Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Toxicol ; 37(5): 1231-1243, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35112775

RESUMO

Cigarette smoke (CS) substances are known to induce diverse ailments such as cancer, decreased immunity, and lung diseases. Although some studies have been actively conducted to evaluate cigarette toxicity, the current animal exposure methods, that is, exposure of 28- or 90-days, require considerable research cost and lead to obscure results of the CS effects. In a previous study, we compared the effects of CS in a rat model of bleomycin (BLM) and lipopolysaccharide (LPS) induced lung disease. We determined that compared to the LPS-induced rat model, the BLM-induced rat model was more sensitive to alterations in secreting cytokines and total cell number. In the current study, we further confirmed the time-point of effective inhalation exposure by CS in the BLM-induced lung injury rat model. Using an automatic video instillator, rats were administered a single dose of 2.5 mg/kg BLM (day 1), and subsequently exposed to CS via inhalation (nose-only) 4 h/day, for 1, 2, 3, and 4 weeks. The bronchoalveolar lavage fluid (BALF) was obtained from the right lung lobes, total cell numbers were counted, and chemokine and cytokine expressions were evaluated using Enzyme-Linked Immunosorbent Assay. For the 1-week exposure, we observed a greater increase of neutrophils in the BLM + CS 300 µg/L group than in the BLM or CS 300 µg/L groups. Exposure of CS in the BLM-induced lung injury rat model enhanced the secretions of chemokines and cytokines, such as CCL2/MCP-1, CXCL2/MIP-2 and TNF-α, at 1 week. Immunohistochemistry and Hematoxylin and Eosin staining of lungs at 1-2 weeks after exposure clearly confirmed this tendency in the increased levels of CCL2/MCP-1 and TNF-α. Taken together, these results indicate that the rat model of BLM-induced lung injury is more sensitive to CS exposure than other rat models, and may be an appropriate model to evaluate the effect of CS exposure at 1-2 weeks.


Assuntos
Fumar Cigarros , Lesão Pulmonar , Animais , Bleomicina/toxicidade , Líquido da Lavagem Broncoalveolar/química , Fumar Cigarros/efeitos adversos , Pulmão , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Ratos
2.
Biomol Ther (Seoul) ; 30(2): 151-161, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34261818

RESUMO

This study elucidates the anti-cancer potential of gallic acid (GA) as a promising therapeutic agent that exerts its effect by regulating the PI3K/Akt pathway. To prove our research rationale, we used diverse experimental methods such as cell viability assay, colony formation assay, tumor spheroid formation assay, cell cycle analysis, TUNEL assay, Western blot analysis, xenograft mouse model and histological analysis. Treatment with GA inhibited cell proliferation in dose-dependent manner as measured by cell viability assay at 48 h. GA and cisplatin (CDDP) also inhibited colony formation and tumor spheroid formation. In addition, GA and CDDP induced apoptosis, as determined by the distribution of early and late apoptotic cells and DNA fragmentation. Western blot analysis revealed that inhibition of the PI3K/Akt pathway induced upregulation of p53 (tumor suppressor protein), which in turn regulated cell cycle related proteins such as p21, p27, Cyclin D1 and E1, and intrinsic apoptotic proteins such as Bax, Bcl-2 and cleaved caspase-3. The anti-cancer effect of GA was further confirmed in an in vivo mouse model. Intraperitoneal injection with GA for 4 weeks in an A549-derived tumor xenograft model reduced the size of tumor mass. Injection of them downregulated the expression of proliferating cell nuclear antigen and p-Akt, but upregulated the expression of cleaved caspase-3 in tumor tissues. Taken together, these results indicated that GA hindered lung cancer progression by inducing cell cycle arrest and apoptosis, suggesting that GA would be a potential therapeutic agent against non-small cell lung cancer.

3.
Reprod Toxicol ; 104: 76-84, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34280493

RESUMO

Fludioxnil is extensively used as a fungicide in agricultural application, but its possible impact on embryonic development is not yet well understood. In this study, the potential effect of fludioxonil on cardiac differentiation was evaluated in mouse embryonic stem cells (mESCs). The water-soluble tetrazolium (WST) and colony formation assays were conducted to confirm the effect of fludioxonil on proliferation of mESCs. The effect of fludioxonil on the ability of mESCs to form mouse embryoid bodies (mEBs) was determined by the hanging drop assay, whereas the ability of cardiomyocyte differentiation in the early stage was evaluated by determining the beating ratio (ratio of the number of contracting cells to the number of attached EBs) of cardiomyocytes. The viability of mESCs was significantly decreased (less than 50 %) at 10-5 M fludioxonil. Results of the colony formation assay revealed suppressed colony formation at 10-5 M fludioxonil (about 50 % at 5 days). Furthermore, the expressions of cell-cycle related proteins, i.e., cyclin D1, cyclin E, p21 and p27, were altered and trending towards inhibiting cell growth. Exposure to fludioxonil also resulted in reduced size of the mEB and induced increasing expression levels of the pluripotency markers Oct4, Sox2 and Nanog. Development of the beating ratio in the process of differentiation to cardiomyocytes derived from mESCs was completely inhibited after exposure to 10-5 M fludioxonil during the early stage of differentiation (day 5), whereas the beating ratio gradually increased after 5-day treatment. Simultaneously, expressions of the cardiomyocyte-related proteins, Gata4, Hand1 and cTnI, were inhibited after exposure to 10-5 M fludioxonil. Taken together, these results imply that fludioxonil may impact on the developmental process of mESCs, particularly the cardiac lineage.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dioxóis/toxicidade , Fungicidas Industriais/toxicidade , Pirróis/toxicidade , Animais , Linhagem Celular , Proliferação de Células , Corpos Embrioides/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Organogênese
5.
J Nutr Biochem ; 84: 108444, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32615369

RESUMO

Gallic acid (GA) is known to possess diverse biological activities, including anticancer. Histone deacetylase (HDACs) are controlled by tumor suppressor gene transcription and are overexpressed in various tumors, resulting in tumor development, progression and poor prognosis. This study aims to demonstrate the effect of GA on inhibition of prostate cancer (PCa) progression by modulating the expression of HDAC1 and 2 in PCa cells. To prove our research rationale, we used diverse experimental methods. GA decreased the cell viability of only PCa cell lines and not normal cells (contrary to another HDAC inhibitor, suberoylanilide hydroxamic acid) and also inhibited colony and tumor spheroid formation. Exposure to GA decreased the mitochondrial membrane potential (ΔΨm), increased the number of apoptotic cells and induced DNA fragmentation. Western blot analysis revealed down-regulated expression of HDAC1 and 2, leading to up-regulation of acetyl-p53 expression at the protein level, subsequent to down-regulating the expression of cell-cycle-related genes, i.e., proliferating cell nuclear antigen (PCNA), Cyclin D1 and E1, up-regulating the expression of cell cycle arrest gene p21 and regulating the expression of apoptosis intrinsic pathway-related genes, such as Bax, Bcl-2, cleaved Caspase-3 and poly (ADP-ribose) polymerase 1 in both PCa cell lines. Furthermore, oral administration of GA for 8 weeks on PC-3 cells-derived tumor xenograft mice model decreases the tumor size, damages the tumor structure and down-regulates the expression of HDAC1 and 2 and PCNA in tumor mass, as confirmed by histological analysis. These results indicated that GA may hinder the PCa progression by inhibiting HDAC1 and 2 expression, thereby demonstrating the potential of GA to be used as HDACs inhibitor and anti-PCa therapeutics.


Assuntos
Antineoplásicos/farmacologia , Ácido Gálico/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Ácido Gálico/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 1/análise , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/genética , Histona Desacetilase 2/análise , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/genética , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Masculino , Camundongos Endogâmicos BALB C , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
6.
Reprod Toxicol ; 95: 75-85, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32454085

RESUMO

Maternal smoking during the perinatal period is linked to adverse neonatal outcomes such as low birth weight and birth defects. Numerous studies have shown that cigarette smoke or nicotine exposure has a widespread effect on fetal nerve development. However, there exists a lack of understanding of what specific changes occur at the cellular level on persistent exposure to cigarette smoke during the differentiation of embryonic stem cells (ESCs) into neural cells. We previously investigated the effects of cigarette smoke extract (CSE) and its major component, nicotine, on the neural differentiation of mouse embryonic stem cells (mESCs). Differentiation of mESCs into neural progenitor cells (NPCs) or neural crest cells (NCCs) was induced with chemically defined media, and the cells were continuously exposed to CSE or nicotine during neural differentiation and development. Disturbed balance of the pluripotency state was observed in the NPCs, with consequent inhibition of neurite outgrowth and glial fibrillary acidic protein (Gfap) expression. These inhibitions correlated with the altered expression of proteins involved in the Notch-1 signaling pathways. The migration ability of NCCs was significantly decreased by CSE or nicotine exposure, which was associated with reduced protein expression of migration-related proteins. Taken together, we concluded that CSE and nicotine inhibit differentiation of mESCs into NPCs or NCCs, and may disrupt functional development of neural cells. These results imply that cigarette smoking during the perinatal period potentially inhibits neural differentiation and development of ESCs cells, leading to neonatal abnormal brain development and behavioral abnormalities.


Assuntos
Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Nicotiana , Fumaça/efeitos adversos , Produtos do Tabaco/efeitos adversos , Animais , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Environ Toxicol ; 35(1): 66-77, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31507073

RESUMO

The heart is the first organ formed in the developing fetus, and abnormal development of the heart is a major cause of fetal death. The adverse effects of cigarette smoke on the heart have been well established, but it is not well understood how cigarette smoke components regulate signaling molecules and cardiac specific functions during the early differentiation stage of the embryonic heart. In this study, we identified changes in the size of mouse embryoid bodies (mEBs) in response to treatment with cigarette smoke extract (CSE) via regulation of HDAC2, p53, p21, and cyclin D1 protein expression, which are cardiac differentiation and cell-cycle markers, respectively. In addition, exposure of mouse embryonic stem cells (mESCs) to cigarette smoke components inhibited myocardial differentiation and development through the expression of HDAC1, HDAC2, GATA4, NKX2-5, TBX5, HAND1, and Troponin I. Long-term exposure studies showed that CSE and nicotine may delay the development of mouse cardiomyocytes from mESCs and inhibit the contractibility, which is a fundamental function of the heart. Taken together, these findings suggest that cigarette smoke components, including nicotine, may affect abnormal myocardial differentiation and development.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Nicotiana/toxicidade , Fumaça/efeitos adversos , Animais , Ciclo Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Histona Desacetilase 2/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fumar/efeitos adversos
8.
Reprod Toxicol ; 90: 15-23, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31425785

RESUMO

Pesticides, antibiotics, and industrial excipients are widely used in agriculture, medicine, and chemical industry, respectively. They often end up in the environment, not only being not easily decomposed but also being accumulated. Moreover, they may cause serious toxic problems such as reproductive and developmental defects, immunological toxicity, and carcinogenesis. Hence, they are called environmental pollutants. It is known that the environmental pollutants easily enter the body through various channels such as respiration, ingestion of food, and skin contact etc. in everyday life. If they enter the mother through the placenta, they can cause the disturbance in embryo development as well as malfunction of organs after birth because early prenatal developmental process is highly sensitive to toxic chemicals and stress. Embryonic stem cells (ESCs) that consist of inner cell mass of blastocyst differentiate into distinct cell lineages via three germ layers such as the ectoderm, mesoderm, and endoderm due to their pluripotency. The differentiation process initiated from ESCs reflects dynamic nature of embryonic development. Therefore, ESCs have been used as a useful tool to investigate early developmental toxicities of a variety of stress. Based on relatively recent scientific results, this review would address toxicity of a few chemical substances that have been widely used as pesticide, antibiotics, and industrial excipient on ESCs based-prenatal developmental process. This review further suggests how they act on the viability of ESCs and/or early stages of cardiac and neuronal development derived from ESCs as well as on expression of pluripotency and/or differentiation markers through diverse mechanisms.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Coração/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Desenvolvimento Embrionário/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Humanos , Testes de Toxicidade/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...