundancies are maintained during cell cycle recovery after a DNA damage-induced arrest in G1. Using non-transformed RPE-1 cells, we find that while Cdk4 and Cdk6 act redundantly during normal S-phase entry, they both become essential for S-phase entry after DNA damage in G1. We show that this is due to a greater overall dependency for Cdk4/6 activity, rather than to independent functions of either kinase. In addition, we show that inactivation of pocket proteins is sufficient to overcome the inhibitory effects of complete Cdk4/6 inhibition in otherwise unperturbed cells, but that this cannot revert the effects of Cdk4/6 inhibition in DNA damaged cultures. Indeed, we could confirm that, in addition to inactivation of pocket proteins, Cdh1-dependent anaphase-promoting complex/cyclosome (APC/CCdh1) activity needs to be inhibited to promote S-phase entry in damaged cultures. Collectively, our data indicate that DNA damage in G1 creates a unique situation where high levels of Cdk4/6 activity are required to inactivate pocket proteins and APC/CCdh1 to promote the transition from G1 to S phase.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Dano ao DNA/genética , Fase G1/fisiologia , Humanos , Transfecção
10.
Cell Mol Life Sci ; 78(10): 4507-4519, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33884444

RESUMO

During early embryonic development both the rapid increase in cell number and the expression of genes that control developmental decisions are tightly regulated. Accumulating evidence has indicated that these two seemingly independent processes are mechanistically intertwined. The picture that emerges from studies on the cell cycle of embryonic stem cells is one in which proteins that promote cell cycle progression prevent differentiation and vice versa. Here, we review which transcription factors and signalling pathways play a role in both maintenance of pluripotency as well as cell cycle progression. We will not only describe the mechanism behind their function but also discuss the role of these regulators in different states of mouse pluripotency. Finally, we elaborate on how canonical cell cycle regulators impact on the molecular networks that control the maintenance of pluripotency and lineage specification.


Assuntos
Fase G1/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Humanos , Transdução de Sinais/fisiologia
11.
Cancer Res ; 81(8): 2029-2043, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33574086

RESUMO

Despite recent promising advances in targeted therapies and immunotherapies, patients with melanoma incur substantial mortality. In particular, inhibitors targeting BRAF-mutant melanoma can lead to resistance, and no targeted therapies exist for NRAS-mutant melanoma, motivating the search for additional therapeutic targets and vulnerable pathways. Here we identify a regulator of Wnt/ß-catenin signaling, PLEKHA4, as a factor required for melanoma proliferation and survival. PLEKHA4 knockdown in vitro decreased Dishevelled levels, attenuated Wnt/ß-catenin signaling, and blocked progression through the G1-S cell-cycle transition. In mouse xenograft and allograft models, inducible PLEKHA4 knockdown attenuated tumor growth in BRAF- and NRAS-mutant melanomas and exhibited an additive effect with the clinically used inhibitor encorafenib in a BRAF-mutant model. As an E3 ubiquitin ligase regulator with both lipid- and protein-binding partners, PLEKHA4 presents several opportunities for targeting with small molecules. Our work identifies PLEKHA4 as a promising drug target for melanoma and clarifies a controversial role for Wnt/ß-catenin signaling in the control of melanoma proliferation. SIGNIFICANCE: This study establishes that melanoma cell proliferation requires the protein PLEKHA4 to promote pathologic Wnt signaling for proliferation, highlighting PLEKHA4 inhibition as a new avenue for the development of targeted therapies.


Assuntos
Proliferação de Células/fisiologia , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/patologia , Via de Sinalização Wnt/fisiologia , Animais , Carbamatos/farmacologia , Linhagem Celular Tumoral , Proteínas Desgrenhadas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Fase G1/fisiologia , GTP Fosfo-Hidrolases/genética , Xenoenxertos , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/mortalidade , Proteínas de Membrana/genética , Camundongos , Terapia de Alvo Molecular , Mutação , Transplante de Neoplasias , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , RNA Interferente Pequeno/metabolismo , Fase S/fisiologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/mortalidade , Sulfonamidas/farmacologia , Ensaio Tumoral de Célula-Tronco
12.
Science ; 369(6502): 466-471, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32703881

RESUMO

Cell size is fundamental to cell physiology. For example, cell size determines the spatial scale of organelles and intracellular transport and thereby affects biosynthesis. Although some genes that affect mammalian cell size have been identified, the molecular mechanisms through which cell growth drives cell division have remained elusive. We show that cell growth during the G1 phase of the cell division cycle dilutes the cell cycle inhibitor Retinoblastoma protein (Rb) to trigger division in human cells. RB overexpression increased cell size and G1 duration, whereas RB deletion decreased cell size and removed the inverse correlation between cell size at birth and the duration of the G1 phase. Thus, Rb dilution through cell growth in G1 provides one of the long-sought molecular mechanisms that promotes cell size homeostasis.


Assuntos
Divisão Celular/fisiologia , Proteína do Retinoblastoma/fisiologia , Pontos de Checagem do Ciclo Celular/fisiologia , Proliferação de Células , Tamanho Celular , Fase G1/fisiologia , Humanos
13.
Nat Commun ; 11(1): 1836, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32296067

RESUMO

Studies on multisite phosphorylation networks of cyclin-dependent kinase (CDK) targets have opened a new level of signaling complexity by revealing signal processing routes encoded into disordered proteins. A model target, the CDK inhibitor Sic1, contains linear phosphorylation motifs, docking sites, and phosphodegrons to empower an N-to-C terminally directed phosphorylation process. Here, we uncover a signal processing mechanism involving multi-step competition between mutually diversional phosphorylation routes within the S-CDK-Sic1 inhibitory complex. Intracomplex phosphorylation plays a direct role in controlling Sic1 degradation, and provides a mechanism to sequentially integrate both the G1- and S-CDK activities while keeping S-CDK inhibited towards other targets. The competing phosphorylation routes prevent premature Sic1 degradation and demonstrate how integration of MAPK from the pheromone pathway allows one to tune the competition of alternative phosphorylation paths. The mutually diversional phosphorylation circuits may be a general way for processing multiple kinase signals to coordinate cellular decisions in eukaryotes.


Assuntos
Fase G1/fisiologia , Fase S/efeitos da radiação , Transdução de Sinais/fisiologia , Western Blotting , Divisão Celular/genética , Divisão Celular/fisiologia , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fase G1/genética , Imunoprecipitação , Espectrometria de Massas , Fosforilação , Fase S/genética , Transdução de Sinais/genética
14.
Cell Rep ; 31(1): 107464, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32268083

RESUMO

N6-Methyladenosine (m6A) modification is the major chemical modification in mRNA that controls fundamental biological processes, including cell proliferation. Herein, we demonstrate that fat mass and obesity-associated (FTO) demethylates m6A modification of cyclin D1, the key regulator for G1 phase progression and controls cell proliferation in vitro and in vivo. FTO depletion upregulates cyclin D1 m6A modification, which in turn accelerates the degradation of cyclin D1 mRNA, leading to the impairment of G1 progression. m6A modification of cyclin D1 oscillates in a cell-cycle-dependent manner; m6A levels are suppressed during the G1 phase and enhanced during other phases. Low m6A levels during G1 are associated with the nuclear translocation of FTO from the cytosol. Furthermore, nucleocytoplasmic shuttling of FTO is regulated by casein kinase II-mediated phosphorylation of FTO. Our results highlight the role of m6A in regulating cyclin D1 mRNA stability and add another layer of complexity to cell-cycle regulation.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Ciclina D1/metabolismo , RNA Mensageiro/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/fisiologia , Animais , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Linhagem Celular , Ciclina D1/genética , Quinases Ciclina-Dependentes/metabolismo , Desmetilação , Fase G1/fisiologia , Xenoenxertos , Humanos , Masculino , Camundongos , Fosforilação , Estabilidade de RNA , RNA Mensageiro/genética
15.
Biosystems ; 191-192: 104128, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32165312

RESUMO

Biological systems are difficult to understand complex systems. Scientists continue to create models to simulate biological systems but these models are complex too; for this reason, new reduction methods to simplify complex biological models into simpler ones are increasingly needed. In this paper, we present a way of reducing complex quantitative (continuous) models into logical models based on time windows of system activity and logical (Boolean) models. Time windows were used to define slow and fast activity areas. We use the proposed approach to reduce a continuous ODE model into a logical model describing the G1/S checkpoint with and without DNA damage as a case study. We show that the temporal unfolding of this signalling system can be broken down into three time windows where only two display high level of activity and the other shows little or no activity. The two active windows represent a cell committing to cell cycle and making the G1/S transition, respectively, the two most important high level functions of cell cycle in the G1 phase. Therefore, we developed two models to represent these time windows to reduce time complexity and used Boolean approach to reduce interaction complexity in the ODE model in the respective time windows. The developed reduced models correctly produced the commitment to cell cycle and G1/S transfer through the expected behavior of signalling molecules involved in these processes. As most biological models have a large number of fast reactions and a relatively smaller number of slow reactions, we believe that the proposed approach could be suitable for representing many, if not all biological signalling networks. The approach presented in this study greatly helps in simplifying complex continuous models (ODE models) into simpler models. Moreover, it will also assist scientists build models concentrating on understanding and representing system behavior rather than setting values for a large number of kinetic parameters.


Assuntos
Algoritmos , Dano ao DNA , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Simulação por Computador , Fase G1/genética , Fase G1/fisiologia , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Redes Reguladoras de Genes , Mapas de Interação de Proteínas , Fase S/genética , Fase S/fisiologia , Transdução de Sinais/genética , Fatores de Tempo
16.
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
17.
Biosci Biotechnol Biochem ; 84(2): 297-304, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31661371

RESUMO

Glioma is the most common highly malignant primary brain tumor. MicroRNA-519d-3p exerts important effects in several tumors, but its functional role in glioma remained poorly understood. In this study, we found miR-519d-3p expression was significantly decreased in glioma tissues and cell lines. Moreover, the in vitro experiments showed that overexpression of miR-519d-3p suppressed cell proliferation and induced cell cycle G0/G1 phase arrest using MTT and flow cytometry assays in glioma cell lines, U87 and U251. Mechanistically, Cyclin D1 (CCND1) was predicted and confirmed as the direct target genes of miR-519d-3p using luciferase report assay. In addition, knockdown of CCND1 imitated the suppressive effects of miR-519d-3p on cell proliferation and cell cycle progression. Furthermore, restoration of CCND1 reversed the effects of miR-519d-3p overexpression in glioma cells. Taken together, these data demonstrate that suppression of CCND1 by miR-519d-3p might be a therapeutic target for glioma.Abbreviations miR-519d-3p: microRNA-519d-3p; CCND1: Cyclin D1; ATCC: American Type Culture Collection; MTT: 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide; PI: propidium iodide; WT: wild type; MUT: mutant type; SD: standard deviation.


Assuntos
Neoplasias Encefálicas/patologia , Proliferação de Células/fisiologia , Ciclina D1/antagonistas & inibidores , Fase G1/fisiologia , Glioma/patologia , MicroRNAs/fisiologia , Regiões 3' não Traduzidas , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Regulação para Baixo , Técnicas de Silenciamento de Genes , Glioma/metabolismo , Humanos , Prognóstico
18.
Exp Cell Res ; 387(1): 111776, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31838060

RESUMO

Microtubule-binding proteins provide an alternative and vital pathway to the functional diversity of microtubules. Considerable work is still required to understand the complexities of microtubule-associated cellular processes and to identify novel microtubule-binding proteins. In this study, we identify Bcl2-associated athanogene cochaperone 6 (BAG6) as a novel microtubule-binding protein and reveal that it is crucial for primary ciliogenesis. By immunofluorescence we show that BAG6 largely colocalizes with intracellular microtubules and by co-immunoprecipitation we demonstated that it can interact with α-tubulin. Additionally, both the UBL and BAG domains of BAG6 are indispensable for its interaction with α-tubulin. Moreover, the assembly of primary cilia in RPE-1 cells is significantly inhibited upon the depletion of BAG6. Notably, BAG6 inhibition leads to an abnormal G0/G1 transition during the cell cycle. In addition, BAG6 colocalizes and interactes with the centrosomal protein γ-tubulin, suggesting that BAG6 might regulate primary ciliogenesis through its action in centrosomal function. Collectively, our findings suggest that BAG6 is a novel microtubule-bindng protein crucial for primary ciliogenesis.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Fase G1/fisiologia , Células HEK293 , Células HeLa , Humanos , Ligação Proteica/fisiologia , Fase de Repouso do Ciclo Celular/fisiologia
19.
Biomed Pharmacother ; 121: 109598, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31733572

RESUMO

Lung cancer remains the leading cause of cancer mortality because of highly malignant and metastatic potential. The current status of lung cancer treatment is limited, and more treatment options are needed. Interesting, antipsychotic drugs have been reported to show anti-cancer effects. In this present study, we investigated the anticancer potential of penfluridol (PF), an anti-schizophrenic drug, in lung cancer and its underlying mechanism in vitro and in vivo. In vitro, it could inhibit the viability of various lung cancer cells with G0/G1 phase arrest via increasing the expression level of p21/p27 and decreasing the expression levels of cyclin-CDK complex. Meanwhile, cell-cycle arrest causes DNA repair in the nucleus, which was associated with the upregulation of H2A.X and p-H2A.X. Moreover, PF could also decrease mitochondrial membrane potential and increase reactive oxygen species levels in the lung cancer cells. These results implied that PF might induce the mitochondria-mediated intrinsic apoptosis. In addition, PF inhibits the migration and invasion of lung cancer cells via downregulation of FAK-MMP signaling. In vivo, oral administration of PF at concentration of 10 mg/kg inhibited tumor growth in A549 xenograft model. Notably, PF is an approved drug and the price is exceedingly cheap, so this study demonstrates the potential of PF to treat lung cancer.


Assuntos
Antipsicóticos/uso terapêutico , Apoptose/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Penfluridol/uso terapêutico , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Células A549 , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antipsicóticos/farmacologia , Apoptose/fisiologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Fase G1/fisiologia , Inibidores do Crescimento/farmacologia , Inibidores do Crescimento/uso terapêutico , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/patologia , Penfluridol/farmacologia , Fase de Repouso do Ciclo Celular/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
Nat Commun ; 10(1): 4436, 2019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31570711

RESUMO

CENP-A is an essential histone H3 variant that epigenetically marks the centromeric region of chromosomes. Here we show that CENP-A nucleosomes form characteristic clusters during the G1 phase of the cell cycle. 2D and 3D super-resolution microscopy and segmentation analysis reveal that these clusters encompass a globular rosette-like structure, which evolves into a more compact structure in late G1. The rosette-like clusters contain numerous CENP-A molecules and form a large cellular structure of ∼250-300 nm diameter with remarkably similar shapes for each centromere. Co-localization analysis shows that HJURP, the CENP-A chaperone, is located in the center of the rosette and serves as a nucleation point. The discovery of an HJURP-mediated CENP-A nucleation in human cells and its structural description provide important insights into the mechanism of CENP-A deposition and the organization of CENP-A chromatin in the centromeric region.


Assuntos
Proteína Centromérica A/metabolismo , Proteína Centromérica A/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Fase G1/fisiologia , Nucleossomos/metabolismo , Animais , Ciclo Celular/fisiologia , Linhagem Celular , Centrômero/metabolismo , Centrômero/ultraestrutura , Cromatina , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas de Ligação a DNA/química , Epigenômica , Células HeLa , Humanos , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL/embriologia , Chaperonas Moleculares/química , Nucleossomos/ultraestrutura , Imagem Óptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...