Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 16(23): 29844-29855, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38829261

RESUMEN

Copper plays critical roles as a metal active site cofactor and metalloallosteric signal for enzymes involved in cell proliferation and metabolism, making it an attractive target for cancer therapy. In this study, we investigated the efficacy of polydopamine nanoparticles (PDA NPs), classically applied for metal removal from water, as a therapeutic strategy for depleting intracellular labile copper pools in triple-negative breast cancer models through the metal-chelating groups present on the PDA surface. By using the activity-based sensing probe FCP-1, we could track the PDA-induced labile copper depletion while leaving total copper levels unchanged and link it to the selective MDA-MB-231 cell death. Further mechanistic investigations revealed that PDA NPs increased reactive oxygen species (ROS) levels, potentially through the inactivation of superoxide dismutase 1 (SOD1), a copper-dependent antioxidant enzyme. Additionally, PDA NPs were found to interact with the mitochondrial membrane, resulting in an increase in the mitochondrial membrane potential, which may contribute to enhanced ROS production. We employed an in vivo tumor model to validate the therapeutic efficacy of PDA NPs. Remarkably, in the absence of any additional treatment, the presence of PDA NPs alone led to a significant reduction in tumor volume by a factor of 1.66 after 22 days of tumor growth. Our findings highlight the potential of PDA NPs as a promising therapeutic approach for selectively targeting cancer by modulating copper levels and inducing oxidative stress, leading to tumor growth inhibition as shown in these triple-negative breast cancer models.


Asunto(s)
Cobre , Indoles , Nanopartículas , Polímeros , Especies Reactivas de Oxígeno , Neoplasias de la Mama Triple Negativas , Cobre/química , Cobre/farmacología , Polímeros/química , Polímeros/farmacología , Indoles/química , Indoles/farmacología , Humanos , Animales , Ratones , Nanopartículas/química , Femenino , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , Oxidación-Reducción , Nanomedicina , Proliferación Celular/efectos de los fármacos , Homeostasis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Superóxido Dismutasa-1/metabolismo
2.
Cells ; 12(9)2023 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-37174738

RESUMEN

The induction of pluripotency by enforced expression of different sets of genes in somatic cells has been achieved with reprogramming technologies first described by Yamanaka's group. Methodologies for generating induced pluripotent stem cells are as varied as the combinations of genes used. It has previously been reported that the adenoviral E1a gene can induce the expression of two of the Yamanaka factors (c-Myc and Oct-4) and epigenetic changes. Here, we demonstrate that the E1a-12S over-expression is sufficient to induce pluripotent-like characteristics closely to epiblast stem cells in mouse embryonic fibroblasts through the activation of the pluripotency gene regulatory network. These findings provide not only empirical evidence that the expression of one single factor is sufficient for partial reprogramming but also a potential mechanistic explanation for how viral infection could lead to neoplasia if they are surrounded by the appropriate environment or the right medium, as happens with the tumorogenic niche.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas , Animales , Ratones , Reprogramación Celular/genética , Diferenciación Celular , Fibroblastos/metabolismo , Factor 4 Similar a Kruppel , Células Madre Pluripotentes Inducidas/metabolismo
3.
Acta Biomater ; 134: 593-604, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34325075

RESUMEN

Adenoviral (Ad) vectors have proven to be important tools for gene and cell therapy, although some issues still need to be addressed, such as undesired interactions with blood components and off-target sequestration that ultimately hamper efficacy. In the past years, several organic and inorganic materials have been developed to reduce immunogenicity and improve biodistribution of Ad vectors. Here we investigated the influence of the functionalization of 14 nm PEGylated gold nanoparticles (AuNPs) with quaternary ammonium groups and an arginine-glycine-aspartic acid (RGD)-motif on the uptake and biodistribution of Ad vectors. We report the formation of Ad@AuNPs complexes that promote cell attachment and uptake, independently of the presence of the coxsackievirus and adenovirus receptor (CAR) and αvß3 and αvß5 integrins, significantly improving transduction without limiting Ad bioactivity. Besides, the presence of the RGD peptide favors tumor targeting and decreases Ad sequestration in the liver. Additionally, tumor delivery of a coated Ad vector expressing the human sodium iodide symporter (hNIS) by mesenchymal stem cells induces increased accumulation of radioactive iodine (131I) and tumor volume reduction compared to naked Ad-hNIS, highlighting the promising potential of our coating formulation in cancer gene therapy. STATEMENT OF SIGNIFICANCE: Modification of adenoviral vectors with lipids and polymers can reduce interactions with blood components and increase tumor accumulation; however, increased toxicity and reduced transduction efficiency were indicated. Coating with gold nanoparticles has proven to be a successful strategy for increasing the efficiency of transduction of receptor-defective cell lines. Here we explore the contribution of cell surface receptors on the mechanisms of entry of Ad vectors coated with gold nanoparticles in cell lines with varying degrees of resistance to infection. The enhancement of the anti-tumoral effect shown in this work provides new evidence for the potential of our formulation.


Asunto(s)
Nanopartículas del Metal , Neoplasias de la Tiroides , Adenoviridae/genética , Línea Celular Tumoral , Vectores Genéticos , Oro , Humanos , Radioisótopos de Yodo , Distribución Tisular
4.
Nat Commun ; 12(1): 3660, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135321

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) integrates cellular nutrient signaling and hormonal cues to control metabolism. We have previously shown that constitutive nutrient signaling to mTORC1 by means of genetic activation of RagA (expression of GTP-locked RagA, or RagAGTP) in mice resulted in a fatal energetic crisis at birth. Herein, we rescue neonatal lethality in RagAGTP mice and find morphometric and metabolic alterations that span glucose, lipid, ketone, bile acid and amino acid homeostasis in adults, and a median lifespan of nine months. Proteomic and metabolomic analyses of livers from RagAGTP mice reveal a failed metabolic adaptation to fasting due to a global impairment in PPARα transcriptional program. These metabolic defects are partially recapitulated by restricting activation of RagA to hepatocytes, and revert by pharmacological inhibition of mTORC1. Constitutive hepatic nutrient signaling does not cause hepatocellular damage and carcinomas, unlike genetic activation of growth factor signaling upstream of mTORC1. In summary, RagA signaling dictates dynamic responses to feeding-fasting cycles to tune metabolism so as to match the nutritional state.


Asunto(s)
Ayuno/metabolismo , Hígado/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Glucosa/metabolismo , Homeostasis , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Proteínas de Unión al GTP Monoméricas/genética , Nutrientes/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Fenotipo , Proteómica , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/farmacología , Transcripción Genética/efectos de los fármacos , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo
5.
Nat Metab ; 1(8): 775-789, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31579886

RESUMEN

The humoral immune response demands that B cells undergo a sudden anabolic shift and high cellular nutrient levels which are required to sustain the subsequent proliferative burst. Follicular lymphoma (FL) originates from B cells that have participated in the humoral response, and 15% of FL samples harbor point, activating mutations in RRAGC, an essential activator of mTORC1 downstream of the sensing of cellular nutrients. The impact of recurrent RRAGC mutations in B cell function and lymphoma is unexplored. RRAGC mutations, targeted to the endogenous locus in mice, confer a partial insensitivity to nutrient deprivation, but strongly exacerbate B cell responses and accelerate lymphomagenesis, while creating a selective vulnerability to pharmacological inhibition of mTORC1. This moderate increase in nutrient signaling synergizes with paracrine cues from the supportive T cell microenvironment that activates B cells via the PI3K-Akt-mTORC1 axis. Hence, Rragc mutations sustain induced germinal centers and murine and human FL in the presence of decreased T cell help. Our results support a model in which activating mutations in the nutrient signaling pathway foster lymphomagenesis by corrupting a nutrient-dependent control over paracrine signals from the T cell microenvironment.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Activación de Linfocitos , Linfoma Folicular/tratamiento farmacológico , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Humanos , Linfoma Folicular/patología , Ratones , Ratones Transgénicos
6.
Nat Commun ; 9(1): 3012, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30069007

RESUMEN

Polo-like kinase 1 (Plk1) is overexpressed in a wide spectrum of human tumors, being frequently considered as an oncogene and an attractive cancer target. However, its contribution to tumor development is unclear. Using a new inducible knock-in mouse model we report here that Plk1 overexpression results in abnormal chromosome segregation and cytokinesis, generating polyploid cells with reduced proliferative potential. Mechanistically, these cytokinesis defects correlate with defective loading of Cep55 and ESCRT complexes to the abscission bridge, in a Plk1 kinase-dependent manner. In vivo, Plk1 overexpression prevents the development of Kras-induced and Her2-induced mammary gland tumors, in the presence of increased rates of chromosome instability. In patients, Plk1 overexpression correlates with improved survival in specific breast cancer subtypes. Therefore, despite the therapeutic benefits of inhibiting Plk1 due to its essential role in tumor cell cycles, Plk1 overexpression has tumor-suppressive properties by perturbing mitotic progression and cytokinesis.


Asunto(s)
Carcinogénesis/genética , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Centrosoma/metabolismo , Segregación Cromosómica , Citocinesis , Modelos Animales de Enfermedad , Embrión de Mamíferos/citología , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Ratones , Proteínas Nucleares/metabolismo , Oncogenes , Quinasa Tipo Polo 1
7.
Clin Cancer Res ; 24(15): 3550-3559, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29588308

RESUMEN

Purpose: Despite the wide use of antiangiogenic drugs in the clinical setting, predictive biomarkers of response to these drugs are still unknown.Experimental Design: We applied whole-exome sequencing of matched germline and basal plasma cell-free DNA samples (WES-cfDNA) on a RAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patient with primary resistance to standard treatment regimens, including inhibitors to the VEGF:VEGFR2 pathway. We performed extensive functional experiments, including ectopic expression of VEGFR2 mutants in different cell lines, kinase and drug sensitivity assays, and cell- and patient-derived xenografts.Results: WES-cfDNA yielded a 77% concordance rate with tumor exome sequencing and enabled the identification of the KDR/VEGFR2 L840F clonal, somatic mutation as the cause of therapy refractoriness in our patient. In addition, we found that 1% to 3% of samples from cancer sequencing projects harbor KDR somatic mutations located in protein residues frequently mutated in other cancer-relevant kinases, such as EGFR, ABL1, and ALK. Our in vitro and in vivo functional assays confirmed that L840F causes strong resistance to antiangiogenic drugs, whereas the KDR hot-spot mutant R1032Q confers sensitivity to strong VEGFR2 inhibitors. Moreover, we showed that the D717V, G800D, G800R, L840F, G843D, S925F, R1022Q, R1032Q, and S1100F VEGFR2 mutants promote tumor growth in mice.Conclusions: Our study supports WES-cfDNA as a powerful platform for portraying the somatic mutation landscape of cancer and discovery of new resistance mechanisms to cancer therapies. Importantly, we discovered that VEGFR2 is somatically mutated across tumor types and that VEGFR2 mutants can be oncogenic and control sensitivity/resistance to antiangiogenic drugs. Clin Cancer Res; 24(15); 3550-9. ©2018 AACR.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Neoplasias Colorrectales/genética , Neovascularización Patológica/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Quinasa de Linfoma Anaplásico/genética , Inhibidores de la Angiogénesis/efectos adversos , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular/genética , Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Receptores ErbB/genética , Exoma/genética , Femenino , Xenoinjertos , Humanos , Ratones , Mutación , Neovascularización Patológica/sangre , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Conformación Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-abl/genética , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Secuenciación del Exoma
8.
Graefes Arch Clin Exp Ophthalmol ; 256(3): 535-546, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29353344

RESUMEN

PURPOSE: To study the safety and biocompatibility of Laponite clay (LAP) within an intravitreal and suprachoroidal administration in rabbit eyes. METHODS: Thirty-two New Zealand albino rabbits were divided into two experimental groups to test intravitreal (IVT group) and suprachoroidal (SCS group) administration of a 100-µl and 50-µl Laponite suspension respectively. Following injection, the eyes were monitored by ocular tonometry, slit-lamp eye examination and indirect ophthalmoscopy, at 24 h, 1, 4, 12, and 14 weeks post administration. Histological examination was also performed to determine whether any ocular pathological change had occurred. Throughout the study, LAP presence in vitreous was estimated by complexometric titration with ethylenediaminetetraacetic acid (EDTA), taking advantage of the Laponite high content of magnesium ions. RESULTS: Neither significant differences in the intraocular pressure, nor relevant ocular complications were found in the two experimental groups after LAP administration. The histology of the retina remained unchanged. LAP presence in vitreous could be indirectly confirmed by complexometric titration until 14 weeks post administration in eyes of IVT group. CONCLUSION: Laponite could be considered as a vehicle for potential clinical use in ocular drug administration, due to its proven ocular biocompatibility and its transparency in gel state.


Asunto(s)
Retina/patología , Enfermedades de la Retina/tratamiento farmacológico , Silicatos/administración & dosificación , Visión Ocular , Silicatos de Aluminio/administración & dosificación , Animales , Materiales Biocompatibles/administración & dosificación , Arcilla , Modelos Animales de Enfermedad , Electrorretinografía , Femenino , Inyecciones Intravítreas , Oftalmoscopía , Conejos , Retina/efectos de los fármacos , Retina/fisiopatología , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/fisiopatología
9.
Aging Cell ; 17(2)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29280266

RESUMEN

Cellular senescence is a damage response aimed to orchestrate tissue repair. We have recently reported that cellular senescence, through the paracrine release of interleukin-6 (IL6) and other soluble factors, strongly favors cellular reprogramming by Oct4, Sox2, Klf4, and c-Myc (OSKM) in nonsenescent cells. Indeed, activation of OSKM in mouse tissues triggers senescence in some cells and reprogramming in other cells, both processes occurring concomitantly and in close proximity. In this system, Ink4a/Arf-null tissues cannot undergo senescence, fail to produce IL6, and cannot reprogram efficiently; whereas p53-null tissues undergo extensive damage and senescence, produce high levels of IL6, and reprogram efficiently. Here, we have further explored the genetic determinants of in vivo reprogramming. We report that Ink4a, but not Arf, is necessary for OSKM-induced senescence and, thereby, for the paracrine stimulation of reprogramming. However, in the absence of p53, IL6 production and reprogramming become independent of Ink4a, as revealed by the analysis of Ink4a/Arf/p53 deficient mice. In the case of the cell cycle inhibitor p21, its protein levels are highly elevated upon OSKM activation in a p53-independent manner, and we show that p21-null tissues present increased levels of senescence, IL6, and reprogramming. We also report that Il6-mutant tissues are impaired in undergoing reprogramming, thus reinforcing the critical role of IL6 in reprogramming. Finally, young female mice present lower efficiency of in vivo reprogramming compared to male mice, and this gender difference disappears with aging, both observations being consistent with the known anti-inflammatory effect of estrogens. The current findings regarding the interplay between senescence and reprogramming may conceivably apply to other contexts of tissue damage.


Asunto(s)
Reprogramación Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Interleucina-6/metabolismo , Animales , Senescencia Celular , Femenino , Humanos , Factor 4 Similar a Kruppel , Ratones
10.
Cell Rep ; 19(5): 928-938, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467906

RESUMEN

Mammalian DNA replication origins are "licensed" by the loading of DNA helicases, a reaction that is mediated by CDC6 and CDT1 proteins. After initiation of DNA synthesis, CDC6 and CDT1 are inhibited to prevent origin reactivation and DNA overreplication before cell division. CDC6 and CDT1 are highly expressed in many types of cancer cells, but the impact of their deregulated expression had not been investigated in vivo. Here, we have generated mice strains that allow the conditional overexpression of both proteins. Adult mice were unharmed by the individual overexpression of either CDC6 or CDT1, but their combined deregulation led to DNA re-replication in progenitor cells and lethal tissue dysplasias. This study offers mechanistic insights into the necessary cooperation between CDC6 and CDT1 for facilitation of origin reactivation and describes the physiological consequences of DNA overreplication.


Asunto(s)
Replicación del ADN , Diarrea Infantil/genética , Mucosa Intestinal/metabolismo , Síndromes de Malabsorción/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Diarrea Infantil/metabolismo , Femenino , Mucosa Intestinal/patología , Síndromes de Malabsorción/metabolismo , Masculino , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transgenes
11.
Science ; 354(6315)2016 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-27884981

RESUMEN

Reprogramming of differentiated cells into pluripotent cells can occur in vivo, but the mechanisms involved remain to be elucidated. Senescence is a cellular response to damage, characterized by abundant production of cytokines and other secreted factors that, together with the recruitment of inflammatory cells, result in tissue remodeling. Here, we show that in vivo expression of the reprogramming factors OCT4, SOX2, KLF4, and cMYC (OSKM) in mice leads to senescence and reprogramming, both coexisting in close proximity. Genetic and pharmacological analyses indicate that OSKM-induced senescence requires the Ink4a/Arf locus and, through the production of the cytokine interleukin-6, creates a permissive tissue environment for in vivo reprogramming. Biological conditions linked to senescence, such as tissue injury or aging, favor in vivo reprogramming by OSKM. These observations may be relevant for tissue repair.


Asunto(s)
Reprogramación Celular/genética , Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Madre Pluripotentes Inducidas/citología , Factores de Transcripción/metabolismo , Compuestos de Anilina/farmacología , Animales , Antineoplásicos/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Regulación de la Expresión Génica , Sitios Genéticos , Células Madre Pluripotentes Inducidas/metabolismo , Interleucina-6/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Sulfonamidas/farmacología , Teratoma/genética , Teratoma/patología , Factores de Transcripción/genética
12.
Dig Dis Sci ; 61(10): 2896-2907, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27343037

RESUMEN

BACKGROUND AND AIM: Recent observational studies have shown therapeutic benefits of acetylsalicylic acid (ASA) in several types of cancer. We examined whether ASA exerts antitumor activity in esophageal adenocarcinoma (EAC). METHODS: Human EAC cells (OE33) were treated with ASA (0-5 mM) to evaluate proliferation, apoptosis, and migration. In vivo model: OE33-derived tumors were subcutaneously implanted into athymic mice which were allocated to ASA (5 or 50 mg/kg/day)/vehicle (5-6 animals/group). Tumor growth was assessed every 2-3 days, and after 40 days, mice were euthanized. Plasma drug levels were determined by high-performance liquid chromatography. Histological and immunohistochemical (Ki67, activated caspase-3) analysis of tumors were performed. The effect of ASA on tumor prostaglandin E2 (PGE2) levels was also evaluated. RESULTS: In vitro cell proliferation and migration were significantly inhibited while apoptosis increased (p < 0.05) by ASA. Although ASA did not induce tumor remission, tumor progression was significantly lower in ASA-treated mice when compared to non-treated animals (478 % in mice treated with 5 mg/kg/day ASA vs. 2696 % control; 748 % in mice treated with 50 mg/kg/day ASA vs. 2670 % control). Maximum tumor inhibition was 92 and 85 %, respectively. This effect was associated with a significant decrease of proliferation index in tumors. ASA 5 mg/kg/day did not modify tumor PGE2 levels. Whereas tumor PGE2 content in mice treated with ASA 50 mg/kg was lower than in control mice, the difference was not significant. CONCLUSION: Although these results need to be confirmed in other EAC cells, our data suggest a role for ASA in the treatment of this tumor.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Aspirina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Dinoprostona/metabolismo , Progresión de la Enfermedad , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Humanos , Técnicas In Vitro , Antígeno Ki-67/efectos de los fármacos , Antígeno Ki-67/metabolismo , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Pathol ; 239(2): 206-17, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27174785

RESUMEN

The Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, craniofacial dysmorphism, and congenital heart defects. A significant proportion of NS patients may also develop myeloproliferative disorders (MPDs), including juvenile myelomonocytic leukaemia (JMML). Surprisingly, scarce information is available in relation to other tumour types in these patients. We have previously developed and characterized a knock-in mouse model that carries one of the most frequent KRAS-NS-related mutations, the K-Ras(V14I) substitution, which recapitulates most of the alterations described in NS patients, including MPDs. The K-Ras(V14I) mutation is a mild activating K-Ras protein; thus, we have used this model to study tumour susceptibility in comparison with mice expressing the classical K-Ras(G12V) oncogene. Interestingly, our studies have shown that these mice display a generalized tumour predisposition and not just MPDs. In fact, we have observed that the K-Ras(V14I) mutation is capable of cooperating with the p16Ink4a/p19Arf and Trp53 tumour suppressors, as well as with other risk factors such as pancreatitis, thereby leading to a higher cancer incidence. In conclusion, our results illustrate that the K-Ras(V14I) activating protein is able to induce cancer, although at a much lower level than the classical K-Ras(G12V) oncogene, and that it can be significantly modulated by both genetic and non-genetic events. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Cardiopatías Congénitas/genética , Neoplasias Pulmonares/genética , Neoplasias/genética , Síndrome de Noonan/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Alelos , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Tamización de Portadores Genéticos , Cardiopatías Congénitas/patología , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Neoplasias/patología , Síndrome de Noonan/patología , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
14.
Mol Cell Biol ; 35(20): 3566-78, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26240282

RESUMEN

Aurora kinase B, one of the three members of the mammalian Aurora kinase family, is the catalytic component of the chromosomal passenger complex, an essential regulator of chromosome segregation in mitosis. Aurora B is overexpressed in human tumors although whether this kinase may function as an oncogene in vivo is not established. Here, we report a new mouse model in which expression of the endogenous Aurkb locus can be induced in vitro and in vivo. Overexpression of Aurora B in cultured cells induces defective chromosome segregation and aneuploidy. Long-term overexpression of Aurora B in vivo results in aneuploidy and the development of multiple spontaneous tumors in adult mice, including a high incidence of lymphomas. Overexpression of Aurora B also results in a reduced DNA damage response and decreased levels of the p53 target p21(Cip1) in vitro and in vivo, in line with an inverse correlation between Aurora B and p21(Cip1) expression in human leukemias. Thus, overexpression of Aurora B may contribute to tumor formation not only by inducing chromosomal instability but also by suppressing the function of the cell cycle inhibitor p21(Cip1).


Asunto(s)
Aneuploidia , Aurora Quinasa B/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/metabolismo , Expresión Génica , Silenciador del Gen , Ratones Endogámicos C57BL , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
15.
Nat Commun ; 5: 5863, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25519492

RESUMEN

Coronary heart disease is one of the main causes of death in the developed world, and treatment success remains modest, with high mortality rates within 1 year after myocardial infarction (MI). Thus, new therapeutic targets and effective treatments are necessary. Short telomeres are risk factors for age-associated diseases, including heart disease. Here we address the potential of telomerase (Tert) activation in prevention of heart failure after MI in adult mice. We use adeno-associated viruses for cardiac-specific Tert expression. We find that upon MI, hearts expressing Tert show attenuated cardiac dilation, improved ventricular function and smaller infarct scars concomitant with increased mouse survival by 17% compared with controls. Furthermore, Tert treatment results in elongated telomeres, increased numbers of Ki67 and pH3-positive cardiomyocytes and a gene expression switch towards a regeneration signature of neonatal mice. Our work suggests telomerase activation could be a therapeutic strategy to prevent heart failure after MI.


Asunto(s)
Terapia Genética/métodos , Insuficiencia Cardíaca/prevención & control , Infarto del Miocardio/genética , Telomerasa/genética , Telómero/genética , Animales , Dependovirus/genética , Expresión Génica , Perfilación de la Expresión Génica , Vectores Genéticos , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Anotación de Secuencia Molecular , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Telomerasa/metabolismo , Telómero/metabolismo , Telómero/patología , Remodelación Ventricular
16.
Nat Genet ; 46(9): 1028-33, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25129145

RESUMEN

Neutrophils are key innate immune effector cells that are essential to fighting bacterial and fungal pathogens. Here we report that mice carrying a hematopoietic lineage-specific deletion of Jagn1 (encoding Jagunal homolog 1) cannot mount an efficient neutrophil-dependent immune response to the human fungal pathogen Candida albicans. Global glycobiome analysis identified marked alterations in the glycosylation of proteins involved in cell adhesion and cytotoxicity in Jagn1-deficient neutrophils. Functional analysis confirmed marked defects in neutrophil migration in response to Candida albicans infection and impaired formation of cytotoxic granules, as well as defective myeloperoxidase release and killing of Candida albicans. Treatment with granulocyte/macrophage colony-stimulating factor (GM-CSF) protected mutant mice from increased weight loss and accelerated mortality after Candida albicans challenge. Notably, GM-CSF also restored the defective fungicidal activity of bone marrow cells from humans with JAGN1 mutations. These data directly identify Jagn1 (JAGN1 in humans) as a new regulator of neutrophil function in microbial pathogenesis and uncover a potential treatment option for humans.


Asunto(s)
Candidiasis/inmunología , Proteínas de la Membrana/inmunología , Neutrófilos/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/microbiología , Candida albicans , Candidiasis/tratamiento farmacológico , Candidiasis/metabolismo , Candidiasis/microbiología , Glicosilación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Neutrófilos/microbiología
17.
Cell Rep ; 8(2): 420-9, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-25017060

RESUMEN

During bacterial sepsis, proinflammatory cytokines contribute to multiorgan failure and death in a process regulated in part by cytolytic cell granzymes. When challenged with a sublethal dose of the identified mouse pathogen Brucella microti, wild-type (WT) and granzyme A (gzmA)(-/-) mice eliminate the organism from liver and spleen in 2 or 3 weeks, whereas the bacteria persist in mice lacking perforin or granzyme B as well as in mice depleted of Tc cells. In comparison, after a fatal challenge, only gzmA(-/-) mice exhibit increased survival, which correlated with reduced proinflammatory cytokines. Depletion of natural killer (NK) cells protects WT mice from sepsis without influencing bacterial clearance and the transfer of WT, but not gzmA(-/-) NK, cells into gzmA(-/-) recipients restores the susceptibility to sepsis. Therefore, infection-related pathology, but not bacterial clearance, appears to require gzmA, suggesting the protease may be a therapeutic target for the prevention of bacterial sepsis without affecting immune control of the pathogen.


Asunto(s)
Bacteriemia/metabolismo , Granzimas/metabolismo , Animales , Brucella , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Granzimas/genética , Células Asesinas Naturales/metabolismo , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Bazo/metabolismo , Bazo/patología
18.
Eur J Gastroenterol Hepatol ; 26(8): 871-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24922356

RESUMEN

INTRODUCTION: The current staging systems for hepatocellular carcinoma (HCC) do not sufficiently predict outcomes after liver transplantation (LT). The present study assessed whether some tissue markers related to proliferation and angiogenesis have prognostic value. PATIENTS AND METHODS: The expression of CD34, vascular endothelial growth factor (VEGF), VEGFR2, VEGFR1, angiopoietin-1, angiopoietin-2, TIE2, COX-2, and proliferating cell nuclear antigen (PCNA) in tumor and adjacent cirrhotic tissue samples from 36 patients with HCC (n=10 with tumor recurrence after LT) was determined by immunochemistry. Microvessel density was assessed by CD34 staining and the PCNA labeling index calculated as the percentage of positive cells among at least 1000 hepatocyte nuclei studied in each sample using the computer program ContimUZ. VEGF, VEGFR2, VEGFR-1, angiopoietin-1, angiopoietin-2, TIE2, and COX-2 staining were evaluated by two blinded pathologists. The tumor recurrence rate was analyzed after a minimum follow-up of 36 months. RESULTS: A higher proliferation index in both tumor and adjacent cirrhotic tissue was related to HCC recurrence. The proliferation index in tumor tissue was also related to microvascular invasion. High expression (staining in ≥50% of hepatocytes) of COX2 [P=0.025, odds ratio (OR)=7.5, 95% confidence interval (CI) 1.3-43.4], VEGF (P=0.01, OR=12, 95% CI 1.8-80.4), and its receptor VEGFR-2 (P=0.02, OR=8.5, 95% CI 1.4-49.5) in cirrhotic liver tissue, but not tumor tissue, was related to HCC recurrence after LT. CONCLUSION: A high proliferation index in tumor and cirrhotic tissue and high expression levels of some angiogenic markers in adjacent cirrhotic tissue could be predictive of tumor recurrence after LT.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/cirugía , Neoplasias Hepáticas/cirugía , Trasplante de Hígado , Neovascularización Patológica/metabolismo , Anciano , Proteínas Angiogénicas/metabolismo , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Femenino , Estudios de Seguimiento , Humanos , Cirrosis Hepática/complicaciones , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Microvasos/patología , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/patología , Pronóstico , Antígeno Nuclear de Célula en Proliferación/metabolismo , Recurrencia , Resultado del Tratamiento
19.
J Biomed Nanotechnol ; 10(3): 529-42, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24730248

RESUMEN

We examined the effects of intravenous administration of purified arc-discharge single-walled carbon nanotubes (SWCNTs) on rabbit ileum to establish the possibility of using these SWCNTs as cell markers or drug carriers for the treatment of intestinal diseases. The SWCNT purification process eliminated carbonaceous impurities and decreased the amount of metals. SWCNTs increased the contractile responses induced by KCl, acetylcholine (ACh), and serotonin (5-HT) in rabbit ileum. Verapamil, apamin, glibenclamide, quinine and charybdotoxin reduced the contractile responses induced by ACh and 5-HT in ileum from rabbits treated with SWCNTs, indicating that voltage-dependent Ca2+ channels and small, intermediate, and large-conductance Ca(2+)-activated, ATP-sensitive, and voltage-dependent K+ channels are involved in these effects. Atropine and hexamethonium reduced the ACh response, indicating that muscarinic and nicotinic receptors are involved in this effect. Ondansetron and GR 113808 reduced the 5-HT response, indicating that serotonin 5-HT3 and 5-HT4 receptors are involved in this effect. SWCNTs increased the malondialdehyde plus 4-hydroxyalkenals and carbonyl levels in rabbit plasma and ileum, indicating that SWCNTs produce oxidative stress. SWCNTs did not produce relevant histological changes or modify the levels of the inflammatory mediators iNOS and COX-2 in the ileum. In conclusion, this study demonstrates that the intravenous administration of SWCNTs can evoke oxidative stress and affect contractility in rabbit ileum. These effects could reduce the possibility of using the arc-discharge SWCNTs as cell markers or drug carriers to treat intestinal diseases.


Asunto(s)
Acetilcolina/farmacología , Íleon/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Nanotubos de Carbono , Estrés Oxidativo/efectos de los fármacos , Cloruro de Potasio/farmacología , Serotonina/farmacología , Animales , Sinergismo Farmacológico , Motilidad Gastrointestinal/efectos de los fármacos , Íleon/fisiología , Masculino , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiología , Nanotubos de Carbono/química , Nanotubos de Carbono/ultraestructura , Conejos
20.
Stem Cells ; 32(2): 447-61, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24123596

RESUMEN

Matrix metalloproteinases (MMPs), a family of endopeptidases that are involved in the degradation of extracellular matrix components, have been implicated in skeletal muscle regeneration. Among the MMPs, MMP-2 and MMP-9 are upregulated in Duchenne muscular dystrophy (DMD), a fatal X-linked muscle disorder. However, inhibition or overexpression of specific MMPs in a mouse model of DMD (mdx) has yielded mixed results regarding disease progression, depending on the MMP studied. Here, we have examined the role of MMP-10 in muscle regeneration during injury and muscular dystrophy. We found that skeletal muscle increases MMP-10 protein expression in response to damage (notexin) or disease (mdx mice), suggesting its role in muscle regeneration. In addition, we found that MMP-10-deficient muscles displayed impaired recruitment of endothelial cells, reduced levels of extracellular matrix proteins, diminished collagen deposition, and decreased fiber size, which collectively contributed to delayed muscle regeneration after injury. Also, MMP-10 knockout in mdx mice led to a deteriorated dystrophic phenotype. Moreover, MMP-10 mRNA silencing in injured muscles (wild-type and mdx) reduced muscle regeneration, while addition of recombinant human MMP-10 accelerated muscle repair, suggesting that MMP-10 is required for efficient muscle regeneration. Furthermore, our data suggest that MMP-10-mediated muscle repair is associated with VEGF/Akt signaling. Thus, our findings indicate that MMP-10 is critical for skeletal muscle maintenance and regeneration during injury and disease.


Asunto(s)
Metaloproteinasa 10 de la Matriz/genética , Músculo Esquelético/crecimiento & desarrollo , Distrofias Musculares/genética , Regeneración/genética , Animales , Modelos Animales de Enfermedad , Humanos , Metaloproteinasa 10 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA