Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Strahlenther Onkol ; 200(8): 691-697, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38416163

RESUMEN

PURPOSE: To evaluate the impact of sustained hypogonadism after androgen deprivation therapy (ADT) associated with radiotherapy in prostate cancer (PCa) patients with biochemical relapse-free survival (bRFS). METHODS: A retrospective cohort analysis of 213 consecutive PCa patients referred for radiotherapy plus ADT was carried out. Follow-up times including time to testosterone recovery (TTR) and bRFS were calculated from the end of ADT. Univariate and multivariate Cox regression analyses predicting bRFS were used. The optimal cutoffs for TTR and duration of ADT were determined using the maximally selected rank statistics (MSRS). RESULTS: After a median follow-up of 104 months, 18 patients relapsed among those who had recovered testosterone levels and 9 among those who did not. Median ADT duration was 36 months. The optimal cutoff for TTR was determined using MSRS. TTR >48 months was significantly associated with better bRFS (logrank, p < 0.0027). Five-year bRFS was 100% for >48 months vs. 85% for <48 months. TTR was the only significant variable for bRFS in multivariate Cox analysis. CONCLUSION: Our data show an association between longer TTR and bRFS values among PCa patients treated with ADT.


Asunto(s)
Antagonistas de Andrógenos , Neoplasias de la Próstata , Testosterona , Humanos , Masculino , Neoplasias de la Próstata/radioterapia , Neoplasias de la Próstata/tratamiento farmacológico , Antagonistas de Andrógenos/uso terapéutico , Anciano , Testosterona/sangre , Testosterona/uso terapéutico , Persona de Mediana Edad , Estudios Retrospectivos , Hipogonadismo/etiología , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Resultado del Tratamiento , Estudios de Cohortes , Terapia Combinada , Estudios de Seguimiento
2.
Prostate ; 83(11): 1068-1075, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37150842

RESUMEN

PURPOSE: To compare the clinical outcomes of single-fraction high-dose-rate (HDR) brachytherapy and single-fraction low-dose-rate (LDR) brachytherapy as the sole treatment for primary prostate cancer. MATERIAL AND METHODS: A quasi-randomized study that allocated, from March 2008 to February 2012, 129 low and intermediate risk prostate cancer patients to one single-fraction HDR of 19 Gy (61 patients) or to a 145 Gy 125 I LDR permanent implant (68 patients. Biochemical relapse-free survival (bRFS) and overall survival (OS) were compared using the Kaplan-Meier method and Cox regression analysis. RESULTS: After a median follow-up of 72 months in the HDR group, 26 patients relapsed, and after a median follow-up of 84 months in the LDR group, 7 patients relapsed (p < 0.0001). The 5-year bRFS was significantly better for the LDR group than for the HDR group (93.7% and 61.1%, respectively) (p < 0.0001). The 5-year OS also was significantly better in the LDR group (95.5% vs. 89.9%) (p = 0.0436). CONCLUSIONS: Permanent LDR prostate implant brachytherapy offers better clinical outcomes than single-fraction HDR for prostate cancer.


Asunto(s)
Braquiterapia , Neoplasias de la Próstata , Masculino , Humanos , Próstata/patología , Estudios Prospectivos , Braquiterapia/métodos , Dosificación Radioterapéutica , Recurrencia Local de Neoplasia/radioterapia
3.
J Biol Inorg Chem ; 28(6): 531-547, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37458856

RESUMEN

In the treatment of hormone-dependent cancers, aromatase inhibitors (AI) are receiving increased attention due to some undesirable effects such as the risk of endometrial cancer and thromboembolism of SERMs (selective estrogen receptor modulators). Letrozole is the most active AI with 99% aromatase inhibition. Unfortunately, this compound also exhibits some adverse effects such as hot flashes and fibromyalgias. Therefore, there is an urgent need to explore new types of AIs that retain the same-or even increased-antitumor ability. Inspired by the letrozole structure, a set of new derivatives has been synthesized that include a ferrocenyl moiety and different heterocycles. The derivative that contains a benzimidazole ring, namely compound 6, exhibits a higher aromatase inhibitory activity than letrozole and it also shows potent cytostatic behavior when compared to other well-established aromatase inhibitors, as demonstrated by dose-response, cell cycle, apoptosis and time course experiments. Furthermore, 6 promotes the inhibition of cell growth in both an aromatase-dependent and -independent fashion, as indicated by the study of A549 and MCF7 cell lines. Molecular docking and molecular dynamics calculations on the interaction of 6 or letrozole with the aromatase binding site revealed that the ferrocene moiety increases the van der Waals and hydrophobic interactions, thus resulting in an increase in binding affinity. Furthermore, the iron atom of the ferrocene fragment can form a metal-acceptor interaction with a propionate fragment, and this results in a stronger coupling with the heme group-a possibility that is consistent with the strong aromatase inhibition of 6.


Asunto(s)
Neoplasias de la Mama , Citostáticos , Humanos , Femenino , Letrozol/farmacología , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/química , Aromatasa/metabolismo , Metalocenos , Simulación del Acoplamiento Molecular , Nitrilos/farmacología , Triazoles/farmacología , Células MCF-7
4.
Ann Rheum Dis ; 2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35788494

RESUMEN

OBJECTIVES: S100A9, an alarmin that can form calprotectin (CP) heterodimers with S100A8, is mainly produced by keratinocytes and innate immune cells. The contribution of keratinocyte-derived S100A9 to psoriasis (Ps) and psoriatic arthritis (PsA) was evaluated using mouse models, and the potential usefulness of S100A9 as a Ps/PsA biomarker was assessed in patient samples. METHODS: Conditional S100A9 mice were crossed with DKO* mice, an established psoriasis-like mouse model based on inducible epidermal deletion of c-Jun and JunB to achieve additional epidermal deletion of S100A9 (TKO* mice). Psoriatic skin and joint disease were evaluated in DKO* and TKO* by histology, microCT, RNA and proteomic analyses. Furthermore, S100A9 expression was analysed in skin, serum and synovial fluid samples of patients with Ps and PsA. RESULTS: Compared with DKO* littermates, TKO* mice displayed enhanced skin disease severity, PsA incidence and neutrophil infiltration. Altered epidermal expression of selective pro-inflammatory genes and pathways, increased epidermal phosphorylation of STAT3 and higher circulating TNFα were observed in TKO* mice. In humans, synovial S100A9 levels were higher than the respective serum levels. Importantly, patients with PsA had significantly higher serum concentrations of S100A9, CP, VEGF, IL-6 and TNFα compared with patients with only Ps, but only S100A9 and CP could efficiently discriminate healthy individuals, patients with Ps and patients with PsA. CONCLUSIONS: Keratinocyte-derived S100A9 plays a regulatory role in psoriatic skin and joint disease. In humans, S100A9/CP is a promising marker that could help in identifying patients with Ps at risk of developing PsA.

5.
J Cell Mol Med ; 25(22): 10591-10603, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34655447

RESUMEN

Sorafenib is a multikinase inhibitor widely used in cancer therapy with an antitumour effect related to biological processes as proliferation, migration or invasion, among others. Initially designed as a Raf inhibitor, Sorafenib was later shown to also block key molecules in tumour progression such as VEGFR and PDGFR. In addition, sorafenib has been connected with key signalling pathways in cancer such as EGFR/EGF. However, no definitive clue about the molecular mechanism linking sorafenib and EGF signalling pathway has been established so far. Our data in HeLa, U2OS, A549 and HEK293T cells, based on in silico, chemical and genetic approaches demonstrate that the MEK5/ERK5 signalling pathway is a novel target of sorafenib. In addition, our data show how sorafenib is able to block MEK5-dependent phosphorylation of ERK5 in the Ser218/Tyr220, affecting the transcriptional activation associated with ERK5. Moreover, we demonstrate that some of the effects of this kinase inhibitor onto EGF biological responses, such as progression through cell cycle or migration, are mediated through the effect exerted onto ERK5 signalling pathway. Therefore, our observations describe a novel target of sorafenib, the ERK5 signalling pathway, and establish new mechanistic insights for the antitumour effect of this multikinase inhibitor.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Sorafenib/farmacología , Biomarcadores de Tumor , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular , Susceptibilidad a Enfermedades , Factor de Crecimiento Epidérmico/metabolismo , Citometría de Flujo , Humanos , Proteína Quinasa 7 Activada por Mitógenos/química , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/química , Transducción de Señal/efectos de los fármacos , Sorafenib/química , Relación Estructura-Actividad
6.
Mol Cell ; 48(5): 681-91, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23103253

RESUMEN

The mammalian target of rapamycin (mTOR) pathway, which is essential for cell proliferation, is repressed in certain cell types in hypoxia. However, hypoxia-inducible factor 2α (HIF2α) can act as a proliferation-promoting factor in some biological settings. This paradoxical situation led us to study whether HIF2α has a specific effect on mTORC1 regulation. Here we show that activation of the HIF2α pathway increases mTORC1 activity by upregulating expression of the amino acid carrier SLC7A5. At the molecular level we also show that HIF2α binds to the Slc7a5 proximal promoter. Our findings identify a link between the oxygen-sensing HIF2α pathway and mTORC1 regulation, revealing the molecular basis of the tumor-promoting properties of HIF2α in von Hippel-Lindau-deficient cells. We also describe relevant physiological scenarios, including those that occur in liver and lung tissue, wherein HIF2α or low-oxygen tension drive mTORC1 activity and SLC7A5 expression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Proteínas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sitios de Unión , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Transportador de Aminoácidos Neutros Grandes 1/genética , Hígado/metabolismo , Pulmón/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Ratones SCID , Complejos Multiproteicos , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Proteínas/genética , Interferencia de ARN , Transducción de Señal , Serina-Treonina Quinasas TOR , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
7.
Int J Mol Sci ; 21(20)2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053909

RESUMEN

The p38 mitogen-activated protein kinase (MAPK) signaling pathway is implicated in cancer biology and has been widely studied over the past two decades as a potential therapeutic target. Most of the biological and pathological implications of p38MAPK signaling are often associated with p38α (MAPK14). Recently, several members of the p38 family, including p38γ and p38δ, have been shown to play a crucial role in several pathologies including cancer. However, the specific role of p38ß (MAPK11) in cancer is still elusive, and further investigation is needed. Here, we summarize what is currently known about the role of p38ß in different types of tumors and its putative implication in cancer therapy. All evidence suggests that p38ß might be a key player in cancer development, and could be an important therapeutic target in several pathologies, including cancer.


Asunto(s)
Susceptibilidad a Enfermedades , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína Quinasa 11 Activada por Mitógenos/genética , Familia de Multigenes , Neoplasias/patología , Transducción de Señal
8.
J Cell Mol Med ; 21(2): 244-253, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27641066

RESUMEN

Glioblastoma is the most lethal brain tumour with a poor prognosis. Cancer stem cells (CSC) were proposed to be the most aggressive cells allowing brain tumour recurrence and aggressiveness. Current challenge is to determine CSC signature to characterize these cells and to develop new therapeutics. In a previous work, we achieved a screening of glycosylation-related genes to characterize specific genes involved in CSC maintenance. Three genes named CHI3L1, KLRC3 and PRUNE2 were found overexpressed in glioblastoma undifferentiated cells (related to CSC) compared to the differentiated ones. The comparison of their roles suggest that KLRC3 gene coding for NKG2E, a protein initially identified in NK cells, is more important than both two other genes in glioblastomas aggressiveness. Indeed, KLRC3 silencing decreased self-renewal capacity, invasion, proliferation, radioresistance and tumourigenicity of U87-MG glioblastoma cell line. For the first time we report that KLRC3 gene expression is linked to glioblastoma aggressiveness and could be a new potential therapeutic target to attenuate glioblastoma.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Carcinogénesis/genética , Carcinogénesis/patología , Glioblastoma/genética , Glioblastoma/patología , Subfamília C de Receptores Similares a Lectina de Células NK/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Células Clonales , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Invasividad Neoplásica , ARN Interferente Pequeño/metabolismo , Tolerancia a Radiación , Transducción de Señal/genética
9.
Antimicrob Agents Chemother ; 60(6): 3540-50, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27021315

RESUMEN

Hepatitis C virus (HCV) interacts with cellular components and modulates their activities for its own benefit. These interactions have been postulated as a target for antiviral treatment, and some candidate molecules are currently in clinical trials. The multifunctional cellular kinase Akt/protein kinase B (PKB) must be activated to increase the efficacy of HCV entry but is rapidly inactivated as the viral replication cycle progresses. Viral components have been postulated to be responsible for Akt/PKB inactivation, but the underlying mechanism remained elusive. In this study, we show that HCV polymerase NS5B interacts with Akt/PKB. In the presence of transiently expressed NS5B or in replicon- or virus-infected cells, NS5B changes the cellular localization of Akt/PKB from the cytoplasm to the perinuclear region. Sequestration of Akt/PKB by NS5B could explain its exclusion from its participation in early Akt/PKB inactivation. The NS5B-Akt/PKB interaction represents a new regulatory step in the HCV infection cycle, opening possibilities for new therapeutic options.


Asunto(s)
Hepacivirus/enzimología , Hepatitis C/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Línea Celular Tumoral , Hepatitis C/virología , Humanos , Fosforilación , Replicación Viral/fisiología
10.
Eur J Immunol ; 45(9): 2615-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26115479

RESUMEN

Delta-like protein 1 (DLK1) is a noncanonical ligand that inhibits NOTCH1 receptor activity and regulates multiple differentiation processes. In macrophages, NOTCH signaling increases TLR-induced expression of key pro-inflammatory mediators. We have investigated the role of DLK1 in macrophage activation and inflammation using Dlk1-deficient mice and Raw 264.7 cells overexpressing Dlk1. In the absence of Dlk1, NOTCH1 expression is increased and the activation of macrophages with TLR3 or TLR4 agonists leads to higher production of IFN-ß and other pro-inflammatory cytokines, including TNF-α, IL-12, and IL-23. The expression of key proteins involved in IFN-ß signaling, such as IRF3, IRF7, IRF1, or STAT1, as well as cRel, or RelB, which are responsible for the generation of IL-12 and IL-23, is enhanced in Dlk1 KO macrophages. Consistently, Dlk1 KO mice are more sensitive to LPS-induced endotoxic shock. These effects seem to be mediated through the modulation of NOTCH1 signaling. TLR4 activation reduces DLK1 expression, whereas increases NOTCH1 levels. In addition, DLK1 expression diminishes during differentiation of human U937 cells to macrophages. Overall, these results reveal a novel role for DLK1 as a regulator of NOTCH-mediated, pro-inflammatory macrophage activation, which could help to ensure a baseline level preventing constant tissue inflammation.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/inmunología , Macrófagos/inmunología , Receptor Notch1/inmunología , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 4/inmunología , Animales , Proteínas de Unión al Calcio , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Activación de Macrófagos , Macrófagos/citología , Ratones , Ratones Noqueados , Receptor Notch1/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 4/genética , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Células U937
11.
Cancers (Basel) ; 15(3)2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36765819

RESUMEN

Over the last 30 years, the study of the cellular response to ionizing radiation (IR) has increased exponentially. Among the various signaling pathways affected by IR, p38 MAPK has been shown to be activated both in vitro and in vivo, with involvement in key processes triggered by IR-mediated genotoxic insult, such as the cell cycle, apoptosis or senescence. However, we do not yet have a definitive clue about the role of p38 MAPK in terms of radioresistance/sensitivity and its potential use to improve current radiotherapy. In this review, we summarize the current knowledge on this family of MAPKs in response to IR as well as in different aspects related to radiotherapy, such as their role in the control of REDOX, fibrosis, and in the radiosensitizing effect of several compounds.

12.
Exp Mol Med ; 55(6): 1247-1257, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37332046

RESUMEN

Sarcomas constitute a heterogeneous group of rare and difficult-to-treat tumors that can affect people of all ages, representing one of the most common forms of cancer in childhood and adolescence. Little is known about the molecular entities involved in sarcomagenesis. Therefore, the identification of processes that lead to the development of the disease may uncover novel therapeutic opportunities. Here, we show that the MEK5/ERK5 signaling pathway plays a critical role in the pathogenesis of sarcomas. By developing a mouse model engineered to express a constitutively active form of MEK5, we demonstrate that the exclusive activation of the MEK5/ERK5 pathway can promote sarcomagenesis. Histopathological analyses identified these tumors as undifferentiated pleomorphic sarcomas. Bioinformatic studies revealed that sarcomas are the tumors in which ERK5 is most frequently amplified and overexpressed. Moreover, analysis of the impact of ERK5 protein expression on overall survival in patients diagnosed with different sarcoma types in our local hospital showed a 5-fold decrease in median survival in patients with elevated ERK5 expression compared with those with low expression. Pharmacological and genetic studies revealed that targeting the MEK5/ERK5 pathway drastically affects the proliferation of human sarcoma cells and tumor growth. Interestingly, sarcoma cells with knockout of ERK5 or MEK5 were unable to form tumors when engrafted into mice. Taken together, our results reveal a role of the MEK5/ERK5 pathway in sarcomagenesis and open a new scenario to be considered in the treatment of patients with sarcoma in which the ERK5 pathway is pathophysiologically involved.


Asunto(s)
MAP Quinasa Quinasa 5 , Sarcoma , Animales , Humanos , Ratones , MAP Quinasa Quinasa 5/genética , MAP Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas , Pronóstico , Sarcoma/genética
13.
Glia ; 60(7): 1130-44, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22511272

RESUMEN

We previously reported that addition of extracellular matrix (ECM) extracts to rat Schwann cell-dorsal root ganglion neuron (DRGN) co-cultures activated mitogen-activated protein kinase (MAPK) p38, whereas inhibition blocked myelination. Here, we used p38 pharmacological inhibitors and gene silencing to assess their effects on downstream kinases and key transcription factors. We show that p38α regulates expression of the master transcription factor, Krox-20, required for the onset of myelination in Schwann cell-DRGNs, as assessed by immunocytochemistry and qRT-PCR. p38 activity is also required for the expression of the cell cycle inhibitor p27(kip1) , associated with Schwann cell differentiation. Three potential effectors of p38 were explored: MAPK-activated protein kinase-2 (MK2), mitogen and stress-activated protein kinase-1 (MSK-1), and the transcription factor cAMP response element-binding protein (CREB). Inhibition of MK2 with CMPD1 or gene knockdown with siRNAs reduced numbers of Krox-20-positive Schwann cells and expression of myelin proteins MBP and MAG. ECM activated CREB and increased Krox-20 expression, whereas CREB1 gene silencing reduced Krox-20. Furthermore, two nonselective inhibitors of MSK-1 (H89 and R0-318820) decreased ECM-induced CREB phosphorylation and, similar to anti-MSK-1 siRNAs, reduced Krox-20-positive cells. In addition, p38 modulated the expression of two transcription factors involved in the regulation of Krox-20 [suppressed cAMP-inducible protein (SCIP) and Sox10], but not Sox2, an antagonist of Krox-20. Collectively, our results show that p38 primarily directs Schwann cell differentiation and peripheral myelination by regulating Krox-20 expression through its downstream effectors, MK2 and MSK-1/CREB, and transcription factors SCIP and Sox10.


Asunto(s)
Diferenciación Celular/fisiología , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Vaina de Mielina/metabolismo , Células de Schwann/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Técnicas de Cocultivo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Vaina de Mielina/genética , Neuronas/metabolismo , Factor 6 de Transcripción de Unión a Octámeros/genética , Factor 6 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
Scand J Urol Nephrol ; 46(5): 358-64, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22571179

RESUMEN

OBJECTIVE: This study aimed to evaluate the usefulness of carbonic anhydrase IX (CA-IX) expression in clear cell renal cell carcinoma (CCRCC) using two different techniques to detect protein expression. MATERIAL AND METHODS: An experimental, cross-sectional, analytical study was conducted to analyse proteins in renal tumour and healthy tissue specimens from 38 consecutive patients who underwent nephrectomy for renal cancer. CA-IX protein expression was measured by immunohistochemistry and Western blot analysis and quantified. Statistical analysis was performed with the positive and negative specific agreements and kappa coefficient. The sensitivity and specificity of both techniques were assessed. Statistical tests were conducted to analyse the association between CA-IX expression quantitation and normal prognosis factors (TNM stage and Fuhrman nuclear grade), only in CCRCC. RESULTS: The mean patient age was 65 years, 78.9% of patients were men and 57.9% of tumours were CCRCC. CA-IX protein expression was positive in 63.2% of tumours by immunohistochemistry and in 60.5% by Western blot. Both techniques detected CA-IX expression only in CCRCC and unclassifiable tumours. High concordance indices were observed for CCRCC diagnosis. Western blot and immunohistochemistry had a sensitivity of 95.5% and 100%, respectively; the specificity was 100% in both techniques. CA-IX expression quantitation did not correlate with tumour stage or Fuhrman nuclear grade. CONCLUSIONS: Immunochemistry and Western blot techniques can be used to detect abnormal CA-IX protein expression in CCRCC and to support morphology-based diagnostic techniques.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Anhidrasas Carbónicas/metabolismo , Carcinoma de Células Renales/diagnóstico , Neoplasias Renales/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Anhidrasa Carbónica IX , Carcinoma de Células Renales/enzimología , Estudios de Cohortes , Estudios Transversales , Femenino , Humanos , Inmunohistoquímica , Neoplasias Renales/enzimología , Masculino , Persona de Mediana Edad , Nefrectomía , Pronóstico , Sensibilidad y Especificidad
15.
Cancers (Basel) ; 14(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35884568

RESUMEN

Sarcomas are a heterogeneous group of tumors in which the role of ERK5 is poorly studied. To clarify the role of this MAPK in sarcomatous pathology, we used a murine 3-methyl-cholanthrene (3MC)-induced sarcoma model. Our data show that 3MC induces pleomorphic sarcomas with muscle differentiation, showing an increased expression of ERK5. Indeed, this upregulation was also observed in human sarcomas of muscular origin, such as leiomyosarcoma or rhabdomyosarcoma. Moreover, in cell lines derived from these 3MC-induced tumors, abrogation of Mapk7 expression by using specific shRNAs decreased in vitro growth and colony-forming capacity and led to a marked loss of tumor growth in vivo. In fact, transcriptomic profiling in ERK5 abrogated cell lines by RNAseq showed a deregulated gene expression pattern for key biological processes such as angiogenesis, migration, motility, etc., correlating with a better prognostic in human pathology. Finally, among the various differentially expressed genes, Klf2 is a key mediator of the biological effects of ERK5 as indicated by its specific interference, demonstrating that the ERK5-KLF2 axis is an important determinant of sarcoma biology that should be further studied in human pathology.

16.
J Inorg Biochem ; 218: 111403, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33730639

RESUMEN

The problems of resistance and side effects associated with cisplatin and other chemotherapeutic drugs have boosted research aimed at finding new compounds with improved properties. The use of platinum(IV) prodrugs is one alternative, although there is some controversy regarding the predictive ability of the peak reduction potentials. In the work described here a series of fourteen chloride Pt(II) and Pt(IV) compounds was synthesised and fully characterised. The compounds contain different bidentate arylazole heterocyclic ligands. Their cytotoxic properties against human lung carcinoma (A549), human breast carcinoma (MCF7) and human colon carcinoma (HCT116 and HT29) cell lines were studied. A clear relationship between the type of ligand and the anti-proliferative properties was found, with the best results obtained for the Pt(II) compound that contains an aniline fragment, (13), thus evidencing a positive effect of the NH2 group. Stability and aquation studies in DMSO, DMF and DMSO/water mixtures were carried out on the active complexes and an in-depth analysis of the two aquation processes, including DFT analysis, of 13 was undertaken. It was verified that DNA was the target and that cell death occurred by apoptosis in the case of 13. Furthermore, the cytotoxic derivatives did not exhibit haemolytic activity. The reduction of the Pt(IV) compounds whose Pt(II) congeners were active was studied by several techniques. It was concluded that the peak reduction potential was not useful to predict the ability for reduction. However, a correlation between the cytotoxic activity and the standard reduction potential was found.


Asunto(s)
Compuestos de Anilina/química , Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Compuestos Organoplatinos/farmacología , Platino (Metal)/química , Profármacos/farmacología , Células A549 , Antineoplásicos/química , Apoptosis , Proliferación Celular , Células HCT116 , Humanos , Células MCF-7 , Neoplasias/patología , Compuestos Organoplatinos/química , Profármacos/química
17.
Cell Rep ; 26(9): 2257-2265.e4, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30811976

RESUMEN

Cellular aspartate drives cancer cell proliferation, but signaling pathways that rewire aspartate biosynthesis to control cell growth remain largely unknown. Hypoxia-inducible factor-1α (HIF1α) can suppress tumor cell proliferation. Here, we discovered that HIF1α acts as a direct repressor of aspartate biosynthesis involving the suppression of several key aspartate-producing proteins, including cytosolic glutamic-oxaloacetic transaminase-1 (GOT1) and mitochondrial GOT2. Accordingly, HIF1α suppresses aspartate production from both glutamine oxidation as well as the glutamine reductive pathway. Strikingly, the addition of aspartate to the culture medium is sufficient to relieve HIF1α-dependent repression of tumor cell proliferation. Furthermore, these key aspartate-producing players are specifically repressed in VHL-deficient human renal carcinomas, a paradigmatic tumor type in which HIF1α acts as a tumor suppressor, highlighting the in vivo relevance of these findings. In conclusion, we show that HIF1α inhibits cytosolic and mitochondrial aspartate biosynthesis and that this mechanism is the molecular basis for HIF1α tumor suppressor activity.


Asunto(s)
Ácido Aspártico/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias/metabolismo , Proteínas Supresoras de Tumor/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Aspartato Aminotransferasa Citoplasmática/metabolismo , Aspartato Aminotransferasa Mitocondrial/metabolismo , Ácido Aspártico/farmacología , Carcinoma de Células Renales/enzimología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glutamina/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/enzimología , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/antagonistas & inhibidores , Neoplasias/patología , Oxidación-Reducción , Proteínas Supresoras de Tumor/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
18.
Int J Cancer ; 122(2): 289-97, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17893873

RESUMEN

Activation of p38 MAPK is a critical requisite for the therapeutics activity of the antitumor agent cisplatin. In this sense, a growing body of evidences supports the role of c-Abl as a major determinant of p38 MAPK activation, especially in response to genotoxic stress when triggered by cisplatin. Here, we demonstrate that p38 MAPK activation in response to cisplatin does not require the tyrosine kinase activity of c-Abl. Indeed, c-Abl can activate the p38 MAPK signaling pathway by a mechanism that is independent of its tyrosine kinase activity, but that instead involves the ability of c-Abl to increase the stability of MKK6. Similar results were obtained in chronic myeloid leukemia-derived cell lines, in which a chimeric Bcr/Abl protein mimics the effects of c-Abl overexpression on p38 MAPK activation. These findings may explain why a clinically used c-Abl inhibitor, imatinib mesylate, fails to inhibit the p38 MAPK pathway alone or in combination with cisplatin, and provide evidence of a novel signaling mechanism in which these antitumor agents act.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Benzamidas , Línea Celular Tumoral , Cicloheximida/farmacología , Regulación Leucémica de la Expresión Génica , Humanos , Mesilato de Imatinib , MAP Quinasa Quinasa 6/metabolismo , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Piperazinas/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Pirimidinas/farmacología
19.
Front Oncol ; 8: 214, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29938199

RESUMEN

The most common type of the renal cancers detected in humans is clear cell renal cell carcinomas (ccRCCs). These tumors are usually initiated by biallelic gene inactivation of the Von Hippel-Lindau (VHL) factor in the renal epithelium, which deregulates the hypoxia-inducible factors (HIFs) HIF1α and HIF2α, and provokes their constitutive activation irrespective of the cellular oxygen availability. While HIF1α can act as a ccRCC tumor suppressor, HIF2α has emerged as the key HIF isoform that is essential for ccRCC tumor progression. Indeed, preclinical and clinical data have shown that pharmacological inhibitors of HIF2α can efficiently combat ccRCC growth. In this review, we discuss the molecular basis underlying the oncogenic potential of HIF2α in ccRCC by focusing on those pathways primarily controlled by HIF2α that are thought to influence the progression of these tumors.

20.
PLoS One ; 13(7): e0200878, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30048489

RESUMEN

OBJECTIVES: To fully clarify the role of Mitogen Activated Protein Kinase in the therapeutic response to Sorafenib in Renal Cell Carcinoma as well as the cell death mechanism associated to this kinase inhibitor, we have evaluated the implication of several Mitogen Activated Protein Kinases in Renal Cell Carcinoma-derived cell lines. MATERIALS AND METHODS: An experimental model of Renal Cell Carcinoma-derived cell lines (ACHN and 786-O cells) was evaluated in terms of viability by MTT assay, induction of apoptosis by caspase 3/7 activity, autophagy induction by LC3 lipidation, and p62 degradation and kinase activity using phospho-targeted antibodies. Knock down of ATG5 and ERK5 was performed using lentiviral vector coding specific shRNA. RESULTS: Our data discard Extracellular Regulated Kinase 1/2 and 5 as well as p38 Mitogen Activated Protein Kinase pathways as mediators of Sorafenib toxic effect but instead indicate that the inhibitory effect is exerted through the PI3K/Akt signalling pathway. Furthermore, we demonstrate that inhibition of Akt mediates cell death associated to Sorafenib without caspase activation, and this is consistent with the induction of autophagy, as indicated by the use of pharmacological and genetic approaches. CONCLUSION: The present report demonstrates that Sorafenib exerts its toxic effect through the induction of autophagy in an Akt-dependent fashion without the implication of Mitogen Activated Protein Kinase. Therefore, our data discard the use of inhibitors of the RAF-MEK-ERK1/2 signalling pathway in RCC and support the use of pro-autophagic compounds, opening new therapeutic opportunities for Renal Cell Carcinoma.


Asunto(s)
Autofagia/efectos de los fármacos , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sorafenib/farmacología , Western Blotting , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA