Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Adv Mater ; 36(26): e2403588, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38490170

RESUMO

A low-generation lysine dendrimer, SPr-G2, responds to intracellular glutathione to initiate bioorthogonal in situ polymerization, resulting in the formation of large assemblies in mouse breast cancer cells. The intracellular large assemblies of SPr-G2 can interact with lysosomes to induce lysosome expansion and enhance lysosomal membrane permeabilization, leading to major histocompatibility complex class I upregulation on tumor cell surfaces and ultimately tumor cell death. Moreover, the use of the SPr-G2 dendrimer to conjugate the chemotherapeutic drug, camptothecin (CPT), can boost the therapeutic potency of CPT. Excellent antitumor effects in vitro and in vivo are obtained from the combinational treatment of the SPr-G2 dendrimer and CPT. This combinational effect also enhances antitumor immunity through promoting activation of cytotoxic T cells in tumor tissues and maturation of dendritic cells. This study can shed new light on the development of peptide dendritic agents for cancer therapy.


Assuntos
Apresentação de Antígeno , Dendrímeros , Lisossomos , Polimerização , Lisossomos/metabolismo , Lisossomos/química , Animais , Dendrímeros/química , Camundongos , Linhagem Celular Tumoral , Apresentação de Antígeno/efeitos dos fármacos , Camptotecina/farmacologia , Camptotecina/química , Humanos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química
2.
Part Fibre Toxicol ; 21(1): 9, 2024 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-38419076

RESUMO

BACKGROUND: Zinc oxide nanoparticles (ZnONPs) are common materials used in skin-related cosmetics and sunscreen products due to their whitening and strong UV light absorption properties. Although the protective effects of ZnONPs against UV light in intact skin have been well demonstrated, the effects of using ZnONPs on damaged or sunburned skin are still unclear. In this study, we aimed to reveal the detailed underlying mechanisms related to keratinocytes and macrophages exposed to UVB and ZnONPs. RESULTS: We demonstrated that ZnONPs exacerbated mouse skin damage after UVB exposure, followed by increased transepidermal water loss (TEWL) levels, cell death and epithelial thickness. In addition, ZnONPs could penetrate through the damaged epithelium, gain access to the dermis cells, and lead to severe inflammation by activation of M1 macrophage. Mechanistic studies indicated that co-exposure of keratinocytes to UVB and ZnONPs lysosomal impairment and autophagy dysfunction, which increased cell exosome release. However, these exosomes could be taken up by macrophages, which accelerated M1 macrophage polarization. Furthermore, ZnONPs also induced a lasting inflammatory response in M1 macrophages and affected epithelial cell repair by regulating the autophagy-mediated NLRP3 inflammasome and macrophage exosome secretion. CONCLUSIONS: Our findings propose a new concept for ZnONP-induced skin toxicity mechanisms and the safety issue of ZnONPs application on vulnerable skin. The process involved an interplay of lysosomal impairment, autophagy-mediated NLRP3 inflammasome and macrophage exosome secretion. The current finding is valuable for evaluating the effects of ZnONPs for cosmetics applications.


Assuntos
Exossomos , Nanopartículas , Óxido de Zinco , Camundongos , Animais , Óxido de Zinco/toxicidade , Proteína 3 que Contém Domínio de Pirina da Família NLR , Raios Ultravioleta/efeitos adversos , Citocinas , Inflamassomos , Nanopartículas/toxicidade , Células Epiteliais
3.
Ecotoxicol Environ Saf ; 257: 114947, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37105094

RESUMO

Due to the rapid production growth and a wide range of applications, safety concerns are being raised about the genotoxic properties of silver nanoparticles (AgNPs). In this research, we found AgNPs induced a size-dependent genotoxicity via lysosomal-autophagy dysfunction in human-hamster hybrid (AL) cells. Compared with 25 nm and 75 nm particles, 5 nm AgNPs could accentuate the genotoxic responses, including DNA double-strand breaks (DSBs) and multi-locus deletion mutation, which could be significantly enhanced by autophagy inhibitors 3-methyl adenine (3-MA), Bafilomycin A1 (BFA), and cathepsin inhibitors, respectively. The autophagy dysfunction was closely related to the accumulation of 5 nm AgNPs in the lysosomes and the interruption of lysosome-autophagosome fusion. With lysosomal protective agent 3-O-Methylsphingomyelin (3-O-M) and endocytosis inhibitor wortmannin, the reactivation of lysosomal function and the recovery of autophagy significantly attenuated AgNP-induced genotoxicity. Our data provide clear evidence to illustrate the role of subcellular targets in the genotoxicity of AgNPs in mammalian cells, which laid the basis for better understanding the health risk of AgNPs and their related products.


Assuntos
Nanopartículas Metálicas , Prata , Animais , Humanos , Prata/toxicidade , Nanopartículas Metálicas/toxicidade , Autofagia/genética , Lisossomos , Deleção de Sequência , Mamíferos
4.
Int J Mol Sci ; 24(6)2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36982262

RESUMO

Although silica nanoparticles (SNPs) are generally thought to be biocompatible and safe, the adverse effects of SNPs were also reported in previous studies. SNPs cause follicular atresia via the induction of ovarian granulosa cell apoptosis. However, the mechanisms for this phenomenon are not well understood. This study focuses on exploring the relationship between autophagy and apoptosis induced by SNPs in ovarian granulosa cells. Our results showed that 25.0 mg/kg body weight (b.w.)/intratracheal instillation of 110 nm in diameter spherical Stöber SNPs caused ovarian granulosa cell apoptosis in follicles in vivo. We also found that SNPs mainly internalized into the lumens of the lysosomes in primary cultured ovarian granulosa cells in vitro. SNPs induced cytotoxicity via a decrease in viability and an increase in apoptosis in a dose-dependent manner. SNPs increased BECLIN-1 and LC3-II levels, leading to the activation of autophagy and increased P62 level, resulting in the blockage of autophagic flux. SNPs increased the BAX/BCL-2 ratio and cleaved the caspase-3 level, resulting in the activation of the mitochondrial-mediated caspase-dependent apoptotic signaling pathway. SNPs enlarged the LysoTracker Red-positive compartments, decreased the CTSD level, and increased the acidity of lysosomes, leading to lysosomal impairment. Our results reveal that SNPs cause autophagy dysfunction via lysosomal impairment, resulting in follicular atresia via the enhancement of apoptosis in ovarian granulosa cells.


Assuntos
Atresia Folicular , Nanopartículas , Feminino , Humanos , Atresia Folicular/fisiologia , Células da Granulosa/metabolismo , Apoptose , Autofagia/fisiologia
5.
Small ; 18(27): e2201585, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35644863

RESUMO

To overcome the autophagy compromised mechanism of protective cellular processes by "eating"/"digesting" damaged organelles or potentially toxic materials with autolysosomes in tumor cells, lysosomal impairment can be utilized as a traditional autophagy dysfunction route for tumor therapy; however, this conventional one-way autophagy dysfunction approach is always limited by the therapeutic efficacy. Herein, an innovative pharmacological strategy that can excessively provoke autophagy via endoplasmic reticulum (ER) stress is implemented along with lysosomal impairment to enhance autophagy dysfunction. In this work, the prepared tellurium double-headed nanobullets (TeDNBs) with controllable morphology are modified with human serum albumin (HSA) which facilitates internalization by tumor cells. On the one hand, ER stress can be stimulated by upregulating the phosphorylation eukaryotic translation initiation factor 2 (P-eIF2α) owing to the production of tellurite (TeO32- ) in the specifical hydrogen peroxide-rich tumor environment; thus, autophagy overstimulation occurs. On the other hand, OME can deacidify and impair lysosomes by downregulating lysosomal-associated membrane protein 1 (LAMP1), therefore blocking autolysosome formation. Both in vitro and in vivo results demonstrate that the synthesized TeDNBs-HSA/OME (TeDNBs-HO) exhibit excellent therapeutic efficacy by autophagy dysfunction through ER stress induction and lysosomal damnification. Thus, TeDNBs-HO is verified to be a promising theranostic nanoagent for effective tumor therapy.


Assuntos
Lisossomos , Telúrio , Autofagia , Estresse do Retículo Endoplasmático , Humanos , Lisossomos/metabolismo , Fosforilação
6.
Food Chem Toxicol ; 161: 112819, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35038498

RESUMO

Beauvericin (BEA) and enniatin B (ENNB) are emerging mycotoxins frequently detected in plant-based fish feed. With ionophoric properties, they have shown cytotoxic potential in mammalian models. Sensitivity in fish is still largely unknown. Primary hepatocytes isolated from Atlantic salmon (Salmo salar) were used as a model and exposed to BEA and ENNB (0.05-10 µM) for 48 h. Microscopy, evaluation of cell viability, total ATP, total H2O2, total iron content, total Gpx enzyme activity, and RNA sequencing were used to characterize the toxicodynamics of BEA and ENNB. Both mycotoxins became cytotoxic at ≥ 5 µM, causing condensation of the hepatocytes followed by formation of blister-like protrusions on the cell's membrane. RNA sequencing analysis at sub-cytotoxic levels indicated BEA and ENNB exposed hepatocytes to experience increased energy expenditure, elevated oxidative stress, and iron homeostasis disturbances sensitizing the hepatocytes to ferroptosis. The present study provides valuable knowledge disclosing the toxic action of these mycotoxins in Atlantic salmon primary hepatocytes.


Assuntos
Depsipeptídeos/toxicidade , Ferroptose/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Ferro/metabolismo , Fígado/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Depsipeptídeos/administração & dosagem , Relação Dose-Resposta a Droga , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Lisossomos/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Salmo salar
7.
Biomed Pharmacother ; 136: 111227, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33485070

RESUMO

Hydroxysafflor yellow A (HSYA) extracted from the herb Cathartics tinctorius L. negatively regulates liver cancer growth. However, the exact mechanism of HSYA action in liver cancer remains largely unknown. In this study, HSYA inhibited liver cancer cell growth in vivo and in vitro, evidenced by cell proliferation inhibition detected by CCK8, numerous apoptotic cells shown by flow cytometry assay, and expression of apoptosis-related proteins determined by western blot. Importantly, our data revealed that HSYA triggered autophagic response and autophagosome accumulation considering the increased levels of LC3II-conversion examined by western blot, LC3 puncta visualized by immunofluorescence, and expression of autophagy-related genes shown by quantitative real-time PCR. Furthermore, HSYA blocked the late-phase of autophagic flux via impairing the lysosomal acidification and downregulating LAMP1 expression, thereby likely inducing apoptosis. In addition, HSYA inhibited PI3K/AKT/mTOR signaling pathway. Taken together, as HSYA might inhibit cell proliferation and promote apoptosis via blocking autophagic flux in liver cancer, it may be considered a promising candidate for liver cancer therapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Chalcona/análogos & derivados , Neoplasias Hepáticas/tratamento farmacológico , Quinonas/farmacologia , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/patologia , Proliferação de Células/efeitos dos fármacos , Chalcona/farmacologia , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
8.
Sci Total Environ ; 761: 143290, 2021 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-33243499

RESUMO

Autophagy is a dynamic process for waste disposal and cell equilibrium. Previous studies have demonstrated that atmospheric particulate matter (APM) induces autophagy and enhances LC3II expression in human vascular endothelial cells. However, the underlying mechanism of autophagosome accumulation in human vascular endothelial cells under the exposure to APM has not been understood. In principle, the upregulation of LC3II or autophagosomes accumulation is presumably caused by the enhancement of autophagic ability, or alternatively, by the abnormal autophagic degradation. Therefore, in the current study, autophagic ability and autophagic flux are systemically studied to decipher the exact cause of autophagosomes accumulation in human umbilical vein endothelial cells (HUVECs) in response to a standard urban particulate matter, PM SRM1648a. As a result, it was observed that after 24 h of exposure, PM SRM1648a significantly increases LC3II expression with apparent autophagosomes accumulation in HUVECs. Compared with the control group, there is a time-dependent increase in p62, a protein of autophagic substrate that can be preliminarily used to evaluate the autophagic degradation, in the PM SRM1648a-exposed HUVECs, which suggested that normal function of autophagic degradation was probably impaired. Additionally, mRFP-GFP-LC3 assay and LAMP-2/LC3B co-localization suggested that autolysosomes (fusion between autophagosomes and lysosomes) were partially inhibited in PM SRM1648a-treated HUVECs. Furthermore, LC3II turn-over assay hinted that after 24 h, LC3II upregulation is attributed to the blockage of autophagic flux instead of the enhancement of autophagic induction. Mechanistically, the blockade of autophagic flux can be explained by the detrimental effects of PM SRM1648a on lysosomal function, including lysosomal destabilization, lysosomal alkalization and hydrolase inactivation, which are involved in the blockade of fusion between autophagosomes and lysosomes, further disrupting autophagic degradation and waste disposal. These observations provide evidence that PM SRM1648a destroys the equilibrium of lysosomal stability and thus results in the dysfunction of autophagic flux, eventually contributing to endothelial cell damage.


Assuntos
Lisossomos , Material Particulado , Autofagossomos , Autofagia , Células Endoteliais da Veia Umbilical Humana , Humanos , Material Particulado/toxicidade
9.
Part Fibre Toxicol ; 17(1): 23, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32513195

RESUMO

BACKGROUND: Wide applications of nanoparticles (NPs) have raised increasing concerns about safety to humans. Oxidative stress and inflammation are extensively investigated as mechanisms for NPs-induced toxicity. Autophagy and lysosomal dysfunction are emerging molecular mechanisms. Inhalation is one of the main pathways of exposing humans to NPs, which has been reported to induce severe pulmonary inflammation. However, the underlying mechanisms and, more specifically, the interplays of above-mentioned mechanisms in NPs-induced pulmonary inflammation are still largely obscure. Considered that NPs exposure in modern society is often unavoidable, it is highly desirable to develop effective strategies that could help to prevent nanomaterials-induced pulmonary inflammation. RESULTS: Pulmonary inflammation induced by intratracheal instillation of silica nanoparticles (SiNPs) in C57BL/6 mice was prevented by PJ34, a poly (ADP-ribose) polymerase (PARP) inhibitor. In human lung bronchial epithelial (BEAS-2B) cells, exposure to SiNPs reduced cell viability, and induced ROS generation, impairment in lysosome function and autophagic flux. Inhibition of ROS generation, PARP and TRPM2 channel suppressed SiNPs-induced lysosome impairment and autophagy dysfunction and consequent inflammatory responses. Consistently, SiNPs-induced pulmonary inflammation was prevented in TRPM2 deficient mice. CONCLUSION: The ROS/PARP/TRPM2 signaling is critical in SiNPs-induced pulmonary inflammation, providing novel mechanistic insights into NPs-induced lung injury. Our study identifies TRPM2 channel as a new target for the development of preventive and therapeutic strategies to mitigate nanomaterials-induced lung inflammation.


Assuntos
Autofagia/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Nanopartículas/toxicidade , Pneumonia/induzido quimicamente , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Dióxido de Silício/toxicidade , Canais de Cátion TRPM/metabolismo , Animais , Exposição por Inalação , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Pneumonia/metabolismo , Pneumonia/patologia , Transdução de Sinais , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA