Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.083
Filtrar
1.
Development ; 149(11)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35588250

RESUMEN

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.


Asunto(s)
Células-Madre Neurales , Ciclo Celular/fisiología , División Celular , Fase G1/fisiología , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
2.
J Neurosci ; 43(7): 1154-1165, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36596698

RESUMEN

During development, cortical neurons are produced in a temporally regulated sequence from apical progenitors, directly or indirectly, through the production of intermediate basal progenitors. The balance between these major progenitor types is critical for the production of the proper number and types of neurons, and it is thus important to decipher the cellular and molecular cues controlling this equilibrium. Here we address the role of a cell cycle regulator, the CDC25B phosphatase, in this process. We show that, in the developing mouse neocortex of both sex, deleting CDC25B in apical progenitors leads to a transient increase in the production of TBR1+ neurons at the expense of TBR2+ basal progenitors. This phenotype is associated with lengthening of the G2 phase of the cell cycle, the total cell cycle length being unaffected. Using in utero electroporation and cortical slice cultures, we demonstrate that the defect in TBR2+ basal progenitor production requires interaction with CDK1 and is because of the G2 phase lengthening in CDC25B mutants. Together, this study identifies a new role for CDC25B and G2 phase length in direct versus indirect neurogenesis at early stages of cortical development.SIGNIFICANCE STATEMENT This study is the first analysis of the function of CDC25B, a G2/M regulator, in the developing neocortex. We show that removing CDC25B function leads to a transient increase in neuronal differentiation at early stages, occurring simultaneously with a decrease in basal intermediate progenitors (bIPs). Conversely, a CDC25B gain of function promotes production of bIPs, and this is directly related to CDC25B's ability to regulate CDK1 activity. This imbalance of neuron/progenitor production is linked to a G2 phase lengthening in apical progenitors; and using pharmacological treatments on cortical slice cultures, we show that shortening the G2 phase is sufficient to enhance bIP production. Our results reveal the importance of G2 phase length regulation for neural progenitor fate determination.


Asunto(s)
Neocórtex , Células-Madre Neurales , Neurogénesis , Animales , Ratones , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/metabolismo
3.
J Biol Chem ; 299(3): 102957, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36717077

RESUMEN

Cyclin A and CDC25A are both activators of cyclin-dependent kinases (CDKs): cyclin A acts as an activating subunit of CDKs and CDC25A a phosphatase of the inhibitory phosphorylation sites of the CDKs. In this study, we uncovered an inverse relationship between the two CDK activators. As cyclin A is an essential gene, we generated a conditional silencing cell line using a combination of CRISPR-Cas9 and degron-tagged cyclin A. Destruction of cyclin A promoted an acute accumulation of CDC25A. The increase of CDC25A after cyclin A depletion occurred throughout the cell cycle and was independent on cell cycle delay caused by cyclin A deficiency. Moreover, we determined that the inverse relationship with cyclin A was specific for CDC25A and not for other CDC25 family members or kinases that regulate the same sites in CDKs. Unexpectedly, the upregulation of CDC25A was mainly caused by an increase in transcriptional activity instead of a change in the stability of the protein. Reversing the accumulation of CDC25A severely delayed G2-M in cyclin A-depleted cells. Taken together, these data provide evidence of a compensatory mechanism involving CDC25A that ensures timely mitotic entry at different levels of cyclin A.


Asunto(s)
Ciclina A , Quinasas Ciclina-Dependientes , Fosfatasas cdc25 , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo , Ciclo Celular , División Celular , Ciclina A/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Fosforilación
4.
Mol Cell ; 62(2): 307-313, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27067599

RESUMEN

One recurring theme in drug development is to exploit synthetic lethal properties as means to preferentially damage the DNA of cancer cells. We and others have previously developed inhibitors of the ATR kinase, shown to be particularly genotoxic for cells expressing certain oncogenes. In contrast, the mechanisms of resistance to ATR inhibitors remain unexplored. We report here on a genome-wide CRISPR-Cas9 screen that identified CDC25A as a major determinant of sensitivity to ATR inhibition. CDC25A-deficient cells resist high doses of ATR inhibitors, which we show is due to their failure to prematurely enter mitosis in response to the drugs. Forcing mitotic entry with WEE1 inhibitors restores the toxicity of ATR inhibitors in CDC25A-deficient cells. With ATR inhibitors now entering the clinic, our work provides a better understanding of the mechanisms by which these compounds kill cells and reveals genetic interactions that could be used for their rational use.


Asunto(s)
Antineoplásicos/farmacología , Sistemas CRISPR-Cas , Resistencia a Antineoplásicos/genética , Células Madre Embrionarias/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Fosfatasas cdc25/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/patología , Estudio de Asociación del Genoma Completo , Humanos , Mitosis/efectos de los fármacos , Terapia Molecular Dirigida , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transfección , Fosfatasas cdc25/genética
5.
Environ Toxicol ; 39(5): 3225-3237, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38357781

RESUMEN

Lung cancer (LC) is the most prevalent cancer type, with a high mortality rate worldwide. The current treatment options for LC have not been particularly successful in improving patient outcomes. Yifei Sanjie (YFSJ), a well-applicated traditional Chinese medicine formula, is widely used to treat pulmonary diseases, especially LC, yet little is known about its molecular mechanisms. This study was conducted to explore the molecular mechanism by which YFSJ ameliorated LC progression. The A549, NCI-H1975, and Calu-3 cells were treated with the YFSJ formula and observed for colony number, apoptosis, migration, and invasion properties recorded via corresponding assays. The PRMT6-YBX1-CDC25A axis was tested and verified through luciferase reporter, RNA immunoprecipitation, and chromatin immunoprecipitation assays and rescue experiments. Our results demonstrated that YFSJ ameliorated LC cell malignant behaviors by increasing apoptosis and suppressing proliferation, migration, and invasion processes. We also noticed that the xenograft mouse model treated with YFSJ significantly reduced tumor growth compared with the control untreated group in vivo. Mechanistically, it was found that YFSJ suppressed the expression of PRMT6, YBX1, and CDC25A, while the knockdown of these proteins significantly inhibited colony growth, migration, and invasion, and boosted apoptosis in LC cells. In summary, our results suggest that YFSJ alleviates LC progression via the PRMT6-YBX1-CDC25A axis, confirming its efficacy in clinical use. The findings of our study provide a new regulatory network for LC growth and metastasis, which could shed new insights into pulmonary medical research.


Asunto(s)
Neoplasias Pulmonares , MicroARNs , Humanos , Animales , Ratones , Neoplasias Pulmonares/patología , Proliferación Celular/genética , Movimiento Celular/genética , Pulmón/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteína 1 de Unión a la Caja Y/genética , Proteína 1 de Unión a la Caja Y/metabolismo , Proteínas Nucleares/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/uso terapéutico , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
6.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732131

RESUMEN

Overexpression of the 14-3-3ε protein is associated with suppression of apoptosis in cutaneous squamous cell carcinoma (cSCC). This antiapoptotic activity of 14-3-3ε is dependent on its binding to CDC25A; thus, inhibiting 14-3-3ε - CDC25A interaction is an attractive therapeutic approach to promote apoptosis in cSCC. In this regard, designing peptide inhibitors of 14-3-3ε - CDC25A interactions is of great interest. This work reports the rational design of peptide analogs of pS, a CDC25A-derived peptide that has been shown to inhibit 14-3-3ε-CDC25A interaction and promote apoptosis in cSCC with micromolar IC50. We designed new peptide analogs in silico by shortening the parent pS peptide from 14 to 9 amino acid residues; then, based on binding motifs of 14-3-3 proteins, we introduced modifications in the pS(174-182) peptide. We studied the binding of the peptides using conventional molecular dynamics (MD) and steered MD simulations, as well as biophysical methods. Our results showed that shortening the pS peptide from 14 to 9 amino acids reduced the affinity of the peptide. However, substituting Gln176 with either Phe or Tyr amino acids rescued the binding of the peptide. The optimized peptides obtained in this work can be candidates for inhibition of 14-3-3ε - CDC25A interactions in cSCC.


Asunto(s)
Proteínas 14-3-3 , Simulación de Dinámica Molecular , Unión Proteica , Fosfatasas cdc25 , Fosfatasas cdc25/metabolismo , Fosfatasas cdc25/química , Fosfatasas cdc25/antagonistas & inhibidores , Proteínas 14-3-3/metabolismo , Proteínas 14-3-3/química , Humanos , Péptidos/química , Péptidos/metabolismo , Secuencia de Aminoácidos
7.
Biochem Biophys Res Commun ; 665: 98-106, 2023 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-37149988

RESUMEN

Zebrafish have the ability to fully regenerate their hearts after injury since cardiomyocytes subsequently dedifferentiate, re-enter cell cycle, and proliferate to replace damaged myocardial tissue. Recent research identified the reactivation of dormant developmental pathways during cardiac regeneration in adult zebrafish, suggesting pro-proliferative pathways important for developmental heart growth to be also critical for regenerative heart growth after injury. Histone deacetylase 1 (Hdac1) was recently shown to control both, embryonic as well as adult regenerative cardiomyocyte proliferation in the zebrafish model. Nevertheless, regulatory pathways controlled by Hdac1 are not defined yet. By analyzing RNA-seq-derived transcriptional profiles of the Hdac1-deficient zebrafish mutant baldrian, we here identified DNA damage response (DDR) pathways activated in baldrian mutant embryos. Surprisingly, although the DDR signaling pathway was transcriptionally activated, we found the complete loss of protein expression of the known DDR effector and cell cycle inhibitor p21. Consequently, we observed an upregulation of the p21-downstream target Cdk2, implying elevated G1/S phase transition in Hdac1-deficient zebrafish hearts. Remarkably, Cdk1, another p21-but also Cdc25-downstream target was downregulated. Here, we found the significant downregulation of Cdc25 protein expression, explaining reduced Cdk1 levels and suggesting impaired G2/M phase progression in Hdac1-deficient zebrafish embryos. To finally prove defective cell cycle progression due to Hdac1 loss, we conducted Cytometer-based cell cycle analyses in HDAC1-deficient murine HL-1 cardiomyocytes and indeed found impaired G2/M phase transition resulting in defective cardiomyocyte proliferation. In conclusion, our results suggest a critical role of Hdac1 in maintaining both, regular G1/S and G2/M phase transition in cardiomyocytes by controlling the expression of essential cell cycle regulators such as p21 and Cdc25.


Asunto(s)
Miocitos Cardíacos , Pez Cebra , Animales , Ratones , Ciclo Celular/genética , División Celular , Proliferación Celular , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Miocitos Cardíacos/metabolismo , Pez Cebra/metabolismo , Fosfatasas cdc25/metabolismo , Proteína Quinasa CDC2/metabolismo
8.
Cell Biol Toxicol ; 39(5): 1-18, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-35567596

RESUMEN

Circular RNAs (circRNAs) have been extensively studied in tumor development and treatment. CircZNF609 (hsa_circ_0000615) has been shown to serve as an oncogene in all kinds of solid tumors and may act as the novel biomarker in tumor diagnosis and therapy in tumor early diagnosis and therapy. However, the underlying character and mechanism of circZNF609 in cisplatin chemosensitivity and bladder cancer (BCa) development were unknown. The expression level of cell division cycle 25B (CDC25B), microRNA 1200 (miR-1200), and circZNF609 in BCa cells and tissues depended on quantitative real-time PCR (qRT-PCR). CDC25B protein level was assayed with Western blot. Functional assays in vitro and in vivo had been conducted to inspect the important role of circZNF609 on BCa progression and cisplatin chemosensitivity in BCa. RNA sequencing and online databases were used to predict the interactions among circZNF609, miR-1200, and CDC25B. Mechanistic exploration was confirmed by RNA pull-down assay, RNA fluorescence in situ hybridization (FISH) and Dual luciferase reporter assay. CircZNF609 expression was increased significantly in BCa cell lines and tissues. For BCa patients, increased expression of circZNF609 was correlated with a worse survival. In vitro and in vivo, enforced expression of circZNF609 enhanced BCa cells proliferation, migration, and cisplatin chemoresistance. Mechanistically, circZNF609 alleviated the inhibition effect on target CDC25B expression by sponging miR-1200. CircZNF609 promoted tumor growth through novel circZNF609/miR-1200/CDC25B axis, implying that circZNF609 has significant potential to act as a new diagnostic biomarker and therapeutic target in BCa. Enhancing cisplatin sensitivity is an important direction for bladder cancer management. 1. This research reveals that circZNF609 improves bladder cancer progression and inhibits cisplatin sensitivity by inducing G1/S cell cycle arrest via a novel miR-1200/CDC25B cascades. 2. CircZNF609 was confirmed associated with worse survival of bladder cancer patients. 3. CircZNF609 act as a prognostic biomarker for bladder cancer treatment.


Asunto(s)
MicroARNs , Neoplasias de la Vejiga Urinaria , Humanos , Cisplatino/farmacología , Cisplatino/uso terapéutico , MicroARNs/genética , MicroARNs/metabolismo , Hibridación Fluorescente in Situ , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Proliferación Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
9.
Int J Mol Sci ; 24(2)2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36675024

RESUMEN

Cell division regulators play a vital role in neural progenitor cell (NPC) proliferation and differentiation. Cell division cycle 25C (CDC25C) is a member of the CDC25 family of phosphatases which positively regulate cell division by activating cyclin-dependent protein kinases (CDKs). However, mice with the Cdc25c gene knocked out were shown to be viable and lacked the apparent phenotype due to genetic compensation by Cdc25a and/or Cdc25b. Here, we investigate the function of Cdc25c in developing rat brains by knocking down Cdc25c in NPCs using in utero electroporation. Our results indicate that Cdc25c plays an essential role in maintaining the proliferative state of NPCs during cortical development. The knockdown of Cdc25c causes early cell cycle exit and the premature differentiation of NPCs. Our study uncovers a novel role of CDC25C in NPC division and cell fate determination. In addition, our study presents a functional approach to studying the role of genes, which elicit genetic compensation with knockout, in cortical neurogenesis by knocking down in vivo.


Asunto(s)
Proteínas de Ciclo Celular , Células-Madre Neurales , Neurogénesis , Fosfatasas cdc25 , Animales , Ratas , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo/genética , Neurogénesis/genética , Neurogénesis/fisiología , Células-Madre Neurales/metabolismo
10.
Cancer ; 128(9): 1775-1786, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35143052

RESUMEN

BACKGROUND: The objective of this study was to investigate the role and molecular mechanism of cyclin-dependent kinase 5 (CDK5) in regulating the growth of tongue squamous cell carcinoma (TSCC). METHODS: The authors used multiple methods to detect the levels of CDK5 expression in samples of TSCC and to explore the relation between CDK5 expression and various clinicopathologic factors. In vivo and in vitro cell experiments were performed to detect the proliferation, invasion, and migration of TSCC cells with CDK5 knockdown or overexpression. These studies verified that CDK5 regulates the occurrence and development of TSCC cells through the microRNA 513c-5p/cell division cycle 25B pathway. RESULTS: An elevated level of CDK5 expression in TSCC tissues was identified as an independent risk factor affecting TSCC growth and patient prognosis. Patients who had TSCC with low levels of CDK5 expression had a higher survival rate than those with high levels. Knockdown of CDK5 reduced the proliferation, migration, and invasion of TSCC cells both in vitro and in vivo. In addition, the authors observed that CDK5 regulated the growth of TSCC through the microRNA 513c-5p/cell division cycle C25B pathway. CONCLUSIONS: CDK5 functions as an oncogene in TSCC and might serve as a molecular marker for use in the diagnosis and treatment of TSCC. LAY SUMMARY: Tongue squamous cell carcinoma (TSCC) is 1 of the most common malignant tumors of the head and neck, and the survival rate of patients with tongue cancer has been very low. Therefore, it is important to study the molecular mechanism of TSCC progression to identify biomarkers that can be used to improve its clinical diagnosis and treatment. Cyclin-dependent kinase 5 (CDK5) is an atypical member of the cyclin-dependent kinase family and is involved in regulating the cell cycle. Changes in the cell cycle are of great significance for the occurrence and development of tumor cells; and, in recent years, increasing evidence has suggested that CDK5 exists in a disordered state in cancer cells. In this study, the authors demonstrate that CDK5 functions as an oncogene in TSCC and might serve as a molecular marker for use in the diagnosis and treatment of TSCC.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , MicroARNs , Carcinoma de Células Escamosas de Cabeza y Cuello , Neoplasias de la Lengua , Fosfatasas cdc25 , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/patología , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
11.
Anticancer Drugs ; 33(1): e349-e361, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34407051

RESUMEN

Circular RNAs (circRNAs) play critical roles in regulating the radiosensitivity of various cancers, including esophageal squamous cell carcinoma (ESCC). This research aimed to explore the role and potential mechanism of hsa_circ_0014879 in regulating ESCC radioresistance. The levels of hsa_circ_0014879, microRNA-519-3p (miR-519-3p) and cell division cycle 25A (CDC25A) were measured using quantitative real-time PCR or western blot. Cell proliferation was evaluated by colony formation assay. Cell migration and invasion were assessed by transwell and scratch assays. The levels of epithelial-mesenchymal transition (EMT)-related proteins were detected by western blot. Xenograft assay was used to analyze the effect of hsa_circ_0014879 on radiosensitivity in vivo. The binding relationship among hsa_circ_0014879, miR-519-3p and CDC25A was confirmed by dual-luciferase reporter assay. Hsa_circ_0014879 and CDC25A were upregulated, whereas miR-519-3p was downregulated in radio-resistant ESCC tissues and cells. Depletion of hsa_circ_0014879 suppressed the proliferation, migration and invasion of radio-resistant ESCC cells. Hsa_circ_0014879 knockdown elevated radiosensitivity of radio-resistant cells by modulating miR-519-3p. Moreover, miR-519-3p enhanced the radiosensitivity of radio-resistant cells by targeting CDC25A. Also, hsa_circ_0014879 upregulated CDC25A via sponging miR-519-3p. Hsa_circ_0014879 silencing enhanced the radiosensitivity of ESCC via regulating the miR-519-3p/CDC25A pathway.


Asunto(s)
Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , MicroARNs/metabolismo , ARN Circular/genética , Tolerancia a Radiación/genética , Fosfatasas cdc25/metabolismo , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Clin Exp Pharmacol Physiol ; 49(11): 1209-1220, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36184488

RESUMEN

Circular RNA (circRNAs) Fibronectin Type III Domain Containing 3B (FNDC3B) (circFNDC3B) has been revealed to be involved in the progression of oesophageal squamous cell carcinoma (ESCC). Hence, the potential regulatory network of circFNDC3B in ESCC was further investigated. Levels of genes and proteins were examined by qRT-PCR and Western blot. In vitro assays were performed using colony formation assay, 5-Ethynyl-2'-deoxyuridine (EdU) assay, flow cytometry, wound healing assay, and transwell assay. The target relationship between miR-214-3p and circFNDC3B or cell division cycle 25 homologue A (CDC25A) was verified by dual-luciferase reporter and RIP assays. In vivo assay was carried out using the xenograft nude mice model. CircFNDC3B was highly expressed in ESCC, and high circFNDC3B expression was tightly associated with poor prognosis in ESCC patients. Functionally, circFNDC3B knockdown not only suppressed ESCC cell growth, migration and invasion in vitro, but hindered ESCC tumour growth in vivo. Mechanistically, circFNDC3B acted as a sponge for miR-214-3p to up-regulate the expression of its target CDC25A. Rescue experiments showed that miR-214-3p inhibitor reversed the anticancer effects of circFNDC3B knockdown. Moreover, forced expression of miR-214-3p suppressed the malignant phenotypes mentioned above, while this condition was abolished by CDC25A overexpression. CircFNDC3B silencing restrains the tumorigenesis of oesophageal squamous cell carcinoma through miR-214-3p/CDC25A axis, which opens a new window to the development of novel therapeutic strategy for ESCC patients.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , MicroARNs , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
13.
Genes Dev ; 28(4): 384-95, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24478331

RESUMEN

The vertebrate body forms from a multipotent stem cell-like progenitor population that progressively contributes newly differentiated cells to the most posterior end of the embryo. How the progenitor population balances proliferation and other cellular functions is unknown due to the difficulty of analyzing cell division in vivo. Here, we show that proliferation is compartmentalized at the posterior end of the embryo during early zebrafish development by the regulated expression of cdc25a, a key controller of mitotic entry. Through the use of a transgenic line that misexpresses cdc25a, we show that this compartmentalization is critical for the formation of the posterior body. Upon misexpression of cdc25a, several essential T-box transcription factors are abnormally expressed, including Spadetail/Tbx16, which specifically prevents the normal onset of myoD transcription, leading to aberrant muscle formation. Our results demonstrate that compartmentalization of proliferation during early embryogenesis is critical for both extension of the vertebrate body and differentiation of the multipotent posterior progenitor cells to the muscle cell fate.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre/citología , Pez Cebra/embriología , Pez Cebra/genética , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo , Animales , Diferenciación Celular , División Celular , Proliferación Celular , Células Musculares/citología , Fosforilación , Células Madre/enzimología , Proteínas de Dominio T Box/genética , Proteínas de Pez Cebra/genética
14.
Carcinogenesis ; 42(2): 232-242, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32816038

RESUMEN

More than a million cases of cutaneous squamous cell carcinoma are diagnosed in the USA each year, and its incidence is increasing. Most of these malignancies arise from premalignant lesions, providing an opportunity for intervention before malignant progression. We previously documented how cytoplasmic mislocalization of CDC25A in premalignant and malignant skin cancers confers resistance to apoptotic cell death via a mechanism that depends on its interaction with 14-3-3ε. From these data, we hypothesized that 14-3-3ε overexpression drives skin tumor development and progression, such that targeting 14-3-3ε may be a useful strategy for skin cancer treatment. Like CDC25A, 14-3-3ε was overexpressed and mislocalized to the cytoplasm of both benign and malignant human skin cancer. Skin-targeted deletion of the 14-3-3ε gene reduced skin tumor development by 75% and blocked malignant progression. 14-3-3ε suppressed apoptosis through activation of Akt, leading to inhibition of BCL2 associated agonist of cell death and upregulation of Survivin. Using virtual tetrapeptide libraries, we developed a novel peptide that specifically blocked 14-3-3ε heterodimerization and thereby prevented its interaction with CDC25A. The peptide reduced prosurvival signaling, killed skin cancer cells and reduced skin tumor growth in xenograft. Normal skin keratinocytes were unaffected by inhibition or deletion of 14-3-3ε. Thus, targeting of 14-3-3ε dimerization is a promising strategy for the treatment of premalignant skin lesions.


Asunto(s)
Proteínas 14-3-3/antagonistas & inhibidores , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Fosfatasas cdc25/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Femenino , Humanos , Queratinocitos , Masculino , Ratones , Ratones Noqueados , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Multimerización de Proteína/efectos de los fármacos , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol/administración & dosificación , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
15.
EMBO J ; 36(24): 3666-3681, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29150431

RESUMEN

Mutations of microcephalin (MCPH1) can cause the neurodevelopmental disorder primary microcephaly type 1. We previously showed that MCPH1 deletion in neural stem cells results in early mitotic entry that distracts cell division mode, leading to exhaustion of the progenitor pool. Here, we show that MCPH1 interacts with and promotes the E3 ligase ßTrCP2 to degrade Cdc25A independent of DNA damage. Overexpression of ßTrCP2 or the knockdown of Cdc25A remedies the high mitotic index and rescues the premature differentiation of Mcph1-deficient neuroprogenitors in vivo MCPH1 itself is degraded by APC/CCdh1, but not APC/CCdc20, in late mitosis and G1 phase. Forced MCPH1 expression causes cell death, underlining the importance of MCPH1 turnover after mitosis. Ectopic expression of Cdh1 leads to premature differentiation of neuroprogenitors, mimicking differentiation defects of Mcph1-knockout neuroprogenitors. The homeostasis of MCPH1 in association with the ubiquitin-proteasome system ensures mitotic entry independent of cell cycle checkpoint. This study provides a mechanistic understanding of how MCPH1 controls neural stem cell fate and brain development.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Fosfatasas cdc25/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Línea Celular , Proteínas del Citoesqueleto , Daño del ADN , Técnicas de Inactivación de Genes , Homeostasis , Humanos , Ratones , Mitosis , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/enzimología , Células-Madre Neurales/fisiología , Neurogénesis/genética , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas/genética , Proteínas con Repetición de beta-Transducina/genética , Fosfatasas cdc25/genética
16.
J Cell Sci ; 132(2)2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30635443

RESUMEN

Chk1 (encoded by CHEK1 in mammals) is an evolutionarily conserved protein kinase that transduces checkpoint signals from ATR to Cdc25A during the DNA damage response (DDR). In mammals, Chk1 also controls cellular proliferation even in the absence of exogenous DNA damage. However, little is known about how Chk1 regulates unperturbed cell cycle progression, and how this effect under physiological conditions differs from its regulatory role in DDR. Here, we have established near-diploid HCT116 cell lines containing endogenous Chk1 protein tagged with a minimum auxin-inducible degron (mAID) through CRISPR/Cas9-based gene editing. Establishment of these cells enabled us to induce specific and rapid depletion of the endogenous Chk1 protein, which resulted in aberrant accumulation of DNA damage factors that induced cell cycle arrest at S or G2 phase. Cdc25A was stabilized upon Chk1 depletion before the accumulation of DNA damage factors. Simultaneous depletion of Chk1 and Cdc25A partially suppressed the defects caused by Chk1 single depletion. These results indicate that, similar to its function in DDR, Chk1 controls normal cell cycle progression mainly by inducing Cdc25A degradation.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Daño del ADN , Puntos de Control de la Fase G2 del Ciclo Celular , Proteolisis , Puntos de Control de la Fase S del Ciclo Celular , Fosfatasas cdc25/metabolismo , Sistemas CRISPR-Cas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Edición Génica , Células HCT116 , Humanos , Fosfatasas cdc25/genética
17.
Biochem Biophys Res Commun ; 571: 96-103, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34314996

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) has been verified as a really common cancer worldwide. Several studies have suggested that the suppression of malignancy growth can be traced to miR-199a-5p. Even though CDC25A could activate the tumorigenesis of various cancer by modulating cell cycle, the modulation of the miR-199a-5p/CDC25A axis is still not clear in HCC. Our aim is to identify the modulation of the miR-199a-5p/CDC25A axis in HCC. METHODS: The expression of CDC25A and miR-199a-5p in HCC cells and tissues was assessed using qRT-PCR. After using western blot assay to confirm the protein level, luciferase reporter and RNA pull-down assays were performed to explore the relation between CDC25A and miR-199a-5p. Functional assays such as CCK8 assay, BrdU proliferation assay and flow cytometry analysis identified the cell progression. RESULTS: Experimental findings indicated the downregulation of miR-199a-5p in HCC samples. It was also found that miR-199a-5p overexpression declined the development of the cells with HCC and that it could bind to CDC25A to suppress the progression of HCC. CONCLUSION: Research suggested that miR-199a-5p could restrain the proliferation ability of HCC cells by regulating CDC25A, thus inducing cell-cycle arrest.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Fosfatasas cdc25/metabolismo , Carcinoma Hepatocelular/patología , Puntos de Control del Ciclo Celular , Proliferación Celular , Humanos , Neoplasias Hepáticas/patología , MicroARNs/genética , Células Tumorales Cultivadas , Fosfatasas cdc25/genética
18.
Biochem Biophys Res Commun ; 553: 17-24, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33756341

RESUMEN

Breast cancer (BC) threatens the life and health of women worldwide because of its high morbidity and mortality. The present study aimed to explore the biological functions and potential mechanism of BTNL9 in BC. RNA sequence and clinical data extracted from the Kaplan-Meier plotter database and The Cancer Genome Atlas (TCGA) were utilized to analyze the relationship between the expression level of BTNL9 in BC tissues and clinicopathological features and the effects of BTNL9 expression on the prognosis of BC. The diagnostic efficacy of BTNL9 expression was estimated by receiver operating characteristic (ROC) curve analysis. The mRNA and protein expression levels of BTNL9 in BC cell lines and in BC tissue were determined by quantitative real-time PCR (qPCR) and western blotting, respectively. The functions of BTNL9 were measured by colony formation, CCK-8, Transwell, flow cytometry and EdU assays. Western blotting analysis was also performed to explore the latent mechanism of BTNL9. The results showed that the expression of BTNL9 declined in BC tissues and cell lines. Low expression of BTNL9 was significantly associated with early progression of T stage, relapse-free survival (RFS), and poor overall survival (OS). Ectopic expression of BTNL9 inhibited cell proliferation, colony formation and metastasis and induced apoptosis in BC, while knockdown of BTNL9 had the opposite result. Furthermore, BTNL9 blocked BC cells in the G2/M phase via the P53/CDC25C and P53/GADD45 pathways. Our results suggest that BTNL9 may play a tumor-suppressive role in BC and has the potency to become a new biomarker for early BC diagnosis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Butirofilinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Regulación hacia Abajo , Metástasis de la Neoplasia , Proteína p53 Supresora de Tumor/metabolismo , Fosfatasas cdc25/metabolismo , Apoptosis , Neoplasias de la Mama/patología , Butirofilinas/deficiencia , Butirofilinas/genética , Línea Celular Tumoral , Movimiento Celular , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Puntos de Control de la Fase M del Ciclo Celular , Invasividad Neoplásica/patología , Pronóstico , Linfocitos T/inmunología , Proteínas Supresoras de Tumor/metabolismo
19.
Biochem Biophys Res Commun ; 585: 103-110, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34800881

RESUMEN

OBJECTIVE: This study aimed to screen pyroptosis-related genes influencing the therapeutic effect of dehydroabietic acid in liver cancer and to construct an effective survival prognostic nomogram model. METHODS: Differentially expressed genes (DEGs) between liver cancer tissues and normal tissues were analyzed with The Cancer Genome Atlas database, weighted gene coexpression network analysis and a genetic expression compilation database. The targets of dehydroabietic acid were screened with databases such as TCMSP and pharmacy. Spearman correlation analysis was analyzed. The prognosis model was built through one-factor Cox analysis and LASSO regression. The final core targets were screened by prognosis-related genes combined with a protein-protein interaction (PPI) network. On this basis, the survival nomogram was constructed. The effects of different concentrations of dehydroabietic acid on the growth of HepG2 liver cancer cells were detected by CCK8. Moreover, the expression of related genes was further verified through real-time fluorescence quantitative PCR and Western blot. RESULTS: Venn diagram analysis of DEGs of liver cancer in three databases was performed, through which 890 genes related to the genesis and development of liver cancer were acquired. According to Venn diagram analysis of targets of dehydroabietic acid and related genes of liver cancer, 44 intersecting targets for liver cancer treatment with dehydroabietic acid were acquired. Then, 7 prognosis-related genes were identified through one-factor Cox analysis and LASSO regression of 25 related genes. Next, 10 targets were screened through the PPI network, and the intersection was processed, thus obtaining 3 ultimate core targets of KIF11, CCNA2 and CDC25A. The IC50 of dehydroabietic acid is 23.22 ± 0.98 µg/mL. According to further verification of related genes, the mRNA and protein levels of KIF11, CCNA2 and CDC25A decrease significantly after treatment with dehydroabietic acid. The nomogram shows that T stage is an independent risk factor, and the postoperative survival C-index of the model group was 0.709. CONCLUSIONS: Three pyroptosis-related genes that influence the therapeutic effect of dehydroabietic acid in liver cancer were screened through bioinformatics methods. The survival prognostic nomogram model, which is built based on independent risk factors that influence the postoperative survival of patients in the T stage, has good accuracy and can provide references for clinical and fundamental studies in the future.


Asunto(s)
Abietanos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Detección Precoz del Cáncer , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Piroptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Ciclina A2/genética , Ciclina A2/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Células Hep G2 , Humanos , Estimación de Kaplan-Meier , Cinesinas/genética , Cinesinas/metabolismo , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Masculino , Nomogramas , Piroptosis/genética , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
20.
Bioorg Med Chem Lett ; 48: 128265, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34273487

RESUMEN

Cdc25B phosphatase catalyzes the dephosphorylation and activation of cyclin-dependent kinases 2 (CDK2/CycA) and their overexpression has been reported in cancers. Although Cdc25B has received much attention as a drug target, its flat and featureless surface makes it challenging to develop new agents targeting this protein. In this study, we investigated the rational design of a series of bivalent triazine-based derivatives with the aim of simultaneously targeting the active site and the remote hotspot critical for the interaction with CDK2/CycA. Compounds 1e and 10, containing aromatic residues, were shown to inhibit Cdc25B activity selectively over Cdc25A at low micromolar concentration.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Triazinas/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Dominio Catalítico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Triazinas/síntesis química , Triazinas/química , Fosfatasas cdc25/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA