Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Cell Sci ; 134(3)2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33536247

RESUMEN

The class I phosphoinositide 3-kinase (PI3K) catalytic subunits p110α and p110ß are ubiquitously expressed but differently targeted in tumours. In cancer, PIK3CB (encoding p110ß) is seldom mutated compared with PIK3CA (encoding p110α) but can contribute to tumorigenesis in certain PTEN-deficient tumours. The underlying molecular mechanisms are, however, unclear. We have previously reported that p110ß is highly expressed in endometrial cancer (EC) cell lines and at the mRNA level in primary patient tumours. Here, we show that p110ß protein levels are high in both the cytoplasmic and nuclear compartments in EC cells. Moreover, high nuclear:cytoplasmic staining ratios were detected in high-grade primary tumours. High levels of phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P3] were measured in the nucleus of EC cells, and pharmacological and genetic approaches showed that its production was partly dependent upon p110ß activity. Using immunofluorescence staining, p110ß and PtdIns(3,4,5)P3 were localised in the nucleolus, which correlated with high levels of 47S pre-rRNA. p110ß inhibition led to a decrease in both 47S rRNA levels and cell proliferation. In conclusion, these results present a nucleolar role for p110ß that may contribute to tumorigenesis in EC.This article has an associated First Person interview with Fatemeh Mazloumi Gavgani, joint first author of the paper.


Asunto(s)
Neoplasias Endometriales , Fosfatidilinositol 3-Quinasa , Proliferación Celular/genética , Neoplasias Endometriales/genética , Femenino , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Regulación hacia Arriba/genética
2.
Biochem Biophys Res Commun ; 656: 23-29, 2023 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-36947963

RESUMEN

Lipids have been implicated in Parkinson's Disease (PD). We therefore studied the lipid profile of the neuroblastoma SH-SY5Y cell line, which is used extensively in PD research and compared it to that of the A431 epithelial cancer cell line. We have isolated whole cell extracts (WC) and plasma membrane (PM) fractions of both cell lines. The isolates were analyzed with 31P NMR. We observed a significant higher abundance of phosphatidylcholine (PC) for SH-SY5Y cells for both WC (55 ± 4.1%) and PM (63.3 ± 3.1%) compared to WC (40.5 ± 2.2%) and PM (43.4 ± 1.3%) of A431. Moreover, a higher abundance of phosphatidylethanolamine was detected for the WC of A431 compared to the SH-SY5Y. Using LC-MS/MS, we also determined the relative abundance of fatty acid (FA) moieties for each phospholipid class, finding that SH-SY5Y had high polyunsaturated FA levels, including arachidonic acid compared to A431 cells. When comparing our results to reported compositions of brain and neural tissues, we note the much higher PC levels, as well as very low levels of docosahexaenoic acid. However, relative levels of arachidonic acid and other polyunsaturated fatty acids were elevated, in line with what is desirable for a neural model system.


Asunto(s)
Neuroblastoma , Fosfolípidos , Humanos , Fosfatidilcolinas , Cromatografía Liquida , Neuroblastoma/metabolismo , Espectrometría de Masas en Tándem , Línea Celular Tumoral , Ácidos Grasos Insaturados , Ácido Araquidónico
3.
Mol Cell Proteomics ; 20: 100102, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34048982

RESUMEN

Polyphosphoinositides (PPIns) play essential roles as lipid signaling molecules, and many of their functions have been elucidated in the cytoplasm. However, PPIns are also intranuclear where they contribute to chromatin remodeling, transcription, and mRNA splicing. The PPIn, phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3), has been mapped to the nucleus and nucleoli, but its role remains unclear in this subcellular compartment. To gain further insights into the nuclear functions of PtdIns(3,4,5)P3, we applied a previously developed quantitative MS-based approach to identify the targets of PtdIns(3,4,5)P3 from isolated nuclei. We identified 179 potential PtdIns(3,4,5)P3-interacting partners, and gene ontology analysis for the biological functions of this dataset revealed an enrichment in RNA processing/splicing, cytokinesis, protein folding, and DNA repair. Interestingly, about half of these interactors were common to nucleolar protein datasets, some of which had dual functions in rRNA processes and DNA repair, including poly(ADP-ribose) polymerase 1 (PARP1, now referred as ADP-ribosyltransferase 1). PARP1 was found to interact directly with PPIn via three polybasic regions in the DNA-binding domain and the linker located N-terminal of the catalytic region. PARP1 was shown to bind to PtdIns(3,4,5)P3 as well as phosphatidylinositol 3,4-bisphosphate in vitro and to colocalize with PtdIns(3,4,5)P3 in the nucleolus and with phosphatidylinositol 3,4-bisphosphate in nucleoplasmic foci. In conclusion, the PtdIns(3,4,5)P3 interactome reported here will serve as a resource to further investigate the molecular mechanisms underlying PtdIns(3,4,5)P3-mediated interactions in the nucleus and nucleolus.


Asunto(s)
Núcleo Celular/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Células HeLa , Humanos , Mapas de Interacción de Proteínas
4.
Int J Mol Sci ; 19(12)2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30544563

RESUMEN

The phosphoinositide 3-kinase (PI3K) signalling pathway is highly dysregulated in cancer, leading to elevated PI3K signalling and altered cellular processes that contribute to tumour development. The pathway is normally orchestrated by class I PI3K enzymes and negatively regulated by the phosphatase and tensin homologue, PTEN. Endometrial carcinomas harbour frequent alterations in components of the pathway, including changes in gene copy number and mutations, in particular in the oncogene PIK3CA, the gene encoding the PI3K catalytic subunit p110α, and the tumour suppressor PTEN. PIK3CB, encoding the other ubiquitously expressed class I isoform p110ß, is less frequently altered but the few mutations identified to date are oncogenic. This isoform has received more research interest in recent years, particularly since PTEN-deficient tumours were found to be reliant on p110ß activity to sustain transformation. In this review, we describe the current understanding of the common and distinct biochemical properties of the p110α and p110ß isoforms, summarise their mutations and highlight how they are targeted in clinical trials in endometrial cancer.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Endometriales/genética , Femenino , Humanos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasa/genética
5.
Biochem J ; 473(14): 2033-47, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27118868

RESUMEN

Polyphosphoinositides (PPIns) are present in the nucleus where they participate in crucial nuclear processes, such as chromatin remodelling, transcription and mRNA processing. In a previous interactomics study, aimed to gain further insight into nuclear PPIns functions, we identified ErbB3 binding protein 1 (EBP1) as a potential nuclear PPIn-binding protein in a lipid pull-down screen. EBP1 is a ubiquitous and conserved protein, located in both the cytoplasm and nucleolus, and associated with cell proliferation and survival. In the present study, we show that EBP1 binds directly to several PPIns via two distinct PPIn-binding sites consisting of clusters of lysine residues and positioned at the N- and C-termini of the protein. Using interaction mutants, we show that the C-terminal PPIn-binding motif contributes the most to the localization of EBP1 in the nucleolus. Importantly, a K372N point mutation, located within the C-terminal motif and found in endometrial tumours, is sufficient to alter the nucleolar targeting of EBP1. Our study reveals also the presence of the class I phosphoinositide 3-kinase (PI3K) catalytic subunit p110ß and its product PtdIns(3,4,5)P3 together with EBP1 in the nucleolus. Using NMR, we further demonstrate an association between EBP1 and PtdIns(3,4,5)P3 via both electrostatic and hydrophobic interactions. Taken together, these results show that EBP1 interacts directly with PPIns and associate with PtdIns(3,4,5)P3 in the nucleolus. The presence of p110ß and PtdIns(3,4,5)P3 in the nucleolus indicates their potential role in regulating nucleolar processes, at least via EBP1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Nucléolo Celular/metabolismo , Proteínas Nucleares/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Unión al ADN , Humanos , Lisina/química , Lisina/metabolismo , Espectroscopía de Resonancia Magnética , Ratones , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética
6.
MicroPubl Biol ; 20242024.
Artículo en Inglés | MEDLINE | ID: mdl-38495585

RESUMEN

AKT is an essential player in the phosphoinositide 3-kinase (PI3K) signalling pathway. Although the mechanisms of its action are well understood at the plasma membrane, AKT can also be found in the nucleus. In adipocytes, this pathway is activated during the process of adipogenesis and solicits both plasma membrane and nuclear AKT activity. However, the endogenous presence of active AKT in the nucleus during adipogenesis has not been shown. Here, we show that the levels of active AKT phosphorylated at Ser-473 increase rapidly after the induction of differentiation in 3T3-L1 cells, both in the cytoplasm and in the nucleus, and tend to remain elevated over the course of differentiation. In conclusion, these results support the notion that nuclear AKT plays an important role in this process.

7.
Mol Cell Proteomics ; 10(2): M110.003376, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21048195

RESUMEN

Considerable insight into phosphoinositide-regulated cytoplasmic functions has been gained by identifying phosphoinositide-effector proteins. Phosphoinositide-regulated nuclear functions however are fewer and less clear. To address this, we established a proteomic method based on neomycin extraction of intact nuclei to enrich for nuclear phosphoinositide-effector proteins. We identified 168 proteins harboring phosphoinositide-binding domains. Although the vast majority of these contained lysine/arginine-rich patches with the following motif, K/R-(X(n= 3-7)-K-X-K/R-K/R, we also identified a smaller subset of known phosphoinositide-binding proteins containing pleckstrin homology or plant homeodomain modules. Proteins with no prior history of phosphoinositide interaction were identified, some of which have functional roles in RNA splicing and processing and chromatin assembly. The remaining proteins represent potentially other novel nuclear phosphoinositide-effector proteins and as such strengthen our appreciation of phosphoinositide-regulated nuclear functions. DNA topology was exemplar among these: Biochemical assays validated our proteomic data supporting a direct interaction between phosphatidylinositol 4,5-bisphosphate and DNA Topoisomerase IIα. In addition, a subset of neomycin extracted proteins were further validated as phosphatidyl 4,5-bisphosphate-interacting proteins by quantitative lipid pull downs. In summary, data sets such as this serve as a resource for a global view of phosphoinositide-regulated nuclear functions.


Asunto(s)
Núcleo Celular/metabolismo , Neomicina/farmacología , Fosfatidilinositol 4,5-Difosfato/química , Proteómica/métodos , Secuencias de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Células Jurkat , Fosfatidilinositoles/química , Unión Proteica , Estructura Terciaria de Proteína
8.
MicroPubl Biol ; 20232023.
Artículo en Inglés | MEDLINE | ID: mdl-37038481

RESUMEN

Polyphosphoinositides (PPIn) play essential functions as lipid signalling molecules and many of their functions have been elucidated in the cytoplasm. However, PPIn are also intranuclear where they contribute to chromatin remodelling, transcription and mRNA splicing. Using quantitative interactomics, we have previously identified PPIn-interacting proteins with roles in RNA processing/splicing including the heterogeneous nuclear ribonucleoprotein U (hnRNPU/SAF-A). In this study, hnRNPU was validated as a direct PPIn-interacting protein via 2 regions located in the N and C termini. Furthermore, deletion of the polybasic motif region located at aa 9-24 in its DNA binding SAP domain prevented PPIn interaction. In conclusion, these results are consistent with hnRNPU harbouring a polybasic region with dual functions in DNA and PPIn interaction.

9.
Exp Cell Res ; 317(3): 293-306, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20974127

RESUMEN

p120 Catenin (p120(ctn)) regulates cadherin stability, and thus facilitates strong cell-cell adhesion. Previously, we demonstrated that Gα(12) interacts with p120(ctn). In the present study, we have delineated a region of p120(ctn) that binds to Gα(12). We report that the N-terminal region of p120(ctn) (amino acids 1-346) is necessary and sufficient for the interaction. While the coiled-coiled domain and a charged region, comprising a.a 102-120, were found to be dispensable, amino acids 121-323 were required for p120(ctn) binding to Gα(12). This region harbors the phosphorylation domain of p120(ctn) and has been postulated as important for RhoA regulation. Downregulation of Src family kinase-induced tyrosine phosphorylation of p120(ctn) was observed in the presence of activated Gα(12). This down-regulation was triggered by three different Gα(12) mutants uncoupled from RhoA signalling. Furthermore, a dominant active form of RhoA did not reduce Src-induced phosphoryaltion of p120(ctn). In summary, our results suggest that Gα(12) binds to p120(ctn) and modulates its phosphorylation status through a Rho-independent mechanism. Gα(12) emerges as an important regulator of p120(ctn) function, and possibly of cadherin-mediated adhesion and/or cell motility.


Asunto(s)
Cateninas/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo , Sitios de Unión , Línea Celular , Regulación hacia Abajo , Humanos , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Tirosina/metabolismo , Catenina delta
10.
Adv Biol Regul ; 83: 100843, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34920983

RESUMEN

The phosphoinositide 3-kinase (PI3K) signalling pathway plays key roles in many cellular processes and is altered in many diseases. The function and mode of action of the pathway have mostly been elucidated in the cytoplasm. However, many of the components of the PI3K pathway are also present in the nucleus at specific sub-nuclear sites including nuclear speckles, nuclear lipid islets and the nucleolus. Nucleoli are membrane-less subnuclear structures where ribosome biogenesis occurs. Processes leading to ribosome biogenesis are tightly regulated to maintain protein translation capacity of cells. This review focuses on nucleolar PI3K signalling and how it regulates rRNA synthesis, as well as on the identification of downstream phosphatidylinositol (3,4,5)trisphosphate effector proteins.


Asunto(s)
Fosfatidilinositol 3-Quinasa , Fosfatidilinositol 3-Quinasas , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Humanos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
11.
Proteomics ; 9(5): 1400-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19253288

RESUMEN

Post-translationally modified peptides present in low concentrations are often not selected for CID, resulting in no sequence information for these peptides. We have developed a software POSTMan (POST-translational Modification analysis) allowing post-translationally modified peptides to be targeted for fragmentation. The software aligns LC-MS runs (MS(1) data) between individual runs or within a single run and isolates pairs of peptides which differ by a user defined mass difference (post-translationally modified peptides). The method was validated for acetylated peptides and allowed an assessment of even the basal protein phosphorylation of phenylalanine hydroxylase (PHA) in intact cells.


Asunto(s)
Péptidos/metabolismo , Procesamiento Proteico-Postraduccional , Programas Informáticos , Acetilación , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Péptidos/análisis , Fenilalanina Hidroxilasa/análisis , Fenilalanina Hidroxilasa/metabolismo , Fosforilación , Proteómica/métodos , Reproducibilidad de los Resultados
12.
Mol Endocrinol ; 22(1): 91-104, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17901130

RESUMEN

The nuclear receptor steroidogenic factor-1 (SF1) is critical for development and function of steroidogenic tissues. Posttranslational modifications are known to influence the transcriptional capacity of SF1, and it was previously demonstrated that serine 203 is phosphorylated. In this paper we report that serine 203 is phosphorylated by a cyclin-dependent kinase 7 (CDK7)-mediated process. As part of the CDK-activating kinase complex, CDK7 is a component of the basal transcription factor TFIIH, and phosphorylation of SF1 as well as SF1-dependent transcription was clearly reduced in cells carrying a mutation that renders the CDK-activating kinase complex unable to interact with the TFIIH core. Coimmunoprecipitation analyses revealed that SF1 and CDK7 reside in the same complex, and kinase assays demonstrated that immunoprecipitated CDK7 and purified TFIIH phosphorylate SF1 in vitro. The CDK inhibitor roscovitine blocked phosphorylation of SF1, and an inactive form of CDK7 repressed the phosphorylation level and the transactivation capacity of SF1. Structural studies have identified phosphoinositides as potential ligands for SF1. Interestingly, we found that mutations designed to block phospholipid binding dramatically decreased the level of SF1 phosphorylation. Together our results suggest a connection between ligand occupation and phosphorylation and association with the basic transcriptional machinery, indicating an intricate regulation of SF1 transactivation.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Factor Esteroidogénico 1/metabolismo , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Línea Celular , Chlorocebus aethiops , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Células HeLa , Humanos , Inmunoprecipitación , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Fosfolípidos/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Purinas/farmacología , Roscovitina , Serina/metabolismo , Factor Esteroidogénico 1/genética , Factor de Transcripción TFIIH/genética , Factor de Transcripción TFIIH/metabolismo , Factores de Transcripción , Transfección
13.
Adv Biol Regul ; 72: 7-21, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31003946

RESUMEN

Biomolecular interactions between proteins and polyphosphoinositides (PPIn) are essential in the regulation of the vast majority of cellular processes. Consequently, alteration of these interactions is implicated in the development of many diseases. PPIn are phosphorylated derivatives of phosphatidylinositol and consist of seven species with different phosphate combinations. PPIn signal by recruiting proteins via canonical domains or short polybasic motifs. Although their actions are predominantly documented on cytoplasmic membranes, six of the seven PPIn are present within the nucleus together with the PPIn kinases, phosphatases and phospholipases that regulate their turnover. Importantly, the contribution of nuclear PPIn in the regulation of nuclear processes has led to an increased recognition of their importance compared to their more accepted cytoplasmic roles. This review summarises our knowledge on the identification and functional characterisation of nuclear PPIn-effector proteins as well as their mode of interactions, which tend to favour polybasic motifs.


Asunto(s)
Núcleo Celular/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Núcleo Celular/enzimología , Núcleo Celular/genética , Humanos , Fosfolipasas/genética , Fosfolipasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo
14.
Endocrinology ; 149(11): 5599-609, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18653709

RESUMEN

Steroidogenic factor 1 (SF1) is expressed in a time- and cell-specific manner in the endocrine system. In this study we present evidence to support that methylation of CpG sites located in the proximal promoter of the gene encoding SF1 contributes to the restricted expression pattern of this nuclear receptor. DNA methylation analyses revealed a nearly perfect correlation between the methylation status of the proximal promoter and protein expression, such that it was hypomethylated in cells that express SF1 but hypermethylated in nonexpressing cells. Moreover, in vitro methylation of this region completely repressed reporter gene activity in transfected steroidogenic cells. Bisulfite sequencing of DNA from embryonic tissue demonstrated that the proximal promoter was unmethylated in the developing testis and ovary, whereas it was hypermethylated in tissues that do not express SF1. Together these results indicate that the DNA methylation pattern is established early in the embryo and stably inherited thereafter throughout development to confine SF1 expression to the appropriate tissues. Chromatin immunoprecipitation analyses revealed that the transcriptional activator upstream stimulatory factor 2 and RNA polymerase II were specifically recruited to this DNA region in cells in which the proximal promoter is hypomethylated, providing functional support for the fact that lack of methylation corresponds to a transcriptionally active gene. In conclusion, we identified a region within the SF1/Sf1 gene that epigenetically directs cell-specific expression of SF1.


Asunto(s)
Metilación de ADN , Factor Esteroidogénico 1/genética , Animales , Secuencia de Bases , Tipificación del Cuerpo/genética , Islas de CpG , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Datos de Secuencia Molecular , Células 3T3 NIH , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Factor Esteroidogénico 1/metabolismo , Distribución Tisular , Células Tumorales Cultivadas
15.
Oncotarget ; 8(3): 3881-3894, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-28002804

RESUMEN

PTEN loss and constitutive activation of the class I phosphoinositide 3-kinase (PI3K) pathway are key drivers of endometrial tumorigenesis. In some cancer types, PTEN-deficient tumors are reliant on class I PI3K p110ß (encoded by PIK3CB) activity but little is known about this contribution in endometrial tumorigenesis. In this study, we find that p110ß is overexpressed in a panel of 7 endometrial cancer cell lines compared to non-transformed cells. Furthermore, in 234 clinically annotated patient samples, PIK3CB mRNA levels increase significantly in the early phase of tumorigenesis from precursors to low grade primary malignant lesions whereas PIK3CA levels are higher in non-endometrioid compared to endometrioid primary tumors. While high levels of either PIK3CA or PIK3CB associate with poor prognosis, only elevated PIK3CB mRNA levels correlate with a high cell cycle signature score in clinical samples. In cancer cell lines, p110α inhibition reduces cell viability by inducing cell death in PIK3CA mutant cells while p110ß inhibition delayed proliferation in PTEN-deficient cells, but not in WT cells. Taken together, our findings suggest that PIK3CB/p110ß contributes to some of the pleiotropic functions of PI3K in endometrial cancer, particularly in the early steps by contributing to cell proliferation.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias Endometriales/patología , Regulación hacia Arriba , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Morfolinas/farmacología , Mutación , Estadificación de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Pirimidinonas/farmacología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia
16.
Sci Rep ; 7(1): 10240, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28860563

RESUMEN

Mutations of the phosphoinositide-3-kinase (PI3K) catalytic subunit alpha gene (PIK3CA) are frequent in endometrial cancer. We sequenced exon9 and exon20 of PIK3CA in 280 primary endometrial cancers to assess the relationship with clinicopathologic variables, patient survival and associations with PIK3CA mRNA and phospho-AKT1 by gene expression and protein data, respectively. While PIK3CA mutations generally had no impact on survival, and were not associated with clinicopathological variables, patients with exon9 charge-changing mutations, providing a positive charge at the substituted amino acid residue, were associated with poor survival (p = 0.018). Furthermore, we characterized PIK3CA mutations in the metastatic setting, including 32 patients with matched primary tumors and metastases, and found a high level of concordance (85.7%; 6 out of 7 patients), suggesting limited heterogeneity. PIK3CA mRNA levels were increased in metastases compared to the primary tumors (p = 0.031), independent of PIK3CA mutation status, which rather associated with reduced PIK3CA mRNA expression. PIK3CA mutated tumors expressed higher p-AKT/AKT protein levels, both within primary (p < 0.001) and metastatic lesion (p = 0.010). Our results support the notion that the PI3K signaling pathway might be activated, both dependent- and independently of PIK3CA mutations, an aspect that should be considered when designing PIK3 pathway targeting strategies in endometrial cancer.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Endometriales/genética , Mutación , Análisis de Secuencia de ADN/métodos , Regulación hacia Arriba , Exones , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Metástasis de la Neoplasia , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Análisis de Supervivencia
17.
Cell Signal ; 17(2): 253-62, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15494216

RESUMEN

Activation of protein kinase C delta (PKCdelta) is believed to be pro-apoptotic. PKCdelta is reported to be reduced in colon cancers. Using a colon cancer cell line, COLO 205, we have examined the roles of PKCdelta in apoptosis and of caspase-3 in the activation and inhibition of PKCdelta. PKCdelta activation with bistratene A and its inhibition with rottlerin induced apoptosis. Effects of PKC activators and inhibitors were additive, suggesting that PKCdelta down-regulation was responsible for the effects on apoptosis. Different apoptotic pathways induced PKCdelta cleavage, but the fragment produced was inactive in kinase assays. Caspase-3 inhibition did not block DNA fragmentation or PKCdelta proteolysis despite blocking intracellular caspase-3 activity. Calpain inhibition with calpeptin did not prevent TPA-induced PKCdelta cleavage. We conclude that in colonocytes, inhibition of PKCdelta is sufficient to lead to caspase-3-independent apoptosis. Caspase-3 does not cleave PKCdelta to an active form, nor does caspase-3 inhibition block apoptosis.


Asunto(s)
Apoptosis , Caspasas/fisiología , Proteína Quinasa C/fisiología , Acetamidas/farmacología , Acetofenonas/farmacología , Alcaloides , Clorometilcetonas de Aminoácidos/farmacología , Antineoplásicos/farmacología , Benzofenantridinas , Benzopiranos/farmacología , Calpaína/antagonistas & inhibidores , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/química , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Inhibidores de Cisteína Proteinasa/farmacología , Fragmentación del ADN/efectos de los fármacos , Dipéptidos/farmacología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Histonas/metabolismo , Humanos , Indometacina/farmacología , Cinética , Fenantridinas/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteína Quinasa C-delta , Piranos/farmacología , Compuestos de Espiro/farmacología , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología
18.
Artículo en Inglés | MEDLINE | ID: mdl-27379015

RESUMEN

Adrenocorticotropic hormone regulates adrenal steroidogenesis mainly via the intracellular signaling molecule cAMP. The effects of cAMP are principally relayed by activating protein kinase A (PKA) and the more recently discovered exchange proteins directly activated by cAMP 1 and 2 (EPAC1 and EPAC2). While the intracellular roles of PKA have been extensively studied in steroidogenic tissues, those of EPACs are only emerging. EPAC1 and EPAC2 are encoded by the genes RAPGEF3 and RAPGEF4, respectively. Whereas EPAC1 is ubiquitously expressed, the expression of EPAC2 is more restricted, and typically found in endocrine tissues. Alternative promoter usage of RAPGEF4 gives rise to three different isoforms of EPAC2 that vary in their N-termini (EPAC2A, EPAC2B, and EPAC2C) and that exhibit distinct expression patterns. EPAC2A is expressed in the brain and pancreas, EPAC2B in steroidogenic cells of the adrenal gland and testis, and EPAC2C has until now only been found in the liver. In this review, we discuss current knowledge on EPAC expression and function with focus on the known roles of EPAC in adrenal gland physiology.

19.
Cell Signal ; 28(10): 1593-603, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27404349

RESUMEN

DNA topoisomerases (Topo) are multifunctional enzymes resolving DNA topological problems such as those arising during DNA replication, transcription and mitosis. Mammalian cells express 2 class II isoforms, Topoisomerases IIα (Topo IIα) and IIß (Topo IIß), which have similar enzymatic properties but are differently expressed, in dividing and pluripotent cells, and in post-mitotic and differentiated cells respectively. Pre-adipocytes re-enter the cell cycle prior to committing to their differentiation and we hypothesised that Topo II could contribute to these processes. We show that Topo IIα expression in 3T3-L1 cells is induced within 16h after the initiation of the differentiation programme, peaks at 24h and rapidly declines thereafter. In contrast Topo IIß was present both in pre-adipocytes and throughout differentiation. Inhibition of PI3K with LY294002, known to prevent adipocyte differentiation, consistently reduced the expression of Topo IIα, whereas a clear effect on Topo IIß was not apparent. In addition, inhibition of mTOR with rapamycin also reduced the protein levels of Topo IIα. Using specific class IA PI3K catalytic subunit inhibitors, we show that p110α inhibition with A66 has the greatest reduction of Topo IIα expression and of differentiation, as measured by triglyceride storage. The timing of Topo IIα expression coincides with the mitotic clonal expansion (MCE) phase of differentiation and inhibition of Topo II with ICRF-187 during this stage decreased PPARγ1 and 2 protein levels and triglyceride storage, whereas inhibition later on has little impact. Moreover, the addition of ICRF-187 had no effect on the incorporation of EdU during S-phase at day 1 but lowered the relative cell numbers on day 2. ICRF-187 also induced an increase in the centri/pericentromeric heterochromatin localisation of Topo IIα, indicating a role for Topo IIα at these locations during MCE. In summary, we present evidence that Topo IIα plays an important role in adipogenesis during MCE and in a PI3K/mTOR-dependent manner. Considering that Topoisomerases II are targets in cancer chemotherapy, our results highlight that treatment of cancer with Topo II inhibitors may alter metabolic processes in the adipose tissue.


Asunto(s)
Adipogénesis , ADN-Topoisomerasas de Tipo II/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Biocatálisis , Proliferación Celular , Células Clonales , ADN/biosíntesis , Isoenzimas/metabolismo , Ratones , Mitosis , PPAR gamma/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba
20.
Mol Endocrinol ; 17(3): 450-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12554771

RESUMEN

Abundant evidence supports the ability of Ras to stimulate thyroid cell proliferation. Stable expression of activated Ras enhances the sensitivity of thyroid cells to apoptosis. We report that apoptosis is a primary and general response of rat thyroid cells to acute expression of activated Ras in the absence or presence of thyrotropin, insulin, and serum, survival factors for thyroid cells. Ras induced apoptosis in quiescent and cycling cells. Concomitantly, Ras stimulated S phase entry in quiescent cells and enhanced G1/S transition in cycling cells. Ras effects on the cell cycle were characterized by delayed progression through S phase and an apparent failure to proceed through G2/M phase. Unlike thyroid cell mitogens, Ras markedly decreased cyclin D1 expression. Although acute expression of Ras decreased cyclin D1 protein levels, cells selected to survive chronic Ras expression exhibited a selective increase in cyclin D1 expression. In summary, thyroid cells harbor an apoptotic program activated by Ras that outstrips the protective effects of thyrotropin, insulin, and serum. Apoptosis is accompanied by dysregulated cell cycle progression, suggesting that cell death may arise, at least in part, as a consequence of inappropriate proliferative cues.


Asunto(s)
Apoptosis/fisiología , Ciclo Celular/fisiología , Glándula Tiroides/citología , Proteínas ras/fisiología , Animales , Western Blotting , Bromodesoxiuridina/metabolismo , Células Cultivadas , Ciclina D1/genética , Ciclina D1/fisiología , Citometría de Flujo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA