Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Physiol ; 234(7): 11188-11199, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30565691

RESUMEN

Clusterin (CLU) is a chaperone-like protein with multiple functions. sCLU is frequently upregulated in prostate tumor cells after chemo- or radiotherapy and after surgical or pharmacological castration. Moreover, CLU has been documented to modulate the cellular homolog of murine thymoma virus akt8 oncogene (AKT) activity. Here, we investigated how CLU overexpression influences phosphatidylinositol 3'-kinase (PI3K)/AKT signaling in human normal and cancer epithelial prostate cells. Human prostate cells stably transfected with CLU were broadly profiled by reverse phase protein array (RPPA), with particular emphasis on the PI3K/AKT pathway. The effect of CLU overexpression on normal and cancer cell motility was also tested. Our results clearly indicate that CLU overexpression enhances phosphorylation of AKT restricted to isoform 2. Mechanistically, this can be explained by the finding that the phosphatase PH domain leucine-rich repeat-containing protein phosphatase 1 (PHLPP1), known to dephosphorylate AKT2 at S474, is markedly downregulated by CLU, whereas miR-190, a negative regulator of PHLPP1, is upregulated. Moreover, we found that phosphatase and tensin homolog (PTEN) was heavily phosphorylated at the inhibitory site S380, contributing to the hyperactivation of AKT signaling. By keeping AKT2 phosphorylation high, CLU dramatically enhances the migratory behavior of prostate epithelial cell lines with different migratory and invasive phenotypes, namely prostate normal epithelial 1A (PNT1A) and prostatic carcinoma 3 (PC3) cells. Altogether, our results unravel for the first time a circuit by which CLU can switch a low migration phenotype toward a high migration phenotype, through miR-190-dependent downmodulation of PHLPP1 expression and, in turn, stabilization of AKT2 phosphorylation.


Asunto(s)
Clusterina/metabolismo , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células 3T3 , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Clusterina/genética , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , MicroARNs/genética , Células PC-3 , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Próstata/patología , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética
2.
Mol Carcinog ; 58(5): 708-721, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30582225

RESUMEN

Cells in non-invasive breast lesions are widely believed to possess molecular alterations that render them either susceptible or refractory to the acquisition of invasive capability. One such alteration could be the ectopic expression of the ß2 isoform of phosphoinositide-dependent phospholipase C (PLC-ß2), known to counteract the effects of hypoxia in low-invasive breast tumor-derived cells. Here, we studied the correlation between PLC-ß2 levels and the propensity of non-invasive breast tumor cells to acquire malignant features. Using archival FFPE samples and DCIS-derived cells, we demonstrate that PLC-ß2 is up-regulated in DCIS and that its forced down-modulation induces an epithelial-to-mesenchymal shift, expression of the cancer stem cell marker CD133, and the acquisition of invasive properties. The ectopic expression of PLC-ß2 in non-transformed and DCIS-derived cells is, to some extent, dependent on the de-regulation of miR-146a, a tumor suppressor miRNA in invasive breast cancer. Interestingly, an inverse relationship between the two molecules, indicative of a role of miR-146a in targeting PLC-ß2, was not detected in primary DCIS from patients who developed a second invasive breast neoplasia. This suggests that alterations of the PLC-ß2/miR-146a relationship in DCIS may constitute a molecular risk factor for the appearance of new breast lesions. Since neither traditional classification systems nor molecular characterizations are able to predict the malignant potential of DCIS, as is possible for invasive ductal carcinoma (IDC), we propose that the assessment of the PLC-ß2/miR-146a levels at diagnosis could be beneficial for identifying whether DCIS patients may have either a low or high propensity for invasive recurrence.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Células Madre Neoplásicas/patología , Fosfolipasa C beta/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/metabolismo , Proliferación Celular , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Fosfolipasa C beta/genética , Pronóstico , Células Tumorales Cultivadas
3.
J Cell Mol Med ; 22(6): 3149-3158, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29532991

RESUMEN

It has been recently demonstrated that high pre-treatment levels of miR-29b positively correlated with the response of patients with acute myeloid leukaemia (AML) to hypomethylating agents. Upmodulation of miR-29b by restoring its transcriptional machinery appears indeed a tool to improve therapeutic response in AML. In cells from acute promyelocytic leukaemia (APL), miR-29b is regulated by PU.1, in turn upmodulated by agonists currently used to treat APL. We explored here the ability of PU.1 to also regulate miR-29b in non-APL cells, in order to identify agonists that, upmodulating PU.1 may be beneficial in hypomethylating agents-based therapies. We found that PU.1 may regulate miR-29b in the non-APL Kasumi-1 cells, showing the t(8;21) chromosomal rearrangement, which is prevalent in AML and correlated with a relatively low survival. We demonstrated that the PU.1-mediated contribution of the 2 miR-29b precursors is cell-related and almost completely dependent on adequate levels of Vav1. Nuclear PU.1/Vav1 association accompanies the transcription of miR-29b but, at variance with the APL-derived NB4 cells, in which the protein is required for the association of PU.1 with both miRNA promoters, Vav1 is part of molecular complexes to the PU.1 consensus site in Kasumi-1. Our results add new information on the transcriptional machinery that regulates miR-29b expression in AML-derived cells and may help in identifying drugs useful in upmodulation of this miRNA in pre-treatment of patients with non-APL leukaemia who can take advantage from hypomethylating agent-based therapies.


Asunto(s)
Leucemia Mieloide Aguda/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Regulación Leucémica de la Expresión Génica/genética , Humanos , Leucemia Mieloide Aguda/patología , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Regiones Promotoras Genéticas
4.
J Cell Physiol ; 233(10): 6440-6454, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29667769

RESUMEN

Despite considerable progress in treatment protocols, B-lineage acute lymphoblastic leukemia (B-ALL) displays a poor prognosis in about 15-20% of pediatric cases and about 60% of adult patients. In addition, life-long irreversible late effects from chemo- and radiation therapy, including secondary malignancies, are a growing problem for leukemia survivors. Targeted therapy holds promising perspectives for cancer treatment as it may be more effective and have fewer side effects than conventional therapies. The phosphatidylinositol 3-phosphate kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) signaling pathway is a key regulatory cascade which controls proliferation, survival and drug-resistance of cancer cells, and it is frequently upregulated in the different subtypes of B-ALL, where it plays important roles in the pathophysiology, maintenance and progression of the disease. Moreover, activation of this signaling cascade portends a poorer prognosis in both pediatric and adult B-ALL patients. Promising preclinical data on PI3K/Akt/mTOR inhibitors have documented their anticancer activity in B-ALL and some of these novel drugs have entered clinical trials as they could lead to a longer event-free survival and reduce therapy-associated toxicity for patients with B-ALL. This review highlights the current status of PI3K/Akt/mTOR inhibitors in B-ALL, with an emphasis on emerging evidence of the superior efficacy of synergistic combinations involving the use of traditional chemotherapeutics or other novel, targeted agents.


Asunto(s)
Fosfatidilinositol 3-Quinasa/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética , Linfocitos B/patología , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Humanos , Terapia Molecular Dirigida , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
5.
BMC Cancer ; 18(1): 1194, 2018 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-30497437

RESUMEN

BACKGROUND: The presence of hypoxic areas is common in all breast lesions but no data clearly correlate low oxygenation with the acquisition of malignant features by non-invasive cells, particularly by cells from ductal carcinoma in situ (DCIS), the most frequently diagnosed tumor in women. METHODS: By using a DCIS-derived cell line, we evaluated the effects of low oxygen availability on malignant features of non-invasive breast tumor cells and the possible role of all-trans retinoic acid (ATRA), a well-known anti-leukemic drug, in counteracting the effects of hypoxia. The involvement of the ß2 isoform of PI-PLC (PLC-ß2), an ATRA target in myeloid leukemia cells, was also investigated by specific modulation of the protein expression. RESULTS: We demonstrated that moderate hypoxia is sufficient to induce, in DCIS-derived cells, motility, epithelial-to-mesenchymal transition (EMT) and expression of the stem cell marker CD133, indicative of their increased malignant potential. Administration of ATRA supports the epithelial-like phenotype of DCIS-derived cells cultured under hypoxia and keeps down the number of CD133 positive cells, abrogating almost completely the effects of poor oxygenation. We also found that the mechanisms triggered by ATRA in non-invasive breast tumor cells cultured under hypoxia is in part mediated by PLC-ß2, responsible to counteract the effects of low oxygen availability on CD133 levels. CONCLUSIONS: Overall, we assigned to hypoxia a role in increasing the malignant potential of DCIS-derived cells and we identified in ATRA, currently used in treatment of acute promyelocytic leukemia (APL), an agonist potentially useful in preventing malignant progression of non-invasive breast lesions showing hypoxic areas.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Hipoxia/metabolismo , Tretinoina/farmacología , Biomarcadores , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica , Humanos , Hipoxia/genética , Inmunohistoquímica , Clasificación del Tumor , Estadificación de Neoplasias , Oxígeno/metabolismo , Fosfolipasa C beta/metabolismo
6.
BMC Cancer ; 17(1): 617, 2017 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-28870198

RESUMEN

BACKGROUND: The malignant potential of triple negative breast cancer (TNBC) is also dependent on a sub-population of cells with a stem-like phenotype. Among the cancer stem cell markers, CD133 and EpCAM strongly correlate with breast tumor aggressiveness, suggesting that simultaneous targeting of the two surface antigens may be beneficial in treatment of TNBC. Since in TNBC-derived cells we demonstrated that PLC-ß2 induces the conversion of CD133high to CD133low cells, here we explored its possible role in down-modulating the expression of both CD133 and EpCAM and, ultimately, in reducing the number of TNBC cells with a stem-like phenotype. METHODS: A magnetic step-by-step cell isolation with antibodies directed against CD133 and/or EpCAM was performed on the TNBC-derived MDA-MB-231 cell line. In the same cell model, PLC-ß2 was over-expressed or down-modulated and cell proliferation and invasion capability were evaluated by Real-time cell assays. The surface expression of CD133, EpCAM and CD44 in the different experimental conditions were measured by multi-color flow cytometry immunophenotyping. RESULTS: A CD133+/EpCAM+ sub-population with high proliferation rate and invasion capability is present in the MDA-MB-231 cell line. Over-expression of PLC-ß2 in CD133+/EpCAM+ cells reduced the surface expression of both CD133 and EpCAM, as well as proliferation and invasion capability of this cellular subset. On the other hand, the up-modulation of PLC-ß2 in the whole MDA-MB-231 cell population reduced the number of cells with a CD44+/CD133+/EpCAM+ stem-like phenotype. CONCLUSIONS: Since selective targeting of the cells with the highest aggressive potential may have a great clinical importance for TNBC, the up-modulation of PLC-ß2, reducing the number of cells with a stem-like phenotype, may be a promising goal for novel therapies aimed to prevent the progression of aggressive breast tumors.


Asunto(s)
Antígeno AC133/metabolismo , Molécula de Adhesión Celular Epitelial/metabolismo , Fosfolipasa C beta/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Biomarcadores , Recuento de Células , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunofenotipificación , Clasificación del Tumor , Células Madre Neoplásicas/metabolismo , Fenotipo , Fosfolipasa C beta/genética
7.
Mol Carcinog ; 55(12): 2210-2221, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26785288

RESUMEN

Limited oxygen availability plays a critical role in the malignant progression of breast cancer by orchestrating a complex modulation of the gene transcription largely dependent on the tumor phenotype. Invasive breast tumors belonging to different molecular subtypes are characterized by over-expression of PLC-ß2, whose amount positively correlates with the malignant evolution of breast neoplasia and supports the invasive potential of breast tumor cells. Here we report that hypoxia modulates the expression of PLC-ß2 in breast tumor cells in a phenotype-related manner, since a decrease of the protein was observed in the BT-474 and MCF7 cell lines while an increase was revealed in MDA-MB-231 cells as a consequence of low oxygen availability. Under hypoxia, the down-modulation of PLC-ß2 was mainly correlated with the decrease of the EMT marker E-cadherin in the BT-474 cells and with the up-regulation of the stem cell marker CD133 in MCF7 cells. The increase of PLC-ß2 induced by low oxygen in MDA-MB-231 cells supports the hypoxia-related reorganization of actin cytoskeleton and sustains invasion capability. In all examined cell lines, but with an opposite role in the ER-positive and ER-negative cells, PLC-ß2 was involved in the hypoxia-induced increase of HIF-1α, known to affect both EMT and CD133 expression. Our data include PLC-ß2 in the complex and interconnected signaling pathways induced by low oxygen availability in breast tumor cells and suggest that the forced modulation of PLC-ß2 programmed on the basis of tumor phenotype may prevent the malignant progression of breast neoplasia as a consequence of intra-tumoral hypoxia. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Hipoxia/genética , Fosfolipasa C beta/genética , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Hipoxia de la Célula , Línea Celular Tumoral , Femenino , Humanos , Hipoxia/complicaciones , Hipoxia/metabolismo , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Fosfolipasa C beta/análisis , Fosfolipasa C beta/metabolismo , Transducción de Señal
8.
Cell Mol Life Sci ; 72(12): 2337-47, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25712020

RESUMEN

OBJECTIVE: Increased activity of PI3K/AKT/mTOR pathway has been observed in a huge number of malignancies. This pathway can function as a prosurvival factor in leukemia stem cells and early committed leukemic precursors and its inhibition is regarded as a therapeutic approach. Accordingly, the aim of this review is to evaluate the PI3K/Akt/mTOR inhibitors used in leukemia models. DISCUSSION: Inhibition of the PI3K/AKT/mTOR pathway has been reported to have beneficial therapeutic effects in leukemias, both in vitro in leukemia cell lines and in vivo in animal models. Overall, the use of dual PI3K/mTOR inhibitor, dual Akt/RTK inhibitor, Akt inhibitor, selective inhibitor of PI3K, mTOR inhibitor and dual PI3K/PDK1 inhibitor in CML, AML, APL, CLL, B-ALL and T-ALL has a better therapeutic effect than conventional treatments. CONCLUSIONS: Targeting the PI3K/Akt/mTOR pathway may have pro-apoptotic and antiproliferative effects on hematological malignancies. Furthermore, modulation of miRNA can be used as a novel therapeutic approach to regulate the PI3K/Akt/mTOR pathway. However, both aspects require further clinical studies.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia/tratamiento farmacológico , Terapia Molecular Dirigida , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Humanos , Leucemia/metabolismo , Leucemia/patología
9.
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
10.
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
11.
Invest New Drugs ; 30(1): 417-22, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20953816

RESUMEN

The multi-kinase inhibitor dasatinib induced a variable but significant decrease of viability in both p53(wild-type) (EHEB, JVM-2, JVM-3) and p53(mutated) (MEC-1, MEC-2, BJAB) prolymphocytic B leukemic cells, due to a combination of cell cycle block in G1 and apoptosis. Antibody phospho-kinase array analysis revealed that dasatinib inhibited the phosphorylation of various kinases, including ERK1/2 and p38/MAPK as well as of STAT3 transcription factors, in both p53(wild-type) and p53(mutated) cells. Therefore, dasatinib might offer a novel therapeutic strategy not only for p53(wild-type), but also for p53(mutated) B malignancies that have the worst prognosis and urgently need innovative therapeutic approaches.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos B/efectos de los fármacos , Células Precursoras de Granulocitos/efectos de los fármacos , Leucemia Prolinfocítica Tipo Células B/metabolismo , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Proteína p53 Supresora de Tumor/genética , Apoptosis/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dasatinib , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/patología , Humanos , Leucemia Prolinfocítica Tipo Células B/genética , Leucemia Prolinfocítica Tipo Células B/patología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Cell Tissue Res ; 345(1): 163-75, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21647562

RESUMEN

Vav1 is a critical signal transducer for both the development and function of normal hematopoietic cells, in which it regulates the acquisition of maturation-related properties, including adhesion, motility, and phagocytosis. Vav1 is also important for the agonist-induced maturation of acute promyelocytic leukemia (APL)-derived promyelocytes, in which it promotes the acquisition of a mature phenotype by playing multiple functions at both cytoplasmic and nuclear levels. We investigated the possible role of Vav1 in the differentiation of leukemic precursors to monocytes/macrophages. Tumoral promyelocytes in which Vav1 was negatively modulated were induced to differentiate into monocytes/macrophages with phorbol-12-myristate-13-acetate (PMA) and monitored for their maturation-related properties. We found that Vav1 was crucial for the phenotypical differentiation of tumoral myeloid precursors to monocytes/macrophages, in terms of CD11b expression, adhesion capability and cell morphology. Confocal analysis revealed that Vav1 may synergize with actin in modulating nuclear morphology of PMA-treated adherent cells. Our data indicate that, in tumoral promyelocytes, Vav1 is a component of lineage-specific transduction machineries that can be recruited by various differentiating agents. Since Vav1 plays a central role in the completion of the differentiation program of leukemic promyelocytes along diverse hematopoietic lineages, it can be considered a common target for developing new therapeutic strategies for the various subtypes of myeloid leukemias.


Asunto(s)
Diferenciación Celular , Leucemia Promielocítica Aguda/patología , Macrófagos/metabolismo , Macrófagos/patología , Monocitos/metabolismo , Monocitos/patología , Proteínas Proto-Oncogénicas c-vav/metabolismo , Actinas/metabolismo , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Forma del Núcleo Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/metabolismo , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Fenotipo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Regulación hacia Arriba/efectos de los fármacos
13.
Invest New Drugs ; 29(6): 1303-13, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20628892

RESUMEN

The serine/threonine kinase Akt, a downstream effector of phosphatidylinositol 3-kinase (PI3K), is involved in cell survival and anti-apoptotic signaling. Akt has been shown to be constitutively expressed in a variety of human tumors including hepatocellular carcinoma (HCC). In this report we analyzed the status of Akt pathway in three HCC cell lines, and tested cytotoxic effects of Akt pathway inhibitors LY294002, Wortmannin and Inhibitor VIII. In Mahlavu human hepatoma cells Akt was constitutively activated, as demonstrated by its Ser473 phosphorylation, downstream hyperphosphorylation of BAD on Ser136, and by a specific cell-free kinase assay. In contrast, Huh7 and HepG2 did not show hyperactivation when tested by the same criteria. Akt enzyme hyperactivation in Mahlavu was associated with a loss of PTEN protein expression. Akt signaling was inhibited by the upstream kinase inhibitors, LY294002, Wortmannin, as well as by the specific Akt Inhibitor VIII in all three hepatoma cell lines. Cytotoxicity assays with Akt inhibitors in the same cell lines indicated that they were all sensitive, but with different IC50 values as assayed by RT-CES. We also demonstrated that the cytotoxic effect was through apoptotic cell death. Our findings provide evidence for its constitutive activation in one HCC cell line, and that HCC cell lines, independent of their Akt activation status respond to Akt inhibitors by apoptotic cell death. Thus, Akt inhibition may be considered as an attractive therapeutic intervention in liver cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Androstadienos/administración & dosificación , Androstadienos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Cromonas/administración & dosificación , Cromonas/farmacología , Humanos , Concentración 50 Inhibidora , Neoplasias Hepáticas/patología , Morfolinas/administración & dosificación , Morfolinas/farmacología , Quinoxalinas/administración & dosificación , Quinoxalinas/farmacología , Transducción de Señal/efectos de los fármacos , Wortmanina
14.
Exp Cell Res ; 316(1): 38-47, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19747912

RESUMEN

Vav1 plays an important role in the all-trans retinoic acid (ATRA)-induced completion of the differentiation program of acute promyelocytic leukemia (APL)-derived cells, in which it strengthens the drug effects and is involved in the regulation of maturation-related proteins, such as the CD11b surface antigen. In both myeloid and lymphoid cells, accumulating data attribute to the multidomain protein Vav1 a functional relevance in the control of gene expression, by direct interaction with chromatin remodeling and/or transcriptional proteins. The present study provides evidence that, in the APL-derived NB4 cell line, Vav1 and the transcription factor PU.1 cooperate in regulating the ATRA-induced CD11b expression. Both chromatin immunoprecipitation (ChIP) experiments and electrophoretic mobility shift assays (EMSA) indicate that Vav1 and PU.1 are recruited to CD11b promoter. Even if the two proteins may participate in diverse protein/DNA complexes, the amounts of complexes including PU.1 seem to be dependent on the interaction of this transcription factor with tyrosine-phosphorylated Vav1. The reported data suggest that the ATRA-induced increase of Vav1 expression and tyrosine phosphorylation may be involved in recruiting PU.1 to its consensus sequence on the CD11b promoter and, ultimately, in regulating CD11b expression during the late stages of neutrophil differentiation of APL-derived promyelocytes.


Asunto(s)
Antígeno CD11b/genética , Diferenciación Celular/fisiología , Células Precursoras de Granulocitos/metabolismo , Regiones Promotoras Genéticas/fisiología , Proteínas Proto-Oncogénicas c-vav/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Tretinoina/farmacología , Antígeno CD11b/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/metabolismo , Inmunoprecipitación de Cromatina , ADN/genética , ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Regulación Neoplásica de la Expresión Génica/fisiología , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia Promielocítica Aguda/patología , Neutrófilos/citología , Neutrófilos/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-vav/genética , ARN Interferente Pequeño/genética , Estilbenos/farmacología , Quinasa Syk , Transactivadores/genética
15.
Stem Cell Rev Rep ; 17(2): 673-684, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33165749

RESUMEN

All-trans retinoic acid (ATRA) promotes the development and the function of insulin producing cells and induces partial differentiation of pancreatic tumor cells. A number of evidences clearly indicate that the ATRA mediated signaling may have a substantial role in therapeutic approaches based on restoration of functional ß-cells. Among the proteins up-regulated by ATRA, Vav1 is involved in maturation and function of haematopoietic cells and is essential for retinoids induced differentiation of tumor promyelocytes. The presence of Vav1 in solid tissues, including pancreas, is considered ectopic and no role in the differentiation of human epithelial cells has so far been described. We demonstrated here that Vav1 sustains the maturation to ß-cells of the normal precursors human Biliary Tree Stem/progenitor Cells (hBTSCs) induced by a differentiation medium containing ATRA and that, in the mature normal pancreas, insulin-producing cells express variable levels of Vav1. Using pancreatic ductal adenocarcinoma (PDAC)-derived cells, we also revealed that the ATRA induced up-modulation of Vav1 is essential for the retinoid-induced trans-differentiation of neoplastic cells into insulin producing cells. The results of this study identify Vav1 as crucial molecule in ATRA induced maturation of insulin producing cells and suggest this protein as a marker for new strategies ended to restore functional ß-cells. Graphical abstract.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/citología , Proteínas Proto-Oncogénicas c-vav , Tretinoina , Humanos , Proteínas Proto-Oncogénicas c-vav/genética , Tretinoina/farmacología
16.
J Proteome Res ; 9(2): 752-60, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20028078

RESUMEN

Vav1, whose physiological expression is restricted to hematopoietic system, is one of the signaling proteins up-regulated by all-trans retinoic acid (ATRA) in acute promyelocytic leukemia (APL)-derived precursors, in which it promotes the overcoming of the differentiation blockade. High levels of tyrosine phosphorylated Vav1 accumulate in differentiating APL-derived cells, suggesting that one or more Vav1 tyrosine residues are involved in neutrophil differentiation of tumoral promyelocytes. Here, we have found that phosphorylation of Vav1 Y174, that is known to regulate Vav1 activity in mature neutrophils, is up-regulated by ATRA in NB4 cells. Nevertheless, this tyrosine residue does not seem crucial for the agonist-induced phenotypical differentiation of APL-derived cells. Mass spectrometry analysis performed on Vav1 from differentiating NB4 cells allowed to identify the highly conserved Y745 residue as a phosphorylated tyrosine that plays crucial roles in the completion of the maturation program of this cell line. In fact, the overexpression of a mutated form of Vav1, in which Y745 was replaced with a phenylalanine, significantly reduced the ATRA-induced CD11b expression and essentially abrogated the differentiation-related acquisition of the migratory capability. Even though the intracellular signaling involving Vav1 phosphorylated in Y745 is unknown, the identification of a tyrosine residue essential for differentiation of tumoral precursors may constitute the basis to identify new specific targets for differentiation therapy of APL.


Asunto(s)
Leucemia Promielocítica Aguda/metabolismo , Espectrometría de Masas/métodos , Proteínas Proto-Oncogénicas c-vav/metabolismo , Tirosina/metabolismo , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Cartilla de ADN , Humanos , Leucemia Promielocítica Aguda/patología , Fosforilación , Proteínas Proto-Oncogénicas c-vav/química , Tretinoina/farmacología
17.
Circ Res ; 100(1): 61-9, 2007 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17138942

RESUMEN

Nutlin-3, a nongenotoxic activator of the p53 pathway, dose-dependently (range 0.1 to 10 micromol/L) inhibited the formation of capillaries in an in vivo matrigel assay, as well as the formation of capillary-like structures in an in vitro coculture system composed of endothelial cells surrounded by fibroblasts. In contrast to the chemotherapeutic agent doxorubicin, nutlin-3 showed no induction of apoptosis in vitro either in the cocultures or in isolated vascular endothelial cells, even when used at the highest concentration (10 micromol/L). However, treatment with pharmacological inhibitors of the nuclear factor kappaB and phosphatidylinositol 3-kinase/Akt pathways sensitized endothelial cells to nutlin-3-induced apoptosis. Although nutlin-3 and doxorubicin induced a comparable p53 accumulation in endothelial cells, nutlin-3 was significantly more efficient than doxorubicin in upregulating the p53 target genes CDKN1A/p21, MDM2, and GDF-15, as well as in inhibiting cell cycle progression. However, the predominant in vitro effect of nutlin-3 was its strong antimigratory activity observed at concentrations significantly lower (0.1 micromol/L) than those required to inhibit endothelial cell cycle progression. Taken together, our data suggest that the antiangiogenic activity of nutlin-3 observed in vivo was mainly attributable to inhibition of endothelial cell migration, to some extent attributable to cell cycle arrest, and to a lesser extent attributable to induction of apoptosis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Imidazoles/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Capilares/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
18.
J Oncol ; 2019: 7512632, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31636668

RESUMEN

Initially correlated with hematopoietic precursors, the surface expression of CD133 was also found in epithelial and nonepithelial cells from adult tissues in which it has been associated with a number of biological events. CD133 is expressed in solid tumors as well, including breast cancer, in which most of the studies have been focused on its use as a surface marker for the detection of cells with stem-like properties (i.e., cancer stem cells (CSCs)). Differently with other solid tumors, very limited and in part controversial are the information about the significance of CD133 in breast cancer, the most common malignancy among women in industrialized countries. In this review, we summarize the latest findings about the implication of CD133 in breast tumors, highlighting its role in tumor cells with a triple negative phenotype in which it directly regulates the expression of proteins involved in metastasis and drug resistance. We provide updates about the prognostic role of CD133, underlining its value as an indicator of increased malignancy of both noninvasive and invasive breast tumor cells. The molecular mechanisms at the basis of the regulation of CD133 levels in breast tumors have also been reviewed, highlighting experimental strategies capable to restrain its level that could be taken into account to reduce malignancy and/or to prevent the progression of breast tumors.

19.
Cell Signal ; 19(8): 1701-12, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17478077

RESUMEN

During both maturation and function, neutrophils are subjected to reorganization of the actin cytoskeleton. Among the molecules that influence cytoskeletal architecture, the amount and subcellular localization of phosphoinositides, regulated by specific kinases and phosphatases, may play a crucial role. In neutrophils, PLC-beta2 is a major phosphoinositide-dependent phospholipase C isoform activated in response to chemoattractants, even though its role in the modifications of cell morphology and motility that occur during the inflammatory process has not been fully elucidated. In APL-derived promyelocytes induced to complete their maturation program, we have found that the expression levels of PLC-beta2 positively correlate with the degree of the reached neutrophil differentiation. Here, we demonstrate that PLC-beta2 modulates the migration capability of promyelocytes induced to differentiate with ATRA. In differentiating cells, the association of PLC-beta2 with actin, mediated by the PH domain, seems crucial for catalytic activity. We conclude that phosphodiesterase activity of PLC-beta2 on the actin-associated PIP2 may be responsible, by modifying the phosphoinositide pools, for the modifications of cytoskeleton architecture that take place during motility of differentiating promyelocytes.


Asunto(s)
Actinas/metabolismo , Movimiento Celular/fisiología , Células Precursoras de Granulocitos/fisiología , Isoenzimas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfolipasas de Tipo C/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Glutatión Transferasa/metabolismo , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/enzimología , Humanos , Inmunohistoquímica , Isoenzimas/genética , Fosfolipasa C beta , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Fosfolipasas de Tipo C/genética
20.
Oncotarget ; 9(17): 14005-14034, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29568412

RESUMEN

Cancer patients experience symptoms and adverse effects of treatments that may last even after the end of treatments. Exercise is a safe, non-pharmacological and cost-effective therapy that can provide several health benefits in cancer patient and survivors, reducing cancer symptoms and cancer treatment side effects. The purpose of this review is to describe how the physical exercise is capable to reduce cancer symptoms and cancer treatment side effects. We realized a pragmatic classification of symptoms, dividing them into physical, psychological and psycho-physical aspects. For each symptom we discuss causes, therapies, we analyse the effects of physical exercise and we summarize the most effective type of exercise to reduce the symptoms. This review also points out what are the difficulties that patients and survivors face during the practice of physical activity and provides some solutions to overcome these barriers. Related to each specific cancer, it emerges that type, frequency and intensity of physical exercise could be prescribed and supervised as a therapeutic program, like it occurs for the type, dose and duration of a drug treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA