Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 327(1): C184-C192, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38826137

RESUMEN

Clinical experience with tyrosine kinase inhibitors (TKIs) over the past two decades has shown that, despite the apparent therapeutic benefit, nearly 30% of patients with chronic myelogenous leukemia (CML) display primary resistance or intolerance to TKIs, and approximately 25% of those treated are forced to switch TKIs at least once during therapy due to acquired resistance. Safe and effective treatment modalities targeting leukemic clones that escape TKI therapy could hence be game changers in the professional management of these patients. Here, we aimed to investigate the efficacy of a novel therapeutic oligonucleotide of unconventional design, called ASP210, to reduce BCR-ABL1 mRNA levels in TKI-resistant CML cells, with the assumption of inducing their apoptosis. Imatinib- and dasatinib-resistant sublines of BCR-ABL1-positive MOLM-7 and CML-T1 cells were established and exposed to 0.25 and 2.5 µM ASP210 for 10 days. RT-qPCR showed a remarkable reduction of the target mRNA level by >99% after a single application. Cell viability was monitored daily by trypan blue staining. In response to the lack of driver oncoprotein BCR-ABL1, TKI-resistant CML cells underwent apoptosis regardless of the presence of the clinically relevant T315I mutation by day 5 after redosing with ASP210. The effect was selective for cancer cells, indicating a favorable safety profile for this therapeutic modality. Furthermore, the spontaneous uptake and high intracellular concentrations of ASP210 suggest its potential to be effective at relatively low doses. The present findings suggest that ASP210 is a promising therapeutic avenue for patients with CML who fail to respond to TKI therapy.NEW & NOTEWORTHY Effective treatment modalities targeting leukemic clones that escape tyrosine kinase inhibitor (TKI) therapy could be game changers in the professional management of patients displaying primary resistance, intolerance, or acquired resistance to TKIs. Although delivering authentic innovations today is more complex than ever, we developed a highly potent and safe oligonucleotide-based modality against BCR-ABL1 mRNA named ASP210 that effectively induces cell death in BCR-ABL1-positive TKI-resistant cells while sparing BCR-ABL1-negative healthy cells.


Asunto(s)
Apoptosis , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva , Oligonucleótidos , Inhibidores de Proteínas Quinasas , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/metabolismo , Línea Celular Tumoral , Oligonucleótidos/farmacología , Apoptosis/efectos de los fármacos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Dasatinib/farmacología , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
Neoplasma ; 70(3): 390-401, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37498075

RESUMEN

Silver nanoparticles (AgNPs) exhibit unique physicochemical properties, making these nanomaterials attractive for various medical applications. Among them, AgNPs have shown great potential in the treatment of cancer by inducing apoptosis in cancer cells, inhibiting tumor growth, and enhancing the efficacy of conventional cancer treatments such as chemotherapy and radiation therapy. Despite the promising therapeutical advantage of AgNPs, there are several challenges that need to be addressed. One of the most important is AgNPs' toxicity, which in case of treatment might be extended to non-cancerous cells and tissues. In our study, we therefore investigated the effects of spherical AgNPs with the silver core size of 10, 30, and 45 nm coated with polyacrylic acid (PAA-AgNPs) in an in vitro model using cancer (A549) and non-cancer (HEL299) cells. We estimated the impact of these nanoparticles on cell viability, cell proliferation, and cell actin cytoskeleton remodeling. Moreover, changes in the expression of TNFA, IL-10, FN1, and SOD1 mRNA induced by PAA-AgNPs were determined. Our results suggest that the smallest (10 nm) PAA-AgNPs are the most effective in apoptosis induction, however, they are also the most toxic from the three AgNPs types to both, cancer and non-cancer cells, while bigger (30 and 45 nm) PAA-AgNPs showed fewer undesirable effects in these pulmonary cells.


Asunto(s)
Neoplasias Pulmonares , Nanopartículas del Metal , Humanos , Plata/farmacología , Plata/química , Nanopartículas del Metal/química , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Apoptosis , Pulmón/metabolismo
3.
Int J Cancer ; 148(1): 128-139, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32621791

RESUMEN

Recently, we reported about exosomes possessing messenger RNA (mRNA) of suicide gene secreted from mesenchymal stem/stromal cells (MSCs) engineered to express the suicide gene-fused yeast cytosine deaminase::uracil phosphoribosyltransferase (yCD::UPRT). The yCD::UPRT-MSC exosomes are internalized by tumor cells and intracellularly convert prodrug 5-fluorocytosine (5-FC) to cytotoxic drug 5-fluorouracil (5-FU). Human tumor cells with the potential to metastasize release exosomes involved in the creation of a premetastatic niche at the predicted organs. We found that cancer cells stably transduced with yCD::UPRT gene by retrovirus infection released exosomes acting similarly like yCD::UPRT-MSC exosomes. Different types of tumor cells were transduced with the yCD::UPRT gene. The homogenous cell population of yCD::UPRT-transduced tumor cells expressed the yCD::UPRT suicide gene and secreted continuously exosomes with suicide gene mRNA in their cargo. All tumor cell suicide gene exosomes upon internalization into the recipient tumor cells induced the cell death by intracellular conversion of 5-FC to 5-FU and to 5-FUMP in a dose-dependent manner. Most of tumor cell-derived suicide gene exosomes were tumor tropic, in 5-FC presence they killed tumor cells but did not inhibit the growth of human skin fibroblast as well as DP-MSCs. Tumor cell-derived suicide gene exosomes home to their cells of origin and hold an exciting potential to become innovative specific therapy for tumors and potentially for metastases.


Asunto(s)
Antineoplásicos/uso terapéutico , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Profármacos/administración & dosificación , Animales , Antineoplásicos/farmacología , Ingeniería Celular/métodos , Línea Celular Tumoral , Medios de Cultivo Condicionados , Citosina Desaminasa/genética , Exosomas/genética , Flucitosina/administración & dosificación , Flucitosina/metabolismo , Fluorouracilo/metabolismo , Proteínas Fúngicas/genética , Vectores Genéticos/genética , Humanos , Ratones , Pentosiltransferasa/genética , Profármacos/metabolismo , Proteínas Recombinantes de Fusión/genética , Retroviridae/genética , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
BMC Cancer ; 18(1): 848, 2018 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143021

RESUMEN

BACKGROUND: Efficiency of colorectal carcinoma treatment by chemotherapy is diminished as the resistance develops over time in patients. The same holds true for 5-fluorouracil, the drug used in first line chemotherapy of colorectal carcinoma. METHODS: Chemoresistant derivative of HT-29 cells was prepared by long-term culturing in increasing concentration of 5-fluorouracil. Cells were characterized by viability assays, flow cytometry, gene expression arrays and kinetic imaging. Immunomagnetic separation was used for isolation of subpopulations positive for cancer stem cells-related surface markers. Aldehyde dehydrogenase expression was attenuated by siRNA. In vivo studies were performed on SCID/bg mice. RESULTS: The prepared chemoresistant cell line labeled as HT-29/EGFP/FUR is assigned with different morphology, decreased proliferation rate and 135-fold increased IC50 value for 5-fluorouracil in comparison to parental counterparts HT-29/EGFP. The capability of chemoresistant cells to form tumor xenografts, when injected subcutaneously into SCID/bg mice, was strongly compromised, however, they formed distant metastases in mouse lungs spontaneously. Derived cells preserved their resistance in vitro and in vivo even without the 5-fluorouracil selection pressure. More importantly, they were resistant to cisplatin, oxaliplatin and cyclophosphamide exhibiting high cross-resistance along with alterations in expression of cancer-stem cell markers such as CD133, CD166, CD24, CD26, CXCR4, CD271 and CD274. We also detected increased aldehyde dehydrogenase (ALDH) activity associated with overexpression of specific ALDH isoform 1A3. Its inhibition by siRNA approach partially sensitized cells to various agents, thus linking for the first time the ALDH1A3 and chemoresistance in colorectal cancer. CONCLUSION: Our study demonstrated that acquired chemoresistance goes along with metastatic and migratory phenotype and can be accompanied with increased activity of aldehyde dehydrogenase. We describe here the valuable model to study molecular link between resistance to chemotherapy and metastatic dissemination.


Asunto(s)
Aldehído Oxidorreductasas/genética , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/genética , Adulto , Anciano , Animales , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , ARN Interferente Pequeño , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mutagenesis ; 32(1): 193-202, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27658822

RESUMEN

Nowadays engineered nanomaterials (ENMs) are increasingly used in a wide range of commercial products and biomedical applications. Despite this, the knowledge of human potential health risk as well as comprehensive biological and toxicological information is still limited. We have investigated the capacity of two frequently used metallic ENMs, nanosilver and magnetite nanoparticles (MNPs), to induce thymidine kinase (Tk +/-) mutations in L5178Y mouse lymphoma cells and transformed foci in Bhas 42 cells. Two types of nanosilver, spherical nanoparticles (AgNM300) and fibrous (AgNM302) nanorods/wires, and MNPs differing in surface modifications [MNPs coated with sodium oleate (SO-MNPs), MNPs coated with SO + polyethylene glycol (SO-PEG-MNPs) and MNPs coated with SO + PEG + poly(lactide-co-glycolic acid) SO-PEG-PLGA-MNPs] were included in this study. Spherical AgNM300 showed neither mutagenic nor carcinogenic potential. In contrast, silver nanorods/wires (AgNM302) increased significantly the number of both gene mutations and transformed foci compared with the control (untreated) cells. Under the same treatment conditions, neither SO-MNPs nor SO-PEG-PLGA-MNPs increased the mutant frequency compared with control cells though an equivocal mutagenic effect was estimated for SO-PEG-MNPs. Although SO-MNPs and SO-PEG-MNPs did not show any carcinogenic potential, SO-PEG-PLGA-MNPs increased concentration dependently the number of transformed foci in Bhas 42 cells compared with the control cells. Our results revealed that fibrous shape underlies the mutagenic and carcinogenic potential of nanosilver while surface chemistry affects the biosafety of MNPs. Considering that both nanosilver and MNPs are prospective ENMs for biomedical applications, further toxicological evaluations are warranted to assess comprehensively the biosafety of these nanomaterials.


Asunto(s)
Nanopartículas del Metal/toxicidad , Mutación , Plata/toxicidad , Timidina Quinasa/efectos de los fármacos , Animales , Carcinógenos/farmacología , Carcinógenos/toxicidad , Compuestos Férricos/farmacología , Compuestos Férricos/toxicidad , Nanopartículas del Metal/química , Ratones , Pruebas de Mutagenicidad , Mutágenos/farmacología , Mutágenos/toxicidad , Plata/farmacología , Timidina Quinasa/genética
6.
Nanomedicine ; 13(1): 69-80, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27593490

RESUMEN

Iron oxide nanoparticles are one of the most promising types of nanoparticles for biomedical applications, primarily in the context of nanomedicine-based diagnostics and therapy; hence, great attention should be paid to their bio-safety. Here, we investigate the ability of surface-modified magnetite nanoparticles (MNPs) to produce chromosome damage in human alveolar A549 cells. Compared to control cells, all the applied MNPs increased the level of micronuclei moderately but did not cause structural chromosomal aberrations in exposed cells. A rise in endoreplication, polyploid and multinuclear cells along with disruption of tubulin filaments, downregulation of Aurora protein kinases and p53 protein activation indicated the capacity of these MNPs to impair the chromosomal passenger complex and/or centrosome maturation. We suppose that surface-modified MNPs may act as aneugen-like spindle poisons via interference with tubulin polymerization. Further studies on experimental animals revealing mechanisms of therapeutic-aimed MNPs are required to confirm their suitability as potential anti-cancer drugs.


Asunto(s)
Aneugénicos/farmacología , Antineoplásicos/farmacología , Nanopartículas de Magnetita/química , Huso Acromático/efectos de los fármacos , Células A549 , Daño del ADN , Humanos , Micronúcleos con Defecto Cromosómico , Nanomedicina , Tubulina (Proteína)/efectos de los fármacos
7.
Bioconjug Chem ; 27(10): 2558-2574, 2016 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-27602782

RESUMEN

Cationic colloidal gold nanorods (GNRs) have a great potential as a theranostic tool for diverse medical applications. GNRs' properties such as cellular internalization and stability are determined by physicochemical characteristics of their surface coating. GNRs modified by (16-mercaptohexadecyl)trimethylammonium bromide (MTAB), MTABGNRs, show excellent cellular uptake. Despite their promise for biomedicine, however, relatively little is known about the cellular pathways that facilitate the uptake of GNRs, their subcellular fate and intracellular persistence. Here we studied the mechanism of cellular internalization and long-term fate of GNRs coated with MTAB, for which the synthesis was optimized to give higher yield, in various human cell types including normal diploid versus cancerous, and dividing versus nondividing (senescent) cells. The process of MTABGNRs internalization into their final destination in lysosomes proceeds in two steps: (1) fast passive adhesion to cell membrane mediated by sulfated proteoglycans occurring within minutes and (2) slower active transmembrane and intracellular transport of individual nanorods via clathrin-mediated endocytosis and of aggregated nanorods via macropinocytosis. The expression of sulfated proteoglycans was the major factor determining the extent of uptake by the respective cell types. Upon uptake into proliferating cells, MTABGNRs were diluted equally and relatively rapidly into daughter cells; however, in nondividing/senescent cells the loss of MTABGNRs was gradual and very modest, attributable mainly to exocytosis. Exocytosed MTABGNRs can again be internalized. These findings broaden our knowledge about cellular uptake of gold nanorods, a crucial prerequisite for future successful engineering of nanoparticles for biomedical applications such as photothermal cancer therapy or elimination of senescent cells as part of the emerging rejuvenation approach.


Asunto(s)
Exocitosis , Oro/química , Oro/farmacocinética , Nanotubos/química , Polilisina/química , Polilisina/farmacocinética , Compuestos de Amonio Cuaternario/química , Compuestos de Sulfhidrilo/química , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Medios de Cultivo , Estabilidad de Medicamentos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Citometría de Flujo , Humanos , Lisosomas/efectos de los fármacos , Microscopía Confocal , Microscopía Electrónica de Rastreo , Nanotubos/análisis , Proteoglicanos/química , Proteoglicanos/metabolismo , Compuestos de Amonio Cuaternario/síntesis química
8.
Genomics ; 106(4): 204-13, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26200819

RESUMEN

Type 2 diabetes induces pathophysiological changes in the liver. The aim of this study was to identify differently expressed genes in the livers of male and female ZSF1 rats (ZDFxSHHF-hybrid, generation F1), a model for type 2 diabetes. Gene expression was investigated using next-generation sequencing (NGS). Selected candidate genes were verified by real-time PCR in the livers of obese and lean rats. 103 sex-different genes, associated to pathways "response to chemical stimulus", "lipid metabolism", and "response to organic substance", were identified. Male-specific genes were involved in hepatic metabolism, detoxification, and secretion, e.g. cytochrome P450 2c11 (Cyp2c11), Cyp4a2, glutathione S-transferases mu 2 (Gstm2), and Slc22a8 (organic anion transporter 3, Oat3). Most female-specific genes were associated to lipid metabolism (e.g. glycerol-3-phosphate acyltransferase 1, Gpam) or glycolysis (e.g. glucokinase, Gck). Our data suggest the necessity to pay attention to sex- and diabetes-dependent changes in pre-clinical testing of hepatic metabolized and secreted drugs.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Hígado/metabolismo , Masculino , Ratas , Ratas Zucker
9.
Int J Nanomedicine ; 19: 4103-4120, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736658

RESUMEN

Introduction: Gold nanoparticles are promising candidates as vehicles for drug delivery systems and could be developed into effective anticancer treatments. However, concerns about their safety need to be identified, addressed, and satisfactorily answered. Although gold nanoparticles are considered biocompatible and nontoxic, most of the toxicology evidence originates from in vitro studies, which may not reflect the responses in complex living organisms. Methods: We used an animal model to study the long-term effects of 20 nm spherical AuNPs coated with bovine serum albumin. Mice received a 1 mg/kg single intravenous dose of nanoparticles, and the biodistribution and accumulation, as well as the organ changes caused by the nanoparticles, were characterized in the liver, spleen, and kidneys during 120 days. Results: The amount of nanoparticles in the organs remained high at 120 days compared with day 1, showing a 39% reduction in the liver, a 53% increase in the spleen, and a 150% increase in the kidneys. The biological effects of chronic nanoparticle exposure were associated with early inflammatory and fibrotic responses in the organs and were more pronounced in the kidneys, despite a negligible amount of nanoparticles found in renal tissues. Conclusion: Our data suggest, that although AuNPs belong to the safest nanomaterial platforms nowadays, due to their slow tissue elimination leading to long-term accumulation in the biological systems, they may induce toxic responses in the vital organs, and so understanding of their long-term biological impact is important to consider their potential therapeutic applications.


Asunto(s)
Oro , Riñón , Hígado , Nanopartículas del Metal , Albúmina Sérica Bovina , Bazo , Animales , Masculino , Ratones , Oro/química , Oro/farmacocinética , Oro/toxicidad , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Nanopartículas del Metal/administración & dosificación , Tamaño de la Partícula , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/farmacocinética , Bazo/efectos de los fármacos , Distribución Tisular
10.
Heliyon ; 8(11): e11595, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36419669

RESUMEN

Fundamental studies investigating the biological effects induced by nanoparticles (NPs) explicitly require the correct assessment of their intracellular concentration. Ultrasensitive atomic absorption spectroscopy (AAS) is perceived as one of the gold standard methods for quantifying internalized NPs. Besides its limitation to metal-based NPs though, AAS also requires specific infrastructure and tedious sample preparation and handling, making it time-consuming and cost-intensive. Herein we report on a reliable, rapid, and affordable alternative to AAS - plate reader spectroscopy (PRS), which offers an accessible option for everyday laboratory use without sophisticated instrumentation. Our results demonstrate, that following a proper methodological approach, data on intracellular concentration of NPs obtained by PRS are fully comparable to AAS results. Specifically, the intracellular concentration of magnetite NPs coated with sodium oleate and bovine serum albumin in human alveolar A549 cells was assessed by PRS and AAS in parallel, with a remarkable correlation coefficient of R = 0.9914.

11.
Viruses ; 14(4)2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-35458415

RESUMEN

In only two years, the coronavirus disease 2019 (COVID-19) pandemic has had a devastating effect on public health all over the world and caused irreparable economic damage across all countries. Due to the limited therapeutic management of COVID-19 and the lack of tailor-made antiviral agents, finding new methods to combat this viral illness is now a priority. Herein, we report on a specific oligonucleotide-based RNA inhibitor targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It displayed remarkable spontaneous cellular uptake, >94% efficiency in reducing RNA-dependent RNA polymerase (RdRp) RNA levels in transfected lung cell lines, and >98% efficiency in reducing SARS-CoV-2 RNA levels in samples from patients hospitalized with COVID-19 following a single application.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Oligonucleótidos , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéutico , Humanos , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , SARS-CoV-2/genética
12.
Biomed Pharmacother ; 147: 112662, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35091237

RESUMEN

Acquired drug resistance and metastasis in breast cancer (BC) are coupled with epigenetic deregulation of gene expression. Epigenetic drugs, aiming to reverse these aberrant transcriptional patterns and sensitize cancer cells to other therapies, provide a new treatment strategy for drug-resistant tumors. Here we investigated the ability of DNA methyltransferase (DNMT) inhibitor decitabine (DAC) to increase the sensitivity of BC cells to anthracycline antibiotic doxorubicin (DOX). Three cell lines representing different molecular BC subtypes, JIMT-1, MDA-MB-231 and T-47D, were used to evaluate the synergy of sequential DAC + DOX treatment in vitro. The cytotoxicity, genotoxicity, apoptosis, and migration capacity were tested in 2D and 3D cultures. Moreover, genome-wide DNA methylation and transcriptomic analyses were employed to understand the differences underlying DAC responsiveness. The ability of DAC to sensitize trastuzumab-resistant HER2-positive JIMT-1 cells to DOX was examined in vivo in an orthotopic xenograft mouse model. DAC and DOX synergistic effect was identified in all tested cell lines, with JIMT-1 cells being most sensitive to DAC. Based on the whole-genome data, we assume that the aggressive behavior of JIMT-1 cells can be related to the enrichment of epithelial-to-mesenchymal transition and stemness-associated pathways in this cell line. The four-week DAC + DOX sequential administration significantly reduced the tumor growth, DNMT1 expression, and global DNA methylation in xenograft tissues. The efficacy of combination therapy was comparable to effect of pegylated liposomal DOX, used exclusively for the treatment of metastatic BC. This work demonstrates the potential of epigenetic drugs to modulate cancer cells' sensitivity to other forms of anticancer therapy.


Asunto(s)
Neoplasias de la Mama/patología , ADN (Citosina-5-)-Metiltransferasa 1/antagonistas & inhibidores , Decitabina/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/análogos & derivados , Transición Epitelial-Mesenquimal , Femenino , Genes erbB-2/genética , Humanos , Concentración 50 Inhibidora , Ratones , Ratones SCID , Pruebas de Mutagenicidad , Polietilenglicoles/farmacología , Distribución Aleatoria , Trastuzumab/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Am J Pathol ; 176(5): 2150-62, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20363918

RESUMEN

Chronic inflammation and fibrosis are the leading causes of chronic allograft failure. The nuclear receptor peroxisome proliferator-activated receptor (PPAR)gamma is a transcription factor known to have antidiabetogenic and immune effects, and PPARgamma forms obligate heterodimers with the retinoid X receptor (RXR). We have reported that a retinoic acid (RAR)/RXR-agonist can potently influence the course of renal chronic allograft dysfunction. In this study, in a Fischer to Lewis rat renal transplantation model, administration of the PPARgamma-agonist, rosiglitazone, independent of dose (3 or 30 mg/kgBW/day), lowered serum creatinine, albuminuria, and chronic allograft damage with a chronic vascular damage score as follows: 35.0 +/- 5.8 (controls) vs. 8.1 +/- 2.4 (low dose-Rosi; P < 0.05); chronic tubulointerstitial damage score: 13.6 +/- 1.8 (controls) vs. 2.6 +/- 0.4 (low dose-Rosi; P < 0.01). The deposition of extracellular matrix proteins (collagen, fibronectin, decorin) was strikingly lower. The expression of transforming growth factor-beta1 was inhibited, whereas that of bone morphogenic protein-7 (BMP-7) was increased. Intragraft mononuclear cells and activated fibroblast numbers were reduced by 50%. In addition, the migratory and proliferative activity of these cells was significantly inhibited in vitro. PPARgamma activation diminished the number of cells expressing the proinflammatory and fibrogenic proteoglycan biglycan. In macrophages its secretion was blocked by rosiglitazone in a predominantly PPARgamma-dependent manner. The combination of PPARgamma- and RAR/RXR-agonists resulted in additive effects in the inhibition of fibrosis. In summary, PPARgamma activation was potently immunosuppressive and antifibrotic in kidney allografts, and these effects were enhanced by a RAR/RXR-agonist.


Asunto(s)
Regulación de la Expresión Génica , Trasplante de Riñón/métodos , PPAR gamma/metabolismo , Animales , Biglicano/metabolismo , Presión Sanguínea , Fibroblastos/metabolismo , Inmunohistoquímica/métodos , Inflamación , Riñón/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratas , Rosiglitazona , Tiazolidinedionas/farmacología
14.
J Immunol ; 181(4): 2831-45, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684975

RESUMEN

The calcineurin inhibitor (CNI)-induced renal fibrosis is attributed to an exaggerated deposition of extracellular matrix, which is mainly due to an increased expression of TGFbeta. Herein we demonstrate that the CNI cyclosporin A and tacrolimus (FK506), independent of TGFbeta synthesis, rapidly activate TGFbeta/Smad signaling in cultured mesangial cells and in whole kidney samples from CNI-treated rats. By EMSA, we demonstrate increased DNA binding of Smad-2, -3, and -4 to a cognate Smad-binding promoter element (SBE) accompanied by CNI-triggered activation of Smad-dependent expression of tissue inhibitor of metalloprotease-1 (TIMP-1) and connective tissue growth factor. Using an activin receptor-like kinase-5 (ALK-5) inhibitor and by small interfering RNA we depict a critical involvement of both types of TGFbeta receptors in CNI-triggered Smad signaling and fibrogenic gene expression, respectively. Mechanistically, CNI cause a rapid activation of latent TGFbeta, which is prevented in the presence of the antioxidant N-acetyl cysteine. A convergent activation of p38 MAPK is indicated by the partial blockade of CNI-induced Smad-2 activation by SB203580; conversely, both TGFbeta-RII and TGFbeta are critically involved in p38 MAPK activation by CNI. Activation of both signaling pathways is similarly triggered by reactive oxygen species. Finally, we show that neutralization of TGFbeta markedly reduced the CNI-dependent Smad activation in vitro and in vivo. Collectively, this study demonstrates that CNI via reactive oxygen species generation activate latent TGFbeta and thereby initiate the canonical Smad pathway by simultaneously activating p38 MAPK, which both synergistically induce Smad-driven gene expression.


Asunto(s)
Inhibidores de la Calcineurina , Ciclosporina/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/efectos de los fármacos , Tacrolimus/administración & dosificación , Animales , Células Cultivadas , Fibrosis , Humanos , Inyecciones Intraperitoneales , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/enzimología , Células Mesangiales/patología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Ratas Wistar , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal/inmunología , Proteína Smad2/metabolismo , Proteína Smad2/fisiología , Tacrolimus/farmacología , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/metabolismo
15.
RSC Adv ; 10(40): 23916-23929, 2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35517346

RESUMEN

Drug-induced nephrotoxicity is a frequent adverse event and a dose-limiting factor in patient treatment and is a leading cause of prospective drug attrition during pharmaceutical development. Despite the obvious benefits of nanotherapeutics in healthcare strategies, the clearance of imaging agents and nanocarriers from the body following their therapeutic or diagnostic application generates concerns about their safety for human health. Considering the potency of nanoparticles and their massive utilization in biomedicine the impact of magnetic nanoparticles (MNPs) on cells forming the filtration apparatus of the kidney was studied. Using primary mouse renal glomerular podocytes and mesangial cells, we investigated their response to exposure to magnetic nanoparticles coated with polyethylene glycol and bovine serum albumin. Cultured podocytes were more sensitive to MNPs than mesangial cells displaying signs of cell damage and stronger inflammatory response. Both types of MNPs induced the remodeling of actin fibers, affected the cell shape and triggered expression of inflammatory cytokines TNFα and IL-6 in podocytes. On the other hand, iNOS was induced in both renal cell types but only by MNPs with a polyethylene glycol coating. Our results have revealed that the type of cell and the type of nanoparticle coating might be the strongest determinants of cellular response toward nanoparticle exposure. Differences in susceptibility of cells to MNPs might be evident also between neighboring renal cell subpopulations integrally forming functional sub-units of this organ.

16.
Food Chem Toxicol ; 136: 110954, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31707033

RESUMEN

Due to the growing number of applications of cadmium oxide nanoparticles (CdO NPs), there is a concern about their potential deleterious effects. The objective of our study was to investigate the effect of CdO NPs on the immune response, renal and intestine oxidative stress, blood antioxidant defence, renal fibrotic response, bone density and mineral content. Six-week-old female ICR mice were exposed to CdO NPs for 6 weeks by inhalation (particle size: 9.82 nm, mass concentration: 31.7 µg CdO/m3, total deposited dose: 0.195 µg CdO/g body weight). CdO NPs increased percentage of thymus CD3e+CD8a+ cells and moderately enhanced splenocyte proliferation and production of cytokines and chemokines. CdO NPs elevated pro-fibrotic factors (TGF-ß2, α-SMA and collagen I) in the kidney, and concentrations of AGEs in the intestine. The ratio of GSH and GSSG in blood was slightly reduced. Exposure to CdO NPs resulted in 10-fold higher Cd concentration in tibia bones. No differences were found in bone mass density, mineral content, bone area values, bone concentrations of Ca, P, Mg and Ca/P ratio. Our findings indicate stimulation of immune/inflammatory response, oxidative stress in the intestine, starting fibrotic response in kidneys and accumulation of CdO NPs in bones of mice.


Asunto(s)
Compuestos de Cadmio/toxicidad , Fibrosis/inducido químicamente , Inmunidad Celular/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Estrés Oxidativo/efectos de los fármacos , Óxidos/toxicidad , Tibia/efectos de los fármacos , Administración por Inhalación , Animales , Compuestos de Cadmio/administración & dosificación , Citocinas/metabolismo , Femenino , Intestinos/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/patología , Ganglios Linfáticos/efectos de los fármacos , Nanopartículas del Metal/administración & dosificación , Ratones Endogámicos ICR , Óxidos/administración & dosificación , Bazo/efectos de los fármacos , Timo/efectos de los fármacos
17.
Kidney Int ; 76(8): 857-67, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19657322

RESUMEN

Transforming growth factor-beta2 (TGF-beta2) stimulates the expression of pro-fibrotic connective tissue growth factor (CTGF) during the course of renal disease. Because sphingosine kinase-1 (SK-1) activity is also upregulated by TGF-beta, we studied its effect on CTGF expression and on the development of renal fibrosis. When TGF-beta2 was added to an immortalized human podocyte cell line we found that it activated the promoter of SK-1, resulting in upregulation of its mRNA and protein expression. Further, depletion of SK-1 by small interfering RNA or its pharmacological inhibition led to accelerated CTGF expression in the podocytes. Over-expression of SK-1 reduced CTGF induction, an effect mediated by intracellular sphingosine-1-phosphate. In vivo, SK-1 expression was also increased in the podocytes of kidney sections of patients with diabetic nephropathy when compared to normal sections of kidney obtained from patients with renal cancer. Similarly, in a mouse model of streptozotocin-induced diabetic nephropathy, SK-1 and CTGF were upregulated in podocytes. In SK-1 deficient mice, exacerbation of disease was detected by increased albuminuria and CTGF expression when compared to wild-type mice. Thus, SK-1 activity has a protective role in the fibrotic process and its deletion or inhibition aggravates fibrotic disease.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/enzimología , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Podocitos/enzimología , Esfingosina/análogos & derivados , Factor de Crecimiento Transformador beta2/metabolismo , Albuminuria/enzimología , Albuminuria/etiología , Animales , Línea Celular , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Regulación hacia Abajo , Fibrosis , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Podocitos/efectos de los fármacos , Podocitos/patología , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Esfingosina/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
18.
J Clin Invest ; 115(8): 2223-33, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16025156

RESUMEN

Biglycan, a small leucine-rich proteoglycan, is a ubiquitous ECM component; however, its biological role has not been elucidated in detail. Here we show that biglycan acts in macrophages as an endogenous ligand of TLR4 and TLR2, which mediate innate immunity, leading to rapid activation of p38, ERK, and NF-kappaB and thereby stimulating the expression of TNF-alpha and macrophage inflammatory protein-2 (MIP-2). In agreement, the stimulatory effects of biglycan are significantly reduced in TLR4-mutant (TLR4-M), TLR2-/-, and myeloid differentiation factor 88-/- (MyD88-/-) macrophages and completely abolished in TLR2-/-/TLR4-M macrophages. Biglycan-null mice have a considerable survival benefit in LPS- or zymosan-induced sepsis due to lower levels of circulating TNF-alpha and reduced infiltration of mononuclear cells in the lung, which cause less end-organ damage. Importantly, when stimulated by LPS-induced proinflammatory factors, macrophages themselves are able to synthesize biglycan. Thus, biglycan, upon release from the ECM or from macrophages, can boost inflammation by signaling through TLR4 and TLR2, thereby enhancing the synthesis of TNF-alpha and MIP-2. Our results provide evidence for what is, to our knowledge, a novel role of the matrix component biglycan as a signaling molecule and a crucial proinflammatory factor. These findings are potentially relevant for the development of new strategies in the treatment of sepsis.


Asunto(s)
Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanos/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Biglicano , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide , FN-kappa B/metabolismo , Proteoglicanos/genética , Receptores de Superficie Celular/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Sepsis/inducido químicamente , Sepsis/metabolismo , Sepsis/patología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like , Factor de Necrosis Tumoral alfa/biosíntesis , Zimosan/toxicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Biomaterials ; 154: 275-290, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29149721

RESUMEN

The exceptionally high cellular uptake of gold nanorods (GNRs) bearing cationic surfactants makes them a promising tool for biomedical applications. Given the known specific toxic and stress effects of some preparations of cationic nanoparticles, the purpose of this study was to evaluate, in an in vitro and in vivo in mouse, the potential harmful effects of GNRs coated with (16-mercaptohexadecyl)trimethylammonium bromide (MTABGNRs). Interestingly, even after cellular accumulation of high amounts of MTABGNRs sufficient for induction of photothermal effect, no genotoxicity (even after longer-term accumulation), induction of autophagy, destabilization of lysosomes (dominant organelles of their cellular destination), alterations of actin cytoskeleton, or in cell migration could be detected in vitro. In vivo, after intravenous administration, the majority of GNRs accumulated in mouse spleen followed by lungs and liver. Microscopic examination of the blood and spleen showed that GNRs interacted with white blood cells (mononuclear and polymorphonuclear leukocytes) and thrombocytes, and were delivered to the spleen red pulp mainly as GNR-thrombocyte complexes. Importantly, no acute toxic effects of MTABGNRs administered as 10 or 50 µg of gold per mice, as well as no pathological changes after their high accumulation in the spleen were observed, indicating good tolerance of MTABGNRs by living systems.


Asunto(s)
Oro/metabolismo , Nanotubos/química , Compuestos de Amonio Cuaternario/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Autofagia/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/patología , Plaquetas/ultraestructura , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Daño del ADN , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Mutágenos/toxicidad , Nanotubos/toxicidad , Nanotubos/ultraestructura , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Bazo/efectos de los fármacos , Bazo/patología , Distribución Tisular
20.
Mater Sci Eng C Mater Biol Appl ; 70(Pt 1): 161-168, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27770877

RESUMEN

Comprehensive characterization of nanoparticles associated with investigation of their cellular uptake creates the basis on which fundamental in vitro and in vivo studies can be built. In this work, a complex analysis of various surface-modified magnetite nanoparticles in biologically relevant environment is reported and the promotion of incorrect characterization into the results obtained from model biological experiments leading to false conclusions is demonstrated. Via a bottom-up approach from particle characterization by DLS towards interpretation of biological data based on cellular uptake, this work draws attention to the systematic propagation of errors stemming from inaccurate determination of input parameters for DLS, improper selection of particle size distribution, inadequate sampling, unknown colloidal behavior and the omission of fraction of particles complying with the internalization threshold. In addition, cellular uptake depending on the number of treated cells is shown. The definition of cellular uptake efficacy reflecting the size distribution of particles beside their absolute internalization is postulated.


Asunto(s)
Fenómenos Químicos , Espacio Intracelular/metabolismo , Nanopartículas de Magnetita/química , Células A549 , Coloides/química , Dispersión Dinámica de Luz , Endocitosis , Humanos , Hidrodinámica , Nanopartículas de Magnetita/ultraestructura , Tamaño de la Partícula , Soluciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA