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.
Artículo en Inglés | MEDLINE | ID: mdl-37770147

RESUMEN

Telomere repeat-containing RNAs (TERRA) are transcribed from telomeres as long non-coding RNAs and are part of the telomere structure with protective function. The genetic stability of cells requires telomeric repeats at the ends of chromosomes. Maintenance of telomere length (TL) is essential for proliferative capacity and chromosomal integrity. In contrast, telomere shortening is a recognized risk factor for carcinogenesis and a biomarker of aging due to the cumulative effects of environmental exposures and life experiences such as trauma or stress. In this context, telomere repeats are lost due to cell proliferation, but are also susceptible to stress factors including reactive oxygen species (ROS) inducing oxidative base damage. Quantitative PCR (qPCR) of genomic DNA is an established method to analyze TL as a tool to detect genotoxic events. That same qPCR method can be applied to RNA converted into cDNA to quantify TERRA as a useful tool to perform high-throughput screenings. This short review summarizes relevant qPCR studies using both TL and TERRA quantification, provides an overall view of the molecular mechanisms of telomere protection against ROS by TERRA, and summarizes the presented studies comparing the results at DNA and RNA levels, which indicate that fluctuations at transcript level might reflect a short-term response. Therefore, we conclude that performing both of these measurements together will improve genotoxicity studies.


Asunto(s)
ARN Largo no Codificante , Telómero , Especies Reactivas de Oxígeno , Telómero/genética , ARN Largo no Codificante/genética , ADN , Daño del ADN , Medición de Riesgo
2.
Int J Mol Sci ; 24(12)2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37373232

RESUMEN

Higher organisms, such as humans, are made up of trillions of cells that have to act as a unit in a finely tuned way to ensure the functioning of the living being that is composed of them [...].


Asunto(s)
Transducción de Señal , Humanos , Transducción de Señal/fisiología
3.
Neurobiol Dis ; 175: 105927, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36379394

RESUMEN

BACKGROUND: C9orf72 repeat expansions have been observed in a wide variety of neurodegenerative disorders. The cut-off between normal and pathogenic alleles is not well established as repeat sizing methods are often semi-quantitative. However, intermediate alleles might influence disease prevalence and phenotype, as seen for other repeat expansion disorders. We aimed to further delineate the prevalence of small, intermediate and expanded C9orf72 alleles and elucidate their potential influence on the disease phenotype. METHODS: DNA derived from patients (n = 1804) and healthy individuals (n = 643) was obtained from multiple collectives in Austria. Genotyping was performed using a two-step PCR assay followed by Southern blotting. RESULTS: 3.4% of clinically diagnosed frontotemporal dementia (FTD; n = 5/147) cases and 0.8% of clinically diagnosed Alzheimer's disease (AD; n = 5/602) cases were carriers of a pathological C9orf72 repeat expansion. A significantly earlier disease onset was detected in expansion carriers compared to non-carriers in the FTD and AD cohorts (median 50 years, range 39-64 vs. median 64 years, range 36-92, p = 0.018 and median 63 years, range 54-71 vs. median 74 years, range 45-92, p = 0.006, respectively). C9orf72 intermediate alleles were significantly associated with cerebellar symptoms (p = 0.0004) and sensory deficits in the dementia cohort (p = 0.01). CONCLUSIONS: C9orf72 repeat expansion carriers showed earlier disease onset compared to non-carriers with clinical diagnosis of FTD and AD. Furthermore, C9orf72 intermediate repeats might modify the phenotypic expression in dementia.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Humanos , Expansión de las Repeticiones de ADN/genética , Proteína C9orf72/genética , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Proteínas/genética , Fenotipo , Esclerosis Amiotrófica Lateral/genética
4.
Cancers (Basel) ; 14(19)2022 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-36230526

RESUMEN

Exosomes are membrane-structured extracellular vesicles (EVs) with nano-scale size that are released from different cell types [...].

5.
Int J Mol Sci ; 22(21)2021 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-34769378

RESUMEN

Sprouty proteins are widely accepted modulators of receptor tyrosine kinase-associated pathways and fulfill diversified roles in cancerogenesis dependent on the originating cells. In this study we detected a high expression of Sprouty3 in osteosarcoma-derived cells and addressed the question of whether Sprouty3 and Sprouty1 influence the malignant phenotype of this bone tumor entity. By using adenoviruses, the Sprouty proteins were expressed in two different cell lines and their influence on cellular behavior was assessed. Growth curve analyses and Scratch assays revealed that Sprouty3 accelerates cell proliferation and migration. Additionally, more colonies were grown in Soft agar if the cells express Sprouty3. In parallel, Sprouty1 had no significant effect on the measured endpoints of the study in osteosarcoma-derived cells. The promotion of the tumorigenic capacities in the presence of Sprouty3 coincided with an increased activation of signaling as measured by evaluating the phosphorylation of extracellular signal-regulated kinases (ERKs). Ectopic expression of a mutated Sprouty3 protein, in which the tyrosine necessary for its activation was substituted, resulted in inhibited migration of the treated cells. Our findings identify Sprouty3 as a candidate for a tumor promoter in osteosarcoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Osteosarcoma/patología , Fosfoproteínas/metabolismo , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Movimiento Celular , Proliferación Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Fosfoproteínas/genética , Células Tumorales Cultivadas
6.
J Mol Med (Berl) ; 99(8): 1151-1171, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34018016

RESUMEN

Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. We recently identified LIM domain only 3 (LMO3) in human mature visceral adipocytes; however, its function in these cells is currently unknown. The aim of this study was to determine the potential involvement of LMO3-dependent pathways in the modulation of key functions of mature adipocytes during obesity. Based on a recently engineered hybrid rAAV serotype Rec2 shown to efficiently transduce both brown adipose tissue (BAT) and white adipose tissue (WAT), we delivered YFP or Lmo3 to epididymal WAT (eWAT) of C57Bl6/J mice on a high-fat diet (HFD). The effects of eWAT transduction on metabolic parameters were evaluated 10 weeks later. To further define the role of LMO3 in insulin-stimulated glucose uptake, insulin signaling, adipocyte bioenergetics, as well as endocrine function, experiments were conducted in 3T3-L1 adipocytes and newly differentiated human primary mature adipocytes, engineered for transient gain or loss of LMO3 expression, respectively. AAV transduction of eWAT results in strong and stable Lmo3 expression specifically in the adipocyte fraction over a course of 10 weeks with HFD feeding. LMO3 expression in eWAT significantly improved insulin sensitivity and healthy visceral adipose tissue expansion in diet-induced obesity, paralleled by increased serum adiponectin. In vitro, LMO3 expression in 3T3-L1 adipocytes increased PPARγ transcriptional activity, insulin-stimulated GLUT4 translocation and glucose uptake, as well as mitochondrial oxidative capacity in addition to fatty acid oxidation. Mechanistically, LMO3 induced the PPARγ coregulator Ncoa1, which was required for LMO3 to enhance glucose uptake and mitochondrial oxidative gene expression. In human mature adipocytes, LMO3 overexpression promoted, while silencing of LMO3 suppressed mitochondrial oxidative capacity. LMO3 expression in visceral adipose tissue regulates multiple genes that preserve adipose tissue functionality during obesity, such as glucose metabolism, insulin sensitivity, mitochondrial function, and adiponectin secretion. Together with increased PPARγ activity and Ncoa1 expression, these gene expression changes promote insulin-induced GLUT4 translocation, glucose uptake in addition to increased mitochondrial oxidative capacity, limiting HFD-induced adipose dysfunction. These data add LMO3 as a novel regulator improving visceral adipose tissue function during obesity. KEY MESSAGES: LMO3 increases beneficial visceral adipose tissue expansion and insulin sensitivity in vivo. LMO3 increases glucose uptake and oxidative mitochondrial activity in adipocytes. LMO3 increases nuclear coactivator 1 (Ncoa1). LMO3-enhanced glucose uptake and mitochondrial gene expression requires Ncoa1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Adipocitos/metabolismo , Metabolismo Energético , Grasa Intraabdominal/metabolismo , Proteínas con Dominio LIM/genética , Obesidad/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Insulina/metabolismo , Grasa Intraabdominal/citología , Proteínas con Dominio LIM/metabolismo , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Biológicos , Obesidad/etiología , Oxidación-Reducción , Fosforilación Oxidativa , PPAR gamma/metabolismo , Unión Proteica
7.
Int J Mol Sci ; 22(4)2021 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-33670044

RESUMEN

Kallmann syndrome is the result of innate genetic defects in the fibroblast growth factor (FGF) regulated signaling network causing diminished signal transduction. One of the rare mutations associated with the syndrome alters the Sprouty (Spry)4 protein by converting the serine at position 241 into a tyrosine. In this study, we characterize the tyrosine Spry4 mutant protein in the primary human embryonic lung fibroblasts WI-38 and osteosarcoma-derived cell line U2OS. As demonstrated in a cell signaling assay, Spry4 gains the capability of inhibiting FGF, but not epithelial growth factor (EGF)-induced signaling as a consequence of the tyrosine substitution. Additionally, migration of normal embryonic lung fibroblasts and osteosarcoma-derived cells is potently inhibited by the tyrosine Spry4 variant, while an effect of the wildtype Spry4 protein is hardly measureable. Concerning cell proliferation, the unaltered Spry4 protein is ineffective to influence the WI-38 cells, while the mutated Spry4 protein decelerates the cell doubling. In summary, these data emphasize that like the other mutations associated with Kallmann syndrome the described Spry4 mutation creates a hyperactive version of a selective inhibitory molecule and can thereby contribute to a weakened FGF signaling. Additionally, the study pinpoints a Spry4 variation expanding the applicability of Spry4 in a potential cancer therapy.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Síndrome de Kallmann/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Pulmón/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Osteosarcoma/patología , Fosforilación/efectos de los fármacos , Transducción de Señal , Tirosina/metabolismo
8.
Cells ; 8(8)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31374860

RESUMEN

Dysregulation of receptor tyrosine kinase-induced pathways is a critical step driving the oncogenic potential of brain cancer. In this study, we investigated the role of two members of the Sprouty (Spry) family in brain cancer-derived cell lines. Using immunoblot analyses we found essential differences in the pattern of endogenous Spry3 and Spry4 expression. While Spry4 expression was mitogen-dependent and repressed in a number of cells from higher malignant brain cancers, Spry3 levels neither fluctuated in response to serum withdrawal nor were repressed in glioblastoma (GBM)-derived cell lines. In accordance to the well-known inhibitory role of Spry proteins in fibroblast growth factor (FGF)-mediated signaling, both Spry proteins were able to interfere with FGF-induced activation of the MAPK pathway although to a different extent. In response to serum solely, Spry4 exerts its role as a negative regulator of MAPK activation. Ectopic expression of Spry4 inhibited proliferation and migration of GBM-originated cells, positioning it as a tumor suppressor in brain cancer. In contrast, elevated Spry3 levels accelerated both proliferation and migration of these cell lines, while repression of Spry3 levels using shRNA caused a significant diminished growth and migration velocity rate of a GBM-derived cell line. This argues for a tumor-promoting function of Spry3 in GBMs. Based on these data we conclude that Spry3 and Spry4 fulfill different if not opposing roles within the cancerogenesis of brain malignancies.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Carcinogénesis/metabolismo , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas del Tejido Nervioso/fisiología , Línea Celular Tumoral , Proliferación Celular , Humanos , ARN Interferente Pequeño/metabolismo , Transducción de Señal
9.
Expert Opin Ther Targets ; 15(7): 829-46, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21375471

RESUMEN

INTRODUCTION: Fibroblast growth factors (FGF) exert a combination of biological effects that contribute to four of the six essential hallmarks of cancer. It is no surprise that FGF-dependent signaling has increasingly moved to the center of cancer therapy research during the past decade. This is illustrated by the large number of publications focusing on various aspects of this theme that have been published in the past 5 years. AREAS COVERED: Information from these sources as well as ongoing work from the authors' groups is used to outline the physiological functions of FGF signaling and to highlight how the high oncogenic effects of deregulated FGFs and FGFRs derive from their physiological functions. The biological effect of deregulated FGFR signaling in malignant diseases is described and the current state of therapeutic targeting of FGFR is summarized. EXPERT OPINION: Strategies for targeting FGFR-signaling for cancer therapy are very promising, but need to be carefully developed based on the physiological roles of FGF signaling. Preventive measures may be necessary for protection from FGF-related side effects. Combined targeting of several receptor tyrosine kinases or combination with other therapies may be a useful way of avoiding or ameliorating side effects. FGF-related markers of prognosis and therapy response still need to be investigated.


Asunto(s)
Neoplasias/tratamiento farmacológico , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Transición Epitelial-Mesenquimal , Humanos , Ligandos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Pronóstico
10.
Cell Mol Life Sci ; 67(19): 3299-311, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20461437

RESUMEN

Sprouty2 is an important inhibitor of cell proliferation and signal transduction. In this study, we found a bimodal expression of Sprouty2 protein during cell cycle progression after exit from quiescence, whereas elevated Sprouty4 expression in the G1 phase stayed high throughout the rest of the cell cycle. Induction of the mitogen-activated protein kinase via activated Ras was crucial for increased Sprouty2 expression at the G0/G1 transition. Following the first peak, accelerated proteasomal protein degradation caused a transient attenuation of Sprouty2 abundance during late G1. Since the decline in its expression was abolished by dominant negative c-Cbl and the timely restricted interaction between Sprouty2 and c-Cbl disappeared at the second peak of Sprouty2 expression, we conclude that the second phase in the cell cycle-specific expression profile of Sprouty2 is solely dependent on ubiquitination by c-Cbl. Our results suggest that Sprouty2 abundance is the result of strictly coordinated activities of Ras and c-Cbl.


Asunto(s)
Ciclo Celular/genética , Genes ras/genética , Línea Celular , Línea Celular Tumoral , Fibroblastos/metabolismo , Humanos , Pulmón/patología
11.
Biol Chem ; 391(7): 813-21, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20482313

RESUMEN

Sprouty (Spry) proteins are well-known negative regulators of receptor tyrosine kinase-mediated signalling. Their expression is controlled by mitogens, implying a negative feedback loop. Correspondingly, the different members of the family fulfil important roles during organogenesis by adjustment of growth factor-induced processes. In addition, Spry4, one member of this protein family, has been shown to regulate angiogenesis by inhibiting vascular endothelial cell growth factor-induced extracellular signalling-regulated kinase (ERK) activation. Because oxygen is an important regulator of angiogenesis, we investigated Spry4 expression patterns under hypoxic conditions. Our data demonstrate that both hypoxia and desferrioxamine (DFO) treatment increased Spry4 expression. Following iron depletion, elevated Spry4 levels were detected in several cell types independent of tissue origin, presence of mitogens, cell differentiation and malignancy. Evaluation of the underlying regulative mechanisms revealed that augmented transcription and increased mRNA stability enhance mRNA levels of Spry4 in response to DFO. This study unveils a growth factor-independent regulation mechanism of Spry4 expression. Because increased Spry4 levels are accompanied by disappearing ERK phosphorylation, Spry4 might be involved in the timely restriction of MAPK signals under hypoxic conditions, similar to its role in mitogen-regulated processes. However, the functional significance of the observed upregulation of Spry4 during iron depletion remains to be clarified.


Asunto(s)
Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas del Tejido Nervioso/genética , Estabilidad del ARN/genética , Transcripción Genética/genética , Hipoxia de la Célula/genética , Células Cultivadas , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Cancer Res ; 69(1): 235-42, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118008

RESUMEN

Recently, we found epigenetic silencing of the Ras effector genes NORE1B and/or RASSF1A in 97% of the hepatocellular carcinoma (HCC) investigated. This is strong evidence that the two genes are of major significance in hepatocarcinogenesis. Although RASSF1A serves as a tumor suppressor gene, the functions of NORE1B are largely unknown. Here, we studied the role of NORE1B for growth and transformation of cells. To understand the molecular mechanisms of action of the gene, we used the wild-type form and deletion mutants without the NH(2) terminus and CENTRAL domain, the Ras association (RA) domain, or the COOH-terminal SARAH-domain. Intact RA and SARAH-domains were found to be necessary for NORE1B (a) to increase the G(0)-G(1) fraction in hepatoma cells, (b) to suppress c-Myc/Ha-Ras-induced cell transformation, and (c) to interact closely with RASSF1A, as determined with fluorescence resonance energy transfer. In further studies, cell cycle delay by NORE1B was equally effective in hepatocyte cell lines with wild-type or mutant Ras suggesting that NORE1B does not interact with either Ras. In conclusion, NORE1B suppresses replication and transformation of cells as effectively as RASSF1A and thus is a putative tumor suppressor gene. NORE1B interacts physically with RASSF1A and functional loss of one of the interacting partners may lead to uncontrolled growth and transformation of hepatocytes. This may explain the frequent epigenetic silencing of NORE1B and/or RASSF1A in HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Transformación Celular Neoplásica/genética , Neoplasias Hepáticas/genética , Proteínas de Unión al GTP Monoméricas/genética , Proteínas Supresoras de Tumor/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Ciclo Celular/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Citoplasma/metabolismo , Eliminación de Gen , Silenciador del Gen , Genes Supresores de Tumor , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas de Unión al GTP Monoméricas/metabolismo , ARN Interferente Pequeño/genética , Transfección , Proteínas Supresoras de Tumor/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
13.
Mol Cancer Res ; 5(5): 509-20, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17510316

RESUMEN

Sprouty (Spry) proteins function as inhibitors of receptor tyrosine kinase signaling mainly by interfering with the Ras/Raf/mitogen-activated protein kinase cascade, a pathway known to be frequently deregulated in human non-small cell lung cancer (NSCLC). In this study, we show a consistently lowered Spry2 expression in NSCLC when compared with the corresponding normal lung epithelium. Based on these findings, we investigated the influence of Spry2 expression on the malignant phenotype of NSCLC cells. Ectopic expression of Spry2 antagonized mitogen-activated protein kinase activity and inhibited cell migration in cell lines homozygous for K-Ras wild type, whereas in NSCLC cells expressing mutated K-Ras, Spry2 failed to diminish extracellular signal-regulated kinase (ERK) phosphorylation. Nonetheless, Spry2 significantly reduced cell proliferation in all investigated cell lines and blocked tumor formation in mice. Accordingly, a Spry2 mutant unable to inhibit ERK phosphorylation reduced cell proliferation significantly but less pronounced compared with the wild-type protein. Therefore, we conclude that Spry2 interferes with ERK phosphorylation and another yet unidentified pathway. Our results suggest that Spry2 plays a role as tumor suppressor in NSCLC by antagonizing receptor tyrosine kinase-induced signaling at different levels, indicating feasibility for the usage of Spry in targeted gene therapy of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación hacia Abajo/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Activación Enzimática , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Proteínas de la Membrana , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Mutantes/metabolismo , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
14.
J Cell Sci ; 119(Pt 3): 459-69, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16418217

RESUMEN

The major vault protein (MVP) is the main component of vaults, large ribonucleoprotein particles implicated in the regulation of cellular signaling cascades and multidrug resistance. Here, we identify MVP as an interferon gamma (IFN-gamma)-inducible protein. Treatment with IFN-gamma resulted in a significant upregulation of MVP promoter activity as well as mRNA and protein levels. Activation of MVP expression by IFN-gamma involved transcriptional upregulation through the JAK/STAT pathway based on an interaction of STAT1 with an interferon-gamma-activated site (GAS) within the proximal MVP promoter. Mutation of this site distinctly reduced basal as well as IFN-gamma-stimulated MVP transcription. IFN-gamma also significantly enhanced the translation rate of MVP. Ectopic MVP overexpression in the MVP-negative lung cancer cell model H65 led to a downregulation of three known IFN-gamma-regulated genes, namely ICAM-1, CD13 and CD36. Additionally, presence of MVP in H65 cells blocked both basal and IFN-gamma-induced ICAM-1 expression whereas downmodulation of endogenous MVP levels by shRNA enhanced IFN-gamma-induced ICAM-1 expression in U373 glioblastoma cells. MVP-mediated IFN-gamma insensitivity was accompanied by significantly reduced STAT1 phosphorylation at Y701 and diminished translocation of STAT1 into the nucleus. Summarizing, we identify MVP as an IFN-gamma-responsive gene interfering with IFN-gamma-activated JAK/STAT signals. These data further substantiate that the vault particle functions as a general interaction platform for cellular signaling cascades.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón gamma/farmacología , Neoplasias Pulmonares/metabolismo , Transducción de Señal/efectos de los fármacos , Partículas Ribonucleoproteicas en Bóveda/biosíntesis , Transporte Activo de Núcleo Celular/efectos de los fármacos , Antineoplásicos/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Interferón gamma/metabolismo , Neoplasias Pulmonares/genética , Factor de Transcripción STAT1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Partículas Ribonucleoproteicas en Bóveda/genética
15.
Biochem Pharmacol ; 71(4): 426-40, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16343446

RESUMEN

Aim of this study was to investigate the anticancer properties of the new lanthanum compound [tris(1,10-phenanthroline)lanthanum(III)]trithiocyanate (KP772; FFC24). In vitro, growth inhibition by KP772 was comparable for >60 tumour cell models with IC50 values generally in the low microM range. KP772 induced tumour cell apoptosis indicated by chromatin condensation, caspase substrate cleavage and mitochondrial membrane depolarisation. DNA is unlikely to represent the primary molecular target of KP772, as no significant interaction or damage of DNA was detectable both in vitro and in living cells. Moreover, we found no evidence for induction of radical species. In contrast, KP772 potently inhibited DNA synthesis paralleled by a massive block of cell cycle in G0/G1 phase and a selective decrease of cyclin B1. Although treatment with KP772 induced expression of p53 and p21Waf1, transfection of wild-type p53 into knock-out cells only marginally enhanced the cytostatic activity of KP772. In vivo, the anticancer activity of KP772 against human DLD-1 colon carcinoma xenografts was comparable to that of cisplatin and methotrexate at doses not causing significant adverse effects. With regard to toxicity, the LD50 and no-observed-adverse-effect levels (NOAEL) of KP772 in Sprague-Dawley rats were 21.6 and 7.5 mg/kg, in outbred albino mice 62 and 10 mg/kg, respectively. In summary, KP772 exerts anticancer activity via potent induction of cell cycle arrest and/or apoptosis and has promising in vivo anticancer activity against a human colon cancer xenograft. Together, these data suggest further development of KP772 as a new anticancer metal-drug.


Asunto(s)
Antineoplásicos/farmacología , Lantano/farmacología , Compuestos Organometálicos/farmacología , Fenantrolinas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Bencimidazoles , Bleomicina/farmacología , Carbocianinas , Caspasas/biosíntesis , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo Cometa , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Fase G1/efectos de los fármacos , Células HL-60 , Humanos , Inyecciones Intravenosas , Sustancias Intercalantes/farmacología , Lantano/química , Lantano/uso terapéutico , Ratones , Ratones Desnudos , Estructura Molecular , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Compuestos Organometálicos/química , Compuestos Organometálicos/uso terapéutico , Fenantrolinas/química , Fenantrolinas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Ratas , Ratas Sprague-Dawley , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Proteína p53 Supresora de Tumor/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...