Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 27(9)2022 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-35566091

RESUMEN

Rhabdomyosarcoma (RMS) is a highly malignant and metastatic pediatric cancer arising from skeletal muscle myogenic progenitors. Recent studies have shown an important role for AKT signaling in RMS progression. Aberrant activation of the PI3K/AKT axis is one of the most frequent events occurring in human cancers and serves to disconnect the control of cell growth, survival, and metabolism from exogenous growth stimuli. In the study reported here, a panel of five compounds targeting the catalytic subunits of the four class I PI3K isoforms (p110α, BYL-719 inhibitor; p110ß, TGX-221 inhibitor; p110γ, CZC24832; p110δ, CAL-101 inhibitor) and the dual p110α/p110δ, AZD8835 inhibitor, were tested on the RMS cell lines RD, A204, and SJCRH30. Cytotoxicity, cell cycle, apoptosis, and the activation of downstream targets were analyzed. Of the individual inhibitors, BYL-719 demonstrated the most anti-tumorgenic properties. BYL-719 treatment resulted in G1/G0 phase cell cycle arrest and apoptosis. When combined with CAL-101, BYL-719 decreased cell viability and induced apoptosis in a synergistic manner, equaling or surpassing results achieved with AZD8835. In conclusion, our findings indicate that BYL-719, either alone or in combination with the p110δ inhibitor, CAL-101, could represent an efficient treatment for human rhabdomyosarcoma presenting with aberrant upregulation of the PI3K signaling pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Rabdomiosarcoma , Apoptosis , Línea Celular Tumoral , Niño , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas , Quinazolinonas , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología
2.
J Cell Physiol ; 235(2): 1103-1119, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31240713

RESUMEN

Osteosarcoma (OS) is a rare, insidious tumor of mesenchymal origin that most often affects children, adolescents, and young adults. While the primary tumor can be controlled with chemotherapy and surgery, it is the lung metastases that are eventually fatal. Multiple studies into the initial drivers of OS development have been undertaken, but few of these have examined innate immune/inflammatory signaling. A central figure in inflammatory signaling is the innate immune/stress-activated kinase double-stranded RNA-dependent protein kinase (PKR). To characterize the role of PKR in OS, U2OS, and SaOS-2 osteosarcoma cell lines were stably transfected with wild-type or dominant-negative (DN) PKR. Overexpression of PKR enhanced colony formation in soft agar (U2OS and SaOS-2), enhanced cellular migration (U2OS), and invasive migration (SaOS-2). In contrast, overexpression of DN-PKR inhibited attachment-independent growth, migration and/or invasion. These data demonstrate a role for inflammatory signaling in OS formation and migration/invasion and suggest the status of PKR expression/activation may have prognostic value.


Asunto(s)
Osteosarcoma/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Antineoplásicos/farmacología , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Doxorrubicina/farmacología , Fibrosarcoma , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Células 3T3 NIH , ARN Bicatenario , Vincristina/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
3.
FASEB J ; 33(8): 9044-9061, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31095429

RESUMEN

Murine thymoma viral oncogene homolog (AKT) kinases target both cytosolic and nuclear substrates for phosphorylation. Whereas the cytosolic substrates are known to be closely associated with the regulation of apoptosis and autophagy or metabolism and protein synthesis, the nuclear substrates are, for the most part, poorly understood. To better define the role of nuclear AKT, potential AKT substrates were isolated from the nuclear lysates of leukemic cell lines using a phosphorylated AKT substrate antibody and identified in tandem mass spectrometry. Among the proteins identified was adenosine deaminase acting on RNA (ADAR)1p110, the predominant nuclear isoform of the adenosine deaminase acting on double-stranded RNA. Coimmunoprecipitation studies and in vitro kinase assays revealed that AKT-1, -2, and -3 interact with both ADAR1p110 and ADAR2 and phosphorylate these RNA editases. Using site-directed mutagenesis of suspected AKT phosphorylation sites, AKT was found to primarily phosphorylate ADAR1p110 and ADAR2 on T738 and T553, respectively, and overexpression of the phosphomimic mutants ADAR1p110 (T738D) and ADAR2 (T553D) resulted in a 50-100% reduction in editase activity. Thus, activation of AKT has a direct and major impact on RNA editing.-Bavelloni, A., Focaccia, E., Piazzi, M., Raffini, M., Cesarini, V., Tomaselli, S., Orsini, A., Ratti, S., Faenza, I., Cocco, L., Gallo, A., Blalock, W. L. AKT-dependent phosphorylation of the adenosine deaminases ADAR-1 and -2 inhibits deaminase activity.


Asunto(s)
Adenosina Desaminasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/química , Adenosina Desaminasa/genética , Sustitución de Aminoácidos , Sitios de Unión/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Activación Enzimática , Células HEK293 , Humanos , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Edición de ARN , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
4.
J Cell Physiol ; 234(7): 10907-10917, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30536897

RESUMEN

Osteosarcoma (OS) is the most common pediatric malignant neoplasia of the skeletal system. It is characterized by a high degree of malignancy and a severe tendency to metastasize. In the past decade, many studies have provided evidence that the phosphoinositide 3-kinase (PI3K) signaling pathway is one of the most frequently altered pathways in human cancer, and has a critical role in driving tumor initiation and progression. Here, we have analyzed the therapeutic potential of the pan-PI3K inhibitor NVP-BKM120, which has recently entered clinical Phase II for treatment of PI3K-dependent cancers on three OS cell lines. We observed a concentration- and time-dependent decrease of Ser473 p-Akt as well as reduced levels of Thr37/46 p-4E-BP1, an indicator of the mammalian target of rapamycin complex 1 activity. All OS cell lines used in this study responded to BKM120 treatment with an arrest of cell proliferation, an increase in cell mortality, and an increase in caspase-3 activity. MG-63 cells were the most responsive cell line, demonstrating a significant increase in sub-G1 cells, and a rapid induction of cell death. Furthermore, we demonstrate that BKM120 is more effective when used in combination with other standard chemotherapeutic drugs. Combining BKM120 with vincristine demonstrated a more synergistic effect than BKM120 with doxorubicin in all the lines. Hence, we suggest that BKM120 may be a novel therapy for the treatment of OS presenting with anomalous upregulation of the PI3K signaling pathway.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Morfolinas/farmacología , Osteosarcoma/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Caspasa 3/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Osteosarcoma/enzimología , Osteosarcoma/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
5.
Int J Mol Sci ; 20(11)2019 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-31163577

RESUMEN

Energetically speaking, ribosome biogenesis is by far the most costly process of the cell and, therefore, must be highly regulated in order to avoid unnecessary energy expenditure. Not only must ribosomal RNA (rRNA) synthesis, ribosomal protein (RP) transcription, translation, and nuclear import, as well as ribosome assembly, be tightly controlled, these events must be coordinated with other cellular events, such as cell division and differentiation. In addition, ribosome biogenesis must respond rapidly to environmental cues mediated by internal and cell surface receptors, or stress (oxidative stress, DNA damage, amino acid depletion, etc.). This review examines some of the well-studied pathways known to control ribosome biogenesis (PI3K-AKT-mTOR, RB-p53, MYC) and how they may interact with some of the less well studied pathways (eIF2α kinase and RNA editing/splicing) in higher eukaryotes to regulate ribosome biogenesis, assembly, and protein translation in a dynamic manner.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas/metabolismo , Transducción de Señal , Animales , Biomarcadores , Ciclo Celular/genética , Susceptibilidad a Enfermedades , Factor 2 Eucariótico de Iniciación/metabolismo , Espacio Extracelular/metabolismo , Genes myc , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Edición de ARN , Empalme del ARN , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Estrés Fisiológico , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética
6.
J Cell Physiol ; 231(3): 623-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26217938

RESUMEN

Bone morphogenetic protein 2 (BMP-2) is a critical growth factor that directs osteoblast differentiation and bone formation. Phosphoinositide-phospholipase Cß 1 (PLCß1) plays a crucial role in the initiation of the genetic program responsible for muscle differentiation. Differentiation of C2C12 mouse myoblasts in response to insulin stimulation is characterized by a marked increase in nuclear PLCß1. Here, the function of PLCß1 in the osteogenic differentiation was investigated. Briefly, in C2C12 cells treated with BMP-2 we assist to a remarkable increase in PLCß1 protein and mRNA expression. The data regarding the influence on differentiation demonstrated that PLCß1 promotes osteogenic differentiation by up-regulating alkaline phosphatase (ALP). Moreover, PLCß1 is present in the nuclear compartment of these cells and overexpression of a cytosolic-PLCß1mutant (cyt-PLCß1), which lacks a nuclear localization sequence, prevented the differentiation of C2C12 cells into osteocytes. Recent evidence indicates that miRNAs act as important post transcriptional regulators in a large number of processes, including osteoblast differentiation. Since miR-214 is a regulator of Osterix (Osx) which is an osteoblast-specific transcription factor that is needful for osteoblast differentiation and bone formation, we further investigated whether PLCß1 could be a potential target of miR-214 in the control of osteogenic differentiation by gain- and loss- of function experiment. The results indicated that inhibition of miR-214 in C2C12 cells significantly enhances the protein level of PLCß1 and promotes C2C12 BMP-2-induced osteogenesis by targeting PLCß1.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Fosfolipasa C beta/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Regulación de la Expresión Génica/genética , Ratones , Mioblastos/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/genética , Fosfolipasa C beta/efectos de los fármacos , Fosfolipasa C beta/genética
7.
FASEB J ; 29(4): 1383-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550457

RESUMEN

The phosphoinositide-dependent signal transduction pathway has been implicated in the control of a variety of biologic processes, such as the regulation of cellular metabolism and homeostasis, cell proliferation and differentiation, and apoptosis. One of the key players in the regulation of inositol lipid signaling is the phospholipase Cß1 (PI-PLCß1), that hydrolyzes phosphatidylinositol 4,5-bisphosphate [PtIns(4,5)P2], giving rise to the second messengers inositol triphosphate and diacylglicerol. PI-PLCß1 has been associated with the regulation of several cellular functions, some of which have not yet been fully understood. In particular, it has been reported that PI-PLCß1 protects murine fibroblasts from oxidative stress-induced cell death. The mediators of oxidative stress, reactive oxygen species (ROS), have been shown to regulate major epigenetic processes, causing the silencing of tumor suppressors and enhancing the proliferation of leukemic cells under oxidative stress. Investigation of the interplay between ROS, PI-PLCß1, and their signaling mediators in leukemia might therefore reveal innovative targets of pharmacological therapy in the treatment for leukemia. In this work, we demonstrate that in pro-B-lymphoblastic cells (Ba/F3), treated with H2O2, PI-PLCß1b conferred resistance to cell death, promoting cell cycle progression and cell proliferation and influencing the expression of cyclin A and E. Interestingly, we found that, expression of PI-PLCß1b affects the activity of caspase-3, caspase-7, and of several protein kinases induced by oxidative stress. In particular, PI-PLCß1b expression completely abolished the phosphorylation of Erk1/2 MAP kinases, down-regulated phosphatase and tensin homolog (PTEN), and up-regulated the phosphorylation of Akt, thereby sustaining cellular proliferation.


Asunto(s)
Ciclina E/metabolismo , Fosfolipasa C beta/metabolismo , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular , Ciclina A/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Peróxido de Hidrógeno/toxicidad , Interleucina-3/metabolismo , Ratones , Estrés Oxidativo , Células Precursoras de Linfocitos B/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal
8.
J Cell Physiol ; 230(3): 587-94, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25160985

RESUMEN

Here we report that both PLCß1a and PLCß1b are relevant regulators of erythropoiesis in that kinamycin F, a potent inducer of γ-globin production in K562 cells, caused a selectively reduction of both PLCß1 isozymes even though the results point out that the effect of the drug is mainly directed toward the expression of the PLCß1a isoform. We have identified a different role for the two isozymes as regulators of K562 differentiation process induced by kinamycin F. The overexpression of PLCß1b induced an increase in γ-globin expression even in the absence of kinamycin F. Moreover during K562 differentiation, cyclin D3 level is regulated by PLCß1 signaling pathway. Namely the amplification of the expression of the PLCß1a, but not of PLCß1b, is able to maintain high levels of expression of cyclin D3 even after treatment with kinamycin F. This could be due to their different distribution in the cell compartments since the amount of PLCß1b is mainly present in the nucleus in respect to PLCß1a. Our data indicate that the amplification of PLCß1a expression, following treatment with kinamycin F, confers a real advantage to K562 cells viability and protects cells themselves from apoptosis.


Asunto(s)
Ciclina D3/genética , Fosfolipasa C beta/biosíntesis , Isoformas de Proteínas/biosíntesis , gamma-Globinas/biosíntesis , Apoptosis , Diferenciación Celular/genética , Línea Celular , Ciclina D3/biosíntesis , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Isoformas de Proteínas/genética , Quinonas/administración & dosificación , Transducción de Señal/efectos de los fármacos
9.
IUBMB Life ; 67(4): 239-54, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25904163

RESUMEN

Prohibitins (PHBs) are a highly conserved class of proteins first discovered as inhibitors of cellular proliferation. Since then PHBs have been found to have a significant role in transcription, nuclear signaling, mitochondrial structural integrity, cell division, and cellular membrane metabolism, placing these proteins among the key regulators of pathologies such as cancer, neuromuscular degeneration, and other metabolic diseases. The human genome encodes two PHB proteins, prohibitin 1 (PHB1) and prohibitin 2 (PHB2), which function not only as a heterodimeric complex, but also independently. While many previous reviews have focused on the better characterized prohibitin, PHB1, this review focuses on PHB2 and new data concerning its cellular functions both in complex with PHB1 and independent of PHB1.


Asunto(s)
Proteínas Represoras/fisiología , Animales , Expresión Génica , Humanos , Prohibitinas , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal
10.
FASEB J ; 28(5): 2009-19, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24522204

RESUMEN

The AKT/PKB kinase is essential for cell survival, proliferation, and differentiation; however, aberrant AKT activation leads to the aggressiveness and drug resistance of many human neoplasias. In the human acute promyelocytic leukemia cell line NB4, nuclear AKT activity increases during all-trans retinoic acid (ATRA)-mediated differentiation. As nuclear AKT activity is associated with differentiation, we sought to identify the nuclear substrates of AKT that were phosphorylated after ATRA treatment. A proteomics-based search for nuclear substrates of AKT in ATRA-treated NB4 cells was undertaken by using 2D-electrophoresis/mass spectrometry (MS) in combination with an anti-AKT phospho-substrate antibody. Western blot analysis, an in vitro kinase assay, and/or site-directed mutagenesis were performed to further characterize the MS findings. MS analysis revealed prohibitin (PHB)-2, a multifunctional protein involved in cell cycle progression and the suppression of oxidative stress, to be a putative nuclear substrate of AKT. Follow-up studies confirmed that AKT phosphorylates PHB2 on Ser-91 and that forced expression of the PHB2(S91A) mutant results in a rapid loss of viability and apoptotic cell death. Activation of nuclear AKT during ATRA-mediated differentiation results in the phosphorylation of several proteins, including PHB2, which may serve to coordinate nuclear-mitochondrial events during differentiation.


Asunto(s)
Diferenciación Celular , Leucemia Promielocítica Aguda/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Tretinoina/metabolismo , Apoptosis , Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Estrés Oxidativo , Fosforilación , Prohibitinas , Proteómica , Transducción de Señal
11.
Mol Cell Proteomics ; 12(8): 2220-35, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23665500

RESUMEN

Two isoforms of inositide-dependent phospholipase C ß1 (PI-PLCß1) are generated by alternative splicing (PLCß1a and PLCß1b). Both isoforms are present within the nucleus, but in contrast to PLCß1a, the vast majority of PLCß1b is nuclear. In mouse erythroid leukemia cells, PI-PLCß1 is involved in the regulation of cell division and the balance between cell proliferation and differentiation. It has been demonstrated that nuclear localization is crucial for the enzymatic function of PI-PLCß1, although the mechanism by which this nuclear import occurs has never been fully characterized. The aim of this study was to characterize both the mechanism of nuclear localization and the molecular function of nuclear PI-PLCß1 by identifying its interactome in Friend's erythroleukemia isolated nuclei, utilizing a procedure that coupled immuno-affinity purification with tandem mass spectrometry analysis. Using this procedure, 160 proteins were demonstrated to be in association with PI-PLCß1b, some of which have been previously characterized, such as the splicing factor SRp20 (Srsf3) and Lamin B (Lmnb1). Co-immunoprecipitation analysis of selected proteins confirmed the data obtained via mass spectrometry. Of particular interest was the identification of the nuclear import proteins Kpna2, Kpna4, Kpnb1, Ran, and Rangap1, as well as factors involved in hematological malignancies and several anti-apoptotic proteins. These data give new insight into possible mechanisms of nuclear trafficking and functioning of this critical signaling molecule.


Asunto(s)
Proteínas Nucleares/metabolismo , Fosfolipasa C beta/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Expresión Génica , Ratones , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Espectrometría de Masas en Tándem/métodos
12.
Biochem J ; 463(1): 115-22, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25005557

RESUMEN

PU.1 is essential for the differentiation of haemopoietic precursors and is strongly implicated in leukaemogenesis, yet the protein interactions that regulate its activity in different myeloid lineages are still largely unknown. In the present study, by combining fluorescent EMSA (electrophoretic mobility-shift assay) with MS, we reveal the presence of hnRNP K (heterogeneous nuclear ribonucleoprotein K) in molecular complexes that PU.1 forms on the CD11b promoter during the agonist-induced maturation of AML (acute myeloid leukaemia)-derived cells along both the granulocytic and the monocytic lineages. Although hnRNP K and PU.1 act synergistically during granulocytic differentiation, hnRNP K seems to have a negative effect on PU.1 activity during monocytic maturation. Since hnRNP K acts as a docking platform, integrating signal transduction pathways to nucleic acid-directed processes, it may assist PU.1 in activating or repressing transcription by recruiting lineage-specific components of the transcription machinery. It is therefore possible that hnRNP K plays a key role in the mechanisms underlying the specific targeting of protein-protein interactions identified as mediators of transcriptional activation or repression and may be responsible for the block of haemopoietic differentiation.


Asunto(s)
Antígeno CD11b/metabolismo , Diferenciación Celular , Granulocitos/metabolismo , Leucemia Mieloide Aguda/metabolismo , Monocitos/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Ribonucleoproteínas/metabolismo , Transactivadores/metabolismo , Antígeno CD11b/genética , Línea Celular Tumoral , Granulocitos/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Monocitos/patología , Proteínas Proto-Oncogénicas/genética , Ribonucleoproteínas/genética , Transactivadores/genética
13.
J Cell Physiol ; 229(8): 1047-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24347309

RESUMEN

The double-strand RNA-dependent protein kinase, PKR, plays a central role in inflammatory/chronic stress-mediated pathologies such as cancer, diabetes, and neuro/muscular degenerative diseases. Although a significant amount of research has been conducted to elucidate the role of PKR signaling in the cytosol, only recently has attention been paid to the role of PKR in the nuclear compartment. Previously our group reported that phosphorylated forms of PKR are present in the nucleus of acute leukemic cell lines, representing a reservoir of active kinase that responds to stress. Using the CCRF-CEM acute T-cell leukemia cell line, a PKR-specific inhibitor, co-immunoprecipitation and a proteomics approach, which included affinity purified mass spectrometry analysis (AP/MS), we identified the proteins present in active and inactive PKR nuclear complexes. Of the proteins identified in the PKR complexes, sixty-nine (69) were specific to the active complex, while thirty-eight (38) were specific to the inactive complex. An additional thirteen (13) proteins associated specifically with both complexes. The majority of the proteins identified are involved in, ribosome biogenesis, RNA splicing, mRNA stability, gene expression, cell cycle, or chromatin organization, including several with known significance to normal hematopoiesis and/or hematological disease. In agreement with the AP/MS data, basal- or over-expression of PKR under normal growth conditions favored cell proliferation in the tested cell lines, whereas pharmacological inhibition of PKR or shRNA-mediated knock-down did not. PKR was also found to influence the isoform and the level of expression of the proto-oncogene MYC.


Asunto(s)
ARN Mensajero/metabolismo , eIF-2 Quinasa/metabolismo , Transporte Activo de Núcleo Celular , Puntos de Control del Ciclo Celular , División Celular , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Proto-Oncogenes Mas , ARN Mensajero/genética , Transcriptoma , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
14.
ACS Nano ; 18(3): 2047-2065, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38166155

RESUMEN

The use of piezoelectric nanomaterials combined with ultrasound stimulation is emerging as a promising approach for wirelessly triggering the regeneration of different tissue types. However, it has never been explored for boosting chondrogenesis. Furthermore, the ultrasound stimulation parameters used are often not adequately controlled. In this study, we show that adipose-tissue-derived mesenchymal stromal cells embedded in a nanocomposite hydrogel containing piezoelectric barium titanate nanoparticles and graphene oxide nanoflakes and stimulated with ultrasound waves with precisely controlled parameters (1 MHz and 250 mW/cm2, for 5 min once every 2 days for 10 days) dramatically boost chondrogenic cell commitment in vitro. Moreover, fibrotic and catabolic factors are strongly down-modulated: proteomic analyses reveal that such stimulation influences biological processes involved in cytoskeleton and extracellular matrix organization, collagen fibril organization, and metabolic processes. The optimal stimulation regimen also has a considerable anti-inflammatory effect and keeps its ability to boost chondrogenesis in vitro, even in an inflammatory milieu. An analytical model to predict the voltage generated by piezoelectric nanoparticles invested by ultrasound waves is proposed, together with a computational tool that takes into consideration nanoparticle clustering within the cell vacuoles and predicts the electric field streamline distribution in the cell cytoplasm. The proposed nanocomposite hydrogel shows good injectability and adhesion to the cartilage tissue ex vivo, as well as excellent biocompatibility in vivo, according to ISO 10993. Future perspectives will involve preclinical testing of this paradigm for cartilage regeneration.


Asunto(s)
Condrogénesis , Proteómica , Nanogeles , Hidrogeles/farmacología , Diferenciación Celular , Ingeniería de Tejidos
15.
Mol Cancer ; 12: 165, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24330829

RESUMEN

BACKGROUND: Beyond its possible correlation with stemness of tumor cells, CD133/prominin1 is considered an important marker in breast cancer, since it correlates with tumor size, metastasis and clinical stage of triple-negative breast cancers (TNBC), to date the highest risk breast neoplasia. METHODS: To study the correlation between the levels of CD133 expression and the biology of breast-derived cells, CD133low and CD133high cell subpopulations isolated from triple negative MDA-MB-231 cells were compared in terms of malignant properties and protein expression. RESULTS: High expression of CD133 characterizes cells with larger adhesion area, lower proliferation rate and reduced migration speed, indicative of a less undifferentiated phenotype. Conversely, when compared with CD133low cells, CD133high cells show higher invasive capability and increased expression of proteins involved in metastasis and drug-resistance of breast tumors. Among the signalling proteins examined, PLC-ß2 expression inversely correlates with the levels of CD133 and has a role in inducing the CD133high cells to CD133low cells conversion, suggesting that, in TNBC cells, the de-regulation of this PLC isoform is responsible of the switch from an early to a mature tumoral phenotype also by reducing the expression of CD133. CONCLUSIONS: Since CD133 plays a role in determining the invasiveness of CD133high cells, it may constitute an attractive target to reduce the metastatic potential of TNBC. In addition, our data showing that the forced up-regulation of PLC-ß2 counteracts the invasiveness of CD133-positive MDA-MB-231 cells might contribute to identify unexplored key steps responsible for the TNBC high malignancy, to be considered for potential therapeutic strategies.


Asunto(s)
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Péptidos/metabolismo , Fosfolipasa C beta/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Antígeno AC133 , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Invasividad Neoplásica , Fenotipo , Fosfolipasa C beta/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología
16.
FASEB J ; 26(1): 203-10, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21974932

RESUMEN

Type 2 diabetes is a heterogeneous disorder caused by concomitant impairment of insulin secretion by pancreatic ß cells and of insulin action in peripheral target tissues. Studies with inhibitors and agonists established a role for PLC in the regulation of insulin secretion but did not distinguish between effects due to nuclear or cytoplasmic PLC signaling pathways that act in a distinct fashion. We report that in MIN6 ß cells, PLCß1 localized in both nucleus and cytoplasm, PLCδ4 in the nucleus, and PLCγ1 in the cytoplasm. By silencing each isoform, we observed that they all affected glucose-induced insulin release both at basal and high glucose concentrations. To elucidate the molecular basis of PLC regulation, we focused on peroxisome proliferator-activated receptor-γ (PPARγ), a nuclear receptor transcription factor that regulates genes critical to ß-cell maintenance and functions. Silencing of PLCß1 and PLCδ4 resulted in a decrease in the PPARγ mRNA level. By means of a PPARγ-promoter-luciferase assay, the decrease could be attributed to a PLC action on the PPARγ-promoter region. The effect was specifically observed on silencing of the nuclear and not the cytoplasmic PLC. These findings highlight a novel pathway by which nuclear PLCs affect insulin secretion and identify PPARγ as a novel molecular target of nuclear PLCs.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , PPAR gamma/metabolismo , Fosfolipasa C beta/metabolismo , Fosfolipasa C delta/metabolismo , Fosfolipasa C gamma/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/enzimología , Citoplasma/enzimología , Diabetes Mellitus Tipo 2/metabolismo , Silenciador del Gen , Glucosa/farmacocinética , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/enzimología , Insulinoma , Ratones , Fosfolipasa C beta/genética , Fosfolipasa C delta/genética , Fosfolipasa C gamma/genética , Sistemas de Mensajero Secundario/fisiología
17.
FASEB J ; 26(7): 3042-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22459146

RESUMEN

Phosphoinositide-phospholipase C ß1 (PLCß1) plays a crucial role in the initiation of the genetic program responsible for muscle differentiation. We previously demonstrated that nuclear PLCß1 activates the cyclin D3 promoter during the differentiation of myoblasts to myotubes, indicating that PLCß1 is essential for cyclin D3 promoter activation and gene transcription, through c-jun/AP1. Myotonic dystrophy (DM) is the most prevalent form of muscular dystrophy in adults. DM type 1 (DM1) and type 2 (DM2) are dominantly inherited multisystem disorders. DM1 is triggered by the pathological expansion of a (CTG)(n) triplet repeat in the gene coding for DMPK, the dystrophia myotonica-protein kinase, whereas a (CCTG)(n) tetranucleotide repeat expansion in the ZNF9 gene, encoding a CCHC-type zinc finger protein, causes DM2. We found that, unlike in normal myotubes, the level of expression of PLCß1 in DM1 and DM2 cells was already elevated in proliferating cells. Treatment with insulin induced a dramatic decrease in the amount of PLCß1. During differentiation, cyclin D3 and myogenin were elevated in normal myotubes, whereas differentiating DM1 and DM2 cells did not increase these proteins. Forced expression of PLCß1 in DM1 and DM2 cells increased the expression of differentiation markers, myogenin and cyclin D3, and enhanced fusion of DM myoblasts. These results highlight again that PLCß1 expression is a key player in myoblast differentiation, functioning as a positive regulator in the correction of delayed differentiation of skeletal muscle in DM human myoblasts.


Asunto(s)
Trastornos Miotónicos/enzimología , Trastornos Miotónicos/genética , Distrofia Miotónica/enzimología , Distrofia Miotónica/genética , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Ciclina D3/genética , Ciclina D3/metabolismo , Perfilación de la Expresión Génica , Humanos , Insulina/farmacología , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/enzimología , Mioblastos Esqueléticos/patología , Miogenina/genética , Miogenina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Regulación hacia Arriba
18.
Genes (Basel) ; 14(7)2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37510291

RESUMEN

The advent of next generation sequencing (NGS) has fostered a shift in basic analytic strategies of a gene expression analysis in diverse pathologies for the purposes of research, pharmacology, and personalized medicine. What was once highly focused research on individual signaling pathways or pathway members has, from the time of gene expression arrays, become a global analysis of gene expression that has aided in identifying novel pathway interactions, the discovery of new therapeutic targets, and the establishment of disease-associated profiles for assessing progression, stratification, or a therapeutic response. But there are significant caveats to this analysis that do not allow for the construction of the full picture. The lack of timely updates to publicly available databases and the "hit and miss" deposition of scientific data to these databases relegate a large amount of potentially important data to "garbage", begging the question, "how much are we really missing?" This brief perspective aims to highlight some of the limitations that RNA binding/modifying proteins and RNA processing impose on our current usage of NGS technologies as relating to cancer and how not fully appreciating the limitations of current NGS technology may negatively affect therapeutic strategies in the long run.


Asunto(s)
Empalme Alternativo , Neoplasias , Humanos , Empalme Alternativo/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Edición de ARN/genética , Perfilación de la Expresión Génica , Neoplasias/genética , Neoplasias/terapia
19.
Nutrients ; 15(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36771452

RESUMEN

Approximately 7% of cancers arising in children and 1% of those arising in adults are soft tissue sarcomas (STS). Of these malignancies, rhabdomyosarcoma (RMS) is the most common. RMS survival rates using current therapeutic protocols have remained largely unchanged in the past decade. Thus, it is imperative that the main molecular drivers in RMS tumorigenesis are defined so that more precise, effective, and less toxic therapies can be designed. Curcumin, a common herbal supplement derived from plants of the Curcuma longa species, has an exceptionally low dietary biotoxicity profile and has demonstrated anti-tumorigenic benefits in vitro. In this study, the anti-tumorigenic activity of curcumin was assessed in rhabdomyosarcoma cell lines and used to identify the major pathways responsible for curcumin's anti-tumorigenic effects. Curcumin treatment resulted in cell cycle arrest, inhibited cell migration and colony forming potential, and induced apoptotic cell death. Proteome profiler array analysis demonstrated that curcumin treatment primarily influenced flux through the AKT-mammalian target of rapamycin (mTOR), signal transducer and activator of transcription (STAT), AMP-dependent kinase (AMPK), and p53 associated pathways in a rhabdomyosarcoma subtype-specific manner. Thus, the strategic, combinational therapeutic targeting of these pathways may present the best option to treat this group of tumors.


Asunto(s)
Antineoplásicos , Curcumina , Rabdomiosarcoma , Adulto , Niño , Humanos , Curcumina/farmacología , Curcumina/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína p53 Supresora de Tumor/genética , Serina-Treonina Quinasas TOR/metabolismo , Antineoplásicos/farmacología , Rabdomiosarcoma/tratamiento farmacológico , Apoptosis , Línea Celular Tumoral
20.
Cancer Gene Ther ; 30(9): 1285-1295, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37353558

RESUMEN

Ewing sarcoma (EWS) is a challenging pediatric cancer characterized by vast intra-tumor heterogeneity. We evaluated the RNA-binding protein IGF2BP3, whose high expression correlates with a poor prognosis and an elevated tendency of metastases, as a possible soluble mediator of inter-cellular communication in EWS. Our data demonstrate that (i) IGF2BP3 is detected in cell supernatants, and it is released inside extracellular vesicles (EVs); (ii) EVs from IGF2BP3-positive or IGF2BP3-negative EWS cells reciprocally affect cell migration but not the proliferation of EWS recipient cells; (iii) EVs derived from IGF2BP3-silenced cells have a distinct miRNA cargo profile and inhibit the PI3K/Akt pathway in recipient cells; (iv) the 11 common differentially expressed miRNAs associated with IGF2BP3-positive and IGF2BP3-negative EVs correctly group IGF2BP3-positive and IGF2BP3-negative clinical tissue specimens. Overall, our data suggest that IGF2BP3 can participate in the modulation of phenotypic heterogeneity.


Asunto(s)
Vesículas Extracelulares , Sarcoma de Ewing , Niño , Humanos , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Vesículas Extracelulares/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA