Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 15: 1351536, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38495105

RESUMEN

Neuroblastoma (NB) is the most common extracranial pediatric solid tumor originating from the abnormal development of cells of the sympathoadrenal lineage of the neural crest. Targeting GD2 ganglioside (GD2), a glycolipid expressed on neuroblastoma cells, with GD2 ganglioside-recognizing antibodies affects several pivotal signaling routes that drive or influence the malignant phenotype of the cells. Previously performed gene expression profiling helped us to identify the PHLDA1 (pleckstrin homology-like domain family A member 1) gene as the most upregulated gene in the IMR-32 human neuroblastoma cells treated with the mouse 14G2a monoclonal antibody. Mass spectrometry-based proteomic analyses were applied to better characterize a role of PHLDA1 protein in the response of neuroblastoma cells to chimeric ch14.18/CHO antibody. Additionally, global protein expression profile analysis in the IMR-32 cell line with PHLDA1 silencing revealed the increase in biological functions of mitochondria, accompanied by differentiation-like phenotype of the cells. Moreover, mass spectrometry analysis of the proteins co-immunoprecipitated using anti-PHLDA1-specific antibody, selected a group of possible PHLDA1 binding partners. Also, a more detailed analysis suggested that PHLDA1 interacts with the DCAF7/AUTS2 complex, a key component of neuronal differentiation in vitro. Importantly, our results indicate that PHLDA1 silencing enhances the EGF receptor signaling pathway and combinatory treatment of gefitinib and ch14.18/CHO antibodies might be beneficial for neuroblastoma patients. Data are available via ProteomeXchange with the identifier PXD044319.

2.
Acta Biochim Pol ; 69(3): 485-494, 2022 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-35810485

RESUMEN

In recent years, immunotherapy has been identified as an effective treatment method for high-risk neuroblastoma. A previous study demonstrated that an anti-GD2 ganglioside (GD2) mouse 14G2a monoclonal antibody (mAb) combined with a small molecule, i.e., an aurora A kinase inhibitor (MK-5108), significantly increased cytotoxicity against human neuroblastoma cells, as compared to monotherapy. This study aimed to demonstrate the mechanism of neuroblastoma cell death in vitro following the addition of an anti-GD2 human-mouse chimeric ch14.18/CHO mAb (presently used in clinics) and two aurora A inhibitors (MK-5108 and MK-8745). The effects of the aforementioned agents on neuroblastoma cells were determined by measuring the level of ATP, the level of apoptotic and necroptotic markers, and the activity of caspase 3/7. The results revealed that the ch14.18/CHO mAb decreased cellular ATP levels in the IMR-32 and CHP-134 neuroblastoma cell lines, similarly to the 14G2a mAb. Regarding ch14.18/CHO mAb treated IMR-32 cells, the observed cytotoxic effect was concomitant with induced caspase 3 cleavage, which indicated the induction of apoptosis in IMR-32 cells, but not in CHP-134 cells. Furthermore, the MK-5108 inhibitor induced apoptosis, as indicated by the increased cleavage of caspase 3 and increased activity of caspase 3/7. However, the presence of necroptosis was ruled out in MK-5108-treated IMR-32 and CHP-134 cells. In summary, the effects of the combination of ch14.18/CHO mAb and aurora A kinase inhibitors (MK-5108 and MK-8745) were shown to enhance apoptosis in IMR-32 cells compared to when used individually.


Asunto(s)
Antineoplásicos , Neuroblastoma , Adenosina Trifosfato , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Apoptosis , Aurora Quinasa A , Caspasa 3 , Línea Celular Tumoral , Ácidos Ciclohexanocarboxílicos , Gangliósidos/metabolismo , Humanos , Ratones , Neuroblastoma/tratamiento farmacológico , Piperazinas , Tiazoles
3.
Mol Carcinog ; 60(4): 227-241, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33544962

RESUMEN

The suppressive activity of monocyte chemoattractant protein 1-induced protein 1 (MCPIP1) an inflammation-related ribonuclease, has been described in a few cancer types but has yet to be assessed in the most common subtype of skin cancer: melanoma. Here, we have evaluated the MCPIP1 expression in melanoma tissues by reanalysis of publicly available transcriptome data from 89 melanoma samples, and immunohistochemical staining of 21 primary and 81 metastatic melanomas. Our data implicated decreased MCPIP1 expression in melanoma tumors compared to normal tissues, and positive correlation between high ribonuclease expression and melanoma-specific survival of patients. To investigate the ribonuclease activity in melanoma cells, MCPIP1 was ectopically expressed in the MV3 human melanoma cell line. Following the transcriptome, proteome, and intracellular signaling of MCPIP1-overexpressing MV3 cells was assessed via real-time quantitative polymerase chain reaction, Western blot analysis, and RNAseq. MV3 cells overexpressing MCPIP1 exhibited a broad range of alterations in the transcriptome and proteome, as well as in the phosphorylation status of a number of proteins, strongly indicating MCPIP1-dependent cell cycle arrest and inhibition of Akt/mTOR signaling in these cells. Moreover, we have shown, that MCPIP1 overexpression downregulates miRNA-193a-3p expression in MV3 cells. Furthermore, the majority of the described effects were dependent on the ribonucleolytic activity of the protein. The presented body of data strongly suggests a potential tumor suppressor role and possible future application as a positive prognostic marker of MCPIP1 protein in melanoma.


Asunto(s)
Regulación hacia Abajo , Melanoma/mortalidad , Ribonucleasas/genética , Ribonucleasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , MicroARNs/genética , Fosforilación , Pronóstico , Proteómica , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de Supervivencia
4.
RNA Biol ; 18(1): 144-156, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32757706

RESUMEN

The role of the inflammation-silencing ribonuclease, MCPIP1 (monocyte chemoattractant protein-induced protein 1), in neoplasia continuous to emerge. The ribonuclease can cleave not only inflammation-related transcripts but also some microRNAs (miRNAs) and viral RNAs. The suppressive effect of the protein has been hitherto suggested in breast cancer, clear cell renal cell carcinoma, osteosarcoma, and neuroblastoma. Our previous results have demonstrated a reduced levels of several oncogenes, as well as inhibited growth of neuroblastoma cells upon MCPIP1 overexpression. Here, we investigate the mechanisms underlying the suppression of MYCN proto-oncogene, bHLH transcription factor (MYCN)-amplified neuroblastoma cells overexpressing the MCPIP1 protein. We showed that the levels of several transcripts involved in cell cycle progression decreased in BE(2)-C and KELLY cells overexpressing MCPIP1 in a ribonucleolytic activity-dependent manner. However, RNA immunoprecipitation indicated that only AURKA mRNA (encoding for Aurora A kinase) interacts with the ribonuclease. Furthermore, the application of a luciferase assay suggested MCPIP1-dependent destabilization of the transcript. Further analyses demonstrated that the entire conserved region of AURKA seems to be indispensable for the interaction with the MCPIP1 protein. Additionally, we examined the effect of the ribonuclease overexpression on the miRNA expression profile in MYCN-amplified neuroblastoma cells. However, no significant alterations were observed. Our data indicate a key role of the binding and cleavage of the AURKA transcript in an MCPIP1-dependent suppressive effect on neuroblastoma cells.


Asunto(s)
Aurora Quinasa A/genética , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/genética , ARN Mensajero/genética , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Regiones no Traducidas 3' , Ciclo Celular/genética , Línea Celular Tumoral , Humanos , MicroARNs/genética , Conformación de Ácido Nucleico , Unión Proteica , Proto-Oncogenes Mas , División del ARN , Interferencia de ARN , Estabilidad del ARN , Ribonucleasas/química , Factores de Transcripción/química
5.
J Cell Biochem ; 121(5-6): 3406-3425, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31919874

RESUMEN

Monocyte chemoattractant protein-1-induced protein 1 (MCPIP1) has a multidomain structure, which assures its pleiotropic activity. The physiological functions of this protein include repression of inflammatory processes and the prevention of immune disorders. The influence of MCPIP1 on the cell cycle of cancer cells has not been sufficiently elucidated. A previous study by our group reported that overexpression of MCPIP1 affects the cell viability, inhibits the activation of the phosphoinositide-3 kinase/mammalian target of rapamycin signalling pathway, and reduces the stability of the MYCN oncogene in neuroblastoma (NB) cells. Furthermore, a decrease in expression and phosphorylation levels of cyclin-dependent kinase (CDK) 1, which has a key role in the M phase of the cell cycle, was observed. On the basis of these previous results, the purpose of our present study was to elucidate the influence of MCPIP1 on the cell cycle of NB cells. It was confirmed that ectopic overexpression of MCPIP1 in two human NB cell lines, KELLY and BE(2)-C, inhibited cell proliferation. Furthermore, flow cytometric analyses and imaging of the cell cycle with a fluorescence ubiquitination cell-cycle indicator test, demonstrated that overexpression of MCPIP1 causes an accumulation of NB cells in the G1 phase of the cell cycle, while the possibility of an increase in G0 phase due to induction of quiescence or senescence was excluded. Additional assessment of the molecular machinery responsible for the transition between the cell-cycle phases confirmed that MCPIP1 overexpression reduced the expression of cyclins A2, B1, D1, D3, E1, and E2 and decreased the phosphorylation of CDK2 and CDK4, as well as retinoblastoma protein. In conclusion, the present results indicated a relevant impact of overexpression of MCPIP1 on the cell cycle, namely a block of the G1/S cell-cycle checkpoint, resulting in arrest of NB cells in the G1 phase.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína Quinasa CDC2/metabolismo , Regulación Neoplásica de la Expresión Génica , Neuroblastoma/metabolismo , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Ciclo Celular , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Perfilación de la Expresión Génica , Humanos , Análisis de los Mínimos Cuadrados , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncogenes , Fosforilación , Programas Informáticos , Transfección , Ubiquitinación
6.
Int J Oncol ; 53(4): 1787-1799, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30066861

RESUMEN

MicroRNAs (miRNAs) are a class of small non­coding RNAs involved in post­transcriptional gene regulation. Furthermore, dysregulation of miRNA expression is an important factor in the pathogenesis of neuroblastoma. Our previous study identified that overexpression of monocyte chemoattractant protein­induced protein 1 protein led to a significant downregulation of a novel miRNA molecule, miRNA­3613­3p. In the present study, the potential involvement of miRNA­3613­3p in the cell biology of neuroblastoma was investigated. It was identified that the expression of miRNA­3613­3p varies among a range of human neuroblastoma cell lines. As the delineation of the functions of a miRNA requires the identification of its target genes, seven putative mRNAs that may be regulated by miRNA­3613­3p were selected. Furthermore, it was identified that overexpression of miRNA­3613­3p causes significant downregulation of several genes exhibiting tumor suppressive potential [encoding apoptotic protease­activating factor 1 (APAF1), Dicer, DNA fragmentation factor subunit ß, von Hippel­Lindau protein and neurofibromin 1] in BE(2)­C human neuroblastoma cells. APAF1 mRNA was the most significantly decreased transcript in the cells with miRNA­3613­3p overexpression. In accordance with the aforementioned results, the downregulation of cleaved caspase-9 and lack of activation of executive caspases in BE(2)­C cells following miRNA­3613­3p overexpression was observed. The results of the present study suggest a potential underlying molecular mechanism of apoptosis inhibition via APAF1 downregulation in human neuroblastoma BE(2)­C cells with miRNA­3613­3p overexpression.


Asunto(s)
Apoptosis/genética , Factor Apoptótico 1 Activador de Proteasas/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neuroblastoma/genética , Regiones no Traducidas 3'/genética , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , MicroARNs/genética , ARN Mensajero/genética
7.
Apoptosis ; 23(9-10): 492-511, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30027525

RESUMEN

The process of autophagy and its role in survival of human neuroblastoma cell cultures was studied upon addition of an anti-GD2 ganglioside (GD2) 14G2a mouse monoclonal antibody (14G2a mAb) and an aurora A kinase specific inhibitor, MK-5108. It was recently shown that combination of these agents significantly potentiates cytotoxicity against IMR-32 and CHP-134 neuroblastoma cells in vitro, as compared to the inhibitor used alone. In this study we gained mechanistic insights on autophagy in the observed cytotoxic effects exerted by both agents using cytotoxicity assays, RT-qPCR, immunoblotting, and autophagy detection methods. Enhancement of the autophagy process in the 14G2a mAb- and MK-5108-treated IMR-32 cells was documented by assessing autophagic flux. Application of a lysosomotropic agent-chloroquine (CQ) affected the 14G2a mAb- and MK-5108-stimulated autophagic flux. It is our conclusion that the 14G2a mAb (40 µg/ml) and MK-5108 inhibitor (0.1 µM) induce autophagy in IMR-32 cells. Moreover, the combinatorial treatment of IMR-32 cells with the 14G2a mAb and CQ significantly potentiates cytotoxic effect, as compared to CQ used alone. Most importantly, we showed that interfering with autophagy at its early and late step augments the 14G2a mAb-induced apoptosis, therefore we can conclude that inhibition of autophagy is the primary mechanism of the CQ-mediated sensitization to the 14G2a mAb-induced apoptosis. Although, there was no virtual stimulation of autophagy in the 14G2a mAb-treated CHP-134 neuroblastoma cells, we were able to show that PHLDA1 protein positively regulates autophagy and this process exists in a mutually exclusive manner with apoptosis in PHLDA1-silenced CHP-134 cells.


Asunto(s)
Apoptosis/genética , Aurora Quinasa A/genética , Autofagia/genética , Neuroblastoma/genética , Factores de Transcripción/genética , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Aurora Quinasa A/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/farmacología , Gangliósidos/antagonistas & inhibidores , Gangliósidos/genética , Humanos , Ratones , Neuroblastoma/patología , Tiazoles/farmacología , Factores de Transcripción/antagonistas & inhibidores
8.
J Cell Biochem ; 118(7): 1741-1755, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27935099

RESUMEN

The main physiological function of MCPIP1 (regnase-1) is negative regulation of inflammation. Moreover, roles of regnase-1 in apoptosis and differentiation have also been described, but its involvement in cancer is yet to be fully recognized. Earlier, we showed a lack of expression of MCPIP1 in both primary tumors and several neuroblastoma cell lines. Additionally, we reported that levels of MCPIP1 and the key neuroblastoma oncoprotein-MYCN were inversely correlated in BE(2)-C clones overexpressing the MCPIP1 gene. Here, we show that exogenous expression of the MCPIP1 protein decreases MYCN mRNA and protein levels without changing the MYCN mRNA half-life. Furthermore, it was shown that MCPIP1-wt exogenous expression affects levels and phosphorylation of MYCN partners such as Aurora A (Thr288), CDC2 (Tyr15 and Thr161), GSK3ß (Ser9), and key cellular components of Akt/mTOR signaling, which regulate MYCN stability and activation. In accordance with the obtained results, we found increased phosphorylation of MYCN protein at Thr58 that causes destabilization of the oncoprotein. Moreover, it is shown that exogenous expression of MCPIP1 does not cause apoptosis. Our data extend knowledge on roles of MCPIP1 in our model and link the protein to regulation of expression and stability of MYCN through decrease of signaling via Akt/mTOR pathway. J. Cell. Biochem. 118: 1741-1755, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/metabolismo , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Aurora Quinasa A/genética , Aurora Quinasa A/metabolismo , Western Blotting , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/genética , Fosforilación/genética , Fosforilación/fisiología , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ribonucleasas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética
9.
Int J Oncol ; 49(2): 823-37, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27278006

RESUMEN

We have recently shown that mRNA and protein of PHLDA1 (pleckstrin-homology-like domain family A, member  1) were by far the most upregulated molecules upon treatment of IMR-32 cells with the anti-GD2 ganglioside monoclonal antibody 14G2a. Hence, we decided to study functions of PHLDA1 using human neuroblastoma IMR-32 cells as a model. Here, we show that constitutive expression of mRNA and protein of the PHLDA1 gene in IMR-32 cells was inversely correlated with transcript of the AURKA gene and Aurora A oncoprotein. Next, we silenced PHLDA1 expression in IMR-32 cells using an shRNA interference method. We report that IMR-32 cells with stable downregulation of PHLDA1 showed enhanced cellular ATP levels and an increase in mitochondrial membrane potential, as compared to control and non-transduced cells. We demonstrated that downregulation of PHLDA1 leads to a significant increase in expression of Aurora A and TRKB that are markers of poor prognosis in neuroblastoma. Also, we measured an increase in Aurora A and Akt kinases phosphorylation in the cells. Most importantly, PHLDA1-silenced cells were less susceptible to apoptosis than control cells, as shown by the lower expression of cleaved caspase-3 and PARP as well as a decreased activity of caspase-3 and -7. Our study negatively correlates expression of PHLDA1 and Aurora A in IMR-32 cells and sheds new light on functions of PHLDA1 in the neuroblastoma tumor cells, suggesting its role as a pro-apoptotic protein. Additionally, our results show possible links of the protein to regulation of features of mitochondria and formation of autophagosomes.


Asunto(s)
Aurora Quinasa A/biosíntesis , Glicoproteínas de Membrana/biosíntesis , Neuroblastoma/genética , Proteínas Tirosina Quinasas/biosíntesis , Factores de Transcripción/genética , Adenosina Trifosfato/biosíntesis , Apoptosis/genética , Aurora Quinasa A/genética , Autofagia/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Glicoproteínas de Membrana/genética , Potencial de la Membrana Mitocondrial/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Neuroblastoma/patología , Proteínas Tirosina Quinasas/genética , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/genética , Receptor trkB , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/biosíntesis
10.
J Cell Biochem ; 117(3): 694-707, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26308737

RESUMEN

The recently discovered MCPIP1 (monocyte chemoattractant protein-induced protein 1), a multidomain protein encoded by the MCPIP1 (ZC3H12A) gene, has been described as a new differentiation factor, a ribonuclease, and a deubiquitination-supporting factor. However, its role in cancer is poorly recognized. Our recent analysis of microarrays data showed a lack of expression of the MCPIP1 transcript in primary neuroblastoma, the most common extracranial solid tumor in children. Additionally, enforced expression of the MCPIP1 gene in BE(2)-C cells caused a significant decrease in neuroblastoma proliferation and viability. Aim of the present study was to further investigate the role of MCPIP1 in neuroblastoma, using expression DNA microarrays and microRNA microarrays. Transient transfections of BE(2)-C cells were used for overexpression of either wild type of MCPIP1 (MCPIP1-wt) or its RN-ase defective mutant (MCPIP1-ΔPIN). We have analyzed changes of transcriptome and next, we have used qRT-PCR to verify mRNA levels of selected genes responding to MCPIP1 overexpression. Additionally, protein levels were determined for some of the selected genes. The choline transporter, CTL1, encoded by the SLC44A1 gene, was significantly repressed at the specific mRNA and protein levels and most importantly this translated into a decreased choline transport in MCPIP1-overexpressing cells. Then, we have found microRNA-3613-3p as the mostly altered in the pools of cells overexpressing the wild type MCPIP1. Next, we analyzed the predicted targets of the miR-3613-3p and validated them using qRT-PCR and western blot. These results indicate that the expression of miR-3613-3p might be regulated by MCPIP1 by cleavage of its precursor form.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neuroblastoma/metabolismo , Ribonucleasas/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Transporte Biológico , Línea Celular Tumoral , Colina/metabolismo , Humanos , MicroARNs/metabolismo , Neuroblastoma/genética , Interferencia de ARN , Ribonucleasas/genética , Factores de Transcripción/genética , Transcriptoma
11.
Acta Biochim Pol ; 62(3): 423-33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26284262

RESUMEN

Neuroblastoma is the most common extra-cranial solid tumor of childhood and it is characterized by the presence of a glycosphingolipid, GD2 ganglioside. Monoclonal antibodies targeting the antigen are currently tested in clinical trials. Additionally, several research groups reported results revealing that ganglioside-specific antibodies can affect cellular signaling and cause direct cytotoxicity against tumor cells. To shed more light on gene expression signatures of tumor cells, we used microarrays to analyze changes of transcriptome in IMR-32 human neuroblastoma cell cultures treated with doxorubicin (DOX) or a mouse monoclonal antibody binding to GD2 ganglioside 14G2a (mAb) for 24 h. The obtained results highlight that disparate cellular pathways are regulated by doxorubicin and 14G2a. Next, we used RT-PCR to verify mRNA levels of selected DOX-responsive genes such as RPS27L, PPM1D, SESN1, CDKN1A, TNFSF10B, and 14G2a-responsive genes such as SVIL, JUN, RASSF6, TLX2, ID1. Then, we applied western blot and analyzed levels of RPS27L, PPM1D, sestrin 1 proteins after DOX-treatment. Additionally, we aimed to measure effects of doxorubicin and topotecan (TPT) and 14G2a on expression of a novel human NDUFAF2 gene encoding for mimitin protein (MYC-induced mitochondrial protein) and correlate it with expression of the MYCN gene. We showed that expression of both genes was concomitantly decreased in the 14G2a-treated IMR-32 cells after 24 h and 48 h. Our results extend knowledge on gene expression profiles after application of DOX and 14G2a in our model and reveal promising candidates for further research aimed at finding novel anti-neuroblastoma targets.


Asunto(s)
Doxorrubicina/química , Gangliósidos/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neuroblastoma/patología , Animales , Antibióticos Antineoplásicos/química , Anticuerpos Monoclonales/química , Línea Celular Tumoral/efectos de los fármacos , Análisis por Conglomerados , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Proteína Proto-Oncogénica N-Myc , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/metabolismo , Programas Informáticos , Topotecan/química , Transcriptoma
12.
Int J Oncol ; 47(3): 1143-59, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26134970

RESUMEN

Mechanisms leading to inhibitory effects of an anti-GD2 ganglioside (GD2) 14G2a mouse monoclonal antibody (mAb) and PI3K/Akt/mTOR pathway inhibitors on human neuroblastoma cell survival were studied in vitro. We have recently shown on IMR-32, CHP­134, and LA-N-1 neuroblastoma cells that targeting GD2 with the mAb decreases cell viability of the cell lines. In this study we used cytotoxicity assays, proteomic arrays and immunoblotting to evaluate the response of the three cell lines to the anti­GD2 14G2a mAb and specific PI3K/Akt/mTOR pathway inhibitors. We show here that the mAb modulates intracellular signal transduction through changes in several kinases and their substrates phosphorylation. More detailed analysis of the PI3K/Akt/mTOR pathway showed significant decrease in activity of Akt, mTOR, p70 S6 and 4E-BP1 proteins and transient increase in PTEN (a suppressor of the pathway), leading to inhibition of the signaling network responsible for stimulation of translation and proliferation. Additionally, combining the GD2-specific 14G2a mAb with an Akt inhibitor (perifosine), dual mTOR/PI3K inhibitors (BEZ-235 and SAR245409), and a pan-PI3K inhibitor (LY294002) was shown to enhance cytotoxic effects against IMR-32, CHP­134 and LA-N-1 cells. Our study extends knowledge on mechanisms of action of the 14G2a mAb on the neuroblastoma cells. Also, it stresses the need for further delineation of molecular signal orchestration aimed at more reasonable selection of drugs to target key cellular pathways in quest for better cure for neuroblastoma patients.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Gangliósidos/antagonistas & inhibidores , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis , Línea Celular Tumoral , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolinas/farmacología , Quinoxalinas/farmacología , Sulfonamidas/farmacología , Serina-Treonina Quinasas TOR/metabolismo
13.
Mol Med Rep ; 12(1): 535-42, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25695841

RESUMEN

Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables, which has been shown to exert an anti-cancer effect when tested in vitro and in vivo. The anti-cancer effects of SFN encompass induction of cytoprotective autophagy; therefore, the present study aimed to determine whether the chemopreventive activity of SFN may be potentiated by inhibition of autophagy. The present study provided detailed insight into the susceptibility of human neuroblastoma cells to treatment with synthetic SFN, in combination with an inhibitor of autophagy, 3-methyladenine (3-MA). The present study confirmed the suppression of the viability of the human neuroblastoma cell line BE(2)-C by SFN and reported the inhibition of DNA synthesis, as determined by a decrease in tritiated thymidine incorporation. Furthermore, the results verified the effectiveness of SFN in inducing apoptosis in the BE(2)-C cell line as demonstrated by caspase activation, increased protein expression levels of B-cell lymphoma 2-associated X protein and loss of mitochondrial membrane potential. Combined treatment of the cells with SFN with 3-MA proved to be effective in decreasing cell viability, through a mechanism that may proceed via the early induction of autophagy by SFN, followed by induction of apoptosis, as well as inhibition of autophagy by 3-MA.


Asunto(s)
Adenina/análogos & derivados , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Adenina/administración & dosificación , Autofagia/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Isotiocianatos/administración & dosificación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neuroblastoma/genética , Neuroblastoma/patología , Especies Reactivas de Oxígeno/metabolismo , Sulfóxidos
14.
Oncol Rep ; 31(5): 2385-92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24626857

RESUMEN

The importance of monocyte chemotactic protein-1-induced protein 1 (MCPIP1) in the negative regulation of inflammatory reactions has already been extensively studied. However, its role in cancer is not yet established. We studied MCPIP1 gene expression in primary human neuroblastomas and several neuroblastoma cell lines. Our results showed a lack of MCPIP1 expression in primary neuroblastoma tumors. Moreover, it was found that the low expression of the protein measured in human neuroblastoma cell lines might be important for neuroblastoma survival, since enforced MCPIP1 gene expression in human neuroblastoma BE(2)-C cells caused a significant decrease in neuroblastoma cell viability and proliferation.


Asunto(s)
Proliferación Celular/genética , Neuroblastoma/genética , Factores de Transcripción/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Endorribonucleasas/metabolismo , Expresión Génica , Humanos , Ribonucleasas , Factores de Transcripción/biosíntesis , Transfección
15.
Cancer Lett ; 341(2): 248-64, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23962557

RESUMEN

The mechanism of the inhibitory effect of anti-GD2 ganglioside (GD2) 14G2a mouse monoclonal antibody (mAb) on human neuroblastoma cells survival was studied in vitro. It was recently shown in IMR-32 cells that death induced by this antibody exhibited several characteristics typical of apoptosis. In this study we used cytotoxixity assays, qRT-PCR and immunoblotting to evaluate the response of several human neuroblastoma cell lines to the anti-GD2 14G2a mAb. We showed that the mAb decreases all three aurora kinases expression and phosphorylation in IMR-32 and LA-N-1 cells. Most importantly, we show, that MK-5108 specific aurora A kinase inhibitor decreases neuroblastoma cell survival, and when used in combination with the mAb, significantly potentiates cytotoxicity against IMR-32, CHP-134, and LA-N-5 neuroblastoma cells in vitro. It was shown that downregulation of aurora A kinase by the therapeutic antibody is associated with decreased levels of MYCN protein in cytoplasm, and induced expression of PHLDA1 and P53 proteins.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Ácidos Ciclohexanocarboxílicos/farmacología , Gangliósidos/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Aurora Quinasa A/genética , Aurora Quinasa A/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Citometría de Flujo , Gangliósidos/inmunología , Gangliósidos/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Ratones , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...