Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Ecotoxicol Environ Saf ; 280: 116520, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38833985

RESUMO

Early studies have shown that the gut microbiota is a critical target during cadmium exposure. The prebiotic activity of epigallocatechin-3-gallate (EGCG) plays an essential role in treating intestinal inflammation and damage. However, the exact intestinal barrier protection mechanism of EGCG against cadmium exposure remains unclear. In this experiment, four-week-old mice were exposed to cadmium (5 mg kg-1) for four weeks. Through 16 S rDNA analysis, we found that cadmium disrupted the gut microbiota and inhibited the indole metabolism pathway of tryptophan (TRP), which serves as the principal microbial production route for endogenous ligands to activate the aryl hydrocarbon receptor (AhR). Additionally, cadmium downregulated the intestinal AhR signaling pathway and harmed the intestinal barrier function. Treatment with EGCG (20 mg kg-1) and the AhR agonist 6-Formylindolo[3,2-b] carbazole (FICZ) (1 µg/d) significantly activated the AhR pathway and alleviated intestinal barrier injury. Notably, EGCG partially restored the gut microbiota and upregulated the TRP-indole metabolism pathway to increase the level of indole-related AhR agonists. Our findings demonstrate that cadmium dysregulates common gut microbiota to disrupt TRP metabolism, impairing the AhR signaling pathway and intestinal barrier. EGCG reduces cadmium-induced intestinal functional impairment by intervening in the intestinal microbiota to metabolize AhR agonists. This study offers insights into the toxic mechanisms of environmental cadmium and a potential mechanism to protect the intestinal barrier with EGCG.


Assuntos
Cádmio , Catequina , Microbioma Gastrointestinal , Receptores de Hidrocarboneto Arílico , Transdução de Sinais , Triptofano , Animais , Catequina/análogos & derivados , Catequina/farmacologia , Receptores de Hidrocarboneto Arílico/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Triptofano/metabolismo , Triptofano/análogos & derivados , Cádmio/toxicidade , Transdução de Sinais/efeitos dos fármacos , Masculino , Intestinos/efeitos dos fármacos , Intestinos/patologia , Camundongos Endogâmicos C57BL , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Indóis/farmacologia , Carbazóis/farmacologia
2.
Lipids Health Dis ; 22(1): 198, 2023 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-37978497

RESUMO

BACKGROUND: Roughly 10 -15% of global populace suffer from Chronic Kidney Disease(CKD). A major secondary disease that can progress to end-stage renal disease (ESRD) is obesity-associated kidney disease (ORG). Although clinical management strategies are currently available, morbidity and mortality rates are increasing. Thus, new solutions are needed. Intestinal permeability, systemic inflammation, and aberrant intestinal metabolites have all been linked to ORG. PURPOSE: ACT001 has anti-inflammatory, redox-regulatory and antitumour activities. The current study was designed to examine how ACT001 affects ORG and analyze the fundamental processes. METHODS: A high-fat diet (HFD) was used to generate ORG in female C57BL/6 J mice. ORG mice were divided into three groups at random: HFD, HFD + ACT001, HFD + polyphosphocholine (PPC). To assess renal and colonic damage, periodic acid-Schiff (PAS) and hematoxylin-eosin (HE) staining were used. Following that, renal inflammation, oxidative stress, lipid deposition, colonic inflammation, and intestinal permeability were evaluated by protein blotting, polymerase chain reaction (PCR), immunohistochemistry, and immunofluorescence staining. Lastly, the SCFAs content was assessed by gas chromatographymass spectrometry. RESULTS: Mice in the HFD group displayed more severe albuminuria, glomerular hypertrophy, renal oxidative damage, inflammation, and lipid accumulation than mice with the normal diet (ND) group, as well as lower levels of intestinal SCFA valproic acid, colonic inflammation, and tight junction protein downregulation. ACT001 treatment restores the content of valproic acid in intestinal SCFAs, promotes the binding of SCFAs to renal GPR43, activates the AMPK signalling pathway. Therefore, it promotes the Nrf2-Keap1 signalling pathway and inhibits the NF-κB signalling pathway. SCFAs, additionally, augment colonic GPR43 concentrations, diminishing NLRP3 inflammasome expression and restoring ZO-1 and occludin protein levels. CONCLUSION: This study is the first to look at ACT001's potential as a treatment for obesity-related kidney disease. Regulating GPR43 and AMPK signalling pathways, By controlling the GPR43 and AMPK signalling pathways, ACT001 improves colitis and the intestinal mucosal barrier, decreases renal lipid deposition, and suppresses inflammation and oxidative stress in the kidneys. According to this study, ACT001 could be a viable ORG therapy option.


Assuntos
Proteínas Quinases Ativadas por AMP , Nefropatias , Feminino , Camundongos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ácido Valproico , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Rim/metabolismo , Inflamação/patologia , Nefropatias/complicações , Nefropatias/patologia , Obesidade/metabolismo
3.
Nutrients ; 15(20)2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37892424

RESUMO

Eriodictyol occurs naturally in a variety of fruits and vegetables, and has drawn significant attention for its potential health benefits. This study aims to look into the effects of eriodictyol on acute liver injury (ALI) induced by LPS/D-GalN and elucidate its potential molecular biological mechanisms. A total of 47 targets were predicted for the treatment of ALI with eriodictyol, and the PI3K/AKT signaling pathway played a key role in the anti-ALI processing of this drug. The in vivo experiment showed that eriodictyol can effectively reduce liver function-related biochemical indicators such as ALT, AST, and AKP. Eriodictyol can also up-regulate the levels of SOD and GSH, and inhibit the release of IL-1ß, IL-6, and TNF-α. Additionally, TUNEL staining, immunohistochemistry, and RT-PCR experiments showed that eriodictyol activated the PI3K/AKT pathway and decreased the expression of Bax, caspase3, and caspase8 while increasing the expression of Bcl-2 m-RNA. Finally, molecular docking experiments and molecular dynamics simulations confirmed the stable binding between eriodictyol and PI3K, AKT molecules. This study showed that eriodictyol can activate the PI3K/AKT signaling pathway to alleviate ALI-related oxidative stress and apoptosis.


Assuntos
Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Lipopolissacarídeos/farmacologia , Simulação de Acoplamento Molecular , Transdução de Sinais , Fígado/metabolismo , Estresse Oxidativo , Apoptose
4.
Aging (Albany NY) ; 15(12): 5887-5916, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37379130

RESUMO

Swertia cincta Burkill is widely distributed along the southwestern region of China. It is known as "Dida" in Tibetan and "Qingyedan" in Chinese medicine. It was used in folk medicine to treat hepatitis and other liver diseases. To understand how Swertia cincta Burkill extract (ESC) protects against acute liver failure (ALF), firstly, the active ingredients of ESC were identified using liquid chromatography-mass spectrometry (LC-MS), and further screening. Next, network pharmacology analyses were performed to identify the core targets of ESC against ALF and further determine the potential mechanisms. Finally, in vivo experiments as well as in vitro experiments were conducted for further validation. The results revealed that 72 potential targets of ESC were identified using target prediction. The core targets were ALB, ERBB2, AKT1, MMP9, EGFR, PTPRC, MTOR, ESR1, VEGFA, and HIF1A. Next, KEGG pathway analysis showed that EGFR and PI3K-AKT signaling pathways could have been involved in ESC against ALF. ESC exhibits hepatic protective functions via anti-inflammatory, antioxidant, and anti-apoptotic effects. Therefore, the EGFR-ERK, PI3K-AKT, and NRF2/HO-1 signaling pathways could participate in the therapeutic effects of ESC on ALF.


Assuntos
Falência Hepática Aguda , Swertia , Humanos , Swertia/metabolismo , Lipopolissacarídeos/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/tratamento farmacológico , Transdução de Sinais , Apoptose , Estresse Oxidativo , Receptores ErbB/metabolismo
5.
Int Immunopharmacol ; 112: 109239, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36113316

RESUMO

Pseudomonas aeruginosa is a dangerous pathogen causing nosocomial pneumonia. P. aeruginosa infection-induced liver damage is another fatal threat, and antibiotic treatment is not effective in relieving P. aeruginosa virulence-triggered damage. We here evaluated the protective effect of epigallocatechin gallate (EGCG), a substance that inhibits virulence of P. aeruginosa through quorum quenching, on liver damage secondary to P. aeruginosa infection. Mice were pretreated with EGCG (20, 40, and 80 mg/kg) for 3 days, and then infected with P. aeruginosa through intratracheal instillation to model acute pneumonia. The mice were sacrificed after 24 h of infection, and samples were harvested for subsequent analysis. EGCG significantly decreased the serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Histopathological changes of liver were significantly ameliorated by EGCG. It also significantly reduced oxidative stress that induced liver damage in P. aeruginosa infection, which relied not on the activation of the Nrf2-HO-1 pathway but on the upregulation of the activity of antioxidative enzymes. Then, the inflammatory response in the liver was tested. EGCG inhibited the release of pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-6) by blocking the inflammation regulating signaling of the TLR4-myD88-NF-κB pathway. EGCG upregulated the activation of nuclear receptors to stronger the liver protective activity against P. aeruginosa infection. Conclusively, EGCG exhibited a significant hepatoprotective effective against P. aeruginosa infection.


Assuntos
Catequina , Hepatopatias , Pneumonia Bacteriana , Infecções por Pseudomonas , Animais , Camundongos , Alanina Transaminase , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Aspartato Aminotransferases , Catequina/uso terapêutico , Catequina/farmacologia , Citocinas/metabolismo , Interleucina-6/metabolismo , Fígado/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Pseudomonas aeruginosa , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Pneumonia Bacteriana/complicações , Pneumonia Bacteriana/tratamento farmacológico , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/tratamento farmacológico , Hepatopatias/microbiologia
6.
J Ethnopharmacol ; 299: 115682, 2022 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-36058478

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: SanHuang XieXin decoction (SXD) is a widely applicated traditional Chinese medicine (TCM) with a significant gut-liver axis regulation effect. AIM OF THE STUDY: To evaluate the therapeutic effect and elucidate the possible underlying molecular mechanisms of SXD on liver damage secondary to ulcerative colitis (UC) in mice. MATERIALS AND METHODS: A model of liver damage secondary to UC was induced by drinking 5% dextran sodium sulfate (DSS) in mice. These mice were treated with one of three doses of SXD or sulfasalazine (SASP), then liver samples were collected and tested. RESULTS: The results reveal that SXD treatment reduced liver cells swelling, and inhibited the accumulation of the hepatic-pro-inflammatory cytokines IL-1ß and tumor necrosis factor-α (TNF-α) in mice with colitis. In addition, SXD reduced the production of nitric oxide (NO) and malondialdehyde (MDA), and increased the activities of superoxide dismutase (SOD). In inflammation regulating, SXD significantly down regulated the protein expression of MyD88 and p-Iκα, but upregulated Iκα. In bile acid metabolism regulating, SXD significantly down regulated the protein expression of FXR, MRP2, BESP and SHP. Therefore, SXD treatment can regulate the TLR4-NF-κB and bile acid metabolism pathways to alleviate liver inflammation and cholestasis. CONCLUSIONS: These results demonstrate that SXD is a potential alternative therapeutic medicine for the treatment of liver damage secondary to colitis.


Assuntos
Colite Ulcerativa , Colite , Animais , Ácidos e Sais Biliares , Colite/induzido quimicamente , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Citocinas/metabolismo , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas , Inflamação/tratamento farmacológico , Fígado/metabolismo , Malondialdeído , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Sulfassalazina/uso terapêutico , Superóxido Dismutase/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Ecotoxicol Environ Saf ; 222: 112518, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34271501

RESUMO

Copper (Cu), as a common chemical contaminant in environment, is known to be toxic at high concentrations. The current research demonstrates the effects of copper upon hepatocyte cell-cycle progression (CCP) in mice. Institute of cancer research (ICR) mice (n = 240) at an age of four weeks were divided randomly into groups treated with different doses of Cu (0, 4, 8, and 16 mg/kg) for 21 and 42 days. Results showed that high Cu exposure caused hepatocellular G0/G1 cell-cycle arrest (CCA) and reduced cell proportion in the G2/M phase. G0/G1 CCA occurred with down-regulation (p < 0.05) of Ras, p-PI3K (Tyr458), p-Akt (Thr308), p-forkhead box O3 (FOXO3A) (Ser253), p-glycogen synthase kinase 3-ß (GSK3-ß) (Ser9), murine double minute 2 (MDM2) protein, and mRNA expression levels, and up-regulation (p < 0.05) of PTEN, p-p53 (Ser15), p27, p21 protein, and mRNA expression levels, which subsequently suppressed (p < 0.05) the protein and mRNA expression levels of CDK2/4 and cyclin E/D. These results indicate that Cu exposure suppresses the Ras/PI3K/Akt signaling pathway to reduce the level of CDK2/4 and cyclin E/D, which are essential for the G1-S transition, and finally causes hepatocytes G0/G1 CCA.


Assuntos
Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Animais , Linhagem Celular Tumoral , Proliferação de Células , Cobre/toxicidade , Pontos de Checagem da Fase G1 do Ciclo Celular , Quinase 3 da Glicogênio Sintase , Camundongos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
8.
Biol Trace Elem Res ; 199(3): 1062-1073, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32666434

RESUMO

Silver nanoparticles (AgNPs) are widely used in industrial products, and they have good antibacterial properties, with potential for prevention and treatment of cow mastitis. However, concerns exist about the cytotoxicity of AgNPs. Thus, we have studied the role of autophagy in AgNP-induced cytotoxicity in mouse HC11 mammary epithelium cells. We found that AgNPs injured HC11 cells, with release of lactate dehydrogenase (LDH). AgNPs also induced autophagy in HC11 cells, which was associated with oxidative stress, as indicated by increased reactive oxygen species (ROS) and increased expression of hemoxygenase-1(HO-1) and Nrf2. Mitochondria were altered by AgNPs: mitochondrial membrane potential (MMP) was decreased and the expression of PINK1 and Parkin was increased. AgNPs also increased the expression of p-AMPK and decreased the expression of p-Akt and p-mTOR. The addition of 3-methyl adenine inhibited autophagy and enhanced the cytotoxicity of AgNPs, indicating that autophagy is protective against AgNP-induced cell death. In summary, AgNPs induced protective autophagy in HC11 cells via the Akt/AMPK/mTOR pathway, associated with cellular oxidative stress and mitochondrial alterations. Our research confirms that AgNPs may damage the breast tissue in clinical applications and should be used with caution. Further research is necessary to clarify whether the damage caused by AgNPs will affect the lactation function of the mammary glands and possible residues in milk.


Assuntos
Nanopartículas Metálicas , Prata , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Autofagia , Feminino , Nanopartículas Metálicas/toxicidade , Camundongos , Mitocôndrias/metabolismo , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Prata/toxicidade , Serina-Treonina Quinases TOR/metabolismo
9.
Environ Sci Pollut Res Int ; 28(5): 4893-4901, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33230792

RESUMO

Nickel (Ni) is a widely distributed metal in the environment and an important pollutant due to its widespread industrial applications. Ni has various toxicity in humans and experimental animals, including carcinogenicity. However, the carcinogenic effects of Ni remain troublesome. Cell cycle dysregulation may be an important carcinogenic mechanism and is also a potential molecular mechanism for Ni complexes anti-cancerous effects. Therefore, we conducted a literature review to summarize the effects of Ni on cell cycle. Up to now, there were three different reports on Ni-induced cell cycle arrest: (i) Ni can induce cell cycle arrest in G0/G1 phase, phosphorylation and degradation of IkappaB kinase-alpha (IKKα)-dependent cyclin D1 and phosphoinositide-3-kinase (PI3K)/serine-threonine kinase (Akt) pathway-mediated down-regulation of expressions of cyclin-dependent kinases 4 (CDK4) play important role in it; (ii) Ni can induce cell cycle arrest in S phase, but the molecular mechanism is not known; (iii) G2/M phase is the target of Ni toxicity, and Ni compounds cause G2/M cell cycle phase arrest by reducing cyclinB1/Cdc2 interaction through the activation of the ataxia telangiectasia mutated (ATM)-p53-p21 and ATM-checkpoint kinase inhibitor 1 (Chk1)/Chk2-cell division cycle 25 (Cdc25) pathways. Revealing the mechanisms of cell cycle dysregulation associated with Ni exposure may help in the prevention and treatment of Ni-related carcinogenicity and toxicology.


Assuntos
Proteínas de Ciclo Celular , Níquel , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Carcinogênese , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Humanos , Níquel/toxicidade , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
10.
BMC Cancer ; 20(1): 1145, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33238981

RESUMO

BACKGROUND: Breast cancer is the most common malignancy in women, and it is also the leading cause of death in female patients; the most common pathological type of BC is infiltrating duct carcinoma (IDC). Some nomograms have been developed to predict bone metastasis (BM) in patients with breast cancer. However, there are no studies on diagnostic and prognostic nomograms for BM in newly diagnosed IDC patients. METHODS: IDC patients with newly diagnosed BM from 2010 to 2016 in the Surveillance, Epidemiology and End Results (SEER) database were reviewed. Multivariate logistic regression analysis was used to identify risk factors for BM in patients with IDC. Univariate and multivariate Cox proportional hazards regression analysis were used to explore the prognostic factors of BM in patients with IDC. We then constructed nomograms to predict the risk and prognosis of BM for patients with IDC. The results were validated using bootstrap resampling and retrospective research on 113 IDC patients with BM from 2015 to 2018 at the Affiliated Hospital of Chengde Medical University. RESULTS: This study included 141,959 patients diagnosed with IDC in the SEER database, of whom 2383 cases were IDC patients with BM. The risk factors for BM in patients with IDC included sex, primary site, grade, T stage, N stage, liver metastasis, race, brain metastasis, breast cancer subtype, lung metastasis, insurance status, and marital status. The independent prognostic factors were brain metastases, race, grade, surgery, chemotherapy, age, liver metastases, breast cancer subtype, insurance status, and marital status. Through calibration, receiver operating characteristic curve and decision curve analyses, we found that the nomogram for predicting the prognosis of IDC patients with BM displayed great performance both internally and externally. CONCLUSION: These nomograms are expected to be a precise and personalized tool for predicting the risk and prognosis for BM in patients with IDC. This will help clinicians develop more rational and effective treatment strategies.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Neoplasias Pulmonares/secundário , Nomogramas , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Ósseas/terapia , Neoplasias Encefálicas/terapia , Neoplasias da Mama/terapia , Carcinoma Ductal de Mama/terapia , Terapia Combinada , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/terapia , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Programa de SEER , Adulto Jovem
11.
Ecotoxicol Environ Saf ; 203: 111006, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32684520

RESUMO

Nickel (Ni) is a widely distributed metal in the environment and an important pollutant because of its many industrial applications. With increasing incidences of Ni contamination, Ni toxicity has become a global public health concern and recent evidence suggests that Ni adversely affects the immune system. Hence, this paper reviews the literature on immune-related effects of Ni exposure, the immunotoxicological effects of Ni, and the underlying mechanism of Ni immunotoxicity. The main focus was on the effect of Ni on the development of organs of immune system, lymphocyte subpopulations, cytokines, immunoglobulins, natural killer (NK) cells, and macrophages. Moreover, Ni toxicity also induces inflammation and several studies demonstrated that Ni could induce immunotoxicity. Excessive Ni exposure can inhibit the development of immune organs by excessively inducing apoptosis and inhibiting proliferation. Furthermore, Ni can decrease T and B lymphocytes, the specific mechanism of which requires further research. The effects of Ni on immunoglobulin A (IgA), IgG, and IgM remain unknown and while Ni inhibited IgA, IgG, and IgM levels in an animal experiment, the opposite result was found in research on humans. Ni inhibits the production of cytokines in non-inflammatory responses. Cytokine levels increased in Ni-induced inflammation responses, and Ni activates inflammation through toll like (TL)4-mediated nuclear factor-κB (NF-κB) and signal transduction cascades mitogen-activated protein kinase (MAPK) pathways. Ni has been indicated to inactivate NK cells and macrophages both in vitro and in vivo. Identifying the mechanisms underlying the Ni-induced immunotoxicity may help to explain the growing risk of infections and cancers in human populations that have been exposed to Ni for a long time. Such knowledge may also help to prevent and treat Ni-related carcinogenicity and toxicology.


Assuntos
Poluentes Ambientais/toxicidade , Sistema Imunitário/efeitos dos fármacos , Níquel/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Citocinas/metabolismo , Humanos , Sistema Imunitário/crescimento & desenvolvimento , Sistema Imunitário/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
12.
Ecotoxicol Environ Saf ; 201: 110806, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32512418

RESUMO

The present study investigated the expressions of signalling molecules and inflammatory cytokines involved in copper-induced inflammatory responses of the mouse liver. A total of 240 institute of cancer research (ICR) mice (half male and half female) aged four weeks were randomly allocated to four groups treated with 0, 4, 8, and 16 mg/kg of [Cu] (Cu2+-CuSO4) for 42 days, respectively. [Cu] exceeding 4 mg/kg was found to induce inflammatory responses of the liver. Results showed significant up-regulation of mRNA and protein levels of apoptosis signal-regulating kinase 1 (ASK1), mitogen-activated protein kinase kinases 3/6 (MEK3/6), c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (p38 MAPK), mitogen-activated protein kinase kinases 4/7 (MEK4/7), mitogen-activated protein kinase kinases 1/2 (MEK1/2), and extracellular signal-regulated protein kinases 1/2 (Erk1/2) due to Cu. By doing so, copper could activate the mitogen-activated protein kinases (MAPKs) signalling pathway. Concurrently, the nuclear factor-kappa B (NF-κB) signalling pathway was also activated in the Cu-treatment, as demonstrated by higher expressions of NF-κB and cyclooxygenase-2 (COX-2), activities of inducible nitric oxide synthase (iNOS), contents of nitric oxide (NO) and prostaglandin E2 (PGE2), and reducing levels of expression of inhibitory kappa B (IκB). High Cu intake also up-regulated expression levels of some pro-inflammatory mediators such as interleukin-2 (IL-2), interleukin-1ß (IL-1ß), and interleukin-8 (IL-8), and down-regulated the levels of expression of transforming growth factor beta (TGF-ß), an anti-inflammatory mediator. Additionally, our results indicated that Cu caused hepatic dysfunction, with evidence of occurrence of histopathological lesions and higher serum activities of alkaline phosphatase (AKP), aspartic acid transferase (AST), alanine amino transferase (ALT), and gamma-glutamyl transpeptidase (GGT), contents of albumin (ALB) and total bilirubin (TBIL). Altogether, the aforementioned results indicate that [Cu], at more than 4 mg/kg, induces the inflammatory responses in the liver via NF-κB and MAPKs signalling pathways, subsequently inducing hepatic dysfunction.


Assuntos
Cobre/toxicidade , Interleucina-1beta/metabolismo , Interleucina-2/metabolismo , Fígado/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Animais , Ciclo-Oxigenase 2/metabolismo , Feminino , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Testes de Função Hepática , Masculino , Camundongos , Camundongos Endogâmicos ICR , Distribuição Aleatória , Transdução de Sinais
13.
Biomed Pharmacother ; 129: 110418, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32570121

RESUMO

(-)-Epigallocatechin-3-gallate (EGCG) is the most abundant polyphenolic compound present in green tea and has been shown to possess bio-activities. In this study, we investigated the protective effects of EGCG against restraint stress (RS)-induced liver injury and immunosuppression. EGCG (10, 20 and 40 mg/kg) was orally administered to mice daily for 7 days before modeling the restraint stress. lood, liver and broncho-alveolar lavage fluid (BALF) samples were collected and tested. We found that EGCG significantly reduced the release of stress hormones to weak restraint stress response. EGCG effectively improved hepatic damage by decreas the serum levels of alanine aminotransaminase (ALT) and aspartate transaminase (AST) in restraint-challenged mice. Furthermore, EGCG also significantly prevented the release of H2O2, NOS and 8-isoprostane, and reduced the levels of interleukin (IL)-1ß, IL-2,and IL-6 restrained mice. EGCG can normal the level of cytochrome P450 (CYP450) 1A2, 2D22, 2E1 and 3A11 that induced by restraint stress., the inhibition status of T cells subsets in serum and gA in BALF were significantly relieved EGCG pretreatment. Taken together, our data suggest that EGCG possesse hepatic- and immune-protective properties against restraint stress through its anti-oxidant, anti-inflammatory and immunomodulatory activities.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Catequina/análogos & derivados , Sistema Enzimático do Citocromo P-450/metabolismo , Fatores Imunológicos/farmacologia , Fígado/efeitos dos fármacos , Restrição Física , Estresse Fisiológico/efeitos dos fármacos , Estresse Psicológico/tratamento farmacológico , Animais , Biomarcadores/sangue , Biotransformação , Catequina/farmacologia , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Imunoglobulina A Secretora/sangue , Mediadores da Inflamação/sangue , Isoenzimas , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos Endogâmicos ICR , Estresse Oxidativo/efeitos dos fármacos , Estresse Psicológico/enzimologia , Estresse Psicológico/etiologia , Estresse Psicológico/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
14.
Oxid Med Cell Longev ; 2020: 1359164, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411316

RESUMO

Copper (Cu) is an essential trace element involved in the normal physiological processes of animals. However, excessive exposure to Cu can produce numerous detrimental impacts. The aim of this study was to investigate the effects of Cu on oxidative stress and apoptosis as well as their relationship in the mouse liver. Four-week-old ICR mice (n = 240) were randomly assigned to different Cu (Cu2+-CuSO4) treatment groups (0, 4, 8, and 16 mg/kg) for periods of 21 and 42 days. The high doses of Cu exposure could induce oxidative stress, by increasing the levels of reactive oxygen species (ROS) and protein carbonyls (PC) and decreasing the activities of antisuperoxide anion (ASA) and antihydroxyl radical (AHR) and content of glutathione (GSH), as well as activities and mRNA expression levels of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). Moreover, high doses of Cu exposure induced hepatic apoptosis via the mitochondrial apoptotic pathway, as characterized by the depolarization of mitochondrial membrane potential (MMP); significantly increased mRNA and protein expression levels of cytosolic cytochrome (Cyt c), apoptosis-inducing factor (AIF), endonuclease G (Endo G), apoptosis protease-activating factor-1 (Apaf-1), cleaved caspase-9, cleaved caspase-3, cleaved PARP, Bcl-2 antagonist killer (Bak), Bcl-2-associated X protein (Bax), and Bcl-2-interacting mediator of cell death (Bim); and decreased mRNA and protein expression levels of B-cell lymphoma-2 (Bcl-2) and Bcl-extra-large (Bcl-xL). Furthermore, the activation of the tumor necrosis factor receptor-1 (TNF-R1) signaling pathway was involved in Cu-induced apoptosis, as characterized by the significantly increased mRNA and protein expression levels of TNF-R1, Fas-associated death domain (FADD), TNFR-associated death domain (TRADD), and cleaved caspase-8. These results indicated that exposure to excess Cu could cause oxidative stress triggered by ROS overproduction and diminished antioxidant function, which in turn promoted hepatic apoptosis via mitochondrial apoptosis and that the TNF-R1 signaling pathway was also involved in the Cu-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cobre/toxicidade , Fígado/patologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Peso Corporal/efeitos dos fármacos , Caspases/metabolismo , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Feminino , Glutationa/metabolismo , Fígado/efeitos dos fármacos , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos ICR , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Modelos Biológicos , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína de Domínio de Morte Associada a Receptor de TNF/genética , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
15.
Ecotoxicol Environ Saf ; 191: 110236, 2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-32001424

RESUMO

Copper (Cu), a transition metal, is an essential trace element in human and animal nutrition at low concentration, but Cu has toxic effects on tissues and organs at high concentration. Endoplasmic reticulum (ER) is a toxicological target in Cu poison. Thus far, no studies have focused on the relationship among copper, endoplasmic reticulum (ER) stress and apoptosis in animal and human livers. In the present study, mice treated with copper sulfate (CuSO4) were used to assess the impacts of copper on ER stress and hepatic apoptosis. A total of 240 mice were orally administered with 0 (control), 10, 20 and 40 mg/kg of CuSO4 for 42 days. The results indicated that CuSO4 at 10 mg/kg markedly induced hepatocyte apoptosis and ER stress. In addition, ER stress was characterized by the increased mRNA and protein levels of glucose-regulated protein 78 (GRP78) and 94 (GRP94). Furthermore, ER stress-triggered 3 apoptotic pathways were also activated by the increased intracellular calcium and up-regulated expression levels of genes involved in growth arrest- and DNA damage-inducible gene 153 (Gadd153/CHOP), c-Jun N-terminal kinase (JNK) and cysteine aspartate-specific protease 12 (caspase-12) signaling pathways in CuSO4-treated mice. In conclusion, CuSO4-induced ER stress can promote hepatic apoptosis in mice by activating CHOP, JNK and caspase-12 signaling pathways.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 12/metabolismo , Sulfato de Cobre/toxicidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/efeitos dos fármacos , Fator de Transcrição CHOP/metabolismo , Animais , Relação Dose-Resposta a Droga , Chaperona BiP do Retículo Endoplasmático , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos ICR , Transdução de Sinais
16.
Aging (Albany NY) ; 11(23): 11659-11672, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31822637

RESUMO

Nickel (Ni), an environmental hazard, widely causes allergic contact hypersensitivity worldwide. Despite that Ni-stimulated pro-inflammatory response is vital in allergy, the underlying molecular mechanisms remain largely unclear. Here, we demonstrated that NiCl2 activated nuclear factor kappa B (NF-κB), mitogen-activated protein kinases (MAPKs) and interferon regulatory factor 3 (IRF3) signaling pathways in primary bone marrow-derived macrophages (BMDMs), leading to the altered transcription levels of interleukin-1ß (IL-1ß), -6, -8, -18, tumor necrosis factor-α (TNF-α) and interferon ß (INF-ß). We also found that nickel chloride (NiCl2) activated Nod-like receptor 3 (NLRP3) inflammasome pathway, resulting in the proteolytic cleavage and release of IL-1ß. NiCl2 induced the accumulation of mitochondrial reactive oxygen species (mtROS) and the release of mitochondrial DNA (mtDNA), thus activating NLRP3 inflammasome pathway. Additionally, NiCl2-induced apoptosis was dependent on the generation of mtROS, and caspase-1 activation might also partly contribute to the apoptotic process. Altogether, abovementioned results indicate that NiCl2 induces inflammatory activation in BMDMs via NF-κB, MAPKs, IRF3 signaling pathways as well as NLRP3 inflammasome pathway, which provides a mechanism to improve the efficiency of treatment against Ni-induced allergic reactions.


Assuntos
Inflamação/induzido quimicamente , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Níquel/farmacologia , Animais , Apoptose , Sobrevivência Celular/efeitos dos fármacos , Poluentes Ambientais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Fator Regulador 3 de Interferon/genética , Macrófagos/metabolismo , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Espécies Reativas de Oxigênio
17.
Int J Mol Sci ; 20(19)2019 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-31546657

RESUMO

Nickel (Ni) is known to be a major carcinogenic heavy metal. Occupational and environmental exposure to Ni has been implicated in human lung and nasal cancers. Currently, the molecular mechanisms of Ni carcinogenicity remain unclear, but studies have shown that Ni-caused DNA damage is an important carcinogenic mechanism. Therefore, we conducted a literature search of DNA damage associated with Ni exposure and summarized known Ni-caused DNA damage effects. In vitro and vivo studies demonstrated that Ni can induce DNA damage through direct DNA binding and reactive oxygen species (ROS) stimulation. Ni can also repress the DNA damage repair systems, including direct reversal, nucleotide repair (NER), base excision repair (BER), mismatch repair (MMR), homologous-recombination repair (HR), and nonhomologous end-joining (NHEJ) repair pathways. The repression of DNA repair is through direct enzyme inhibition and the downregulation of DNA repair molecule expression. Up to now, the exact mechanisms of DNA damage caused by Ni and Ni compounds remain unclear. Revealing the mechanisms of DNA damage from Ni exposure may contribute to the development of preventive strategies in Ni carcinogenicity.


Assuntos
Carcinogênese/induzido quimicamente , Dano ao DNA , Níquel/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Animais , Quebras de DNA de Cadeia Dupla , Dano ao DNA/genética , Reparo de Erro de Pareamento de DNA , Reparo do DNA , Humanos , Níquel/metabolismo
18.
Cell Physiol Biochem ; 51(5): 2421-2433, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30537743

RESUMO

BACKGROUND/AIMS: Excessive fluoride intake can induce cytotoxicity, DNA damage and cell-cycle changes in many tissues and organs, including the kidney. However, the underlying molecular mechanisms of fluoride-induced renal cell-cycle changes are not well understood at present. In this study, we used a mouse model to investigate how sodium fluoride (NaF) induces cell-cycle changes in renal cells. METHODS: Two hundred forty ICR mice were randomly assigned to four equal groups for intragastric administration of NaF (0, 12, 24 and 48 mg/kg body weight/day) for 42 days. Kidneys were taken to measure changes of the cell-cycle at 21 and 42 days of the experiment, using flow cytometry, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot methods. RESULTS: NaF, at more than 12 mg/kg body weight, induced G2/M phase cell-cycle arrest in the renal cells, which was supported by the finding of significantly increased percentages of renal cells in the G2/M phase. We found also that G2/M phase cell-cycle arrest was accompanied by up-regulation of p-ATM, p-Chk2, p-p53, p-Cdc25C, p-CDK1, p21, and Gadd45a protein expression levels; up-regulation of ATM, Chk2, p53, p21, and Gadd45a mRNA expression levels; down-regulation of CyclinB1, mdm2, PCNA protein expression levels; and down-regulation of CyclinB1, CDK1, Cdc25C, mdm2, and PCNA mRNA expression levels. CONCLUSION: In this mouse model, NaF, at more than 12 mg/ kg, induced G2/M phase cell-cycle arrest by activating the ATM-Chk2-p53/Cdc25C signaling pathway, which inhibits the proliferation of renal cells and development of the kidney. Activation of the ATM-Chk2-p53/Cdc25C signaling pathway is the mechanism of NaF-induced renal G2/M phase cell-cycle arrest in this model.


Assuntos
Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Rim/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fluoreto de Sódio/efeitos adversos , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quinase do Ponto de Checagem 2/metabolismo , Feminino , Rim/citologia , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Endogâmicos ICR , Proteína Supressora de Tumor p53/metabolismo , Fosfatases cdc25/metabolismo
19.
Toxicol Res (Camb) ; 7(5): 792-808, 2018 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-30310657

RESUMO

The objective of the present study was to explore the molecular mechanism of apoptosis induced by sodium fluoride (NaF) in the mouse kidney by using the methods of flow cytometry, quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and experimental pathology. 240 four-week-old ICR mice were randomly divided into 4 groups and exposed to different concentrations of NaF (0 mg kg-1, 12 mg kg-1, 24 mg kg-1 and 48 mg kg-1) for a period of 42 days. The results demonstrated that NaF increased cell apoptosis and the depolarization of the mitochondrial membrane potential (MMP), and that the mitochondrial pathway was involved in NaF-induced apoptosis. Alteration of the mitochondrial pathway was characterized by significantly increasing mRNA and protein expression levels of cytosolic cytochrome c (Cyt c), the second mitochondrial activator of caspases/direct inhibitors of the apoptosis binding protein with low pI (Smac/Diablo), the serine protease high-temperature-requirement protein A2/Omi (HtrA2/Omi), the apoptosis inducing factor (AIF), endonuclease G (Endo G), cleaved-cysteine aspartate specific protease-9 (cleaved-caspase-9), cleaved-cysteine aspartate specific protease-3 (cleaved-caspase-3), Bcl-2 antagonist killer (Bak), Bcl-2 associated X protein (Bax), Bcl-2 interacting mediator of cell death (Bim), cleaved-poly-ADP-ribose polymerase (cleaved-PARP), p-p53, and decreasing mRNA and protein expression levels of B-cell lymphoma-2 (Bcl-2), Bcl-extra large (Bcl-xL), and X chromosome-linked inhibitors of apoptosis proteins (XIAPs). To our knowledge, the mitochondrial pathway is reported for the first time in NaF-induced apoptosis of the human or animal kidney. Also, this study provides novel insights for further studying fluoride-induced nephrotoxicity.

20.
Aging (Albany NY) ; 10(7): 1649-1665, 2018 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-30036188

RESUMO

Fluoride is known to impair organism's development and function via adverse effects, and autophagy plays a regulation role in human or animal health and disease. At present, there are no reports focused on fluoride-induced autophagy in the animal and human spleen. The objective of this study was to investigate sodium fluoride (NaF)-induced splenocyte autophagy and the potential mechanism via regulation of p-mTOR in growing mice by using the methods of transmission electron microscopy (TEM), immunohistochemistry (IHC), quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. A total of 240 ICR mice were equally allocated into four groups with intragastric administration of distilled water in the control group and 12, 24, 48 mg/kg NaF solution in the experimental groups for 42 days. Results revealed that NaF increased autophagosomes or autolysosomes in spleen. Simultaneously, the autophagy marker LC3 brown punctate staining was increased with NaF dosage increase. On the other hand, NaF caused inhibition of mTOR activity, which was characterized by down-regulation of PI3K, Akt and mTOR mRNA and protein expression levels. And the suppression of mTOR activity in turn resulted in the significantly increased of ULK1 and Atg13 expression levels. Concurrently, NaF increased the levels of mRNA and protein expression of autophagy markers LC3, Beclin1, Atg16L1, Atg12, Atg5 and decreased the mRNA and protein expression levels of p62. The above-mentioned findings verify that NaF induces autophagy via mTOR signaling pathway. The inhibition of mTOR activity and alteration of autophagy-related genes and proteins are the potential molecular mechanism of NaF-induced splenocyte autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fluoreto de Sódio/farmacologia , Baço/citologia , Homólogo LST8 da Proteína Associada a mTOR/metabolismo , Animais , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Biomarcadores , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação , RNA Mensageiro , Transdução de Sinais , Baço/ultraestrutura , Homólogo LST8 da Proteína Associada a mTOR/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA