Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
PLoS Genet ; 16(11): e1009184, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33137813

RESUMEN

In mitosis and meiosis, chromosome segregation is triggered by the Anaphase-Promoting Complex/Cyclosome (APC/C), a multi-subunit ubiquitin ligase that targets proteins for degradation, leading to the separation of chromatids. APC/C activation requires phosphorylation of its APC3 and APC1 subunits, which allows the APC/C to bind its co-activator Cdc20. The identity of the kinase(s) responsible for APC/C activation in vivo is unclear. Cyclin B3 (CycB3) is an activator of the Cyclin-Dependent Kinase 1 (Cdk1) that is required for meiotic anaphase in flies, worms and vertebrates. It has been hypothesized that CycB3-Cdk1 may be responsible for APC/C activation in meiosis but this remains to be determined. Using Drosophila, we found that mutations in CycB3 genetically enhance mutations in tws, which encodes the B55 regulatory subunit of Protein Phosphatase 2A (PP2A) known to promote mitotic exit. Females heterozygous for CycB3 and tws loss-of-function alleles lay embryos that arrest in mitotic metaphase in a maternal effect, indicating that CycB3 promotes anaphase in mitosis in addition to meiosis. This metaphase arrest is not due to the Spindle Assembly Checkpoint (SAC) because mutation of mad2 that inactivates the SAC does not rescue the development of embryos from CycB3-/+, tws-/+ females. Moreover, we found that CycB3 promotes APC/C activity and anaphase in cells in culture. We show that CycB3 physically associates with the APC/C, is required for phosphorylation of APC3, and promotes APC/C association with its Cdc20 co-activators Fizzy and Cortex. Our results strongly suggest that CycB3-Cdk1 directly activates the APC/C to promote anaphase in both meiosis and mitosis.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Anafase/fisiología , Proteína Quinasa CDC2/metabolismo , Ciclina B/metabolismo , Proteínas de Drosophila/metabolismo , Animales , Animales Modificados Genéticamente , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas Cdc20/metabolismo , Línea Celular , Segregación Cromosómica/fisiología , Ciclina B/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Mutación con Pérdida de Función , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Masculino , Metafase/fisiología , Modelos Animales , Mutagénesis , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación
2.
Nature ; 533(7602): 260-264, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-27120157

RESUMEN

In eukaryotes, the anaphase-promoting complex (APC/C, also known as the cyclosome) regulates the ubiquitin-dependent proteolysis of specific cell-cycle proteins to coordinate chromosome segregation in mitosis and entry into the G1 phase. The catalytic activity of the APC/C and its ability to specify the destruction of particular proteins at different phases of the cell cycle are controlled by its interaction with two structurally related coactivator subunits, Cdc20 and Cdh1. Coactivators recognize substrate degrons, and enhance the affinity of the APC/C for its cognate E2 (refs 4-6). During mitosis, cyclin-dependent kinase (Cdk) and polo-like kinase (Plk) control Cdc20- and Cdh1-mediated activation of the APC/C. Hyperphosphorylation of APC/C subunits, notably Apc1 and Apc3, is required for Cdc20 to activate the APC/C, whereas phosphorylation of Cdh1 prevents its association with the APC/C. Since both coactivators associate with the APC/C through their common C-box and Ile-Arg tail motifs, the mechanism underlying this differential regulation is unclear, as is the role of specific APC/C phosphorylation sites. Here, using cryo-electron microscopy and biochemical analysis, we define the molecular basis of how phosphorylation of human APC/C allows for its control by Cdc20. An auto-inhibitory segment of Apc1 acts as a molecular switch that in apo unphosphorylated APC/C interacts with the C-box binding site and obstructs engagement of Cdc20. Phosphorylation of the auto-inhibitory segment displaces it from the C-box-binding site. Efficient phosphorylation of the auto-inhibitory segment, and thus relief of auto-inhibition, requires the recruitment of Cdk-cyclin in complex with a Cdk regulatory subunit (Cks) to a hyperphosphorylated loop of Apc3. We also find that the small-molecule inhibitor, tosyl-l-arginine methyl ester, preferentially suppresses APC/C(Cdc20) rather than APC/C(Cdh1), and interacts with the binding sites of both the C-box and Ile-Arg tail motifs. Our results reveal the mechanism for the regulation of mitotic APC/C by phosphorylation and provide a rationale for the development of selective inhibitors of this state.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Mitosis , Fosfoproteínas/metabolismo , Secuencias de Aminoácidos , Ciclosoma-Complejo Promotor de la Anafase/química , Ciclosoma-Complejo Promotor de la Anafase/ultraestructura , Antígenos CD , Subunidad Apc1 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc1 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Apoenzimas/metabolismo , Sitios de Unión , Cadherinas/química , Cadherinas/metabolismo , Cadherinas/ultraestructura , Proteínas Cdc20/antagonistas & inhibidores , Proteínas Cdc20/química , Proteínas Cdc20/metabolismo , Proteínas Cdc20/ultraestructura , Microscopía por Crioelectrón , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Activación Enzimática , Humanos , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/ultraestructura , Fosforilación , Unión Proteica , Conformación Proteica , Tosilarginina Metil Éster/farmacología
3.
Metab Brain Dis ; 37(4): 1015-1023, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35098413

RESUMEN

Circular RNA circSLC8A1 is one of the cancer-related circRNAs that is implicated in various cancers. However, studies focusing on the role of circSLC8A1 in glioma is rare. Here we attempted to evaluate the biological function of circSLC8A1 in glioma and explore the potential mechanism. The relative expression of circSLC8A1, miR-214-5p and CDC27 in tissues and cell lines was determined by qRT-PCR. Cell proliferation and invasion were respectively measured by CCK-8 and transwell assays. Protein level of CDC27 was analyzed by western blot. Luciferase reporter assay was performed to confirm the regulatory interaction of cirRNA-miRNA-mRNA. Lowly expressed circSLC8A1 was observed in both glioma tissues and cell lines. Further biological analyses showed that circSLC8A1 inhibits the cell proliferation and invasion of glioma cells. CircSLC8A1 directly sponged miR-214-5p and inhibited miR-214-5p expression in glioma cells. CDC27 was a direct target of miR-214-5p and could be regulated by miR-214-5p. Moreover, miR-214-5p mimics and CDC27 knockdown reversed the inhibitory effects of circSLC8A1 on cell proliferation and invasion. Taken together, our results demonstrated a tumor suppressive role of circSLC8A1 in glioma through regulation of glioma cells proliferation and invasion. The effects of circSLC8A1 were mediated by miR-214-5p/CDC27 axis. Our study provided a new understanding of the occurrence and development of glioma.


Asunto(s)
Glioma , MicroARNs , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética
4.
Nature ; 522(7557): 450-454, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-26083744

RESUMEN

The anaphase-promoting complex (APC/C) is a multimeric RING E3 ubiquitin ligase that controls chromosome segregation and mitotic exit. Its regulation by coactivator subunits, phosphorylation, the mitotic checkpoint complex and interphase early mitotic inhibitor 1 (Emi1) ensures the correct order and timing of distinct cell-cycle transitions. Here we use cryo-electron microscopy to determine atomic structures of APC/C-coactivator complexes with either Emi1 or a UbcH10-ubiquitin conjugate. These structures define the architecture of all APC/C subunits, the position of the catalytic module and explain how Emi1 mediates inhibition of the two E2s UbcH10 and Ube2S. Definition of Cdh1 interactions with the APC/C indicates how they are antagonized by Cdh1 phosphorylation. The structure of the APC/C with UbcH10-ubiquitin reveals insights into the initiating ubiquitination reaction. Our results provide a quantitative framework for the design of future experiments to investigate APC/C functions in vivo.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/ultraestructura , Ubiquitinación , Ciclosoma-Complejo Promotor de la Anafase/química , Antígenos CD , Subunidad Apc1 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc1 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Subunidad Apc1 del Ciclosoma-Complejo Promotor de la Anafase/ultraestructura , Subunidad Apc10 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc10 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Subunidad Apc10 del Ciclosoma-Complejo Promotor de la Anafase/ultraestructura , Subunidad Apc11 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc11 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Subunidad Apc8 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc8 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Subunidad Apc8 del Ciclosoma-Complejo Promotor de la Anafase/ultraestructura , Cadherinas/química , Cadherinas/metabolismo , Cadherinas/ultraestructura , Dominio Catalítico , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/ultraestructura , Microscopía por Crioelectrón , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Proteínas F-Box/química , Proteínas F-Box/metabolismo , Proteínas F-Box/ultraestructura , Humanos , Lisina/metabolismo , Modelos Moleculares , Fosforilación , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitina/ultraestructura , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/ultraestructura
5.
BMC Genomics ; 19(1): 538, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012096

RESUMEN

BACKGROUND: Esophageal squamous cell carcinomas (ESCC) is the fourth most lethal cancer in China. Previous studies reveal several highly conserved mutational processes in ESCC. However, it remains unclear what are the true regulators of the mutational processes. RESULTS: We analyzed the somatic mutational signatures in 302 paired whole-exome sequencing data of ESCC in a Chinese population for potential regulators of the mutational processes. We identified three conserved subtypes based on the mutational signatures with significantly different clinical outcomes. Our results show that patients of different subpopulations of Chinese differ significantly in the activity of the "NpCpG" signature (FDR = 0.00188). In addition, we report ZNF750 and CDC27, of which the somatic statuses and the genetic burdens consistently influence the activities of specific mutational signatures in ESCC: the somatic ZNF750 status is associated with the AID/APOBEC-related mutational process (FDR = 0.0637); the somatic CDC27 copy-number is associated with the "NpCpG" (FDR = 0.00615) and the AID/APOBEC-related mutational processes (FDR = 8.69 × 10- 4). The burdens of germline variants in the two genes also significantly influence the activities of the same somatic mutational signatures (FDR < 0.1). CONCLUSIONS: We report multiple factors that influence the mutational processes in ESCC including: the subpopulations of Chinese; the germline and somatic statuses of ZNF750 and CDC27 and exposure to alcohol and tobacco. Our findings based on the evidences from both germline and somatic levels reveal potential genetic regulators of the somatic mutational processes and provide insights into the biology of esophageal carcinogenesis.


Asunto(s)
Pueblo Asiatico/genética , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Carcinoma de Células Escamosas/patología , China , Variaciones en el Número de Copia de ADN , Bases de Datos Genéticas , Neoplasias Esofágicas/patología , Sitios Genéticos , Predisposición Genética a la Enfermedad , Genoma Humano , Genotipo , Células Germinativas/metabolismo , Humanos , Mutación INDEL , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor
6.
Cell Physiol Biochem ; 50(2): 501-511, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30308498

RESUMEN

BACKGROUND/AIMS: Lymph node metastasis is the primary cause of cancer-related death among patients with gastric cancer (GC), and cell division cycle 27 (CDC27) promotes the metastasis and epithelial-mesenchymal transition in many cancers. Till now, the mechanisms underlying CDC27-induced the epithelial-mesenchymal transition (EMT) of GC are still unclear. METHODS: We analyzed the expression levels of CDC27 and EMT-related biomarkers using immunohistochemistry and Western blot in 60 cases of GC tissues, and then GC cells with CDC27 shRNAs or plasmids were subjected to in vitro and in vivo assays, including CCK-8, wound healing and transwell assays. RESULTS: The CDC27 expression was obviously increased in GC tissues, and significantly correlates with EMT-related biomarkers, lymph node metastasis and poor 5-year overall survival. Additionally, in vitro and in vivo assays demonstrated that silencing of CDC27 expression effectively inhibited GC cell proliferation, invasion and metastasis. Conversely, CDC27 overexpression led to the opposite results. Finally, we demonstrated that Twist shRNA inhibited CDC27-meditated invasion and EMT of GC cells. CONCLUSION: CDC27 facilitates gastric cancer cell proliferation, invasion and metastasis via Twist-induced EMT; thus, this study offered a new therapy method for GC patients.


Asunto(s)
Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Gástricas/patología , Proteína 1 Relacionada con Twist/metabolismo , Adulto , Anciano , Animales , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Humanos , Metástasis Linfática , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad , Tasa de Supervivencia , Proteína 1 Relacionada con Twist/antagonistas & inhibidores , Proteína 1 Relacionada con Twist/genética
7.
J Cell Biochem ; 118(10): 3150-3157, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28004426

RESUMEN

The aim of this study was to investigate the possibility of APC/CCdh1 as a potential therapeutic target in the radiosensitivity of nasopharyngeal carcinoma (NPC) cell CNE-1, and explain the role of APC subunits after silence of Cdh1 combined with radiotherapy. Transfection with Cdh1 shRNA significantly increased the radiosensitivity of CNE-1 cells and the radiation enhancement ratio (RER) of sh-Cdh1 cells was 1.76. Knockdown of Cdh1 in CNE-1 cells increased irradiation induced apoptosis and G2/M phase cell cycle arrest. The levels of CDC20 and CylinB1 increased and the levels of Ku70 and APC3 decreased after irradiation. APC/CCdh1 is involved in regulation of radiosensitivity in human NPC CNE-1 cells. Our study may provide a promising therapeutic strategy for NPC by targeting Cdh1. J. Cell. Biochem. 118: 3150-3157, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Apoptosis , Cadherinas/metabolismo , Carcinoma/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Tolerancia a Radiación , Antígenos CD , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Cadherinas/genética , Carcinoma/genética , Carcinoma/radioterapia , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/radioterapia
8.
Biochem Biophys Res Commun ; 485(4): 820-825, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28257844

RESUMEN

Ubiquitin-conjugating enzyme E2S (UBE2S), a family of E2 protein in the ubiquitin-proteasome system, is highly expressed in several types of cancers; however, its roles in oral squamous cell carcinoma (OSCC) have not yet been well elucidated. The purpose of this study was to clarify the functional activities of UBE2S in OSCCs. We analyzed the expression levels of UBE2S in nine OSCC cell lines and primary OSCC tissues by quantitative reverse transcriptase-polymerase chain reaction, Western blotting, and immunohistochemistry (IHC). The correlations between UBE2S expression and clinical classifications of OSCCs were analyzed using the IHC scoring system. We also used UBE2S knockdown OSCC cells for functional assays (proliferation assay, flow cytometry, and Western blotting). UBE2S was overexpressed in OSCCs in vitro and in vivo and was correlated significantly (P < 0.05) with the primary tumoral size. The cellular growth was decreased and the cell-cycle was arrested in the G2/M phase in the UBE2S knockdown (shUBE2S) cells. The expression level of P21, a target of the ubiquitin-proteasome system, was increased in the shUBE2S cells because of lower anaphase activity that promotes complex subunit 3 (APC3), an E3 ubiquitin ligase, compared with shMock cells. These findings might promote the understanding of the relationship between UBE2S overexpression and oral cancer proliferation, indicating that UBE2S would be a potential biomarker of and therapeutic target in OSCCs.


Asunto(s)
Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias de la Boca/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Anciano , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Citometría de Flujo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética
9.
Biochem Biophys Res Commun ; 471(4): 497-502, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26882976

RESUMEN

Elmo has no intrinsic catalytic activity but coordinate multiple cellular processes via their interactions with other proteins. Studies thus have been focused on identifying Elmo binding partners, but the number of characterized Elmo-interacting proteins remains limited. Here, we report Cdc27 as a novel Elmo1-interacting protein. In yeast and mammalian cells, Cdc27 specifically interacted with the C-terminal region of Elmo1 essential for Dock1 association and function. The interaction of Elmo1 with Dock1 abrogated binding between Elmo1 and Cdc27, but the Dock1-Elmo1 interaction was unaffected by Cdc27. Similarly, cellular phagocytotic functions mediated by the Elmo1-Dock1-Rac module were unaffected by Cdc27 levels. In summary, a novel binding partner, Cdc27, was identified for Elmo1 and they appear to be independent of Elmo-Dock1-Rac-mediated processes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Sitios de Unión , Células HEK293/metabolismo , Humanos , Fagocitosis/fisiología , Mapeo de Interacción de Proteínas/métodos , Proteínas de Unión al GTP rac/metabolismo
10.
Tumour Biol ; 36(7): 5299-304, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25680405

RESUMEN

Cdc27, as a core component of anaphase-promoting complex (APC), is a cell cycle regulator, which participates in control of mitotic checkpoint and surveys the mitotic spindle to maintain chromosomal integrity. It was hypothesized that polymorphisms in cdc27 gene might contribute to the susceptibility of breast cancer (BC) through influencing the mitotic progression of cells. Therefore, a hospital-based case-control study with 463 BC patients and 536 controls was implemented to investigate the association of six single-nucleotide polymorphisms (SNPs) in cdc27 and BC risk in a Chinese Han population. Among the six SNPs, two SNPs of rs11570443 and rs12601027 were positively correlated with BC risk. Individuals carrying rs11570443-CT or CC genotypes showed a higher BC risk with the OR of 1.75 (95 % confidence interval (CI) = 1.13-1.69), compared with those carrying rs11570443-TT genotype. For rs12601027, an increased BC risk was significantly associated with homozygote TT genotype (odds ratio (OR) = 1.49, 95 % CI = 1.12-1.98) compared with homozygote CC and heterozygote CT genotypes. In addition, a significant interaction effect of these two SNPs was found. The rs12601027-TT in combination with rs11570443-CT/CC genotypes showed a strongly elevated risk of BC compared with rs12601027-CC/CT and rs11570443-TT genotype (OR = 1.95, 95 % CI = 1.06-3.59). These findings suggested that polymorphisms in cdc27 may contribute to the susceptibility of BC though functional studies are needed to further elucidate the underling mechanisms of the associations.


Asunto(s)
Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad , Adulto , Anciano , Pueblo Asiatico , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Femenino , Estudios de Asociación Genética , Humanos , Persona de Mediana Edad , Factores de Riesgo
11.
Hum Genomics ; 8: 20, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25496518

RESUMEN

BACKGROUND: Osteosarcoma (OS) is a prevalent primary malignant bone tumour with unknown etiology. These highly metastasizing tumours are among the most frequent causes of cancer-related deaths. Thus, there is an urgent need for different markers, and with our study, we were aiming towards finding novel biomarkers for OS. METHODS: For that, we analysed the whole exome of the tumorous and non-tumour bone tissue from the same patient with OS applying next-generation sequencing. For data analysis, we used several softwares and combined the exome data with RNA-seq data from our previous study. RESULTS: In the tumour exome, we found wide genomic rearrangements, which should qualify as chromotripsis-we detected almost 3,000 somatic single nucleotide variants (SNVs) and small indels and more than 2,000 copy number variants (CNVs) in different chromosomes. Furthermore, the somatic changes seem to be associated to bone tumours, whereas germline mutations to cancer in general. We confirmed the previous findings that the most significant pathway involved in OS pathogenesis is probably the WNT/ß-catenin signalling pathway. Also, the IGF1/IGF2 and IGF1R homodimer signalling and TP53 (including downstream tumour suppressor gene EI24) pathways may have a role. Additionally, the mucin family genes, especially MUC4 and cell cycle controlling gene CDC27 may be considered as potential biomarkers for OS. CONCLUSIONS: The genes, in which the mutations were detected, may be considered as targets for finding biomarkers for OS. As the study is based on a single case and only DNA and RNA analysis, further confirmative studies are required.


Asunto(s)
Exoma , Osteosarcoma/genética , Transcriptoma , Adolescente , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Proteínas Reguladoras de la Apoptosis/genética , Cromosomas Humanos Par 19/genética , Cromosomas Humanos Par 2/genética , Biología Computacional , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Marcadores Genéticos , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Mucina 4/genética , Proteínas Nucleares/genética , Osteosarcoma/diagnóstico , Polimorfismo de Nucleótido Simple , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Análisis de Secuencia de ARN , Transducción de Señal , Programas Informáticos , Proteína p53 Supresora de Tumor/genética , Población Blanca/genética , beta Catenina/genética
12.
Med Sci Monit ; 21: 1297-303, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25943633

RESUMEN

BACKGROUND: MiR-27a is significantly overexpressed in triple-negative breast cancer (TNBC). However, the exact biological function of MiR-27a in TNBC is not fully understood. In this study, we verified miR-27a expression in TNBC cells and explored how its overexpression modulates radiosensitivity of the cells. MATERIAL/METHODS: qRT-PCR analysis was performed to study miR-27a expression in TNBC lines MDA-MB-435 and MDA-MB-231 and in normal human breast epithelial cell line MCF10A. Dual luciferase assay was performed to verify a putative downstream target of miR-27a, CDC27. CCK-8 assay was used to assess the influence of miR-27a-CDC27 axis on cell proliferation under irradiation (IR) treatment. RESULTS: We confirmed significantly higher miR-27a expression in 2 TNBC cell lines--MDA-MB-435 and MDA-MB-231--than in human breast epithelial cell line MCF10A. miR-27a could modulate proliferation and radiosensitivity of TNBC cells. CDC-27 is a direct target of miR-27a and its downregulation conferred increased radioresistance of the cells. CONCLUSIONS: The miR-27a-CDC27 axis might play an important role in modulating response to radiotherapy in TNBC cells. Testing miR-27a expression might be a useful way to identify a subgroup of patients who will benefit from an IR-based therapeutic approach.


Asunto(s)
Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , MicroARNs/fisiología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/radioterapia , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/biosíntesis , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/fisiología , Sitios de Unión , Mama/citología , Línea Celular Tumoral/efectos de la radiación , Células Cultivadas , Secuencia Conservada , Regulación hacia Abajo , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Terapia Molecular Dirigida , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Oligonucleótidos/farmacología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Tolerancia a Radiación/genética , Proteínas Recombinantes de Fusión/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Ensayo de Tumor de Célula Madre
13.
EMBO J ; 29(14): 2407-20, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20526282

RESUMEN

The balance between cell cycle progression and apoptosis is important for both surveillance against genomic defects and responses to drugs that arrest the cell cycle. In this report, we show that the level of the human anti-apoptotic protein Mcl-1 is regulated during the cell cycle and peaks at mitosis. Mcl-1 is phosphorylated at two sites in mitosis, Ser64 and Thr92. Phosphorylation of Thr92 by cyclin-dependent kinase 1 (CDK1)-cyclin B1 initiates degradation of Mcl-1 in cells arrested in mitosis by microtubule poisons. Mcl-1 destruction during mitotic arrest requires proteasome activity and is dependent on Cdc20/Fizzy, which mediates recognition of mitotic substrates by the anaphase-promoting complex/cyclosome (APC/C) E3 ubiquitin ligase. Stabilisation of Mcl-1 during mitotic arrest by mutation of either Thr92 or a D-box destruction motif inhibits the induction of apoptosis by microtubule poisons. Thus, phosphorylation of Mcl-1 by CDK1-cyclin B1 and its APC/C(Cdc20)-mediated destruction initiates apoptosis if a cell fails to resolve mitosis. Regulation of apoptosis, therefore, is linked intrinsically to progression through mitosis and is governed by a temporal mechanism that distinguishes between normal mitosis and prolonged mitotic arrest.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Mitosis/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Secuencia de Aminoácidos , Animales , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Apoptosis/fisiología , Proteína Quinasa CDC2/genética , Caspasa 9/metabolismo , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Línea Celular , Ciclina B1/genética , Humanos , Datos de Secuencia Molecular , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosfopéptidos/genética , Fosfopéptidos/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Interferencia de ARN , Serina/metabolismo , Treonina/metabolismo
14.
Biochem J ; 449(2): 365-71, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23078409

RESUMEN

Mechanistic and structural studies of large multi-subunit assemblies are greatly facilitated by their reconstitution in heterologous recombinant systems. In the present paper, we describe the generation of recombinant human APC/C (anaphase-promoting complex/cyclosome), an E3 ubiquitin ligase that regulates cell-cycle progression. Human APC/C is composed of 14 distinct proteins that assemble into a complex of at least 19 subunits with a combined molecular mass of ~1.2 MDa. We show that recombinant human APC/C is correctly assembled, as judged by its capacity to ubiquitinate the budding yeast APC/C substrate Hsl1 (histone synthetic lethal 1) dependent on the APC/C co-activator Cdh1 [Cdc (cell division cycle) 20 homologue 1], and its three-dimensional reconstruction by electron microscopy and single-particle analysis. Successful reconstitution validates the subunit composition of human APC/C. The structure of human APC/C is compatible with the Saccharomyces cerevisiae APC/C homology model, and in contrast with endogenous human APC/C, no evidence for conformational flexibility of the TPR (tetratricopeptide repeat) lobe is observed. Additional density present in the human APC/C structure, proximal to Apc3/Cdc27 of the TPR lobe, is assigned to the TPR subunit Apc7, a subunit specific to vertebrate APC/C.


Asunto(s)
Complejos Multiproteicos/metabolismo , Proteínas Recombinantes/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Subunidad Apc7 del Ciclosoma-Complejo Promotor de la Anafase , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Humanos , Microscopía Electrónica , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/ultraestructura , Especificidad por Sustrato , Complejos de Ubiquitina-Proteína Ligasa/química , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
15.
Biochemistry ; 52(43): 7618-27, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24073851

RESUMEN

The breast and ovarian cancer susceptibility protein 1 (BRCA1) plays a central role in DNA damage response (DDR). Two tandem BRCA1 C-terminal (BRCT) domains interact with several proteins that function in DDR and contain the generally accepted motif pS-X-X-F (pS denoting phosphoserine and X any amino acid), including the ATR-interacting protein (ATRIP) and the BRCA1-associated protein required for ATM activation-1 (BAAT1). The crystal structures of the BRCA1 BRCTs bound to the phosphopeptides ATRIP (235-PEACpSPQFG-243) and BAAT1 (266-VARpSPVFSS-274) were determined at 1.75 Å and 2.2 Å resolution, respectively. The pSer and Phe(+3) anchor the phosphopeptides into the BRCT binding groove, with adjacent peptide residues contributing to the interaction. In the BRCA1-ATRIP structure, Gln(+2) is accommodated through a conformational change of the BRCA1 E1698 side chain. Importantly, isothermal titration calorimetry experiments showed that the size and charge of the side chains at peptide positions +1 and +2 contribute significantly to the BRCA1 BRCT-peptide binding affinity. In particular, the Asp(+1) and Glu(+2) in the human CDC27 peptide 816-HAAEpSDEF-823 abrogate the interaction with the BRCA1 BRCTs due in large part to electrostatic repulsion between Glu(+2) and E1698, indicating a preference of these domains for specific side chains at positions +1 and +2. These results emphasize the need for a systematic assessment of the contribution of the peptide residues surrounding pSer and Phe(+3) to the binding affinity and specificity of the BRCA1 BRCTs in order to elucidate the molecular mechanisms underlying the hierarchy of target selection by these versatile domains during DDR and tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteína BRCA1/química , Proteínas de Unión al ADN/química , Modelos Moleculares , Proteínas Nucleares/química , Fragmentos de Péptidos/química , Fosfopéptidos/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/química , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ácido Aspártico/química , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Sitios de Unión , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Cinética , Ligandos , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Fenilalanina/química , Fosfopéptidos/metabolismo , Fosfoserina/química , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Difracción de Rayos X
16.
J Biol Chem ; 287(4): 2854-62, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22139841

RESUMEN

Microcephalin (MCPH1), the first gene identified as causative for primary recessive autosomal microcephaly, is aberrantly expressed in autism-like disorders and human malignancy of breast and ovarian origin. MCPH1, the encoded protein product, has been implicated in various cellular processes including the DNA damage checkpoint, DNA repair, and transcription. Although our understanding of the cellular context in which MCPH1 operates continues to develop, a structural understanding of the C-terminal tandem BRCT domains of MCPH1 remains unexplored. Here, we identify cell division cycle protein 27 (Cdc27), a component of the anaphase-promoting complex (APC/C), as a novel interacting partner of MCPH1. We provide in vitro and in vivo evidence that the C-terminal tandem BRCT domains of MCPH1 (C-BRCTs) bind Cdc27 in a phosphorylation-dependent manner. To characterize this interaction further, we determined the structure of MCPH1 C-BRCTs in complex with a phosphorylated Cdc27 peptide (pCdc27) using x-ray crystallography. Based on this structure, we identified single amino acid mutations targeted at the binding interface that disrupted the MCPH1-pCdc27 interaction. Collectively, our data define the biochemical, structural, and cellular determinants of the novel interaction between MCPH1 and Cdc27 and suggest that this interaction may occur within the larger context of MCPH1-APC/C.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Sustitución de Aminoácidos , Ciclosoma-Complejo Promotor de la Anafase , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Proteínas de Ciclo Celular/genética , Línea Celular , Proteínas del Citoesqueleto , Humanos , Mutación Missense , Proteínas del Tejido Nervioso/genética , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Complejos de Ubiquitina-Proteína Ligasa/genética
17.
Proc Natl Acad Sci U S A ; 107(20): 9210-5, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20439707

RESUMEN

The transcription factor CCAAT/enhancer binding protein delta (C/EBPdelta, CEBPD, NFIL-6beta) has tumor suppressor function; however, the molecular mechanism(s) by which C/EBPdelta exerts its effect are largely unknown. Here, we report that C/EBPdelta induces expression of the Cdc27 (APC3) subunit of the anaphase promoting complex/cyclosome (APC/C), which results in the polyubiquitination and degradation of the prooncogenic cell cycle regulator cyclin D1, and also down-regulates cyclin B1, Skp2, and Plk-1. In C/EBPdelta knockout mouse embryo fibroblasts (MEF) Cdc27 levels were reduced, whereas cyclin D1 levels were increased even in the presence of activated GSK-3beta. Silencing of C/EBPdelta, Cdc27, or the APC/C coactivator Cdh1 (FZR1) in MCF-10A breast epithelial cells increased cyclin D1 protein expression. Like C/EBPdelta, and in contrast to cyclin D1, Cdc27 was down-regulated in several breast cancer cell lines, suggesting that Cdc27 itself may be a tumor suppressor. Cyclin D1 is a known substrate of polyubiquitination complex SKP1/CUL1/F-box (SCF), and our studies show that Cdc27 directs cyclin D1 to alternative degradation by APC/C. These findings shed light on the role and regulation of APC/C, which is critical for most cellular processes.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Western Blotting , Proteína delta de Unión al Potenciador CCAAT/genética , Línea Celular Tumoral , Ciclina B1/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Inmunoprecipitación , Ratones , Ratones Noqueados , Microscopía Fluorescente , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Quinasa Tipo Polo 1
18.
J Physiol Biochem ; 79(1): 163-174, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36399312

RESUMEN

C-terminal tensin-like (CTEN) is a tensin family protein typically localized to the cytoplasmic side of focal adhesions, and primarily contributes to cell adhesion and migration. Elevated expression and nuclear accumulation of CTEN have been reported in several types of cancers and found to be associated with malignant behaviors. However, the function of nuclear CTEN remains elusive. In this study, we report for the first time that nuclear CTEN associates with chromatin DNA and occupies the region proximal to the transcription start site in several genes. The mRNA expression level of CTEN positively correlates with that of one of its putative target genes, cell division cycle protein 27 (CDC27), in a clinical colorectal cancer dataset, suggesting that CTEN may play a role in the regulation of CDC27 gene expression. Furthermore, we demonstrated that CTEN is recruited to the promoter region of the CDC27 gene and that the mRNA expression and promoter activity of CDC27 are both reduced when CTEN is downregulated. In addition, we found that enhanced nuclear accumulation of CTEN in HCT116 cells by overexpression of CTEN fused with nuclear localization signals increases CDC27 transcript levels and promoter activity. The increased nuclear-localized CTEN also significantly promotes cell migration, and the migratory ability is suppressed when CDC27 is knocked down. These results demonstrate that nuclear CTEN regulates CDC27 expression transcriptionally and promotes cell migration through CDC27. Our findings provide new insights into CTEN moonlighting in the nucleus as a DNA-associated protein and transcriptional regulator involved in modulating cancer cell migration.


Asunto(s)
Proteínas de Microfilamentos , Neoplasias , Humanos , Tensinas/genética , Tensinas/metabolismo , Proteínas de Microfilamentos/genética , Movimiento Celular , Adhesión Celular/fisiología , ARN Mensajero/genética , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase
19.
Epigenetics ; 18(1): 2195305, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36994860

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with an unclear pathogenesis. This study aimed to elucidate the function and potential mechanisms of TUG1 in IPF progression. Cell viability and migration were detected by CCK-8 and transwell assays. Autophagy, fibrosis, or EMT-related proteins were measured by Western blotting. Pro-inflammatory cytokine levels were assessed by ELISA kits. The subcellular localization of TUG1 was observed by FISH assay. RIP assay detected the interaction between TUG1 and CDC27. TUG1 and CDC27 was up-regulated in TGF-ß1-induced RLE-6TN cells. TUG1 depletion suppressed pulmonary fibrosis via attenuating inflammation, EMT, inducing autophagy and inactivating PI3K/Akt/mTOR pathway in vitro and in vivo. TUG1 knockdown prevented CDC27 expression. TUG1 silencing ameliorated pulmonary fibrosis by reducing CDC27 expression and inhibiting PI3K/Akt/mTOR pathway.


Asunto(s)
Fibrosis Pulmonar , ARN Largo no Codificante , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/genética , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Metilación de ADN , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Animales
20.
J Biol Chem ; 286(12): 10041-50, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21209074

RESUMEN

Loss of TGF-ß-induced growth inhibition is a hallmark of many human tumors. Previous studies implied that activation of the anaphase-promoting complex (APC/cyclosome) is involved in the TGF-ß signaling pathway, which facilitates the destruction of SnoN, a transcriptional co-suppressor, which leads in turn to the transactivation of TGF-ß-responsive genes for cell cycle arrest. The function of APC was demonstrated in TGF-ß signal transduction, but the mechanism by which it is activated in response to TGF-ß signaling remains unclear. We report here that phosphorylation of Cdc27, a core subunit of APC, in response to TGF-ß signaling can facilitate the activation of APC. We have demonstrated that casein kinase II (CKII) is involved in the phosphorylation of Cdc27 in response to TGF-ß signaling. Depletion of CKII by shRNA abolishes the TGF-ß-induced phosphorylation of Cdc27 and subsequent degradation of SnoN. Disruptive mutation of Cdc27 (S154A) attenuates TGF-ß-induced SnoN degradation. In addition, expression of a phosphorylation-resistant Cdc27 mutant significantly attenuates TGF-ß-induced growth inhibition. Together, the results suggest that phosphorylation of Cdc27 by CKII is involved in TGF-ß-induced activation of APC.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Sustitución de Aminoácidos , Ciclosoma-Complejo Promotor de la Anafase , Animales , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Visón , Mutación Missense , Fosforilación/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factor de Crecimiento Transformador beta/genética , Complejos de Ubiquitina-Proteína Ligasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA