Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Nanomedicine ; 19: 4103-4120, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38736658

RESUMO

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.


Assuntos
Ouro , Rim , Fígado , Nanopartículas Metálicas , Soroalbumina Bovina , Baço , Animais , Ouro/química , Ouro/farmacocinética , Ouro/toxicidade , Ouro/administração & dosagem , Nanopartículas Metálicas/química , Nanopartículas Metálicas/toxicidade , Nanopartículas Metálicas/administração & dosagem , Baço/efeitos dos fármacos , Soroalbumina Bovina/química , Soroalbumina Bovina/farmacocinética , Rim/efeitos dos fármacos , Rim/metabolismo , Distribuição Tecidual , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Masculino , Tamanho da Partícula
2.
Neoplasma ; 70(3): 390-401, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37498075

RESUMO

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.


Assuntos
Neoplasias Pulmonares , Nanopartículas Metálicas , Humanos , Prata/farmacologia , Prata/química , Nanopartículas Metálicas/química , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Apoptose , Pulmão/metabolismo
3.
Heliyon ; 8(11): e11595, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36419669

RESUMO

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.

4.
Viruses ; 14(4)2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35458415

RESUMO

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.


Assuntos
Tratamento Farmacológico da COVID-19 , Oligonucleotídeos , SARS-CoV-2 , Antivirais/farmacologia , Antivirais/uso terapêutico , Humanos , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , SARS-CoV-2/genética
5.
Biomed Pharmacother ; 147: 112662, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35091237

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , DNA (Citosina-5-)-Metiltransferase 1/antagonistas & inibidores , Decitabina/farmacologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/análogos & derivados , Transição Epitelial-Mesenquimal , Feminino , Genes erbB-2/genética , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos SCID , Testes de Mutagenicidade , Polietilenoglicóis/farmacologia , Distribuição Aleatória , Trastuzumab/farmacologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Cancer ; 148(1): 128-139, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32621791

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Genes Transgênicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Pró-Fármacos/administração & dosagem , Animais , Antineoplásicos/farmacologia , Engenharia Celular/métodos , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Citosina Desaminase/genética , Exossomos/genética , Flucitosina/administração & dosagem , Flucitosina/metabolismo , Fluoruracila/metabolismo , Proteínas Fúngicas/genética , Vetores Genéticos/genética , Humanos , Camundongos , Pentosiltransferases/genética , Pró-Fármacos/metabolismo , Proteínas Recombinantes de Fusão/genética , Retroviridae/genética , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
RSC Adv ; 10(40): 23916-23929, 2020 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-35517346

RESUMO

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.

8.
Food Chem Toxicol ; 136: 110954, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31707033

RESUMO

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.


Assuntos
Compostos de Cádmio/toxicidade , Fibrose/induzido quimicamente , Imunidade Celular/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Óxidos/toxicidade , Tíbia/efeitos dos fármacos , Administração por Inalação , Animais , Compostos de Cádmio/administração & dosagem , Citocinas/metabolismo , Feminino , Intestinos/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/patologia , Linfonodos/efeitos dos fármacos , Nanopartículas Metálicas/administração & dosagem , Camundongos Endogâmicos ICR , Óxidos/administração & dosagem , Baço/efeitos dos fármacos , Timo/efeitos dos fármacos
9.
BMC Cancer ; 18(1): 848, 2018 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143021

RESUMO

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.


Assuntos
Aldeído Oxirredutases/genética , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Adulto , Idoso , Animais , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica , RNA Interferente Pequeno , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Biomaterials ; 154: 275-290, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29149721

RESUMO

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.


Assuntos
Ouro/metabolismo , Nanotubos/química , Compostos de Amônio Quaternário/metabolismo , Compostos de Sulfidrila/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Autofagia/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Plaquetas/ultraestrutura , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Dano ao DNA , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mutagênicos/toxicidade , Nanotubos/toxicidade , Nanotubos/ultraestrutura , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Baço/efeitos dos fármacos , Baço/patologia , Distribuição Tecidual
11.
Nanomedicine ; 13(1): 69-80, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27593490

RESUMO

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.


Assuntos
Aneugênicos/farmacologia , Antineoplásicos/farmacologia , Nanopartículas de Magnetita/química , Fuso Acromático/efeitos dos fármacos , Células A549 , Dano ao DNA , Humanos , Micronúcleos com Defeito Cromossômico , Nanomedicina , Tubulina (Proteína)/efeitos dos fármacos
12.
Mater Sci Eng C Mater Biol Appl ; 70(Pt 1): 161-168, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770877

RESUMO

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.


Assuntos
Fenômenos Químicos , Espaço Intracelular/metabolismo , Nanopartículas de Magnetita/química , Células A549 , Coloides/química , Difusão Dinâmica da Luz , Endocitose , Humanos , Hidrodinâmica , Nanopartículas de Magnetita/ultraestrutura , Tamanho da Partícula , Soluções
13.
Mutagenesis ; 32(1): 193-202, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27658822

RESUMO

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.


Assuntos
Nanopartículas Metálicas/toxicidade , Mutação , Prata/toxicidade , Timidina Quinase/efeitos dos fármacos , Animais , Carcinógenos/farmacologia , Carcinógenos/toxicidade , Compostos Férricos/farmacologia , Compostos Férricos/toxicidade , Nanopartículas Metálicas/química , Camundongos , Testes de Mutagenicidade , Mutagênicos/farmacologia , Mutagênicos/toxicidade , Prata/farmacologia , Timidina Quinase/genética
14.
Bioconjug Chem ; 27(10): 2558-2574, 2016 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-27602782

RESUMO

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.


Assuntos
Exocitose , Ouro/química , Ouro/farmacocinética , Nanotubos/química , Polilisina/química , Polilisina/farmacocinética , Compostos de Amônio Quaternário/química , Compostos de Sulfidrila/química , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Técnicas de Química Sintética , Meios de Cultura , Estabilidade de Medicamentos , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Citometria de Fluxo , Humanos , Lisossomos/efeitos dos fármacos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Nanotubos/análise , Proteoglicanas/química , Proteoglicanas/metabolismo , Compostos de Amônio Quaternário/síntese química
15.
Endocrinology ; 157(10): 4032-4040, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27399878

RESUMO

1α,25-dihydroxy-vitamin D3 (1,25D) exerts protective effects in the vascular system and promotes myeloid cell differentiation, which are important sources of reactive oxygen species. Given that myeloid cell reactive oxygen species derives from Nox-family NADPH oxidases, we hypothesized that this enzyme family contributes to the beneficial effects of 1,25D on vascular regeneration. The function of Nox enzymes in this context was studied in the murine carotid artery electric injury regeneration model. Male mice were treated with daily injections of 1,25D (100 ng/kg · d) for 5 days and carotid injury was induced after 3 days. After injury, 1,25D increased the expression of Nox2 in the carotid artery. As determined by Evans blue staining on day 6, 1,25D improved vascular regeneration in a Nox2-dependent manner. Healing was lost in mice knockout for Nox2, but not in Nox1 or Nox4, knockout mice. Tissue specific knockouts demonstrated that the myeloid, but not the endothelial Nox2, was required for this effect. Mechanistically, the combination of injury and 1,25D induced the mobilization of angiogenic myeloid cells (AMCs) and increased the vascular expression of the cytokine stem cell derived factor (SDF)1, which attracts AMCs to the site of injury. Vitamin D in a Nox2-dependent manner activated MAPKs, and these are known to contribute to SDF1 induction. Accordingly, SDF1 induction was lost after deletion of Nox2. By inducing SDF1 and enhancing the number of AMCs, VitD3 is a novel approach to promote vascular repair.


Assuntos
Calcitriol/uso terapêutico , Lesões das Artérias Carótidas/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Regeneração/efeitos dos fármacos , Animais , Calcitriol/farmacologia , Lesões das Artérias Carótidas/enzimologia , Quimiocina CXCL12/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Masculino , Camundongos Endogâmicos C57BL , NADPH Oxidase 2 , Espécies Reativas de Oxigênio/metabolismo , Remodelação Vascular
16.
Genomics ; 106(4): 204-13, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26200819

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2/genética , Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Fígado/metabolismo , Masculino , Ratos , Ratos Zucker
17.
J Diabetes Res ; 2015: 483238, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25710042

RESUMO

The aim of this study was to identify sex-dependent expression of renal transporter mRNA in lean and obese Zucker spontaneously hypertensive fatty (ZSF1) rats and to investigate the interaction of the most altered transporter, organic anion transporter 2 (Oat2), with diabetes-relevant metabolites and drugs. Higher incidence of glomerulosclerosis, tubulointerstitial fibrosis, and protein casts in Bowman's space and tubular lumen was detected by PAS staining in obese male compared to female ZSF1 rats. Real-time PCR on RNA isolated from kidney cortex revealed that Sglt1-2, Oat1-3, and Oct1 were higher expressed in kidneys of lean females. Oct2 and Mrp2 were higher expressed in obese males. Renal mRNA levels of transporters were reduced with diabetic nephropathy in females and the expression of transcription factors Hnf1ß and Hnf4α in both sexes. The highest difference between lean and obese ZSF1 rats was found for Oat2. Therefore, we have tested the interaction of human OAT2 with various substances using tritium-labeled cGMP. Human OAT2 showed no interaction with diabetes-related metabolites, diabetic drugs, and ACE-inhibitors. However, OAT2-dependent uptake of cGMP was inhibited by furosemide. The strongly decreased expression of Oat2 and other transporters in female diabetic ZSF1 rats could possibly impair renal drug excretion, for example, of furosemide.


Assuntos
Ratos Zucker/genética , Fatores Sexuais , Fatores de Transcrição/metabolismo , Animais , Transporte Biológico , Pressão Sanguínea , GMP Cíclico/metabolismo , Diabetes Mellitus/metabolismo , Nefropatias Diabéticas/metabolismo , Feminino , Furosemida/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células HEK293 , Humanos , Rim/metabolismo , Masculino , Obesidade/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR
18.
Environ Mol Mutagen ; 56(4): 388-403, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25421724

RESUMO

7H-Dibenzo[c,g]carbazole (DBC) is a heterocyclic aromatic hydrocarbon that is carcinogenic in many species and tissues. DBC is a common environmental pollutant, and is therefore constantly exposed to sunlight. However, there are limited data exploring the toxicity of DBC photoexcitation products. Here, we investigated the impact of ultraviolet (UV) A radiation on the biological activity of DBC and its methyl derivatives, 5,9-dibenzo[c,g]carbazole and N-methyl dibenzo[c,g]carbazole, on human skin HaCaT keratinocytes. Co-exposure of HaCaT cells to UVA and DBC derivatives resulted in a sharp dose-dependent decrease in cell survival and apparent changes in cell morphology. Under the same treatment conditions, significant increases in DNA strand breaks, intracellular reactive oxygen species, and oxidative damage to DNA were observed in HaCaT cells. Consistent with these results, an apparent inhibition in superoxide dismutase, but not glutathione peroxidase activity, was detected in cells treated with DBC and its derivatives under UVA irradiation. The photoactivation-induced toxicity of individual DBC derivatives correlated with the electron excitation energies approximately expressed as the energy difference between the highest occupied and the lowest vacant molecular orbital. Our data provide the first evidence that UVA can enhance the toxicity of DBC and its derivatives. Photoactivation-induced conversion of harmless chemical compounds to toxic photoproducts associated with reactive oxygen species generation may substantially amplify the adverse health effects of UVA radiation and contribute to increased incidence of skin cancer.


Assuntos
Carbazóis/toxicidade , Queratinócitos/efeitos dos fármacos , Raios Ultravioleta/efeitos adversos , Apoptose/efeitos dos fármacos , Carbazóis/metabolismo , Carcinógenos/toxicidade , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Espécies Reativas de Oxigênio/metabolismo , Pele/citologia , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Glutationa Peroxidase GPX1
19.
Toxicol Lett ; 226(3): 303-13, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24614527

RESUMO

The generation of reactive oxygen species (ROS) has been proposed as the underlying mechanism involved in the genotoxicity of iron oxide nanoparticles. The data published to date are, however, inconsistent, and the mechanism underlying ROS formation has not been completely elucidated. Here, we investigated the capacity of several surface-modified magnetite nanoparticles (MNPs) to generate ROS in A549 human lung adenocarcinoma epithelial cells and HEL 12469 human embryonic lung fibroblasts. All MNPs, regardless of the coating, induced significant levels of DNA breakage in A549 cells but not in HEL 12469 cells. Under the same treatment conditions, variable low levels of intracellular ROS were detected in both A549 and HEL 12469 cells, but compared with control treatment, none of the coated MNPs produced any significant increase in oxidative damage to DNA in either of these cell lines. Indeed, no significant changes in the total antioxidant capacity and intracellular glutathione levels were observed in MNPs-treated human lung cell lines regardless of surface coating. In line with these results, none of the surface-modified MNPs increased significantly the GPx activity in A549 cells and the SOD activity in HEL 12469 cells. The GPx activity was significantly increased only in SO-Fe3O4-treated HEL 12469 cells. The SOD activity was significantly increased in SO-PEG-PLGA-Fe3O4-treated A549 cells but significantly decreased in SO-Fe3O4-treated A549 cells. Our data indicate that oxidative stress plays, at most, only a marginal role in the genotoxicity of surface-modified MNPs considered in this study in human lung cells.


Assuntos
Dano ao DNA , Pulmão/efeitos dos fármacos , Nanopartículas de Magnetita/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Células Cultivadas , Glutationa/análise , Glutationa Peroxidase/metabolismo , Humanos , Pulmão/metabolismo , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA