Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.248
Filtrar
1.
Future Med Chem ; 14(14): 1087-1105, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35703127

RESUMEN

Colorectal cancer (CRC) is a common cancer in the world and its prevalence is increasing in developing countries. Deregulated cell cycle traverse is a hallmark of malignant transformation and is often observed in CRC as a result of imprecise activity of cell cycle regulatory components, viz. cyclins and cyclin-dependent kinases (CDKs). Apart from cell cycle regulation, some CDKs also regulate processes such as transcription and have also been shown to be involved in colorectal carcinogenesis. This article aims to review cyclin-dependent kinases as potential targets for CRC. Furthermore, therapeutic candidates to target CDKs are also discussed.


Asunto(s)
Neoplasias Colorrectales , Quinasas Ciclina-Dependientes , Ciclo Celular , Transformación Celular Neoplásica , Neoplasias Colorrectales/tratamiento farmacológico , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/fisiología , Humanos
2.
PLoS Biol ; 19(12): e3001496, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34928937

RESUMEN

Magnesium is essential for cellular life, but how it is homeostatically controlled still remains poorly understood. Here, we report that members of CNNM family, which have been controversially implicated in both cellular Mg2+ influx and efflux, selectively bind to the TRPM7 channel to stimulate divalent cation entry into cells. Coexpression of CNNMs with the channel markedly increased uptake of divalent cations, which is prevented by an inactivating mutation to the channel's pore. Knockout (KO) of TRPM7 in cells or application of the TRPM7 channel inhibitor NS8593 also interfered with CNNM-stimulated divalent cation uptake. Conversely, KO of CNNM3 and CNNM4 in HEK-293 cells significantly reduced TRPM7-mediated divalent cation entry, without affecting TRPM7 protein expression or its cell surface levels. Furthermore, we found that cellular overexpression of phosphatases of regenerating liver (PRLs), known CNNMs binding partners, stimulated TRPM7-dependent divalent cation entry and that CNNMs were required for this activity. Whole-cell electrophysiological recordings demonstrated that deletion of CNNM3 and CNNM4 from HEK-293 cells interfered with heterologously expressed and native TRPM7 channel function. We conclude that CNNMs employ the TRPM7 channel to mediate divalent cation influx and that CNNMs also possess separate TRPM7-independent Mg2+ efflux activities that contribute to CNNMs' control of cellular Mg2+ homeostasis.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Ciclinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPM/metabolismo , Proteínas de Transporte de Catión/fisiología , Cationes Bivalentes/metabolismo , Línea Celular Tumoral , Ciclinas/fisiología , Células HEK293 , Humanos , Magnesio/metabolismo , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/fisiología , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/fisiología
3.
Biol Reprod ; 101(3): 591-601, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31078132

RESUMEN

Cyclins and cyclin-dependent kinases (CDKs) are key regulators of the cell cycle. Most of our understanding of their functions has been obtained from studies in single-cell organisms and mitotically proliferating cultured cells. In mammals, there are more than 20 cyclins and 20 CDKs. Although genetic ablation studies in mice have shown that most of these factors are dispensable for viability and fertility, uncovering their functional redundancy, CCNA2, CCNB1, and CDK1 are essential for embryonic development. Cyclin/CDK complexes are known to regulate both mitotic and meiotic cell cycles. While some mechanisms are common to both types of cell divisions, meiosis has unique characteristics and requirements. During meiosis, DNA replication is followed by two successive rounds of cell division. In addition, mammalian germ cells experience a prolonged prophase I in males or a long period of arrest in prophase I in females. Therefore, cyclins and CDKs may have functions in meiosis distinct from their mitotic functions and indeed, meiosis-specific cyclins, CCNA1 and CCNB3, have been identified. Here, we describe recent advances in the field of cyclins and CDKs with a focus on meiosis and early embryogenesis.


Asunto(s)
Quinasas Ciclina-Dependientes/fisiología , Ciclinas/fisiología , Gametogénesis/genética , Células Germinativas/fisiología , Animales , Embrión de Mamíferos , Femenino , Humanos , Masculino , Mamíferos , Meiosis , Ratones
4.
Oncogene ; 37(25): 3329-3339, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29551768

RESUMEN

Chronic liver injury triggers liver fibrosis and hepatocellular carcinoma (HCC), the third leading cause of cancer-related mortality. Cyclin E1 (CcnE1, formerly designated Cyclin E) is a regulatory subunit of the Cyclin-dependent kinase 2 (CDK2). It is overexpressed in approximately 70% of human HCCs correlating with poor prognosis, while the relevance of its orthologue Cyclin E2 (CcnE2) is unclear. Hepatocyte-specific deletion of NF-kappa-B essential modulator (NEMOΔhepa) leads to chronic hepatitis, liver fibrosis, and HCC as well as CcnE upregulation. To this end, we generated NEMOΔhepa/CcnE1-/- and NEMOΔhepa/CcnE2-/- double knockout mice and investigated age-dependent liver disease progression in these animals. Deletion of CcnE1 in NEMOΔhepa mice decreased basal liver damage and reduced spontaneous liver inflammation in young mice. In contrast, loss of CcnE2 did not affect liver injury in NEMOΔhepa livers pointing to a unique, non-redundant function of CcnE1 in chronic hepatitis. Accordingly, basal compensatory hepatocyte proliferation in NEMOΔhepa mice was reduced by concomitant ablation of CcnE1, but not after loss of CcnE2. In aged NEMOΔhepa mice, loss of CcnE1 resulted in significant reduction of liver tumorigenesis, while deletion of CcnE2 had no effect on HCC formation. CcnE1, but not its orthologue CcnE2, substantially contributes to hepatic inflammatory response, liver disease progression, and hepatocarcinogenesis in NEMOΔhepa mice.


Asunto(s)
Carcinoma Hepatocelular/prevención & control , Ciclina E/fisiología , Ciclinas/fisiología , Modelos Animales de Enfermedad , Hepatitis/prevención & control , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neoplasias Hepáticas Experimentales/prevención & control , Hígado/lesiones , Hígado/patología , Proteínas Oncogénicas/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Hepatitis/etiología , Hepatitis/patología , Neoplasias Hepáticas Experimentales/etiología , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
PLoS One ; 12(6): e0180097, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28658303

RESUMEN

Chronic inflammation is a well-known precursor for cancer development and proliferation. We have recently demonstrated that high salt (NaCl) synergizes with sub-effective interleukin (IL)-17 to induce breast cancer cell proliferation. However, the exact molecular mechanisms mediating this effect are unclear. In our current study, we adopted a phosphoproteomic-based approach to identify salt modulated kinase-proteome specific molecular targets. The phosphoprotemics based binary comparison between heavy labelled MCF-7 cells treated with high salt (Δ0.05 M NaCl) and light labelled MCF-7 cells cultured under basal conditions demonstrated an enhanced phosphorylation of Serine-493 of SIK3 protein. The mRNA transcript and protein expression analysis of SIK3 in MCF-7 cells demonstrated a synergistic enhancement following co-treatment with high salt and sub-effective IL-17 (0.1 ng/mL), as compared to either treatments alone. A similar increase in SIK3 expression was observed in other breast cancer cell lines, MDA-MB-231, BT20, and AU565, while non-malignant breast epithelial cell line, MCF10A, did not induce SIK3 expression under similar conditions. Biochemical studies revealed mTORC2 acted as upstream mediator of SIK3 phosphorylation. Importantly, cell cycle analysis by flow cytometry demonstrated SIK3 induced G0/G1-phase release mediated cell proliferation, while SIK3 silencing abolished this effect. Also, SIK3 induced pro-inflammatory arginine metabolism, as evidenced by upregulation of the enzymes iNOS and ASS-1, along with downregulation of anti-inflammatory enzymes, arginase-1 and ornithine decarboxylase. Furthermore, gelatin zymography analysis has demonstrated that SIK3 induced expression of tumor metastatic CXCR4 through MMP-9 activation. Taken together, our data suggests a critical role of SIK3 in mediating three important hallmarks of cancer namely, cell proliferation, inflammation and metastasis. These studies provide a mechanistic basis for the future utilization of SIK3 as a key drug discovery target to improve breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Proliferación Celular/fisiología , Interleucina-17/fisiología , Proteínas Quinasas/fisiología , Sodio en la Dieta/efectos adversos , Línea Celular Tumoral , Ciclinas/fisiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Óxido Nítrico/metabolismo , Receptores CXCR4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Urea/metabolismo
6.
J Biol Chem ; 292(13): 5349-5363, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28184007

RESUMEN

Cyclin F protein, also known as FBXO1, is the largest among all cyclins and oscillates in the cell cycle like other cyclins. Apart from being a G2/M cyclin, cyclin F functions as the substrate-binding subunit of SCFcyclin F E3 ubiquitin ligase. In a gene expression analysis performed to identify novel gene modulations associated with cell cycle dysregulation during HIV-1 infection in CD4+ T cells, we observed down-regulation of the cyclin F gene (CCNF). Later, using gene overexpression and knockdown studies, we identified cyclin F as negatively influencing HIV-1 viral infectivity without any significant impact on virus production. Subsequently, we found that cyclin F negatively regulates the expression of viral protein Vif (viral infectivity factor) at the protein level. We also identified a novel host-pathogen interaction between cyclin F and Vif protein in T cells during HIV-1 infection. Mutational analysis of a cyclin F-specific amino acid motif in the C-terminal region of Vif indicated rescue of the protein from cyclin F-mediated down-regulation. Subsequently, we showed that Vif is a novel substrate of the SCFcyclin F E3 ligase, where cyclin F mediates the ubiquitination and proteasomal degradation of Vif through physical interaction. Finally, we showed that cyclin F augments APOBEC3G expression through degradation of Vif to regulate infectivity of progeny virions. Taken together, our results demonstrate that cyclin F is a novel F-box protein that functions as an intrinsic cellular regulator of HIV-1 Vif and has a negative regulatory effect on the maintenance of viral infectivity by restoring APOBEC3G expression.


Asunto(s)
Ciclinas/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Virión/patogenicidad , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasa APOBEC-3G/metabolismo , Linfocitos T CD4-Positivos , Células Cultivadas , Ciclinas/genética , Ciclinas/metabolismo , Proteínas F-Box , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Procesamiento Proteico-Postraduccional
7.
Mol Pharmacol ; 92(3): 240-245, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28119482

RESUMEN

Cdk5 and Abl enzyme substrate 1 (Cables1) is an adaptor protein that links cyclin-dependent kinase (Cdks) with nonreceptor tyrosine kinases and regulates the activity of Cdks by enhancing their Y15 phosphorylation. Emerging evidence also shows that Cables1 can interact with, for example, p53 family proteins, 14-3-3, and ß-catenin, suggesting that Cables1 may be a signaling hub for the regulation of cell growth. Abnormal expression of Cables1 has been observed in multiple types of cancers and other diseases. In this review, we summarize the characteristics of Cables1 and highlight the molecular mechanisms through which Cables1 regulates the development of cancer and other diseases. Finally, we discuss future challenges in demonstrating the role and potential application of Cables1 in cancer and other diseases.


Asunto(s)
Proteínas Portadoras/fisiología , Ciclinas/fisiología , Neoplasias/terapia , Fosfoproteínas/fisiología , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Ciclinas/química , Ciclinas/genética , Humanos , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilación , Proteína p53 Supresora de Tumor/fisiología
9.
Sci Rep ; 6: 28297, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27323909

RESUMEN

Cyclins play a central role in cell-cycle regulation; in mammals, the D family of cyclins consists of cyclin D1, D2, and D3. In Xenopus, only homologs of cyclins D1 and D2 have been reported, while a novel cyclin, cyclin Dx (ccndx), was found to be required for the maintenance of motor neuron progenitors during embryogenesis. It remains unknown whether zebrafish possess cyclin D3 or cyclin Dx. In this study, we identified a zebrafish ccndx gene encoding a protein which can form a complex with Cdk4. Through whole-mount in situ hybridization, we observed that zccndx mRNA is expressed in the motor neurons of hindbrain and spinal cord during development. Analysis of a 4-kb promoter sequence of the zccndx gene revealed the presence of HRE sites, which can be regulated by HIF2α. Morpholino knockdown of zebrafish Hif2α and cyclin Dx resulted in the abolishment of isl1 and oligo2 expression in the precursors of motor neurons, and also disrupted axon growth. Overexpression of cyclin Dx mRNA in Hif2α morphants partially rescued zccndx expression. Taken together, our data indicate that zebrafish cyclin Dx plays a role in maintaining the precursors of motor neurons.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Ciclinas/fisiología , Neuronas Motoras/fisiología , Células-Madre Neurales/fisiología , Animales , Células COS , Proliferación Celular , Chlorocebus aethiops , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones , Neurogénesis , Pez Cebra/embriología
10.
Nat Rev Mol Cell Biol ; 17(5): 280-92, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27033256

RESUMEN

The roles of cyclins and their catalytic partners, the cyclin-dependent kinases (CDKs), as core components of the machinery that drives cell cycle progression are well established. Increasing evidence indicates that mammalian cyclins and CDKs also carry out important functions in other cellular processes, such as transcription, DNA damage repair, control of cell death, differentiation, the immune response and metabolism. Some of these non-canonical functions are performed by cyclins or CDKs, independently of their respective cell cycle partners, suggesting that there was a substantial divergence in the functions of these proteins during evolution.


Asunto(s)
Quinasas Ciclina-Dependientes/fisiología , Ciclinas/fisiología , Animales , Ciclo Celular , Diferenciación Celular , Roturas del ADN de Doble Cadena , Reparación del ADN , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal , Activación Transcripcional
11.
Tumour Biol ; 37(8): 10161-75, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26831658

RESUMEN

This study is designated to investigate the roles of cyclin Y (CCNY) and Wnt signaling pathway in regulating ovarian cancer (OC) cell proliferation, migration, and invasion. Quantitative real-time PCR (qRT-PCR), Western blot, MTT assay, cell scratch, and transwell test were used in our study, and transplanted tumor model was constructed on nude mice. C-Myc, cyclin D1, PFTK1, ki67, OGT, and ß-catenin protein expressions in tumor tissues were detected. CCNY was significantly upregulated in OC cell lines and tissues (both P < 0.05); significant association was observed between CCNY expression and clinicopathological stage, lymph node metastasis (LNM) (P < 0.05); and the CCNY expression in stages III to IV was higher than that in stages I to II, and patients with LNM had higher CCNY expression when compared with those in patients without LNM (P < 0.05); expressions of c-Myc, cyclin D, PFTK1, ki67, and OGT were upregulated in OC tissues compared with ovarian benign tissues, suggesting that these expressions were significantly different between the two groups (P < 0.05); CCNY significantly exacerbated proliferation, migration, and invasion of A2780 cells; c-Myc and cyclin D1 protein expressions increased as the expression of CCNY increased (P < 0.001); ß-catenin expressions in A2780 cells with over-expression of CCNY were significantly increased in the nucleus, but significantly decreased in the cytoplasm (both P < 0.05); high expressions of CCNY exacerbated the proliferation of A2780 cells in nude mice and significantly increased c-Myc, cyclin D1, PFTK1, ki67, and OGT protein expressions in tumor tissues which were transplanted into nude mice (P < 0.01). CCNY might exacerbate the proliferation, migration, and invasion of OC cells via activating the Wnt signaling pathway. Thus, this study provides a theoretical foundation for the development of therapeutic drugs that are able to cure OC by targeting CCNY.


Asunto(s)
Ciclinas/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Ováricas/patología , Vía de Señalización Wnt , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/secundario , Adulto , Anciano , Animales , Diferenciación Celular , División Celular , Línea Celular Tumoral , Movimiento Celular , Núcleo Celular/metabolismo , Proliferación Celular , Ciclinas/biosíntesis , Ciclinas/genética , Citoplasma/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Distribución Aleatoria , Proteínas Recombinantes de Fusión/metabolismo
12.
Int Microbiol ; 19(3): 133-141, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28494083

RESUMEN

Phosphorus is a pivotal element in all biochemical systems: it serves to store metabolic energy as ATP, it forms the backbone of genetic material such as RNA and DNA, and it separates cells from the environment as phospholipids. In addition to this "big hits", phosphorus has recently been shown to play an important role in other important processes such as cell cycle regulation. In the present review, we briefly summarize the biological processes in which phosphorus is involved in the yeast Saccharomyces cerevisiae before discussing our latest findings on the role of this element in the regulation of DNA replication in this eukaryotic model organism. We describe both the role of phosphorus in the regulation of G1 progression by means of the Cyclin Dependent Kinase (CDK) Pho85 and the stabilization of the cyclin Cln3, as well as the role of other molecule composed of phosphorus-the polyphosphate-in cell cycle progression, dNTP synthesis, and genome stability. Given the eminent role played by phosphorus in life, we outline the future of phosphorus in the context of one of the main challenges in human health: cancer treatment. [Int Microbiol 19(3):133-141 (2016)].


Asunto(s)
Puntos de Control del Ciclo Celular , Fosfatos/fisiología , Fósforo/fisiología , Saccharomyces cerevisiae/fisiología , Quinasas Ciclina-Dependientes/fisiología , Ciclinas/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología
13.
PLoS Genet ; 11(8): e1005485, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26305884

RESUMEN

Cyclin Y-like 1 (Ccnyl1) is a newly-identified member of the cyclin family and is highly similar in protein sequences to Cyclin Y (Ccny). However, the function of Ccnyl1 is poorly characterized in any organism. Here we found that Ccnyl1 was most abundantly expressed in the testis of mice and was about seven times higher than the level of Ccny. Male Ccnyl1-/- mice were infertile, whereas both male and female Ccny-/- mice displayed normal fertility. These results suggest that Ccnyl1, but not Ccny, is indispensable for male fertility. Spermatozoa obtained from Ccnyl1-/- mice displayed significantly impaired motility, and represented a thinned annulus region and/or a bent head. We found that the protein, but not the mRNA, level of cyclin-dependent kinase 16 (CDK16) was decreased in the testis of Ccnyl1-/- mice. Further study demonstrated that CCNYL1 interacted with CDK16 and this interaction mutually increased the stability of these two proteins. Moreover, the interaction increased the kinase activity of CDK16. In addition, we observed an alteration of phosphorylation levels of CDK16 in the presence of CCNYL1. We identified the phosphorylation sites of CDK16 by mass spectrometry and revealed that several phosphorylation modifications on the N-terminal region of CDK16 were indispensable for the CCNYL1 binding and the modulation of CDK16 kinase activity. Our results therefore reveal a previously unrecognized role of CCNYL1 in regulating spermatogenesis through the interaction and modulation of CDK16.


Asunto(s)
Quinasas Ciclina-Dependientes/fisiología , Ciclinas/metabolismo , Ciclinas/fisiología , Espermatogénesis , Animales , Femenino , Fertilidad , Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Motilidad Espermática
16.
J Virol ; 89(10): 5450-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25741012

RESUMEN

UNLABELLED: Walleye dermal sarcoma virus (WDSV) infection is associated with the seasonal development and regression of walleye dermal sarcoma. Previous work showed that the retroviral cyclin (RV-cyclin), encoded by WDSV, has separable cyclin box and transcription activation domains. It binds to cyclin-dependent kinase 8 (CDK8) and enhances its kinase activity. CDK8 is evolutionarily conserved and is frequently overexpressed in human cancers. It is normally activated by cyclin C and is required for transcription elongation of the serum response genes (immediate early genes [IEGs]) FOS, EGR1, and cJUN. The IEGs drive cell proliferation, and their expression is brief and highly regulated. Here we show that constitutive expression of RV-cyclin in the HCT116 colon cancer cell line significantly increases the level of IEG expression in response to serum stimulation. Quantitative reverse transcription-PCR (RT-PCR) and nuclear run-on assays provide evidence that RV-cyclin does not alter the initiation of IEG transcription but does enhance the overall rate of transcription elongation and maintains transcription reinitiation. RV-cyclin does not increase activating phosphorylation events in the mitogen-activated protein kinase pathway and does not inhibit decay of IEG mRNAs. At the EGR1 gene locus, RV-cyclin increases and maintains RNA polymerase II (Pol II) occupancy after serum stimulation, in conjunction with increased and extended EGR1 gene expression. The RV-cyclin increases CDK8 occupancy at the EGR1 gene locus before and after serum stimulation. Both of RV-cyclin's functional domains, i.e., the cyclin box and the activation domain, are necessary for the overall enhancement of IEG expression. RV-cyclin presents a novel and ancient mechanism of retrovirus-induced oncogenesis. IMPORTANCE: The data reported here are important to both virology and cancer biology. The novel mechanism pinpoints CDK8 in the development of walleye dermal sarcoma and sheds light on CDK8's role in many human cancers. CDK8 controls expression from highly regulated genes, including the interferon-stimulated genes. Its function is likely the target of many viral interferon-resistance mechanisms. CDK8 also controls cellular responses to metabolic stimuli, stress, and hypoxia, in addition to the serum response. The retroviral cyclin (RV-cyclin) represents a highly selected probe of CDK8 function. RV-cyclin does not control CDK8 specificity but instead enhances CDK8's effects on regulated genes, an important distinction for its use to delineate natural CDK8 targets. The outcomes of this research are applicable to investigations of normal and abnormal CDK8 functions. The mechanisms defined here will contribute directly to the dermal sarcoma model in fish and clarify an important path for oncogenesis and innate resistance to viruses.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/metabolismo , Ciclinas/fisiología , Epsilonretrovirus/fisiología , Proteínas de los Retroviridae/fisiología , Animales , Carcinogénesis , Ciclinas/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Epsilonretrovirus/genética , Epsilonretrovirus/patogenicidad , Enfermedades de los Peces/genética , Enfermedades de los Peces/virología , Genes Inmediatos-Precoces , Genes fos , Genes jun , Células HCT116 , Interacciones Huésped-Patógeno , Humanos , Percas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/veterinaria , Infecciones por Retroviridae/virología , Proteínas de los Retroviridae/genética , Elongación de la Transcripción Genética , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/veterinaria , Infecciones Tumorales por Virus/virología
17.
EMBO J ; 34(8): 1078-89, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25712475

RESUMEN

Mucociliary clearance and fluid transport along epithelial surfaces are carried out by multiciliated cells (MCCs). Recently, human mutations in Cyclin O (CCNO) were linked to severe airway disease. Here, we show that Ccno expression is restricted to MCCs and the genetic deletion of Ccno in mouse leads to reduced numbers of multiple motile cilia and characteristic phenotypes of MCC dysfunction including severe hydrocephalus and mucociliary clearance deficits. Reduced cilia numbers are caused by compromised generation of centrioles at deuterosomes, which serve as major amplification platform for centrioles in MCCs. Ccno-deficient MCCs fail to sufficiently generate deuterosomes, and only reduced numbers of fully functional centrioles that undergo maturation to ciliary basal bodies are formed. Collectively, this study implicates CCNO as first known regulator of deuterosome formation and function for the amplification of centrioles in MCCs.


Asunto(s)
Centriolos/fisiología , Ciclinas/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Centriolos/ultraestructura , Cilios/fisiología , Cilios/ultraestructura , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Hidrocefalia/embriología , Hidrocefalia/genética , Ratones , Ratones Transgénicos , Depuración Mucociliar/genética , Organogénesis/genética , Tráquea/citología , Tráquea/embriología , Tráquea/metabolismo
18.
Mol Biol Cell ; 25(18): 2807-16, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25057017

RESUMEN

The yeast cyclin C-Cdk8 kinase forms a complex with Med13p to repress the transcription of genes involved in the stress response and meiosis. In response to oxidative stress, cyclin C displays nuclear to cytoplasmic relocalization that triggers mitochondrial fission and promotes programmed cell death. In this report, we demonstrate that Med13p mediates cyclin C nuclear retention in unstressed cells. Deleting MED13 allows aberrant cytoplasmic cyclin C localization and extensive mitochondrial fragmentation. Loss of Med13p function resulted in mitochondrial dysfunction and hypersensitivity to oxidative stress-induced programmed cell death that were dependent on cyclin C. The regulatory system controlling cyclin C-Med13p interaction is complex. First, a previous study found that cyclin C phosphorylation by the stress-activated MAP kinase Slt2p is required for nuclear to cytoplasmic translocation. This study found that cyclin C-Med13p association is impaired when the Slt2p target residue is substituted with a phosphomimetic amino acid. The second step involves Med13p destruction mediated by the 26S proteasome and cyclin C-Cdk8p kinase activity. In conclusion, Med13p maintains mitochondrial structure, function, and normal oxidative stress sensitivity through cyclin C nuclear retention. Releasing cyclin C from the nucleus involves both its phosphorylation by Slt2p coupled with Med13p destruction.


Asunto(s)
Núcleo Celular/metabolismo , Ciclina C/metabolismo , Ciclinas/fisiología , Complejo Mediador/fisiología , Dinámicas Mitocondriales , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/fisiología , Factores de Transcripción/fisiología , Apoptosis , Transporte de Proteínas , Saccharomyces cerevisiae/citología
19.
J Cell Biol ; 205(5): 633-41, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24891606

RESUMEN

Meiotic crossovers (COs) are crucial for ensuring accurate homologous chromosome segregation during meiosis I. Because the double-strand breaks (DSBs) that initiate meiotic recombination greatly outnumber eventual COs, this process requires exquisite regulation to narrow down the pool of DSB intermediates that may form COs. In this paper, we identify a cyclin-related protein, CNTD1, as a critical mediator of this process. Disruption of Cntd1 results in failure to localize CO-specific factors MutLγ and HEI10 at designated CO sites and also leads to prolonged high levels of pre-CO intermediates marked by MutSγ and RNF212. These data show that maturation of COs is intimately coupled to deselection of excess pre-CO sites to yield a limited number of COs and that CNTD1 coordinates these processes by regulating the association between the RING finger proteins HEI10 and RNF212 and components of the CO machinery.


Asunto(s)
Intercambio Genético , Ciclinas/genética , Ciclinas/fisiología , Meiosis , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Segregación Cromosómica , Cromosomas/ultraestructura , Roturas del ADN de Doble Cadena , Ligasas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Recombinación Genética , Recuento de Espermatozoides , Espermatocitos/citología , Espermatocitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
20.
Hepatology ; 59(2): 651-60, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23787781

RESUMEN

UNLABELLED: The liver has a strong regenerative capacity. After injury, quiescent hepatocytes can reenter the mitotic cell cycle to restore tissue homeostasis. This G(0) /G(1) -S cell-cycle transition of primed hepatocytes is regulated by complexes of cyclin-dependent kinase 2 (Cdk2) with E-type cyclins (CcnE1 or CcnE2). However, single genetic ablation of either E-cyclin or Cdk2 does not affect overall liver regeneration. Here, we systematically investigated the contribution of CcnE1, CcnE2, and Cdk2 for liver regeneration after partial hepatectomy (PH) by generating corresponding double- and triple-knockout (KO) mouse mutants. We demonstrate that conditional deletion of Cdk2 alone in hepatocytes resulted in accelerated induction of CcnE1, but otherwise normal initiation of S phase in vivo and in vitro. Excessive CcnE1 did not contribute to a noncanonical kinase activity, but was located at chromatin together with components of the pre-replication complex (pre-RC), such as the minichromosome maintenance (MCM) helicase. Concomitant ablation of Cdk2 and CcnE1 in hepatocytes caused a defect in pre-RC formation and further led to dramatically impaired S-phase progression by down-regulation of cyclin A2 and cell death in vitro and substantially reduced hepatocyte proliferation and liver regeneration after PH in vivo. Similarly, combined loss of CcnE1 and CcnE2, but also the Cdk2/CcnE1/CcnE2 triple KO in liver, significantly inhibited S-phase initiation and liver mass reconstitution after PH, whereas concomitant ablation of CcnE2 and Cdk2 had no effect. CONCLUSION: In the absence of Cdk2, CcnE1 performs crucial kinase-independent functions in hepatocytes, which are capable of driving MCM loading on chromatin, cyclin A2 expression, and S-phase progression. Thus, combined inactivation of Cdk2 and CcnE1 is the minimal requirement for blocking S-phase machinery in vivo.


Asunto(s)
Ciclina E/deficiencia , Quinasa 2 Dependiente de la Ciclina/deficiencia , Replicación del ADN/fisiología , Hepatocitos/patología , Hepatocitos/fisiología , Regeneración Hepática/fisiología , Proteínas Oncogénicas/deficiencia , Animales , Apoptosis/fisiología , Ciclo Celular/fisiología , Células Cultivadas , Cromatina/fisiología , Ciclina E/genética , Ciclina E/fisiología , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/fisiología , Ciclinas/deficiencia , Ciclinas/genética , Ciclinas/fisiología , Femenino , Homeostasis/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/fisiología , Fase S/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...