Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Molecules ; 26(9)2021 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067016

RESUMO

Shanxi-aged vinegar, a traditional Chinese grain-fermented food that is rich in polyphenols, has been shown to have therapeutic effects on a variety of diseases. However, there has been no comprehensive evaluation of the anti-inflammatory activity of polyphenols extracted from Shanxi-aged vinegar (SAVEP) to date. The anti-inflammatory activities of SAVEP, both in RAW 264.7 macrophages and mice, were extensively investigated for the potential application of SAVEP as a novel anti-inflammatory agent. In order to confirm the notion that polyphenols could improve inflammatory symptoms, SAVEP was firstly detected by gas chromatography mass spectrometry (GC-MS). In total, 19 polyphenols were detected, including 12 phenolic acids. The study further investigated the protective effect of SAVEP on lipopolysaccharide-induced inflammation in RAW264.7 macrophages and ICR mice. The results showed that compared with those of the model group, SAVEP could remarkably recover the inflammation of macrophage RAW264.7 and ICR mice. SAVEP can normalise the expression of related proteins via the suppression of MAPK/NF-κB pathway activation, inhibiting the expression of iNOS and COX-2 proteins, and consequently the production of inflammatory factors, thus alleviating inflammatory stress. These results suggest that SAVEP may have a potential function against inflammation.


Assuntos
Ácido Acético/química , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/patologia , NF-kappa B/metabolismo , Polifenóis/isolamento & purificação , Polifenóis/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Forma do Núcleo Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Fígado/efeitos dos fármacos , Fígado/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Células RAW 264.7
2.
Int J Mol Sci ; 22(9)2021 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067074

RESUMO

The recently discovered interleukin (IL)- 32 isoform IL-32θ exerts anti-metastatic effects in the breast tumor microenvironment. However, the involvement of IL-32θ in breast cancer cell proliferation is not yet fully understood; therefore, the current study aimed to determine how IL-32θ affects cancer cell growth and evaluated the responses of IL-32θ-expressing cells to other cancer therapy. We compared the functions of IL-32θ in triple-negative breast cancer MDA-MB-231 cells that stably express IL-32θ, with MDA-MB-231 cells transfected with a mock vector. Slower growth was observed in cells expressing IL-32θ than in control cells, and changes were noted in nuclear morphology, mitotic division, and nucleolar size between the two groups of cells. Interleukin-32θ significantly reduced the colony-forming ability of MDA-MB-231 cells and induced permanent cell cycle arrest at the G1 phase. Long-term IL-32θ accumulation triggered permanent senescence and chromosomal instability in MDA-MB-231 cells. Genotoxic drug doxorubicin (DR) reduced the viability of MDA-MB-231 cells not expressing IL-32θ more than in cells expressing IL-32θ. Overall, these findings suggest that IL-32θ exerts antiproliferative effects in breast cancer cells and initiates senescence, which may cause DR resistance. Therefore, targeting IL-32θ in combination with DR treatment may not be suitable for treating metastatic breast cancer.


Assuntos
Senescência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Interleucinas/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Instabilidade Genômica , Humanos , Fenótipo , Ploidias
3.
ACS Appl Mater Interfaces ; 13(1): 1943-1955, 2021 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-33373205

RESUMO

In an in vitro nanotoxicity system, cell-nanoparticle (NP) interaction leads to the surface adsorption, uptake, and changes into nuclei/cell phenotype and chemistry, as an indicator of oxidative stress, genotoxicity, and carcinogenicity. Different types of nanomaterials and their chemical composition or "corona" have been widely studied in context with nanotoxicology. However, rare reports are available, which delineate the details of the cell shape index (CSI) and nuclear area factors (NAFs) as a descriptor of the type of nanomaterials. In this paper, we propose a machine-learning-based graph modeling and correlation-establishing approach using tight junction protein ZO-1-mediated alteration in the cell/nuclei phenotype to quantify and propose it as indices of cell-NP interactions. We believe that the phenotypic variation (CSI and NAF) in the epithelial cell is governed by the physicochemical descriptors (e.g., shape, size, zeta potential, concentration, diffusion coefficients, polydispersity, and so on) of the different classes of nanomaterials, which critically determines the intracellular uptake or cell membrane interactions when exposed to the epithelial cells at sub-lethal concentrations. The intrinsic and extrinsic physicochemical properties of the representative nanomaterials (NMs) were measured using optical (dynamic light scattering, NP tracking analysis) methods to create a set of nanodescriptors contributing to cell-NM interactions via phenotype adjustments. We used correlation function as a machine-learning algorithm to successfully predict cell and nuclei shapes and polarity functions as phenotypic markers for five different classes of nanomaterials studied herein this report. The CSI and NAF as nanodescriptors can be used as intuitive cell phenotypic parameters to define the safety of nanomaterials extensively used in consumer products and nanomedicine.


Assuntos
Forma do Núcleo Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Aprendizado de Máquina , Nanopartículas Metálicas/toxicidade , Citoesqueleto de Actina/metabolismo , Animais , Carbono/química , Proliferação de Células/efeitos dos fármacos , Dendrímeros/química , Cães , Células Epiteliais/citologia , Ouro/química , Células Madin Darby de Rim Canino , Nanopartículas Metálicas/química , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
4.
Int J Nanomedicine ; 15: 9025-9047, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33235450

RESUMO

BACKGROUND: Linalool is a monoterpene compound with various potential therapeutic applications in several medical fields. Previous studies have indicated the activity of linalool against cell lines; however, its high level of toxicity restricts its use. The aim of this study was to design and manufacture compounds with a novel structure that can be used for loading linalool, to reduce its toxicity and improve its reachable ability. METHODS: We synthesized and characterized a new molecule for loading linalool onto gold nanoparticles (GNPs) capped with glutathione and conjugated with a CALNN peptide. Linalool was loaded onto the GNPs via the reaction of the surface groups of both linalool and the GNPs. Moreover, the target peptide could be loaded onto the surface of the GNPs via a chemical reaction. The cytotoxic effects of linalool-GNP (LG) and linalool-GNP-CALNN peptide (LGC) conjugates against ovarian cancer cells were investigated, as were the possible mechanisms underlying the induction of apoptosis. RESULTS: Our findings illustrated the significant antiproliferative effect of LG and LGC on SKOV-3 cells. The cytotoxicity assay demonstrated that LG and LGC were selectively toxic in cancer cells and induced apoptosis by activating caspase-8, the p53 protein, and various proteins involved in apoptosis. The present data demonstrated that LG and LGC have a high therapeutic potential and should be given particular consideration as anticancer drug-delivery systems, as LG and LGC were remarkably more cytotoxic against a cancer cell line than were linalool and GNPs alone. CONCLUSION: We concluded that LG and LGC are promising compounds that can be used for treating ovarian cancer (SKOV-3) cells via the induction of apoptosis through extrinsic and intrinsic pathways.


Assuntos
Monoterpenos Acíclicos/química , Apoptose , Glutationa/química , Ouro/química , Nanopartículas Metálicas/química , NF-kappa B/metabolismo , Peptídeos/farmacologia , Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Dano ao DNA , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Nanopartículas Metálicas/ultraestrutura , Mutagênicos/toxicidade , Transporte Proteico/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
5.
Mol Syst Biol ; 16(6): e9442, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32500953

RESUMO

Microscopy is a powerful tool for characterizing complex cellular phenotypes, but linking these phenotypes to genotype or RNA expression at scale remains challenging. Here, we present Visual Cell Sorting, a method that physically separates hundreds of thousands of live cells based on their visual phenotype. Automated imaging and phenotypic analysis directs selective illumination of Dendra2, a photoconvertible fluorescent protein expressed in live cells; these photoactivated cells are then isolated using fluorescence-activated cell sorting. First, we use Visual Cell Sorting to assess hundreds of nuclear localization sequence variants in a pooled format, identifying variants that improve nuclear localization and enabling annotation of nuclear localization sequences in thousands of human proteins. Second, we recover cells that retain normal nuclear morphologies after paclitaxel treatment, and then derive their single-cell transcriptomes to identify pathways associated with paclitaxel resistance in cancers. Unlike alternative methods, Visual Cell Sorting depends on inexpensive reagents and commercially available hardware. As such, it can be readily deployed to uncover the relationships between visual cellular phenotypes and internal states, including genotypes and gene expression programs.


Assuntos
Células/citologia , Microscopia de Fluorescência/instrumentação , Linhagem Celular , Forma do Núcleo Celular/efeitos dos fármacos , Citometria de Fluxo , Testes Genéticos , Humanos , Sinais de Localização Nuclear/metabolismo , Paclitaxel/farmacologia , Fenótipo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
6.
Macromol Biosci ; 20(7): e1900438, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32406183

RESUMO

Targeting the distinct cholesterol metabolism of tumor cells is proposed as a novel way to treat tumors. Blocking acyl-CoA cholesterol acyltransferase-1 (ACAT-1) by the inhibitor avasimible (Ava), which elevates intracellular free cholesterol levels, is shown to effectively induce apoptosis. However, Ava faces disadvantages of poor water solubility, a short half-life, and no capability for fluorescence detection, which have greatly limited its application. Herein, a fluorescent hyperstar polymer (FHSP) is developed to encapsulate Ava to improve its ability to inhibit HeLa cells and K562 cells. The results of this study show that the obtained Ava-FHSP micelles possess a high drug loading capacity of 22.7% and bright green fluorescence. Ava and Ava-FHSP are cytotoxic to both HeLa and K562 cells and cause reductions in cell size, nuclear lysis, and chromatin condensation and hindered proliferation of both cell types by causing S phase cell cycle arrest. Further mechanistic analysis indicates that Ava-FHSP reduces the protein and messenger RNA expression of ACAT-1 and significantly increases intracellular free cholesterol levels, which can increase endoplasmic reticulum stress and finally cause cell apoptosis. All these results suggest that this fluorescent hyperstar polymer represents a potential therapeutic tumor strategy by changing the cholesterol metabolism of tumor cells.


Assuntos
Neoplasias/tratamento farmacológico , Polímeros/química , Esterol O-Aciltransferase/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Forma do Núcleo Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Células HeLa , Humanos , Células K562 , Neoplasias/patologia , Polímeros/síntese química , Espectroscopia de Prótons por Ressonância Magnética , Espectrometria de Fluorescência , Esterol O-Aciltransferase/metabolismo
7.
Molecules ; 25(7)2020 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-32276430

RESUMO

The global incidence of breast cancer has increased. However, there are many impediments to the development of safe and effective anticancer drugs. The aim of the present study was to evaluate the effect of aviculin isolated from Lespedeza cuneata (Dum. Cours.) G. Don. (Fabaceae) on MCF-7 human breast cancer cells and determine the underlying mechanism. Using the bioassay-guided isolation by water soluble tetrazolium salt (WST-1)-based Ez-Cytox assay, nine compounds (four lignan glycosides (1-4), three flavonoid glycosides (5-7), and two phenolic compounds (8 and 9)) were isolated from the ethyl acetate (EA) fraction of the L. cuneata methanolic extract. Of these, aviculin (2), a lignan glycoside, was the only compound that reduced metabolic activity on MCF-7 cells below 50% (IC50: 75.47 ± 2.23 µM). The underlying mechanism was analyzed using the annexin V Alexa Fluor 488 binding assay and Western blotting. Aviculin (2) was found to induce apoptotic cell death through the intrinsic apoptosis pathway, as indicated by the increased expression of initiator caspase-9, executioner caspase-7, and poly (ADP-ribose) polymerase (PARP). Aviculin (2)-induced apoptotic cell death was accompanied by an increase in the Bax/Bcl-2 ratio. These findings demonstrated that aviculin (2) could induce breast cancer cell apoptosis through the intrinsic apoptosis pathway, and it can therefore be considered an excellent candidate for herbal treatment of breast cancer.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Caspases/metabolismo , Glicosídeos/isolamento & purificação , Glicosídeos/farmacologia , Lespedeza/química , Mitocôndrias/metabolismo , Transdução de Sinais , Neoplasias da Mama/metabolismo , Forma do Núcleo Celular/efeitos dos fármacos , Cisplatino/farmacologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Glicosídeos/química , Humanos , Células MCF-7 , Metanol/química , Mitocôndrias/efeitos dos fármacos , Extratos Vegetais/farmacologia , Transdução de Sinais/efeitos dos fármacos
8.
Med Sci Monit ; 26: e919558, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-32005795

RESUMO

BACKGROUND Bauerane is a triterpenoid derived from the dandelion root (Taraxacum officinale). This study aimed to investigate the effects of bauerane on cell proliferation of A549 human lung cancer cells and the molecular mechanisms involved. MATERIAL AND METHODS A549 human lung adenocarcinoma cells and normal MRC-5 lung fibroblasts were grown in culture and treated with increasing doses of bauerane at 0, 2.5, 5, 10, 20, 40, 80, and 160 µM. The MTT assay was used to measure cell proliferation. Cell apoptosis was assessed by 4', 6-diamidino-2-phenylindole (DAPI), and acridine orange/ethidium bromide (AO/EB) staining. The cell cycle was evaluated by flow cytometry. Western blot measured the protein expression levels of cytochrome c, Bax, cyclin B1, Bcl-2, PI3K, p-PI3K, Akt, p-Akt, and STAT3 proteins. RESULTS Bauerane inhibited the proliferation of A549 lung cancer cells in a dose-dependent manner, with an IC50 of 10 µM, with no cytotoxicity for MRC-5 cells. Bauerane treatment induced apoptosis of A549 cells, which was associated with the upregulation of Bax and down-regulation of Bcl-2. Bauerane induced S-phase arrest of A549 cells, which was dose-dependent and associated with reduced expression of cyclin B1. The findings from Western blot showed that bauerane inhibited the phosphorylation of PI3K/AKT and STAT3 signaling pathways. CONCLUSIONS Bauerane inhibited the proliferation of A549 lung cancer cells in vitro and induced cell apoptosis and cell cycle arrest in a dose-dependent manner.


Assuntos
Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fase S , Fator de Transcrição STAT3/metabolismo , Triterpenos/farmacologia , Células A549 , Forma do Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina B1/metabolismo , Humanos , Fase S/efeitos dos fármacos , Transdução de Sinais , Triterpenos/química
9.
Cells ; 8(5)2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31072066

RESUMO

Mechanosensing of the mechanical microenvironment by cells regulates cell phenotype and function. The nucleus is critical in mechanosensing, as it transmits external forces from the cellular microenvironment to the nuclear envelope housing chromatin. This study aims to elucidate how mechanical confinement affects nuclear deformation within several cell types, and to determine the role of cytoskeletal elements in controlling nuclear deformation. Human cancer cells (MDA-MB-231), human mesenchymal stem cells (MSCs), and mouse fibroblasts (L929) were seeded within polydimethylsiloxane (PDMS) microfluidic devices containing microchannels of varying cross-sectional areas, and nuclear morphology and volume were quantified via image processing of fluorescent cell nuclei. We found that the nuclear major axis length remained fairly constant with increasing confinement in MSCs and MDA-MB-231 cells, but increased with increasing confinement in L929 cells. Nuclear volume of L929 cells and MSCs decreased in the most confining channels. However, L929 nuclei were much more isotropic in unconfined channels than MSC nuclei. When microtubule polymerization or myosin II contractility was inhibited, nuclear deformation was altered only in MSCs in wide channels. This work informs our understanding of nuclear mechanics in physiologically relevant spaces, and suggests diverging roles of the cytoskeleton in regulating nuclear deformation in different cell types.


Assuntos
Núcleo Celular/patologia , Animais , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Forma do Núcleo Celular/efeitos dos fármacos , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Miosina Tipo II/metabolismo , Nocodazol/farmacologia , Polimerização , Adulto Jovem
10.
Int J Nanomedicine ; 14: 1177-1191, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30863053

RESUMO

BACKGROUND: Treatment for melanoma is a challenging clinical problem, and some new strategies are worth exploring. PURPOSE: The objective of this study was to investigate the in vitro and in vivo anti-melanoma effects of hydroxyapatite nanoparticles (HANPs) and discuss the involved material factors. MATERIALS AND METHODS: Five types of HANPs, ie, HA-A, HA-B, HA-C, HA-D, and HA-E, were prepared by wet chemical method combining with polymer template and appropriate post-treatments. The in vitro effects of the as-prepared five HANPs on inhibiting the viability of A375 melanoma cells and inducing the apoptosis of the cells were evaluated by Cell Counting Kit-8 analysis, cell nucleus morphology observation, flow cytometer, and PCR analysis. The in vivo anti-melanoma effects of HANPs were studied in the tumor model of nude mice. RESULTS: The five HANPs had different physicochemical properties, including morphology, size, specific surface area (SSA), crystallinity, and so on. By the in vitro cell study, it was found that the material factors played important roles in the anti-melanoma effect of HANPs. Among the as-prepared five HANPs, HA-A with granular shape, smaller size, higher SSA, and lower crystallinity exhibited best effect on inhibiting the viability of A375 cells. At the concentration of 200 µg/mL, HA-A resulted in the lowest cell viability (34.90%) at day 3. All the HANPs could induce the apoptosis of A375 cells, and the relatively higher apoptosis rates of the cells were found in HA-A (20.10%) and HA-B (19.41%) at day 3. However, all the HANPs showed no inhibitory effect on the viability of the normal human epidermal fibroblasts. The preliminary in vivo evaluation showed that both HA-A and HA-C could delay the formation and growth speed of melanoma tissue significantly. Likely, HA-A exhibited better effect on inhibiting the growth of melanoma tissue than HA-C. The inhibition rate of HA-A for tumor tissue growth reached 49.1% at day 23. CONCLUSION: The current study confirmed the anti-melanoma effect of HANPs and provided a new idea for the clinical treatment of melanoma.


Assuntos
Durapatita/farmacologia , Melanoma/patologia , Nanopartículas/química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/genética , Feminino , Fibroblastos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/ultraestrutura , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
12.
Food Funct ; 9(9): 4998-5006, 2018 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-30187896

RESUMO

The anti-lung cancer activity of volatile oil from Alpinia officinarum (VOAO) and the underlying mechanism has not been studied. Herein, VOAO was extracted by steam distillation and its components were identified by GC-MS analysis. Cells viability was measured by an MTT assay and the cell survival capacity was tested via a colony formation assay. Apoptosis cells were detected using the Annexin V-FITC/PI method. Hoechst 33342 cell unclear staining was employed to evaluate the nuclear morphology change. The mitochondrial membrane potential was detected by a JC-1 staining assay. Bcl-2, Mcl-1 and cleaved caspase-3 proteins were quantified by immune blotting. Furthermore, VOAO anti-cancer activity was evaluated on an A549 cell xenograft nude mice model. Our results have revealed that VOAO could inhibit the cell viability of lung carcinoma cells and the colony formation capacity. VOAO downregulates Bcl-2 and Mcl-1 and triggers dysfunction of the mitochondrial membrane potential. VOAO further activates caspase-3 cleavage and induces lung cancer cells apoptosis. In addition, VOAO administration significantly suppresses lung cancer growth in xenograft mice without obvious hepatotoxicity. We conclude that VOAO could be an effective, low cytotoxicity natural component for treatment of lung carcinoma.


Assuntos
Alpinia/química , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Pulmão/efeitos dos fármacos , Óleos Voláteis/uso terapêutico , Animais , Anticarcinógenos/administração & dosagem , Anticarcinógenos/efeitos adversos , Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/farmacologia , Carcinoma/metabolismo , Carcinoma/patologia , Carcinoma/prevenção & controle , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/prevenção & controle , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Óleos Voláteis/administração & dosagem , Óleos Voláteis/efeitos adversos , Óleos Voláteis/farmacologia , Distribuição Aleatória , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
PLoS One ; 13(8): e0201858, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30089134

RESUMO

Triple negative breast cancer (TNBC) cells express increased levels of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which promotes their proliferation and migration. Because TNBC patients are unresponsive to current targeted therapies, new therapeutic strategies are urgently needed. While proteasome inhibition by bortezomib (BZ) or carfilzomib (CZ) has been effective in treating hematological malignancies, it has been less effective in solid tumors, including TNBC, but the mechanisms are incompletely understood. Here we report that proteasome inhibition significantly increases expression of IL-8, and its receptors CXCR1 and CXCR2, in TNBC cells. Suppression or neutralization of the BZ-induced IL-8 potentiates the BZ cytotoxic and anti-proliferative effect in TNBC cells. The IL-8 expression induced by proteasome inhibition in TNBC cells is mediated by IκB kinase (IKK), increased nuclear accumulation of p65 NFκB, and by IKK-dependent p65 recruitment to IL-8 promoter. Importantly, inhibition of IKK activity significantly decreases proliferation, migration, and invasion of BZ-treated TNBC cells. These data provide the first evidence demonstrating that proteasome inhibition increases the IL-8 signaling in TNBC cells, and suggesting that IKK inhibitors may increase effectiveness of proteasome inhibitors in treating TNBC.


Assuntos
Antineoplásicos/farmacologia , Quinase I-kappa B/metabolismo , Interleucina-8/metabolismo , Inibidores de Proteassoma/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Bortezomib/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Forma do Núcleo Celular/fisiologia , Quimioterapia Combinada , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Fator de Transcrição RelA/metabolismo
14.
Toxicol Mech Methods ; 28(3): 157-166, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28849708

RESUMO

The leucine aminopeptidase inhibitor, benzyloxycarbonyl-leucine-chloromethylketone (z-L-CMK), was found to be toxic and readily induce cell death in Jurkat T cells. Dose-response studies show that lower concentration of z-L-CMK induced apoptosis in Jurkat T cells whereas higher concentration causes necrosis. In z-L-CMK-induced apoptosis, both the initiator caspases (-8 and -9) and effector caspases (-3 and -6) were processed to their respective subunits. However, the caspases remained intact in z-L-CMK-induced necrosis. The caspase inhibitor, z-VAD-FMK inhibited z-L-CMK-mediated apoptosis and caspase processing but has no effect on z-L-CMK-induced necrosis in Jurkat T cells. The high mobility group protein B1 (HMGB1) protein was found to be released into the culture medium by the necrotic cells and not the apoptotic cells. These results indicate that the necrotic cell death mediated by z-L-CMK at high concentrations is via classical necrosis rather than secondary necrosis. We also demonstrated that cell death mediated by z-L-CMK was associated with oxidative stress via the depletion of intracellular glutathione (GSH) and increase in reactive oxygen species (ROS), which was blocked by N-acetyl cysteine. Taken together, the results demonstrated that z-L-CMK is toxic to Jurkat T cells and induces apoptosis at low concentrations, while at higher concentrations the cells die of necrosis. The toxic side effects in Jurkat T cells mediated by z-L-CMK are associated with oxidative stress via the depletion of GSH and accumulation of ROS.


Assuntos
Clorometilcetonas de Aminoácidos/toxicidade , Apoptose/efeitos dos fármacos , Leucil Aminopeptidase/antagonistas & inibidores , Necrose/induzido quimicamente , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteases/toxicidade , Linfócitos T/efeitos dos fármacos , Clorometilcetonas de Aminoácidos/antagonistas & inibidores , Biomarcadores/metabolismo , Inibidores de Caspase/farmacologia , Caspases/química , Caspases/metabolismo , Forma do Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Humanos , Células Jurkat , Leucil Aminopeptidase/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Nucleossomos/efeitos dos fármacos , Nucleossomos/imunologia , Nucleossomos/metabolismo , Concentração Osmolar , Fragmentos de Peptídeos/metabolismo , Inibidores de Proteases/química , Proteólise/efeitos dos fármacos , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
J Biol Chem ; 292(51): 21102-21116, 2017 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-29074620

RESUMO

Mensacarcin is a highly oxygenated polyketide that was first isolated from soil-dwelling Streptomyces bacteria. It exhibits potent cytostatic properties (mean of 50% growth inhibition = 0.2 µm) in almost all cell lines of the National Cancer Institute (NCI)-60 cell line screen and relatively selective cytotoxicity against melanoma cells. Moreover, its low COMPARE correlations with known standard antitumor agents indicate a unique mechanism of action. Effective therapies for managing melanoma are limited, so we sought to investigate mensacarcin's unique cytostatic and cytotoxic effects and its mode of action. By assessing morphological and biochemical features, we demonstrated that mensacarcin activates caspase-3/7-dependent apoptotic pathways and induces cell death in melanoma cells. Upon mensacarcin exposure, SK-Mel-28 and SK-Mel-5 melanoma cells, which have the BRAFV600E mutation associated with drug resistance, showed characteristic chromatin condensation as well as distinct poly(ADP-ribose)polymerase-1 cleavage. Flow cytometry identified a large population of apoptotic melanoma cells, and single-cell electrophoresis indicated that mensacarcin causes genetic instability, a hallmark of early apoptosis. To visualize mensacarcin's subcellular localization, we synthesized a fluorescent mensacarcin probe that retained activity. The natural product probe was localized to mitochondria within 20 min of treatment. Live-cell bioenergetic flux analysis confirmed that mensacarcin disturbs energy production and mitochondrial function rapidly. The subcellular localization of the fluorescently labeled mensacarcin together with its unusual metabolic effects in melanoma cells provide evidence that mensacarcin targets mitochondria. Mensacarcin's unique mode of action suggests that it may be a useful probe for examining energy metabolism, particularly in BRAF-mutant melanoma, and represent a promising lead for the development of new anticancer drugs.


Assuntos
Antracenos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Melanoma/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Substituição de Aminoácidos , Antracenos/metabolismo , Antibióticos Antineoplásicos/metabolismo , Transporte Biológico , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Instabilidade Cromossômica/efeitos dos fármacos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Metabolismo Energético/efeitos dos fármacos , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo
16.
Mol Med Rep ; 16(3): 3055-3060, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713949

RESUMO

Acute promyelocytic leukemia (APL) is a special subtype of acute myeloid leukemia that responds to treatment with all­trans retinoic acid and arsenic trioxide. However, severe side effects and drug resistance limit the effectiveness of these treatments. Hence, new drugs for APL are required urgently. Shikonin, an active naphthoquinone derived from the Chinese medical herb Zi Cao exerts antitumor activity in several cancers. In the present study, the effects of shikonin on proliferation and apoptosis in NB4 cells, as well as related mechanisms were assessed. Treatment of NB4 cells with shikonin inhibited proliferation in a concentration­ and time­dependent manner. The cell cycle was arrested in the G1 phase. NB4 cells treated with shikonin exhibited more apoptosis and higher levels of cleaved caspase­3 and poly ADP­ribose polymerase than control cells. Western blotting results demonstrated that the expression of p­p38 mitogen­activated protein kinase (p­p38MAPK) and p­c­Jun N­terminal kinase (p­JNK) was increased significantly by shikonin treatment, while the expression of p­ERK and c­Myc was decreased. In summary, these findings indicated that shikonin inhibited cell proliferation and induced apoptosis partly through modulation of the MAPKs and downregulation of c­Myc.


Assuntos
Apoptose/efeitos dos fármacos , Leucemia Promielocítica Aguda/enzimologia , Leucemia Promielocítica Aguda/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Naftoquinonas/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Microscopia de Fluorescência , Poli(ADP-Ribose) Polimerases/metabolismo
17.
Mol Med Rep ; 16(2): 1133-1138, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627694

RESUMO

The present study aimed to investigate the neuroprotective effect of a novel synthetic compound (5zou) on differentiated PC12 cells against 6­hydroxydopamine (6­OHDA) and L­glutamic acid (L­Glu) neurotoxin­induced cell injury and the potential mechanisms involved. 5zou is a 2, 2­disubstituted 1,2­dihydropyridine. PC12 cells were treated with 6­OHDA and L­Glu to establish neurotoxic cell models. MTT assay, DCFH­DA staining, Fluo­4­AM staining, JC­1 staining and western blotting were used to determine the changes in cell viability, intracellular reactive oxygen species concentration, Ca2+ influx, mitochondrial membrane potential and the protein expressions of B­cell lymphoma­2 (Bcl­2) and B­cell lymphoma­extra large (Bcl­xL). Morphological analysis demonstrated the effect of 5zous on neuritogenesis and differentiation in PC12 cells. The results suggested that 5zou rescued the cell viability, intracellular ROS level, Ca2+ influx, mitochondrial membrane potential, and expression of Bcl­2 and Bcl­xL, which were altered by 6­OHDA and L­Glu. The study confirmed that 5zou has neuroprotective effects on neurotoxin­induced differentiated PC12 cells injury, potentially via the mitochondrial apoptosis pathway.


Assuntos
Mitocôndrias/metabolismo , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Forma do Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ácido Glutâmico , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/química , Neurotoxinas/toxicidade , Oxidopamina , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo , Proteína bcl-X/metabolismo
18.
J Trace Elem Med Biol ; 42: 117-128, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28595784

RESUMO

Cadmium (Cd) is a well-known heavy metal that causes environmental pollution and human health problems. Several studies attempted to assess Cd toxicity in vitro and in vivo. However, the systemic profile of cadmium toxicity has not been studied well. In the present study, we assessed the toxicity of Cd on human peripheral blood lymphocytes and monocytes and gene expression, using a system toxicological approach. Cd effect on cell viability and morphology were analyzed by MTT assay and AO/EB staining respectively. Mitochondrial membrane potential depletion and reactive oxygen singlet generation were assessed by flow cytometry. Effects of Cd treatment on gene expression were also studied. Significant reduction in cell viability and disintegration of nuclear morphology were observed in Cd-treated cells. Cd exposure enhanced the loss of mitochondrial membrane potential through oxidative stress. Dose-dependent upregulation of GSTM3 and downregulation of GSR gene expression were observed. TNF gene expression decreased as the level of Cd exposure increased. We analyzed the toxicological effects of Cd on more than 45 proteins for biological target identification. These system toxicological studies suggested that Cd induced toxicity through various biological processes such as oxidative stress, oxidation-reduction, and ROS and hydrogen peroxide generation. Additionally, Cd affects the cellular component network and modulates the expression level of oxidative stress-related genes.


Assuntos
Cádmio/toxicidade , Linfócitos/metabolismo , Mitocôndrias/metabolismo , Monócitos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Forma do Núcleo Celular/efeitos dos fármacos , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Espaço Intracelular/metabolismo , Linfócitos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Mapas de Interação de Proteínas/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
19.
Toxicol Sci ; 156(1): 275-288, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28115644

RESUMO

Chemotherapy-induced peripheral neuropathy (CIPN) is a major, dose-limiting adverse effect experienced by cancer patients. Advancements in mechanism-based risk mitigation and effective treatments for CIPN can be aided by suitable in vitro assays. To this end, we developed a multiparametric morphology-centered rat dorsal root ganglion (DRG) assay. Morphologic alterations in subcellular structures of neurons and non-neurons were analyzed with an automated microscopy system. Stains for NeuN (a neuron-specific nuclear protein) and Tuj-1 (ß-III tubulin) were used to identify neuronal cell nuclei and neuronal cell bodies/neurites, respectively. Vimentin staining (a component of Schwann cell intermediate filaments) was used to label non-neuronal supporting cells. Nuclei that stained with DAPI, but lacked NeuN represented non-neuronal cells. Images were analyzed following 24 h of continuous exposure to CIPN-inducing agents and 72 h after drug removal to provide a dynamic measure of recovery from initial drug effects. Treatment with bortezomib, cisplatin, eribulin, paclitaxel or vincristine induced a dose-dependent loss of neurite/process areas, mimicking the 'dying back' degeneration of axons, a histopathological hallmark of clinical CIPN in vivo. The IC50 for neurite loss was within 3-fold of the maximal clinical exposure (Cmax) for all five CIPN-inducing drugs, but was >4- or ≥ 28-fold of the Cmax for 2 non-CIPN-inducing agents. Compound-specific effects, eg, neurite fragmentation by cisplatin or bortezomib and enlarged neuronal cell bodies by paclitaxel, were also observed. Collectively, these results support the use of a quantitative, morphologic evaluation and a DRG cell culture model to inform risk and examine mechanisms of CIPN.


Assuntos
Antineoplásicos/efeitos adversos , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Corpo Celular/patologia , Forma do Núcleo Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Eletroforese Capilar , Imunofluorescência , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Processamento de Imagem Assistida por Computador , Cinética , Peso Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neuritos/patologia , Neurônios/metabolismo , Neurônios/patologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Forma das Organelas/efeitos dos fármacos , Tamanho das Organelas/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/etiologia , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Ratos
20.
Sci Rep ; 7: 41340, 2017 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-28112275

RESUMO

Cell death triggered by photodynamic therapy can occur through different mechanisms: apoptosis, necrosis or autophagy. However, recent studies have demonstrated the existence of other mechanisms with characteristics of both necrosis and apoptosis. These new cell death pathways, collectively termed regulated necrosis, include a variety of processes triggered by different stimuli. In this study, we evaluated the cell death mechanism induced by photodynamic treatments with two photosensitizers, meso-tetrakis (4-carboxyphenyl) porphyrin sodium salt (Na-H2TCPP) and its zinc derivative Na-ZnTCPP, in two human breast epithelial cell lines, a non-tumoral (MCF-10A) and a tumoral one (SKBR-3). Viability assays showed that photodynamic treatments with both photosensitizers induced a reduction in cell viability in a concentration-dependent manner and no dark toxicity was observed. The cell death mechanisms triggered were evaluated by several assays and cell line-dependent results were found. Most SKBR-3 cells died by either necrosis or apoptosis. By contrast, in MCF-10A cells, necrotic cells and another cell population with characteristics of both necrosis and apoptosis were predominant. In this latter population, cell death was PARP-dependent and translocation of AIF to the nucleus was observed in some cells. These characteristics are related with parthanatos, being the first evidence of this type of regulated necrosis in the field of photodynamic therapy.


Assuntos
Apoptose , Neoplasias/patologia , Fotoquimioterapia , Fator de Indução de Apoptose/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Forma do Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Citotoxicidade Imunológica/efeitos dos fármacos , Ativação Enzimática , Humanos , Concentração Inibidora 50 , Metaloporfirinas/química , Metaloporfirinas/farmacologia , Necrose , Fosfatidilserinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA