Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.375
Filtrar
1.
Biol Direct ; 19(1): 91, 2024 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-39396994

RESUMEN

The complex network governing self-renewal in epidermal stem cells (EPSCs) is only partially defined. FOXM1 is one of the main players in this network, but the upstream signals regulating its activity remain to be elucidated. In this study, we identify cyclin-dependent kinase 1 (CDK1) as the principal kinase controlling FOXM1 activity in human primary keratinocytes. Mass spectrometry identified CDK1 as a key hub in a stem cell-associated protein network, showing its upregulation and interaction with essential self renewal-related markers. CDK1 phosphorylates FOXM1 at specific residues, stabilizing the protein and enhancing its nuclear localization and transcriptional activity, promoting self-renewal. Additionally, FOXM1 binds to the CDK1 promoter, inducing its expression.We identify the CDK1-FOXM1 feedforward loop as a critical axis sustaining EPSCs during in vitro cultivation. Understanding the upstream regulators of FOXM1 activity offers new insights into the biochemical mechanisms underlying self-renewal and differentiation in human primary keratinocytes.


Asunto(s)
Proteína Quinasa CDC2 , Células Epidérmicas , Proteína Forkhead Box M1 , Queratinocitos , Células Madre , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/genética , Humanos , Queratinocitos/metabolismo , Queratinocitos/citología , Células Madre/metabolismo , Células Madre/citología , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Células Epidérmicas/metabolismo , Fosforilación , Diferenciación Celular , Epidermis/metabolismo , Células Cultivadas
2.
J Med Chem ; 67(20): 18247-18264, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-39388374

RESUMEN

Selective degradation of cyclin-dependent kinases 12 and 13 (CDK12/13) emerges as a new potential therapeutic approach for triple-negative breast cancer (TNBC) and other human cancers. While several proteolysis-targeting chimera (PROTAC) degraders of CDK12/13 were reported, none are orally bioavailable. Here, we report the discovery of ZLC491 as a potent, selective, and orally bioavailable CDK12/13 PROTAC degrader. The compound effectively degraded CDK12 and CDK13 with DC50 values of 32 and 28 nM, respectively, in TNBC MDA-MB-231 cells. Global proteomic assessment and mechanistic studies revealed that ZLC491 selectively induced CDK12/13 degradation in a cereblon- and proteasome-dependent manner. Furthermore, the molecule efficiently suppressed transcription and expression of long genes, predominantly a subset of genes associated with DNA damage response, and significantly inhibited proliferation of multiple TNBC cell lines. Importantly, ZLC491 achieved an oral bioavailability of 46.8% in rats and demonstrated potent in vivo degradative effects on CDK12/13 in an MDA-MB-231 xenografted mouse model.


Asunto(s)
Antineoplásicos , Disponibilidad Biológica , Quinasas Ciclina-Dependientes , Proteolisis , Humanos , Animales , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Ratas , Proteolisis/efectos de los fármacos , Administración Oral , Proliferación Celular/efectos de los fármacos , Ratones , Femenino , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Descubrimiento de Drogas , Ensayos Antitumor por Modelo de Xenoinjerto , Ratas Sprague-Dawley , Ratones Desnudos , Relación Estructura-Actividad , Proteína Quinasa CDC2
3.
Mol Biol Cell ; 35(11): ar141, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39356777

RESUMEN

To ensure genomic fidelity, a series of spatially and temporally coordinated events is executed during prometaphase of mitosis, including bipolar spindle formation, chromosome attachment to spindle microtubules at kinetochores, the correction of erroneous kinetochore-microtubule (k-MT) attachments, and chromosome congression to the spindle equator. Cyclin A/Cdk1 kinase plays a key role in destabilizing k-MT attachments during prometaphase to promote correction of erroneous k-MT attachments. However, it is unknown whether Cyclin A/Cdk1 kinase regulates other events during prometaphase. Here, we investigate additional roles of Cyclin A/Cdk1 in prometaphase by using an siRNA knockdown strategy to deplete endogenous Cyclin A from human cells. We find that depleting Cyclin A significantly extends mitotic duration, specifically prometaphase, because chromosome alignment is delayed. Unaligned chromosomes display erroneous monotelic, syntelic, or lateral k-MT attachments suggesting that bioriented k-MT attachment formation is delayed in the absence of Cyclin A. Mechanistically, chromosome alignment is likely impaired because the localization of the kinetochore proteins BUB1 kinase, KNL1, and MPS1 kinase are reduced in Cyclin A-depleted cells. Moreover, we find that Cyclin A promotes BUB1 kinetochore localization independently of its role in destabilizing k-MT attachments. Thus, Cyclin A/Cdk1 facilitates chromosome alignment during prometaphase to support timely mitotic progression.


Asunto(s)
Proteína Quinasa CDC2 , Segregación Cromosómica , Ciclina A , Cinetocoros , Microtúbulos , Mitosis , Huso Acromático , Humanos , Proteína Quinasa CDC2/metabolismo , Cinetocoros/metabolismo , Mitosis/fisiología , Microtúbulos/metabolismo , Huso Acromático/metabolismo , Ciclina A/metabolismo , Segregación Cromosómica/fisiología , Células HeLa , Prometafase , Proteínas Serina-Treonina Quinasas/metabolismo , Cromosomas Humanos/metabolismo , Proteínas de Ciclo Celular/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/genética
4.
Int J Mol Sci ; 25(19)2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39409153

RESUMEN

Cyclin-dependent kinase 1 (Cdk1) is a master regulator of the G2-M transition between DNA replication and cell division. This study investigates the regulation of cardiomyocyte (CM) proliferation during the early neonatal period and following ischemic injury in adult mice. We analyzed cell cycle dynamics with the assessment of DNA synthesis, and cytokinesis in murine hearts during the first 15 days after birth. A distinct proliferative block was observed at 1 day, followed by a second wave of DNA synthesis at 4 days, leading to CM binucleation (CMBN) by day 5. Genome-wide mRNA profiling revealed the differential expression of cell cycle regulatory genes during this period, with a downregulation of factors involved in cell division and mitosis. The loss of Cdk1 impaired CMBN but extended the neonatal CM proliferation window until day 10 post-birth. In adult hearts, the cardiac-specific ablation of Cdk1 triggered CM proliferation post-myocardial-infarction (MI) in specific zones, driven by the activation of EGFR1 signaling and suppression of the anti-proliferative p38 and p53 signaling. This was accompanied by restoration of fractional shortening, mitochondrial function, and decreased reactive oxygen species. Additionally, cardiac hypertrophy was mitigated, and survival rates post-MI were increased in Cdk1-knockout mice. These findings reveal a novel role of Cdk1 in regulating cell cycle exit and re-entry in differentiated CMs and offer insights into potential strategies for cardiac repair.


Asunto(s)
Proteína Quinasa CDC2 , Proliferación Celular , Ratones Noqueados , Infarto del Miocardio , Miocitos Cardíacos , Animales , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/genética , Ratones , Animales Recién Nacidos , Transducción de Señal
5.
Cell Rep Med ; 5(10): 101752, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39353441

RESUMEN

Cyclin-dependent kinases 12/13 play pivotal roles in orchestrating transcription elongation, DNA damage response, and maintenance of genomic stability. Biallelic CDK12 loss has been documented in various malignancies. Here, we develop a selective CDK12/13 PROTAC degrader, YJ9069, which effectively inhibits proliferation in subsets of prostate cancer cells preferentially over benign immortalized cells. CDK12/13 degradation rapidly triggers gene-length-dependent transcriptional elongation defects, leading to DNA damage and cell-cycle arrest. In vivo, YJ9069 significantly suppresses prostate tumor growth. Modifications of YJ9069 yielded an orally bioavailable CDK12/13 degrader, YJ1206, which exhibits comparable efficacy with significantly less toxicity. To identify pathways synthetically lethal upon CDK12/13 degradation, phosphorylation pathway arrays were performed using cell lines treated with YJ1206. Interestingly, degradation or genetic knockdown of CDK12/13 led to activation of the AKT pathway. Targeting CDK12/13 for degradation, in conjunction with inhibiting the AKT pathway, resulted in a synthetic lethal effect in preclinical prostate cancer models.


Asunto(s)
Quinasas Ciclina-Dependientes , Proteínas Proto-Oncogénicas c-akt , Mutaciones Letales Sintéticas , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Masculino , Quinasas Ciclina-Dependientes/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Ratones , Proliferación Celular/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Administración Oral , Proteolisis/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ratones Desnudos , Daño del ADN/efectos de los fármacos , Disponibilidad Biológica , Proteína Quinasa CDC2
6.
Cell Rep Med ; 5(10): 101758, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39368479

RESUMEN

Biallelic loss of cyclin-dependent kinase 12 (CDK12) defines a metastatic castration-resistant prostate cancer (mCRPC) subtype. It remains unclear, however, whether CDK12 loss drives prostate cancer (PCa) development or uncovers pharmacologic vulnerabilities. Here, we show Cdk12 ablation in murine prostate epithelium is sufficient to induce preneoplastic lesions with lymphocytic infiltration. In allograft-based CRISPR screening, Cdk12 loss associates positively with Trp53 inactivation but negatively with Pten inactivation. Moreover, concurrent Cdk12/Trp53 ablation promotes proliferation of prostate-derived organoids, while Cdk12 knockout in Pten-null mice abrogates prostate tumor growth. In syngeneic systems, Cdk12/Trp53-null allografts exhibit luminal morphology and immune checkpoint blockade sensitivity. Mechanistically, Cdk12 inactivation mediates genomic instability by inducing transcription-replication conflicts. Strikingly, CDK12-mutant organoids and patient-derived xenografts are sensitive to inhibition or degradation of the paralog kinase, CDK13. We therein establish CDK12 as a bona fide tumor suppressor, mechanistically define how CDK12 inactivation causes genomic instability, and advance a therapeutic strategy for CDK12-mutant mCRPC.


Asunto(s)
Quinasas Ciclina-Dependientes , Neoplasias de la Próstata , Mutaciones Letales Sintéticas , Masculino , Animales , Humanos , Quinasas Ciclina-Dependientes/metabolismo , Quinasas Ciclina-Dependientes/genética , Ratones , Mutaciones Letales Sintéticas/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Progresión de la Enfermedad , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Inestabilidad Genómica , Transcripción Genética , Organoides/patología , Organoides/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Proliferación Celular/genética , Replicación del ADN/genética , Ratones Noqueados , Línea Celular Tumoral , Ratones Endogámicos C57BL , Proteína Quinasa CDC2
7.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(10): 1220-1228, 2024 Oct 15.
Artículo en Chino | MEDLINE | ID: mdl-39433496

RESUMEN

Objective: To explore the role and clinical significance of cell-cycle dependent kinase 1 (CDK1) and its upstream and downstream molecules in the development of malignant peripheral nerve sheath tumor (MPNST) through the analysis of clinical tissue samples. Methods: A total of 56 tumor samples from MPNST patients ("Tianjin" dataset) who underwent surgical resection, confirmed by histology and pathology between September 2011 and March 2020, along with 17 normal tissue samples, were selected as the research subjects. MPNST-related hub genes were identified through transcriptome sequencing, bioinformatics analysis, immunohistochemistry staining, and survival analysis, and their expression levels and prognostic associations were analyzed. Results: Transcriptome sequencing and bioinformatics analysis revealed that upregulated genes in MPNST were predominantly enriched in cell cycle-related pathways, with CDK1 occupying a central position among all differentially expressed genes. Further differential analysis demonstrated that CDK1 mRNA expression in sarcoma tissues was significantly higher than in normal tissues [based on searching the cancer genome atlas (TCGA) dataset, P<0.05]. In MPNST tissues, CDK1 mRNA expression was not only significantly higher than in normal tissues (based on Tianjin, GSE141438 datasets, P<0.05), but also significantly higher than in neurofibromatosis (NF) and plexiform neurofibromas (PNF) (based on GSE66743 and GSE145064 datasets, P<0.05). Immunohistochemical staining results indicated that the expression rate of CDK1 protein in MPNST tissues was 40.31%. Survival analysis results demonstrated that CDK1 expression was associated with poor prognosis. The survival time of MPNST patients with high CDK1 mRNA expression was significantly lower than that of the low expression group ( P<0.05), and the overall survival trend of patients with positive CDK1 protein expression was worse than that of patients with negative CDK1 expression. Additionally, differential analysis of CDK family genes (CDK1-8) revealed that only CDK1 was significantly upregulated in MPNST, NF, and PNF. Conclusion: Increased expression of CDK1 is associated with poor prognosis in MPNST patients. Compared to other CDK family members, CDK1 exhibits a unique expression pattern, suggesting its potential as a therapeutic target for MPNST.


Asunto(s)
Proteína Quinasa CDC2 , Neoplasias de la Vaina del Nervio , Humanos , Proteína Quinasa CDC2/metabolismo , Pronóstico , Neoplasias de la Vaina del Nervio/metabolismo , Neoplasias de la Vaina del Nervio/patología , Neoplasias de la Vaina del Nervio/genética , Regulación Neoplásica de la Expresión Génica , Biología Computacional , ARN Mensajero/metabolismo , ARN Mensajero/genética , Perfilación de la Expresión Génica , Transcriptoma , Masculino , Regulación hacia Arriba , Relevancia Clínica
8.
Proc Natl Acad Sci U S A ; 121(37): e2413089121, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39231204

RESUMEN

The ubiquitin ligase Anaphase-Promoting Complex/Cyclosome (APC/C) and its regulatory protein Cdc20 play important roles in the control of different stages of mitosis. APC/C associated with Cdc20 is active and promotes metaphase-anaphase transition by targeting for degradation inhibitors of anaphase initiation. Earlier in mitosis, premature action of APC/C is prevented by the mitotic checkpoint (or spindle assembly checkpoint) system, which ensures that anaphase is not initiated until all chromosomes are properly attached to the mitotic spindle. The active mitotic checkpoint system promotes the assembly of a Mitotic Checkpoint Complex (MCC), which binds to APC/C and inhibits its activity. The interaction of MCC with APC/C is strongly enhanced by Cdc20 bound to APC/C. While the association of Cdc20 with APC/C was known to be essential for both these stages of mitosis, it was not known how Cdc20 remains bound in spite of ongoing processes, phosphorylation and ubiquitylation, that stimulate its release from APC/C. We find that MCC strongly inhibits the release of Cdc20 from APC/C by the action of mitotic protein kinase Cdk1-cyclin B. This is not due to protection from phosphorylation of specific sites in Cdc20 that affect its interaction with APC/C. Rather, MCC stabilizes the binding to APC/C of partially phosphorylated forms of Cdc20. MCC also inhibits the autoubiquitylation of APC/C-bound Cdc20 and its ubiquitylation-promoted release from APC/C. We propose that these actions of MCC to maintain Cdc20 bound to APC/C in mitosis are essential for the control of mitosis during active mitotic checkpoint and in subsequent anaphase initiation.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase , Proteínas Cdc20 , Puntos de Control de la Fase M del Ciclo Celular , Mitosis , Proteínas Cdc20/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Humanos , Mitosis/fisiología , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Células HeLa , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ubiquitinación , Fosforilación , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Unión Proteica , Huso Acromático/metabolismo
9.
Cells ; 13(18)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39329697

RESUMEN

The centrosome of the amoebozoan model Dictyostelium discoideum provides the best-established model for an acentriolar centrosome outside the Opisthokonta. Dictyostelium exhibits an unusual centrosome cycle, in which duplication is initiated only at the G2/M transition and occurs entirely during the M phase. Little is known about the role of conserved centrosomal kinases in this process. Therefore, we have generated knock-in strains for Aurora (AurK), CDK1, cyclin B, Nek2, and Plk, replacing the endogenous genes with constructs expressing the respective green fluorescent Neon fusion proteins, driven by the endogenous promoters, and studied their behavior in living cells. Our results show that CDK1 and cyclin B arrive at the centrosome first, already during G2, followed by Plk, Nek2, and AurK. Furthermore, CDK1/cyclin B and AurK were dynamically localized at kinetochores, and AurK in addition at nucleoli. The putative roles of all four kinases in centrosome duplication, mitosis, cytokinesis, and nucleolar dynamics are discussed.


Asunto(s)
Proteína Quinasa CDC2 , Centrosoma , Dictyostelium , Mitosis , Centrosoma/metabolismo , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Dictyostelium/genética , Dictyostelium/metabolismo , Dictyostelium/enzimología , Quinasas Relacionadas con NIMA/metabolismo , Quinasas Relacionadas con NIMA/genética , Ciclina B/metabolismo , Ciclina B/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Cinetocoros/metabolismo , Aurora Quinasas/metabolismo , Aurora Quinasas/genética , Nucléolo Celular/metabolismo
10.
Phys Chem Chem Phys ; 26(38): 24861-24869, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39291452

RESUMEN

Positive feedback loops exist in many biological circuits important for organismal function. In this work, we investigate how temporal delay affects the dynamics of two canonical positive feedback models. We consider models of a genetic toggle switch and a one-way switch with delay added to the feedback terms. We show that long-lasting transient oscillations exist in both models under general conditions and that the duration depends strongly on the magnitude of the delay and initial conditions. We then show the existence of long-lasting oscillations in specific biological examples: the Cdc2-Cyclin B/Wee1 system and a genetic regulatory network. Our results challenge fundamental assumptions underlying oscillatory behavior in biological systems. While generally delayed negative feedback systems are canonical in generating oscillations, we show that delayed positive feedback systems are a mechanism for generating oscillations as well.


Asunto(s)
Retroalimentación Fisiológica , Redes Reguladoras de Genes , Modelos Biológicos , Ciclina B/metabolismo , Proteína Quinasa CDC2/metabolismo
11.
Radiother Oncol ; 200: 110531, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39270987

RESUMEN

BACKGROUND AND PURPOSE: Overcoming radioresistance is a critical challenge in pancreatic ductal adenocarcinoma (PDAC). Our study investigates the targeting of Cyclin-dependent kinase-1 (CDK1) through genetic and pharmaceutical inhibition to radiosensitize PDAC cells. MATERIALS AND METHODS: Mass spectrometry and phosphoproteomics were used to analyze engineered radiation-resistant PDAC cell lines (MIA PaCa-2 and PANC-1) compared to parental controls. The TCGA PDAC database was queried for clinical outcomes and patients were dichotomized based on the median CDK1 mRNA expression. We generated a microRNA-based TET-on inducible shRNA to inhibit CDK1 expression in two PDAC cell lines. We used an orthotopic model of PDAC to test the radiation sensitivity of PDAC tumors with or without doxycycline treatment. We targeted CDK1 activation with a selective CDK1 inhibitor, RO-3306, followed by in vitro experiments employing immunoblotting, immunocytochemistry, and clonogenic assays. RESULTS: Phosphoproteomics analysis revealed that phospho-CDK1 (Tyr15) was significantly elevated in the resistant clones. We found that high CDK1 expression was associated with worse OS in PDAC patients. Radiation exposure increased CDK1 phosphorylation. In MIA PaCa-2 and PANC-1 cells, CDK1 inhibition synergized with radiation therapy to delay tumor growth in vivo. CDK1 inhibition via. RO-3306 resulted in a significant shift of cells into the G2/M phase and disrupted DNA repair after radiation exposure. In vitro, pre-treatment with RO-3306 led to enhanced radiosensitivity of PDAC cells. CONCLUSION: CDK1 plays a crucial role in PDAC radioresistance. Targeting CDK1 with radiotherapy holds promise for further investigation in PDAC treatment.


Asunto(s)
Proteína Quinasa CDC2 , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Tolerancia a Radiación , Humanos , Tolerancia a Radiación/efectos de los fármacos , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Proteína Quinasa CDC2/metabolismo , Ratones , Carcinoma Ductal Pancreático/radioterapia , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Animales , Quinolinas , Tiazoles
13.
Biol Pharm Bull ; 47(8): 1437-1446, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39143009

RESUMEN

Bisdemethoxycurcumin (BDMC) is one of major forms of curcuminoids found in the rhizomes of turmeric. Docetaxel (DTX) is the standard of care for men diagnosed with androgen-independent prostate cancers. Here we report for the first time that BDMC could reinforce the effect of DTX against prostate cancer in vitro and in vivo. In vitro study, PC3 and LNCaP cells were cultured and treated with BDMC and DTX alone or in combination. The effects on cell viability were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis was assessed by annexin V/propidium iodide (PI) staining, while cell cycle was assessed by PI staining. Bax, Bcl-2, caspase, poly(ADP-ribose)polymerase (PARP), cyclin B1 and CDK1 expression were assayed by Western blot. We found that a combination treatment of BDMC (10 µM) with DTX (10 nM) was more effective in the inhibition of PC3 and LNCaP cell growth and induction of apoptosis as well as G2/M arrest, which is accompanied with the significant inhibition of Bcl-2, cyclin B1, CDK1 expression and significant increase of Bax, cleaved caspase-9, cleaved caspase-3 and cleaved PARP, than those by treatment of BDMC or DTX alone. Moreover, in vivo evaluation further demonstrated the superior anticancer efficacy of BDMC and DTX compared to DTX alone in a murine prostate cancer model. These results suggest that BDMC can be an attractive therapeutic candidate in enhancing the efficacy of DTX in prostate cancer treatment.


Asunto(s)
Antineoplásicos , Apoptosis , Diarilheptanoides , Docetaxel , Neoplasias de la Próstata , Masculino , Diarilheptanoides/farmacología , Diarilheptanoides/uso terapéutico , Humanos , Animales , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Docetaxel/farmacología , Docetaxel/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sinergismo Farmacológico , Ciclina B1/metabolismo , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones , Curcumina/análogos & derivados , Curcumina/farmacología , Curcumina/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Taxoides/farmacología , Taxoides/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratones Endogámicos BALB C , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína Quinasa CDC2/metabolismo
14.
J Cancer Res Ther ; 20(4): 1214-1223, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39206984

RESUMEN

INTRODUCTION: Accumulating evidence suggests the significant involvement of GADD45G in the development of various cancers. This study investigates GADD45G's involvement and methylation status in endometrial cancer (EC), along with molecular mechanisms and potential therapies. METHODS: The expression of GADD45G in EC tissues and controls was evaluated using RNA-seq, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting (WB). Methylation-specific PCR (MSP) evaluated GADD45G's methylation status. Protein-protein interaction (PPI) prediction identified potential interactors of GADD45G, and co-immunoprecipitation (co-IP) confirmed GADD45G interact with Cyclin-dependent kinase 1 (CDK1) and cyclin B1 (CCNB1). Several cell behavior assays were conducted in both in vitro and in vivo settings to comprehensively understand the impact of GADD45G dysregulation in EC. RESULTS: Our findings revealed a significant decrease in the expression of GADD45G in endometrial cancer tissues and cells, which was attributed to its methylation status. Reduced GADD45G expression correlated with increased invasive behaviors in EC cells. Furthermore, GADD45G negatively regulated CDK1 and CCNB1, promoting invasive behaviors at transcript and protein levels. CONCLUSION: This study demonstrated that the downregulation of GADD45G, mediated by methylation, facilitates the invasive behaviors of EC cells through interaction with the CDK1/CCNB1. These findings enhance understanding of the molecular mechanisms underlying endometrial cancer and suggest potential therapeutic strategies targeting GADD45G for treatment.


Asunto(s)
Proteína Quinasa CDC2 , Ciclina B1 , Metilación de ADN , Neoplasias Endometriales , Proteinas GADD45 , Regulación Neoplásica de la Expresión Génica , Animales , Femenino , Humanos , Ratones , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Ciclina B1/genética , Ciclina B1/metabolismo , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Neoplasias Endometriales/metabolismo , Proteinas GADD45/genética , Proteinas GADD45/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Reprod Biol ; 24(3): 100929, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39154626

RESUMEN

This study aims to evaluate the effects of nano-ozone solution (NZS) on canine oocyte nuclear maturation, associated with the alterations of antioxidant and oxidant status and cyclin-dependent kinase 1 (CDK1), cyclin B1 gene expressions. Oocytes were cultured in four distinct concentrations of NZS (0.5, 1, 2, and 5 µg/mL) and parthenogenetically activated. The rates of oocytes arrested at the Germinal Vesicle (GV), Germinal Vesicle Breakdown (GVBD), Metaphase I (MI), and Metaphase II (MII) stages were statistically different among groups (P < 0.05). The oocytes cultured in 1 µg/mL NZS yielded the best oocyte maturation rate at the MI and MII stages; however, the lowest maturation and high degeneration rates were observed in Group E. The measurements of Malondialdehyde (MDA), reduced Glutathione (GSH), Superoxide Dismutase (SOD), and Ferric Reducing/Antioxidant Power assay (FRAP) were performed from IVM culture media. No statistical difference was observed in SOD and MDA results (P > 0.05). GSH levels were statistically significant between Group A-Group E (p = 0.003), Group B-Group E (p = 0.045), and Group E-Group D (p = 0.021). The culture media in Group D and Group E had high FRAP concentrations and significantly differed between groups (P < 0.05). CDK1, and cyclin B1 genes, which are subunits of maturation-promoting factor (MPF), are upregulated in Group B and Group C, while are downregulated in oocytes of Group E. This study showed that low, controlled doses of NZS (1 µg/mL) supplementation could improve the meiotic competence of canine oocytes and lead to positive response in expressions of CDK1 and cyclin B1 on the gene level.


Asunto(s)
Antioxidantes , Proteína Quinasa CDC2 , Ciclina B1 , Técnicas de Maduración In Vitro de los Oocitos , Oocitos , Animales , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ciclina B1/metabolismo , Ciclina B1/genética , Proteína Quinasa CDC2/metabolismo , Perros , Antioxidantes/farmacología , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Femenino , Oxidantes/farmacología
16.
J Nanobiotechnology ; 22(1): 521, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39210346

RESUMEN

Tissue-derived extracellular vesicles (EVs) are emerging as pivotal players to maintain organ homeostasis, which show promise as a next-generation candidate for medical use with extensive source. However, the detailed function and therapeutic potential of tissue EVs remain insufficiently studied. Here, through bulk and single-cell RNA sequencing analyses combined with ultrastructural tissue examinations, we first reveal that in situ liver tissue EVs (LT-EVs) contribute to the intricate liver regenerative process after partial hepatectomy (PHx), and that hepatocytes are the primary source of tissue EVs in the regenerating liver. Nanoscale and proteomic profiling further identify that the hepatocyte-specific tissue EVs (Hep-EVs) are strengthened to release with carrying proliferative messages after PHx. Moreover, targeted inhibition of Hep-EV release via AAV-shRab27a in vivo confirms that Hep-EVs are required to orchestrate liver regeneration. Mechanistically, Hep-EVs from the regenerating liver reciprocally stimulate hepatocyte proliferation by promoting cell cycle progression through Cyclin-dependent kinase 1 (Cdk1) activity. Notably, supplementing with Hep-EVs from the regenerating liver demonstrates translational potential and ameliorates insufficient liver regeneration. This study provides a functional and mechanistic framework showing that the release of regenerative Hep-EVs governs rapid liver regeneration, thereby enriching our understanding of physiological and endogenous tissue EVs in organ regeneration and therapy.


Asunto(s)
Proliferación Celular , Vesículas Extracelulares , Hepatectomía , Hepatocitos , Regeneración Hepática , Hígado , Regeneración Hepática/fisiología , Vesículas Extracelulares/metabolismo , Hepatocitos/metabolismo , Animales , Hígado/metabolismo , Ratones , Humanos , Masculino , Ratones Endogámicos C57BL , Medicina Regenerativa/métodos , Proteína Quinasa CDC2/metabolismo , Proteómica
17.
Exp Eye Res ; 247: 110040, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39134132

RESUMEN

Retinoblastoma (RB) is the most common intraocular malignancy among children and presents a certain mortality risk, especially in low- and middle-income countries. Clarifying the molecular mechanisms underlying the onset and progression of retinoblastoma is vital for devising effective cancer treatment approaches. PRMT1, a major type I PRMT, plays significant roles in cancer development. However, its expression and role in retinoblastoma are still unclear. Our research revealed a marked increase in PRMT1 levels in both retinoblastoma tissues and Y79 cells. The overexpression of PRMT1 in Y79 cells promoted their growth and cell cycle progression. Conversely, the suppression of PRMT1 hindered the growth of Y79 cells and impeded cell cycle progression. Mechanistically, PRMT1 mediated the growth of Y79 retinoblastoma cells by targeting the p53/p21/CDC2/Cyclin B pathway. Additionally, the ability of PRMT1 knockdown to suppress cell proliferation was also observed in vivo. Overall, PRMT1 could function as a potential target for therapeutic treatment in individuals with retinoblastoma.


Asunto(s)
Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteína-Arginina N-Metiltransferasas , Proteínas Represoras , Neoplasias de la Retina , Retinoblastoma , Proteína p53 Supresora de Tumor , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Retinoblastoma/patología , Retinoblastoma/metabolismo , Retinoblastoma/genética , Humanos , Proliferación Celular/fisiología , Neoplasias de la Retina/patología , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Regulación Neoplásica de la Expresión Génica , Animales , Ratones , Western Blotting , Ciclo Celular/fisiología , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Línea Celular Tumoral , Ratones Desnudos
18.
Phytomedicine ; 134: 155959, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39178682

RESUMEN

BACKGROUND: ß,ß-Dimethylacrylalkannin (DMAKN), a natural naphthoquinone found in Zicao, a traditional Chinese medicine (TCM), serves as the designated quantitative marker in the Chinese Pharmacopoeia. Despite its established role in assessing Zicao quality, DMAKN's biological potential remains underexplored in research. METHODS: We investigated DMAKN's involvement in Zicao's anti-hepatocellular carcinoma (HCC) properties using a combination of HPLC content analysis and comprehensive bioinformatics. Subsequently, both in vitro and in vivo experiments were conducted to evaluate DMAKN's efficacy against HCC. Mechanistic investigations focused on elucidating DMAKN's impact on cell cycle regulation and induction of cell death. RESULTS: Integrated HPLC analysis and bioinformatics identified DMAKN as the primary active compound responsible for Zicao's anti-HCC activity. In vitro and in vivo studies confirmed DMAKN's potent efficacy against HCC. Notably, DMAKN demonstrated dual effects on HCC cells: inhibiting proliferation at lower doses and inducing rapid cell death at higher doses. Mechanistic insights revealed that low-dose DMAKN induced G2/M phase cell cycle arrest through modulation of CDK1 and Cdc25C phosphorylation, while high-dose DMAKN triggered necrosis. Importantly, high-dose DMAKN caused a sharp increase in intracellular ROS levels in a short time, while low-dose DMAKN gradually increased ROS levels over a long period. Additionally, low-dose DMAKN-induced ROS activated the JNK pathway, crucial for cell cycle arrest, whereas high-dose DMAKN-induced necrosis was ROS-dependent but JNK-independent. CONCLUSION: This study underscores DMAKN's pivotal role as the principal anti-HCC compound in Zicao, delineating its differential effects and underlying mechanisms. These results demonstrate the potential of DMAKN as a therapeutic agent for the treatment of HCC, providing important information for further study and advancement in cancer therapy.


Asunto(s)
Carcinoma Hepatocelular , Puntos de Control del Ciclo Celular , Neoplasias Hepáticas , Necrosis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/tratamiento farmacológico , Humanos , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Necrosis/tratamiento farmacológico , Naftoquinonas/farmacología , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/química , Línea Celular Tumoral , Antineoplásicos Fitogénicos/farmacología , Ratones Desnudos , Ratones , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/metabolismo , Proliferación Celular/efectos de los fármacos , Masculino , Células Hep G2 , Fosfatasas cdc25/metabolismo , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2
19.
J Cell Biol ; 223(11)2024 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-39105756

RESUMEN

Mitosis in early embryos often proceeds at a rapid pace, but how this pace is achieved is not understood. Here, we show that cyclin B3 is the dominant driver of rapid embryonic mitoses in the C. elegans embryo. Cyclins B1 and B2 support slow mitosis (NEBD to anaphase ∼600 s), but the presence of cyclin B3 dominantly drives the approximately threefold faster mitosis observed in wildtype. Multiple mitotic events are slowed down in cyclin B1 and B2-driven mitosis, and cyclin B3-associated Cdk1 H1 kinase activity is ∼25-fold more active than cyclin B1-associated Cdk1. Addition of cyclin B1 to fast cyclin B3-only mitosis introduces an ∼60-s delay between completion of chromosome alignment and anaphase onset; this delay, which is important for segregation fidelity, is dependent on inhibitory phosphorylation of the anaphase activator Cdc20. Thus, cyclin B3 dominance, coupled to a cyclin B1-dependent delay that acts via Cdc20 phosphorylation, sets the rapid pace and ensures mitotic fidelity in the early C. elegans embryo.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Ciclina B1 , Embrión no Mamífero , Mitosis , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/genética , Proteínas Cdc20/metabolismo , Proteínas Cdc20/genética , Ciclina B/metabolismo , Ciclina B/genética , Ciclina B1/metabolismo , Ciclina B1/genética , Ciclina B2/metabolismo , Ciclina B2/genética , Embrión no Mamífero/metabolismo , Fosforilación
20.
Life Sci ; 356: 123021, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39209249

RESUMEN

Chronic caloric restriction triggers unfavorable alterations in cardiac function albeit responsible scenarios remain unclear. This work evaluated the possible involvement of Akt2 in caloric restriction-evoked cardiac geometric and functional changes and responsible processes focusing on autophagy and mitophagy. Akt2 knockout and WT mice were subjected to caloric restriction for 30 weeks prior to assessment of myocardial homeostasis. Caloric restriction compromised echocardiographic parameters (decreased LV wall thickness, LVEDD, stroke volume, cardiac output, ejection fraction, fractional shortening, and LV mass), cardiomyocyte contractile and intracellular Ca2+ capacity, myocardial atrophy, interstitial fibrosis and mitochondrial injury associated with elevated blood glucocorticoids, autophagy (LC3B, p62, Atg7, Beclin-1), and mitophagy (Pink1, Parkin, TOM20), dampened cardiac ATP levels, mitochondrial protein PGC1α and UCP2, anti-apoptotic protein Bcl2, intracellular Ca2+ governing components Na+-Ca2+ exchanger, phosphorylation of SERCA2a, mTOR (Ser2481) and ULK1 (Ser757), and upregulated Bax, phospholamban, phosphorylation of Akt2, AMPK, and ULK1 (Ser555), the responses except autophagy markers (Beclin-1, Atg7), phosphorylation of AMPK, mTOR and ULK1 were negated by Akt2 ablation. Levels of CDK1 and DRP1 phosphorylation were overtly upregulated with caloric restriction, the response was reversed by Akt2 knockout. Caloric restriction-evoked changes in cardiac remodeling and cardiomyocyte function were alleviated by glucocorticoid receptor antagonism, Parkin ablation and Mdivi-1. In vitro experiment indicated that serum deprivation or glucocorticoids evoked GFP-LC3B accumulation and cardiomyocyte dysfunction, which was negated by inhibition of Akt2, CDK1 or DRP1, whereas mitophagy induction reversed Akt2 ablation-evoked cardioprotection. These observations favor a protective role of Akt2 ablation in sustained caloric restriction-evoked cardiac pathological changes via correction of glucocorticoid-induced mitophagy defect in a CDK1-DRP1-dependent manner.


Asunto(s)
Proteína Quinasa CDC2 , Restricción Calórica , Mitofagia , Miocitos Cardíacos , Proteínas Proto-Oncogénicas c-akt , Animales , Masculino , Ratones , Autofagia/fisiología , Proteína Quinasa CDC2/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Remodelación Ventricular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA