Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 11536, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773201

RESUMO

Advances in modern medicine have extended human life expectancy, leading to a world with a gradually aging society. Aging refers to a natural decline in the physiological functions of a species over time, such as reduced pain sensitivity and reaction speed. Healthy-level physiological pain serves as a warning signal to the body, helping to avoid noxious stimuli. Physiological pain sensitivity gradually decreases in the elderly, increasing the risk of injury. Therefore, geriatric health care receives growing attention, potentially improving the health status and life quality of the elderly, further reducing medical burden. Health food is a geriatric healthcare choice for the elderly with Ganoderma tsuage (GT), a Reishi type, as the main product in the market. GT contains polysaccharides, triterpenoids, adenosine, immunoregulatory proteins, and other components, including anticancer, blood sugar regulating, antioxidation, antibacterial, antivirus, and liver and stomach damage protective agents. However, its pain perception-related effects remain elusive. This study thus aimed at addressing whether GT could prevent pain sensitivity reduction in the elderly. We used a galactose-induced animal model for aging to evaluate whether GT could maintain pain sensitivity in aging mice undergoing formalin pain test, hot water test, and tail flexes. Our results demonstrated that GT significantly improved the sensitivity and reaction speed to pain in the hot water, hot plate, and formalin tests compared with the control. Therefore, our animal study positions GT as a promising compound for pain sensitivity maintenance during aging.


Assuntos
Envelhecimento , Animais , Camundongos , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Masculino , Limiar da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Ganoderma/química , Modelos Animais de Doenças , Medição da Dor
2.
Mol Cell Neurosci ; 120: 103735, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35562037

RESUMO

A traumatic brain injury (TBI) causes abnormal proliferation of neuroglial cells, and over-release of glutamate induces oxidative stress and inflammation and leads to neuronal death, memory deficits, and even death if the condition is severe. There is currently no effective treatment for TBI. Recent interests have focused on the benefits of supplements or natural products like Ganoderma. Studies have indicated that immunomodulatory protein from Ganoderma microsporum (GMI) inhibits oxidative stress in lung cancer cells A549 and induces cancer cell death by causing intracellular autophagy. However, no evidence has shown the application of GMI on TBI. Thus, this study addressed whether GMI could be used to prevent or treat TBI through its anti-inflammation and antioxidative effects. We used glutamate-induced excitotoxicity as in vitro model and penetrating brain injury as in vivo model of TBI. We found that GMI inhibits the generation of intracellular reactive oxygen species and reduces neuronal death in cortical neurons against glutamate excitotoxicity. In neurite injury assay, GMI promotes neurite regeneration, the length of the regenerated neurite was even longer than that of the control group. The animal data show that GMI alleviates TBI-induced spatial memory deficits, expedites the restoration of the injured areas, induces the secretion of brain-derived neurotrophic factors, increases the superoxide dismutase 1 (SOD-1) and lowers the astroglial proliferation. It is the first paper to apply GMI to brain-injured diseases and confirms that GMI reduces oxidative stress caused by TBI and improves neurocognitive function. Moreover, the effects show that prevention is better than treatment. Thus, this study provides a potential treatment in naturopathy against TBI.


Assuntos
Lesões Encefálicas Traumáticas , Disfunção Cognitiva , Ganoderma , Animais , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/farmacologia , Ganoderma/metabolismo , Glutamatos/metabolismo , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Transtornos da Memória , Estresse Oxidativo
3.
PLoS One ; 17(4): e0266331, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35390035

RESUMO

Lingzhi has long been regarded as having life-prolonging effects. Research in recent years has also reported that Lingzhi possesses anti-tumor, anti-inflammatory, immunomodulatory, hepatoprotective, and anti-lipogenic effects. The D-galactose (D-gal, 100 mg/kg/day)-induced aging Long-Evans rats were simultaneously orally administered a DMSO extract of Ganoderma tsugae (GTDE, 200 µg/kg/day) for 25 weeks to investigate the effects of GTDE on oxidative stress and memory deficits in the D-galactose-induced aging rats. We found that GTDE significantly improved the locomotion and spatial memory and learning in the aging rats. GTDE alleviated the aging-induced reduction of dendritic branching in neurons of the hippocampus and cerebral cortex. Immunoblotting revealed a significant increase in the protein expression levels of the superoxide dismutase-1 (SOD-1) and catalase, and the brain-derived neurotrophic factor (BDNF) in rats that received GTDE. D-gal-induced increase in the lipid peroxidation product 4-hydroxynonenal (4-HNE) was significantly attenuated after the administration of GTDE, and pyrin domain-containing 3 protein (NLRP3) revealed a significant decrease in NLRP3 expression after GTDE administration. Lastly, GTDE significantly reduced the advanced glycosylation end products (AGEs). In conclusion, GTDE increases antioxidant capacity and BDNF expression of the brain, protects the dendritic structure of neurons, and reduces aging-induced neuronal damage, thereby attenuating cognitive impairment caused by aging.


Assuntos
Disfunção Cognitiva , Ganoderma , Envelhecimento/metabolismo , Animais , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Galactose/metabolismo , Galactose/toxicidade , Ganoderma/metabolismo , Aprendizagem em Labirinto , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo , Ratos , Ratos Long-Evans
4.
Artigo em Inglês | MEDLINE | ID: mdl-34587401

RESUMO

Tetrabromobisphenol A (TBBPA) is a brominated flame retardant that is used in a variety of consumer products such as electronic equipment, fire extinguishers, furniture, plastics, textiles, and kitchen hoods. Most studies show that the TBBPA production process and TBBPA in industrial and urban sewage waste result in extensive human exposure and environmental contamination. TBBPA can accumulate in organisms, particularly aquatic life, and is classified as a group 2A carcinogen (likely carcinogenic to humans) by the International Agency for Research on Cancer. This compound produces low acute toxicity, but chronic exposure may produce serious consequences. In this review, we focus on TBBPA toxicity by discussing results of various studies that were published in the last two decades. Studies show that TBBPA acts as an endocrine disruptor, causing neurobehavioral and immunotoxic effects, oxidative stress, and apoptosis. Although several experiments were performed in vitro and in vivo, human data are lacking, and thus, chronic toxic effects of TBBPA on humans are not well known, particularly in sensitive populations including pregnant women, newborns, children, and the elderly. Epidemiological studies that comprehensively assess TBBPA levels in biological fluids of different populations and in different pathological conditions are needed. Research on the impact of TBBPA, particularly regarding endocrine disorders and cancer, must also be performed.


Assuntos
Disruptores Endócrinos/toxicidade , Síndromes Neurotóxicas/etiologia , Bifenil Polibromatos/toxicidade , Animais , Organismos Aquáticos/efeitos dos fármacos , Disruptores Endócrinos/farmacocinética , Exposição Ambiental/efeitos adversos , Feminino , Humanos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Bifenil Polibromatos/farmacocinética , Gravidez
5.
Arh Hig Rada Toksikol ; 70(1): 18-29, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30956221

RESUMO

Exposure to alkyl anilines may lead to bladder cancer, which is the second most frequent cancer of the urogenital tract. 3,5-dimethylaniline is highly used in industry. Studies on its primary metabolite 3,5-dimethylaminophenol (3,5-DMAP) showed that this compound causes oxidative stress, changes antioxidant enzyme activities, and leads to death of different mammalian cells. However, there is no in vitro study to show the direct effects of 3,5-DMAP on human bladder and urothelial cells. Selenocompounds are suggested to decrease oxidative stress caused by some chemicals, and selenium supplementation was shown to reduce the risk of bladder cancer. The main aim of this study was to investigate whether selenocompounds organic selenomethionine (SM, 10 µmol/L) or inorganic sodium selenite (SS, 30 nmol/L) could reduce oxidative stress, DNA damage, and apoptosis in UROtsa cells exposed to 3,5-DMAP. 3,5-DMAP caused a dose-dependent increase in intracellular generation of reactive oxygen species, and its dose of 50 µmol/L caused lipid peroxidation, protein oxidation, and changes in antioxidant enzyme activities in different cellular fractions. The comet assay also showed single-strand DNA breaks induced by the 3,5-DMAP dose of 50 µmol/L, but no changes in double-strand DNA breaks. Apoptosis was also triggered. Both selenocompounds provided partial protection against the cellular toxicity of 3,5-DMAP. Low selenium status along with exposure to alkyl anilines can be a major factor in the development of bladder cancer. More mechanistic studies are needed to specify the role of selenium in bladder cancer.


Assuntos
Aminofenóis/toxicidade , Antioxidantes/farmacologia , Dano ao DNA/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Fatores de Proteção , Compostos de Selênio/farmacologia , Urotélio/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Estresse Oxidativo/efeitos dos fármacos
6.
Toxicol Sci ; 168(2): 405-419, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30590852

RESUMO

3,5-Dimethylaniline (3,5-DMA), a monocyclic aromatic amine, is widely present in a spectrum of sources including tobacco, dyes, combustion products, and suspended particulates. 3,5-DMA and its metabolites form superoxides, resulting in apoptosis or oncogenesis. Data of a direct effect of 3,5-DMA on the nervous system, especially the developing brain, are lacking. Therefore, we investigated the effects of 3,5-DMA and its metabolites on fetal neurite growth and brain development using in vitro cell cultures of primary cortical neurons to observe whether these compounds caused neuronal cytotoxicity and affected neurite structural development. With increasing concentrations of 3,5-DMA (10, 50, 100, 500, 1000 µM) and its major metabolite 5-dimethylaminophenol (3,5-DMAP) (10, 50, 100, 500, 1000 µM), reactive oxygen species (ROS), cytotoxicity, and DNA damage increased significantly in the cells and dendritic arborization decreased. The addition of 5 mM N-acetylcysteine, an ROS scavenger, reduced ROS in the cells and alleviated the neuronal damage. In vivo studies in Sprague Dawley pregnant rats suggested that exposure to 3,5-DMA (10, 30, 60, 100 mg/kg/day) subcutaneously from GD15 to GD17 led to fetal cerebral cortex thinning. BrdU labeling showed that 3,5-DMA reduced the number and generation of cortical cells. To detect the laminar position of newly generated neurons, cortex layer markers such as Satb2, Ctip2, and Tbr1 were used. 3,5-DMA perturbed the cortical layer distribution in developing fetal rats. In summary, this is the first study to provide evidence for 3,5-DMA and its metabolites causing anomalies of the fetal central nervous system development through ROS production.


Assuntos
Compostos de Anilina/toxicidade , Córtex Cerebral/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Estresse Oxidativo/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/etiologia , Compostos de Anilina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/embriologia , Córtex Cerebral/patologia , Dano ao DNA , Relação Dose-Resposta a Droga , Feminino , Masculino , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/metabolismo , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Organogênese/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Cultura Primária de Células , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
7.
PLoS One ; 13(10): e0205249, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30307971

RESUMO

Exposure to 3,5-dimethylaminophenol (3,5-DMAP), the metabolite of the 3-5-dimethylaniline, was shown to cause high levels of oxidative stress in different cells. The aim of the present work was to observe whether this metabolite can lead to cytotoxicity, oxidative stress, DNA damage and cell cycle changes in non-small cell lung cancer A549 cells. 3,5-DMAP caused a dose-dependent increase in cytotoxicity, generation of superoxide (O2-.), inductions in the enzyme activities orchestrating cellular antioxidant balance, increases in lipid peroxidation as well as DNA damage. However, 3,5-DMAP showed significantly lower cytotoxicity towards human lung fibroblast (HLF) cells. 3,5-DMAP also led to molecular events, like inducing apoptotic markers (ie. p53, Bad, Bax and cytochrome c); decreasing anti-apoptotic proteins (Bcl-2) and alterations in cell cycle. Our findings indicate that the cytotoxicity caused by this particular alkylaniline metabolite led to initiation of caspase 3-mediated apoptosis. Furthermore, 3,5-DMAP attenuated carcinogenic properties like migration capacity of A549 cells and eventually inhibited growth of A549 cells in an in vivo mouse model. Tumor sections showed that 3,5-DMAP down-regulated c-Myc expression but up-regulated p53 and cytochrome c, all of which might result in tumor growth arrest. Co-treatment with N-acetylcysteine provided reductions in cytotoxicity and positively modulated genetic events induced by 3,5-DMAP in A549 cells. In conclusion, our findings demonstrate 3,5-DMAP may be a potential anti-cancer drug in cancer, due to its self redox cycling properties.


Assuntos
Aminofenóis/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Dano ao DNA/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Células A549 , Acetilcisteína/farmacologia , Aminofenóis/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Fibroblastos , Sequestradores de Radicais Livres/farmacologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biochem Pharmacol ; 151: 1-8, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29499168

RESUMO

IL-17A is implicated in many aspects of pathogenesis of severe asthma, including inducing neutrophilic inflammation, airway hyperresponsiveness, steroid insensitivity and airway remodeling. Diesel exhaust particles (DEP) emission from vehicles has been shown to expand Th17 cells to increase IL-17A release that contributes to DEP-mediated exacerbation of asthma severity. It is not known whether non-immune cells in airways may also release IL-17A in response to DEP exposure. In this study, We found IL-17A expression was upregulated in the epithelium of severe allergic asthma patients from high road traffic pollution areas compared to those in low. Furthermore, we found DEP concentration-dependently increased IL-17A synthesis and release by 122.3 ±â€¯15.72% and 235.5 ±â€¯18.37%, respectively in primary bronchial epithelial cells (PBEC), accompanied with increased ROS production. Pretreatment of ROS scavenger (NAC) significantly inhibited DEP-induced IL-17A mRNA expression. DEP-induced IκBα degradation can be inhibited by NAC. We also found DEP increased p65 and RelB subunits expression, and pretreatment of NF-κB inhibitor (SN50) also inhibited DEP-induced IL-17A expression. We further found DEP increased NF-κB subunit RelB recruitment to IL-17A promoter in PBEC and airway tissue of severe allergic asthma patients from high road traffic pollution areas. These results indicate DEP stimulates IL-17A expression in airway epithelium through ROS/NF-κB pathway, and provide a possible link between traffic pollution exposure and IL-17A-related responses in severe allergic asthma patients.


Assuntos
Asma/imunologia , Interleucina-17/genética , NF-kappa B/metabolismo , Material Particulado/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Emissões de Veículos/toxicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Mucosa Respiratória/imunologia , Índice de Gravidade de Doença , Transdução de Sinais , Regulação para Cima
9.
J Environ Pathol Toxicol Oncol ; 36(2): 131-150, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29199594

RESUMO

The association between inflammation and cancer has long been recognized. Several studies have found that different types of tumors develop at sites of chronic inflammation. It is stated that over 15%-20% of malignancies worldwide can be related to infections caused by viruses, bacteria, and schistosomes. Inflammatory conditions are characterized by overexpression of inducible nitric oxide synthase (iNOS) and overproduction of nitric oxide/reactive nitrogen species (ROSs/RNSs) in epithelial cells. Reactive oxygen species (ROSs) may also lead to cellular alterations and eventually to inflammation. A variety of chronic infectious diseases can generate steady-state levels of ROSs/RNSs within infected cells and possibly lead to different types of DNA lesions. Accumulation of DNA lesions may finally lead to mutations that may activate oncogenes or inactivate tumor suppressor genes. Helicobacter pylori has been shown to generate ROSs/RNSs, induce DNA damage, and lead to chronic inflammation in gastric epithelial cells. A limited number of studies have addressed the effects of Helicobacter pylori on DNA damage, particularly its impact on single-strand and double-strand DNA breaks. This bacterium is classified as a Group I carcinogen by the International Agency for Research on Cancer on the basis of numerous animal and epidemiological studies. Chronic Helicobacter pylori infection can lead to increased risk of gastric cancer and mucosa-associated lymphoid tissue (MALT) lymphoma. This review addresses the DNA-damaging and double-strand break-inducing effects of different microorganisms and their toxins, specifically focusing on Helicobacter pylori.


Assuntos
Quebras de DNA de Cadeia Dupla , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Animais , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos , Dano ao DNA , Humanos , Camundongos , Ratos
10.
J Environ Pathol Toxicol Oncol ; 36(2): 171-190, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29199597

RESUMO

The development of hepatocellular carcinoma (HCC) is a multistep process. In HCC, progressive and morphologically distinct preneoplastic lesions/alterations associated with chronic liver injury, inflammation, hepatocellular degeneration/regeneration, necrosis, and small-cell dysplasia can be observed. The incidence of HCC exhibits regional and ethnic differences. Several cytotoxic and DNA-damaging chemicals are suggested to be the underlying causes of HCC-for example, acrylamide, perfluorooctanoic acid (PFOA), polychlorinated biphenyls (PCBs), benzo(a)pyrene (BaP), perfluorinated chemicals (PFCs), vinyl chloride monomer (VCM), and dietary contaminants (aflatoxins, ochratoxins). Also suggested are substances of abuse (alcohol) and biological agents, such as hepatitis B and C and human immunodeficiency virus 1 (HIV-1). These can act through genetic and/or epigenetic mechanisms. This review will shortly address the genetic and epigenetic mechanisms of HCC and focus on cytotoxic and DNA-damaging chemicals and biological agents, exposure to which are suggested to lead to HCC initiation, promotion, and/or progression.


Assuntos
Carcinoma Hepatocelular/genética , Poluentes Ambientais/toxicidade , Epigênese Genética , Neoplasias Hepáticas/genética , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/epidemiologia , Humanos , Incidência , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/epidemiologia , Camundongos , Ratos , Fatores de Risco
11.
Carcinogenesis ; 38(3): 336-345, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28426879

RESUMO

Di-(2-ethylhexyl) phthalate (DEHP), the common plasticizer used in the production of polyvinyl chloride, can be converted to the more potent metabolite mono-ethylhexyl phthalate (MEHP). Epidemiological studies have shown an association with elevated induction of rat hepatic cancer and reproductive toxicity in response to MEHP exposure. However, the mechanism of genotoxicity and carcinogenicity induced by MEHP treatment remains unclear. As a means to elucidate the mechanisms of action, lethality and mutagenicity in the adenine phosphoribosyltransferase (aprt+/-) gene induced in several CHO cell types by MEHP were assessed. Dose-response relationships were determined in the parental AA8 cell line, its nucleotide repair-deficient UV5 and base repair-deficient EM9 subclones, and also in AS52 cells harboring the bacterial guanine-hypoxanthine phosphoribosyltransferase (gpt) gene and its derived AS52-XPD-knockdown and AS52-PARP-1-knockdown cells. Treatment of AS52 with MEHP led to intracellular production of reactive oxygen species (ROS) and DNA strand breaks in a dose-dependent manner. Separately, mutations in the gpt gene of AS52 cells were characterized and found to be dominated by G:C to A:T and A:T to G:C transitions. Independent AS52-mutant cell (ASMC) clones were collected for the sequential in vivo xenograft tumorigenic studies, 4 of total 20 clones had aggressive tumor growth. Moreover, microarray analysis indicated miR-let-7a and miR-125b downregulated in ASMC, which might raise oncogenic MYC and RAS level and activate ErbB pathway. Comparative evaluation of the results indicates that the principal mechanism of this mutagenic action is probably to be through generation of ROS, causing base excision damage resulting in carcinogenicity.


Assuntos
Dietilexilftalato/análogos & derivados , Dietilexilftalato/metabolismo , Mutagênese/genética , Poli(ADP-Ribose) Polimerase-1/genética , Animais , Células CHO , Cricetinae , Cricetulus , Dano ao DNA/efeitos dos fármacos , Humanos , Mutagênese/efeitos dos fármacos , Testes de Mutagenicidade , Mutação/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo
12.
Cardiovasc Toxicol ; 17(4): 384-392, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-26965709

RESUMO

Epidemiological studies suggest that an increase of diesel exhaust particles (DEP) in ambient air corresponds to an increase in hospital-recorded myocardial infarctions within 48 h after exposure. Among the many theories to explain this data are endothelial dysfunction and translocation of DEP into vasculature. The mechanisms for such DEP-induced vascular permeability remain unknown. One of the major mechanisms underlying the effects of DEP is suggested to be oxidative stress. Experiments have shown that DEP induce the generation of reactive oxygen species (ROS), such as superoxide anion and H2O2 in the HUVEC tube cells. Transcription factor Nrf2 is translocated to the cell nucleus, where it activates transcription of the antioxidative enzyme HO-1 and sequentially induces the release of vascular permeability factor VEGF-A. Furthermore, a recent study shows that DEP-induced intracellular ROS may cause the release of pro-inflammatory TNF-α and IL-6, which may induce endothelial permeability as well by promoting VEGF-A secretion independently of HO-1 activation. These results demonstrated that the adherens junction molecule, VE-cadherin, becomes redistributed from the membrane at cell-cell borders to the cytoplasm in response to DEP, separating the plasma membranes of adjacent cells. DEP were occasionally found in endothelial cell cytoplasm and in tube lumen. In addition, the induced ROS is cytotoxic to the endothelial tube-like HUVEC. Acute DEP exposure stimulates ATP depletion, followed by depolarization of their actin cytoskeleton, which sequentially inhibits PI3K/Akt activity and induces endothelial apoptosis. Nevertheless, high-dose DEP augments tube cell apoptosis up to 70 % but disrupts the p53 negative regulator Mdm2. In summary, exposure to DEP affects parameters influencing vasculature permeability and viability, i.e., oxidative stress and its upregulated antioxidative and pro-inflammatory responses, which sequentially induce vascular permeability factor, VEGF-A release and disrupt cell-cell junction integrity. While exposure to a low dose of DEP actin triggers cytoskeleton depolarization, reduces PI3K/Akt activity, and induces a p53/Mdm2 feedback loop, a high dose causes apoptosis by depleting Mdm2. Addition of ROS scavenger N-acetyl cysteine suppresses DEP-induced oxidative stress efficiently and reduces subsequent damages by increasing endogenous glutathione.


Assuntos
Permeabilidade Capilar/fisiologia , Endotélio Vascular/metabolismo , Mediadores da Inflamação/metabolismo , Material Particulado/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Emissões de Veículos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Permeabilidade Capilar/efeitos dos fármacos , Citotoxinas/toxicidade , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
13.
Toxicol Sci ; 156(1): 72-83, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28013216

RESUMO

Epidemiological studies suggest that an increase of PM2.5 diesel exhaust particles (DEP) in ambient air corresponds to increased myocardial infarctions and atherosclerosis. When exposed to DEP, endothelial cells exhibit increases in oxidative stress and apoptosis, but the role of autophagy in this DEP-induced cell death remains unclear. Here, we suggest that acute DEP exposure produces intracellular reactive oxygen species (ROS) leading to induction of DEP internalization, endothelial dysfunction, and pro-inflammation in an in vitro human umbilical vein endothelial cells (HUVEC) model. This study found that increases in intracellular oxidative stress and cellular internalization of DEP occurred within 2 h of exposure to DEP. After 2 h of DEP exposure, Mdm2 expression was increased, which triggered cellular autophagy after 4 h of DEP exposure and suppressed cellular senescence. Unfortunately, phagocytized DEP could not be eliminated by cellular autophagy, which led to a continuous buildup of ROS, an increased release of cytokines, and an increased expression of anchoring molecules. After 12 h of DEP exposure, HUVEC reduced Mdm2 expression leading to increased p53 expression, which triggered apoptosis and ultimately resulted in endothelial dysfunction. On the other hand, when cells lacked the ability to induce autophagy, DEP was unable to induce cell senescence and most of the cells survived with only a small percentage of the cells undergoing necrosis. The results presented in this study clearly demonstrate the role cellular autophagy plays in DEP-induced atherosclerosis.


Assuntos
Poluentes Atmosféricos/toxicidade , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Material Particulado/toxicidade , Emissões de Veículos/toxicidade , Poluentes Atmosféricos/química , Poluentes Atmosféricos/isolamento & purificação , Poluentes Atmosféricos/metabolismo , Proteína 12 Relacionada à Autofagia/antagonistas & inibidores , Proteína 12 Relacionada à Autofagia/genética , Proteína 12 Relacionada à Autofagia/metabolismo , Biomarcadores/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Tamanho da Partícula , Material Particulado/química , Material Particulado/isolamento & purificação , Material Particulado/metabolismo , Fagocitose/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Fuligem/química , Fuligem/toxicidade , Tóquio , Vasculite/induzido quimicamente , Vasculite/imunologia , Vasculite/metabolismo , Vasculite/patologia , Emissões de Veículos/análise
14.
Sci Rep ; 6: 32373, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27577752

RESUMO

Oxidative stress and inflammatory insults are the major instigating events of bacterial intrauterine infection that lead to fetal brain injury. The purpose of this study is to investigate the remedial effects of N-acetyl-cysteine (NAC) for inflammation-caused deficits in brain development. We found that lipopolysaccharide (LPS) induced reactive oxygen species (ROS) production by RAW264.7 cells. Macrophage-conditioned medium caused noticeable cortical cell damage, specifically in cortical neurons. LPS at 25 µg/kg caused more than 75% fetal loss in rats. An increase in fetal cortical thickness was noted in the LPS-treated group. In the enlarged fetal cortex, laminar positioning of the early born cortical cells expressing Tbr1 and Ctip2 was disrupted, with a scattered distribution. The effect was similar, but minor, in later born Satb2-expressing cortical cells. NAC protected against LPS-induced neuron toxicity in vitro and counteracted pregnancy loss and alterations in thickness and lamination of the neocortex in vivo. Fetal loss and abnormal fetal brain development were due to LPS-induced ROS production. NAC is an effective protective agent against LPS-induced damage. This finding highlights the key therapeutic impact of NAC in LPS-caused abnormal neuronal laminar distribution during brain development.


Assuntos
Acetilcisteína/administração & dosagem , Encéfalo/crescimento & desenvolvimento , Inflamação/tratamento farmacológico , Proteínas do Tecido Nervoso/genética , Proteínas Repressoras/genética , Proteínas com Domínio T/genética , Proteínas Supressoras de Tumor/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Desenvolvimento Fetal/efeitos dos fármacos , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Masculino , Proteínas de Ligação à Região de Interação com a Matriz/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Ratos , Fatores de Transcrição/genética
15.
J Environ Pathol Toxicol Oncol ; 35(1): 43-58, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27279583

RESUMO

Epithelial-mesenchymal transition (EMT) is a process during which epithelial cells lose their polarity and ability to adhere. Instead, they gain properties to move, migrate through the extracellular matrix, become invasive, and finally become mesenchymal stem cells. This trans-differentiation is critical for embryo development, wound healing, and stem cell behavior. However, this same phenomenon is also observed in cancer progression. Phthalates and bisphenol A (BPA) are endocrine-disrupting chemicals (EDCs) that are linked to complex human diseases. These chemicals are suggested to disrupt normal hormonal balance (usually by existing estrogenic/antiandrogenic properties) and stimulate the development of reproductive tumors and steroid hormone-dependent cancers, such as breast cancer. Di(2-ethylhexyl) phthalate (DEHP), the most abundant phthalate, was shown to induce DNA damage in human cells via multiple molecular signals that include altered apoptosis and mitotic rate, increased cell proliferation, tumor mobility, and invasiveness of tumor cells. DEHP was also shown to inhibit gap junction intercellular communication and tight junctions and promote EMT. Phthalates may also cause the proliferation and metastasis of cancer cells and tumor progression via up-regulating histone deacetylase 6 (HDAC6). Phthalates can activate peroxisome proliferator activated receptors (PPARs) that may eventually lead to high proliferation of cancer cells. However, in ovarian cells the expression of Snail, Slug, and vimentin was enhanced by the treatment of BPA, whereas E-cadherin was decreased. Mechanistic studies are needed to show the underlying mechanisms of EMT caused by different EDCs.


Assuntos
Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fenóis/toxicidade , Ácidos Ftálicos/toxicidade , Animais , Humanos , Camundongos , Ratos
16.
PLoS One ; 11(5): e0155469, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27171432

RESUMO

Traditional lung cancer treatments involve chemical or radiation therapies after surgical tumor removal; however, these procedures often kill normal cells as well. Recent studies indicate that chemotherapies, when combined with Traditional Chinese Medicines, may offer a new way to treat cancer. In vitro tests measuring the induction of autophagy and/or apoptosis were used to examine the cytotoxicity of SBPE, commonly used for lung inflammation on A549 cell line. The results indicated that intercellular levels of p62 and Atg12 were increased, LC3-I was cleaved into LC3-II, and autophagy was induced with SBPE only. After 24 hours, the apoptotic mechanism was induced. If the Cisplatin was added after cells reached the autophagy state, we observed synergistic effects of the two could achieve sufficient death of lung cancer cells. Therefore, the Cisplatin dosage used to induce apoptosis could be reduced by half, and the amount of time needed to achieve the inhibitory concentration of 50% was also half that of the original. In addition to inducing autophagy within a shortened period of time, the SBPE and chemotherapy drug combination therapy was able to achieve the objective of rapid low-dosage cancer cell elimination. Besides, SBPE was applied with Gemcitabine or Paclitaxel, and found that the combination treatment indeed achieve improved lung cancer cell killing effects. However, SBPE may also be less toxic to normal cells.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Medicina Tradicional Chinesa , Extratos Vegetais/uso terapêutico , Células A549 , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Humanos , Neoplasias Pulmonares/patologia , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Extratos Vegetais/farmacologia , Reprodutibilidade dos Testes
17.
Am J Chin Med ; 44(2): 355-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27080945

RESUMO

Epidemiological studies show increased particulate matter (PM[Formula: see text]) particles in ambient air are correlated with increased myocardial infarctions. Given the close association of capillaries and alveoli, the dysfunction is caused when inhaled PM[Formula: see text] particles come in close proximity to capillary endothelial cells. We previously suggested that the inhalation of PM[Formula: see text] diesel exhaust particles (DEP) induces oxidative stress and upregulates the Nrf2/HO-1 pathway, inducing vascular permeability factor VEGFA secretion, which results in cell-cell adherens junction disruption and PM[Formula: see text] transmigratation into circulation. Here, we minimized the level that PM[Formula: see text] traveled in the bloodstream by pre-supplementing with a traditional Chinese medicine (TCM) Ganoderma tsugae DMSO extract (GTDE) prior to PM[Formula: see text] exposure. Our results show that PM[Formula: see text] caused alterations in enzyme activities and cellular anti-oxidant balance. We found decreased glutathione levels, a reduced cellular redox ratio, increased ROS generation and cytotoxicity in the cellular fractions. The oxidative stress caused DNA damage and apoptosis, likely causing downstream molecular events that trigger vasculature permeabilization and, eventually, cardiovascular disorders. Our results show long-term GTDE treatment increased endogenous glutathione level, while PM[Formula: see text]-reduced glutathione levels and the cellular redox ratio. GTDE was protective against the genotoxic and apoptotic effects initiated by PM[Formula: see text] oxidative stress. Vascular permeability revealed that PM[Formula: see text] only accumulated on the surface of cells after GTDE treatment; no penetration was detected. After two weeks of GTDE treatment, VEGFA secretion was significantly reduced in human umbilical vein endothelial cells (HUVEC) and endothelial cell migration was blocked. Our results suggest GTDE prevents PM[Formula: see text] transmigration into the bloodstream, and the resultant dysfunction, by inhibiting oxidative stress production and endothelial permeability.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Ganoderma/química , Material Particulado/efeitos adversos , Extratos Vegetais/administração & dosagem , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Glutationa/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/metabolismo , Infarto do Miocárdio/induzido quimicamente , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Material Particulado/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
PLoS One ; 10(7): e0131911, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26148005

RESUMO

Exposure to diesel exhaust particles (DEP) is associated with pulmonary and cardiovascular diseases. Previous studies using in vitro endothelial tubes as a simplified model of capillaries have found that DEP-induced ROS increase vascular permeability with rearrangement or internalization of adherens junctional VE-cadherin away from the plasma membrane. This allows DEPs to penetrate into the cell and capillary lumen. In addition, pro-inflammatory cytokines are up-regulated and mediate vascular permeability in response to DEP. However, the mechanisms through which these DEP-induced pro-inflammatory cytokines increase vascular permeability remain unknown. Hence, we examined the ability of DEP to induce permeability of human umbilical vein endothelial cell tube cells to investigate these mechanisms. Furthermore, supplementation with NAC reduces ROS production following exposure to DEP. HUVEC tube cells contributed to a pro-inflammatory response to DEP-induced intracellular ROS generation. Endothelial oxidative stress induced the release of TNF-α and IL-6 from tube cells, subsequently stimulating the secretion of VEGF-A independent of HO-1. Our data suggests that DEP-induced intracellular ROS and release of the pro-inflammatory cytokines TNF- α and IL-6, which would contribute to VEGF-A secretion and disrupt cell-cell borders and increase vasculature permeability. Addition of NAC suppresses DEP-induced ROS efficiently and reduces subsequent damages by increasing endogenous glutathione.


Assuntos
Cisteína/farmacologia , Citocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Inflamação/metabolismo , Substâncias Protetoras/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Emissões de Veículos/toxicidade , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Capilares/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Heme Oxigenase-1/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/induzido quimicamente , Interleucina-6/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Material Particulado/efeitos adversos , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
J Photochem Photobiol B ; 148: 21-30, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25863440

RESUMO

Phenothiazine and its derivatives are the most effective antipsychotic drugs. They have been used in the treatment of serious mental and emotional symptoms including bipolar disorder, organic psychoses, psychotic depression and schizophrenia. However, these drugs cause serious side effects such as akathisia, hyperprolactinaemia and neuroleptic malignant syndrome. In this work we investigated the molecular recognition of two typical phenothiazine compounds, phenosafranin and safranin O by the most pivotal heme protein hemoglobin using steady state and time-resolved fluorescence, extrinsic 8-anilino-1-naphthalenesulfonic acid (ANS) fluorescent probe, circular dichroism (CD) along with computational modeling. Results show phenothiazines complex with protein via formation of adducts at 298 K with moderate strengths of 3.555×10(4) M(-1) and 2.567×10(4) M(-1) for the hemoglobin-phenosafranin and hemoglobin-safranin O, respectively. We also found phenothiazines were effectors at the protein allosteric site, which affects the allosteric equilibrium. Further, time-resolved fluorescence and hydrophobic ANS experiments showed the static mechanism is dominated for the shrinkage in the fluorescence intensity of ß-37 Trp residue at the α1ß2 interface. The stoichiometric proportion of the protein-drug adduct is 1:1, as derived from Job's plot. Several crucial noncovalent bonds, including hydrogen bonds, π-π stacking and hydrophobic interactions played a major role in stabilizing the noncovalent conjugates. Based on three-dimensional fluorescence, we concluded that the conformation of hemoglobin is partially destabilized after recognition with phenothiazines. These alterations were confirmed by far-UV CD spectra that showed the α-helix of protein decreased from 78.3% in free hemoglobin to 62.8% and 64.8% in hemoglobin-phenosafranin and hemoglobin-safranin O, respectively. Computer-aided molecular docking was consistent, indicating that both phenothiazines are situated within the pocket composed of α1 and ß2 subunits. Affinity of hemoglobin to phenosafranin is superior compared with safranin O. This difference may be explained by the methyl group substituent on A- and C-rings, and by the different molecular volume between phenosafranin and safranin O. Our data provides further explanation of the overall pharmacokinetics of phenothiazines and sheds light on the allosteric regulation of heme proteins.


Assuntos
Antipsicóticos/química , Hemoglobinas/metabolismo , Fenotiazinas/química , Naftalenossulfonato de Anilina/química , Antipsicóticos/metabolismo , Sítios de Ligação , Dicroísmo Circular , Corantes Fluorescentes/química , Hemoglobinas/química , Simulação de Acoplamento Molecular , Fenazinas/química , Fenazinas/metabolismo , Fenotiazinas/metabolismo , Processos Fotoquímicos , Ligação Proteica , Estrutura Terciária de Proteína , Espectrometria de Fluorescência
20.
J Appl Toxicol ; 35(5): 466-77, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25178734

RESUMO

Exposure to monocyclic aromatic alkylanilines (MAAs), namely 2,6-dimethylaniline (2,6-DMA), 3,5-dimethylaniline (3,5-DMA) and 3-ethylaniline (3-EA), was significantly and independently associated with bladder cancer incidence. 3,5-DMAP (3,5-dimethylaminophenol), a metabolite of 3,5-DMA, was shown to induce an imbalance in cytotoxicity cellular antioxidant/oxidant status, and DNA damage in mammalian cell lines. This study was designed to evaluate the protective effect of ascorbic acid (Asc) against the cytotoxicity, reactive oxygen species (ROS) production, genotoxicity and epigenetic changes induced by 3,5-DMAP in AA8 Chinese Hamster Ovary (CHO) cells. In different cellular fractions, 3,5-DMAP caused alterations in the enzyme activities orchestrating a cellular antioxidant balance, decreases in reduced glutathione levels and a cellular redox ratio as well as increases in lipid peroxidation and protein oxidation. We also suggest that the cellular stress caused by this particular alkylaniline leads to both genetic (Aprt mutagenesis) and epigenetic changes in histones 3 and 4 (H3 and H4). This may further cause molecular events triggering different pathological conditions and eventually cancer. In both cytoplasm and nucleus, Asc provided increases in 3,5-DMAP-reduced glutathione levels and cellular redox ratio and decreases in the lipid peroxidation and protein oxidation. Asc was also found to be protective against the genotoxic and epigenetic effects initiated by 3,5-DMAP. In addition, Asc supplied protection against the cell cycle (G1 phase) arrest induced by this particular alkylaniline metabolite.


Assuntos
Aminofenóis/toxicidade , Ácido Ascórbico/farmacologia , Epigênese Genética/efeitos dos fármacos , Compostos de Anilina/toxicidade , Animais , Antioxidantes/metabolismo , Células CHO , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Dano ao DNA/efeitos dos fármacos , Glutationa/metabolismo , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Histonas/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA