Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 72
Filtrer
1.
Ecotoxicol Environ Saf ; 281: 116598, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38896897

RÉSUMÉ

Bisphenol AF (BPAF) is found in high concentrations in aquatic environments due to the increased use of thermal paper and food packaging. However, there have been relatively few toxicological studies and potential risk assessments of BPAF. In this study, the risk quotient (RQ) and hazard quotient (HQ) of BPAF were derived to present the safety standards for environmental risk management and protection in lakes, rivers, bays, and Italian regions. We applied the species sensitivity distribution (SSD) method based on the previous ecotoxicological data and the results of supplementary toxicity tests on BPAF. From the SSD curves, the hazardous concentration for 5 % of the species (HC5) values for the acute and chronic toxicity data were 464.75 µg/L and 3.59 µg/L, respectively, and the acute- and chronic-based predicted no-effect concentration were derived as 154.92 µg/L and 1.20 µg/L, respectively. The acute-based RQ (RQA)values of BPAF in all regions were negligible (RQ < 0.1). The chronic-based RQ (RQC) in the Xitang River (XR) and the Central Italy (CI) showed a considerably high ecological risk (12.77 and 1.29) and the Hangzhou Bay (0.21), the South and North Italy (0.79 and 0.27), and the Tamagawa River (0.13) had a medium ecological risk (0.1 < RQ < 1.0). However, the HQ values based on the tolerable daily intake for BPAF over all age groups in these regions was < 0.1, indicating the low health risk. Nonetheless, the result of this study indicates that BPAF contamination is serious in XR and CI, and their use and emissions require continuous monitoring.

2.
Antioxidants (Basel) ; 13(5)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38790680

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic inflammatory condition caused by the disruption of the intestinal barrier. The intestinal barrier is maintained by tight junctions (TJs), which sustain intestinal homeostasis and prevent pathogens from entering the microbiome and mucosal tissues. Ziziphus jujuba Miller (Z. jujuba) is a natural substance that has been used in traditional medicine as a therapy for a variety of diseases. However, in IBD, the efficacy of Z. jujuba is unknown. Therefore, we evaluated ZJB in Caco2 cells and a dextran sodium sulfate (DSS)-induced mouse model to demonstrate its efficacy in IBD. Z. jujuba extracts were prepared using 70% ethanol and were named ZJB. ZJB was found to be non-cytotoxic and to have excellent antioxidant effects. We confirmed its anti-inflammatory properties via the down-regulation of inflammatory factors, including inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). To evaluate the effects of ZJB on intestinal barrier function and TJ improvement, the trans-epithelial electrical resistance (TEER) and fluorescein isothiocyanate-dextran 4 kDa (FITC-Dextran 4) permeability were assessed. The TEER value increased by 61.389% and permeability decreased by 27.348% in the 200 µg/mL ZJB group compared with the 50 ng/mL IL-6 group after 24 h. Additionally, ZJB alleviated body weight loss, reduced the disease activity index (DAI) score, and induced colon shortening in 5% DSS-induced mice; inflammatory cytokines, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 were down-regulated in the serum. TJ proteins, such as Zonula occludens (ZO)-1 and occludin, were up-regulated by ZJB in an impaired Caco2 mouse model. Additionally, according to the liquid chromatography results, in tandem with mass spectrometry (LC-MS/MS) analysis, seven active ingredients were detected in ZJB. In conclusion, ZJB down-regulated inflammatory factors, protected intestinal barrier function, and increased TJ proteins. It is thus a safe, natural substance with the potential to be used as a therapeutic agent in IBD treatment.

3.
Mol Biol Rep ; 51(1): 305, 2024 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-38361124

RÉSUMÉ

BACKGROUND: Pectolinarigenin (PEC) is a flavone extracted from Cirsium, and because it has anti-inflammatory properties, anti-cancer research is also being conducted. The objective of this work was to find out if PEC is involved in tumor control and which pathways it regulates in vivo and in vitro. METHODS: AGS cell lines were xenografted into BALB/c nude mice to create tumors, and PEC was administered intraperitoneally to see if it was involved in tumor control. Once animal testing was completed, tumor proteins were isolated and identified using LC-MS analysis, and gene ontology of the found proteins was performed. RESULTS: Body weight and hematological measurements on the xenograft mice model demonstrated that PEC was not harmful to non-cancerous cells. We found 582 proteins in tumor tissue linked to biological reactions such as carcinogenesis and cell death signaling. PEC regulated 6 out of 582 proteins in vivo and in vitro in the same way. CONCLUSION: Our findings suggested that PEC therapy may inhibit tumor development in gastric cancer (GC), and proteomic research gives fundamental information about proteins that may have great promise as new therapeutic targets in GC.


Sujet(s)
Apoptose , 4H-1-Benzopyran-4-ones , Tumeurs de l'estomac , Humains , Animaux , Souris , Souris nude , Hétérogreffes , Protéomique , Lignée cellulaire tumorale , Tumeurs de l'estomac/génétique , Protéines proto-oncogènes c-akt/métabolisme , Prolifération cellulaire
4.
Commun Biol ; 6(1): 157, 2023 02 08.
Article de Anglais | MEDLINE | ID: mdl-36750754

RÉSUMÉ

Melatonin protects against Cadmium (Cd)-induced toxicity, a ubiquitous environmental toxicant that causes adverse health effects by increasing reactive oxygen species (ROS) production and mitochondrial dysfunction. However, the underlying mechanism remains unclear. Here, we demonstrate that Cd exposure reduces the levels of mitochondrially-localized signal transducer and activator of transcription 3 (mitoSTAT3) using human prostate stromal cells and mouse embryonic fibroblasts. Melatonin enhances mitoSTAT3 abundance following Cd exposure, which is required to attenuate ROS damage, mitochondrial dysfunction, and cell death caused by Cd exposure. Moreover, melatonin increases mitochondrial levels of GRIM-19, an electron transport chain component that mediates STAT3 import into mitochondria, which are downregulated by Cd. In vivo, melatonin reverses the reduced size of mouse prostate tissue and levels of mitoSTAT3 and GRIM-19 induced by Cd exposure. Together, these data suggest that melatonin regulates mitoSTAT3 function to prevent Cd-induced cytotoxicity and could preserve mitochondrial function during Cd-induced stress.


Sujet(s)
Cadmium , Mélatonine , Mâle , Humains , Animaux , Souris , Cadmium/métabolisme , Mélatonine/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Facteur de transcription STAT-3/métabolisme , Prostate , Fibroblastes/métabolisme , Mitochondries/métabolisme , Stress oxydatif
5.
Molecules ; 27(16)2022 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-36014455

RÉSUMÉ

Cadmium (Cd), a harmful heavy metal, can lead to various pulmonary diseases, including chronic obstructive pulmonary disease (COPD), by inducing cytotoxicity and disturbing redox homeostasis. The aim of the present study was to investigate Cd-mediated cytotoxicity using human lung fibroblasts and the therapeutic potential of 3,3'-diindolylmethane (DIM). Cadmium significantly reduced the cell viability of human embryonic lung (HEL299) cells accompanied by enhanced oxidative stress as evidenced by the increased expression of autophagy-related proteins such as LC3B and p62. However, treatment with DIM significantly suppressed autophagic cell death in Cd-induced HEL299 fibroblasts. In addition, DIM induced antioxidant enzyme activity and decreased intracellular reactive oxygen species (ROS) levels in Cd-damaged HEL299 cells. This study suggests that DIM effectively suppressed Cd-induced lung fibroblast cell death through the upregulation of antioxidant systems and represents a potential agent for the prevention of various diseases related to Cd exposure.


Sujet(s)
Mort cellulaire par autophagie , Cadmium , Antioxydants/métabolisme , Antioxydants/pharmacologie , Apoptose , Autophagie , Cadmium/toxicité , Fibroblastes/métabolisme , Humains , Indoles , Poumon/métabolisme , Stress oxydatif , Espèces réactives de l'oxygène/métabolisme
6.
Article de Anglais | MEDLINE | ID: mdl-35664945

RÉSUMÉ

The transcriptional machinery is composed of numerous factors that help to regulate gene expression in cells. The function and the fundamental role of transcription factors in different human diseases and cancer have been extensively researched. Activator protein-1 (AP-1) is an inducible transcription factor that consists of a diverse group of members including Jun, Fos, Maf, and ATF. AP-1 involves a number of processes such as proliferation, migration, and survival in cells. Dysfunctional AP-1 activity is seen in several diseases, especially cancer and inflammatory disorders. The AP-1 proteins are controlled by mitogen-activated protein kinases (MAPKs) and the NF-κB pathway. AP-1 inhibitors can be actively pursued as drug discovery targets in cancer therapy when used as a treatment to halt tumor progression. The consumption of phytochemicals in the diet is related to decreasing the incidence of cancer and proves to exhibit anticancer properties. Natural product targets AP-1 are effective cancer prevention and treatment options for various cancer types. Targeting AP-1 with natural products is an effective cancer treatment option for different cancer types. This review summarizes AP-1 subunit proteins, their structures, AP-1-related signaling, and its modulation by natural bioactive compounds.

7.
Int J Environ Health Res ; 32(1): 131-140, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-32191530

RÉSUMÉ

The toxicity of cadmium (Cd) occurs through accumulation in the environment. The precise mechanism underlying Cd toxicity remains unclear. Therefore, in the present study, we studied the effects of Cd on MM55.K cells and investigated the mechanisms underlying Cd-induced cell death. CdCl2 significantly elevated apoptotic cell death, mitochondrial membrane potential (ΔΨm) loss, and caspase-dependent cell death. Moreover, immunoblotting results revealed that CdCl2 down-regulated the inhibitor of apoptotic protein such as survivin and Bcl-2 which led to the activation of caspase-3 and the cleavage of PARP in MM55.K cells. Besides, CdCl2 caused the up-regulation of ROS-related proteins such as HO-1 and ER stress-related proteins such as GRP78 and CHOP in MM55.K cells. CdCl2 toxicity resulted in the down-regulation of the AKT pathway that leads to the up-regulation of phosphorylated JNK and p38 in MM55.K cells. Thus, CdCl2 induce toxicity by AKT/MAPK regulation and causing ROS production, ER stress, ΔΨm loss, and apoptotic cell death in normal mouse renal cells.


Sujet(s)
Cadmium , Mitochondries , Animaux , Apoptose , Cadmium/toxicité , Chaperonne BiP du réticulum endoplasmique , Souris , Espèces réactives de l'oxygène
8.
Neoplasia ; 24(2): 98-108, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34954452

RÉSUMÉ

Novel CAR T cells targeting mesothelin (MSLN) expressed on pancreatic cancer cells were developed to overcome the limit of the clinical efficacy of CAR T cell therapy for pancreatic cancer patients. Optimal single-chain variable fragments (scFv) binding to MSLN were selected based on the binding activity and the functional effectiveness of various scFv containing CAR-expressing T cells. Engineered MSLN CAR T cells showed successful anti-tumor activity specific to MSLN expression level. Furthermore, MSLN CAR T cells were evaluated for the anti-cancer efficacy in orthotopic mouse models bearing pancreatic cancer cells, MIA Paca-2, MSLN-overexpressed MIA Paca-2 or endogenously MSLN-expressing AsPC-1. Mice were randomized into control, mock treated, MS501 BBz treated, MS501 28z treated or MS501 28BBz treated group. Mice were monitored by weekly IVIS imaging and tumors were harvested and analyzed by immunohistochemical analyses. MSLN CAR T cells produced the therapeutic effect in orthotopic animal models with complete remission in significant number of mice. Histopathological analysis indicated that CD4+ and CD8+ MSLN CAR T cells infiltrated pancreatic tumor tissue and led to cancer cell eradication. Our results demonstrated the anti-tumor efficacy of MSLN CAR T cell therapy against pancreatic cancer, suggesting its therapeutic potential.


Sujet(s)
Immunothérapie adoptive , Mésothéline/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Anticorps à chaîne unique/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Humains , Souris , Tumeurs du pancréas/thérapie , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Int J Mol Sci ; 22(16)2021 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-34445559

RÉSUMÉ

Scutellarein (SCU) is a well-known flavone with a broad range of biological activities against several cancers. Human hepatocellular carcinoma (HCC) is major cancer type due to its poor prognosis even after treatment with chemotherapeutic drugs, which causes a variety of side effects in patients. Therefore, efforts have been made to develop effective biomarkers in the treatment of HCC in order to improve therapeutic outcomes using natural based agents. The current study used SCU as a treatment approach against HCC using the HepG2 cell line. Based on the cell viability assessment up to a 200 µM concentration of SCU, three low-toxic concentrations of (25, 50, and 100) µM were adopted for further investigation. SCU induced cell cycle arrest at the G2/M phase and inhibited cell migration and proliferation in HepG2 cells in a dose-dependent manner. Furthermore, increased PTEN expression by SCU led to the subsequent downregulation of PI3K/Akt/NF-κB signaling pathway related proteins. In addition, SCU regulated the metastasis with EMT and migration-related proteins in HepG2 cells. In summary, SCU inhibits cell proliferation and metastasis in HepG2 cells through PI3K/Akt/NF-κB signaling by upregulation of PTEN, suggesting that SCU might be used as a potential agent for HCC therapy.


Sujet(s)
Apigénine/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Phosphohydrolase PTEN/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Apoptose , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Mouvement cellulaire , Prolifération cellulaire , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Facteur de transcription NF-kappa B/génétique , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes c-akt/génétique , Cellules cancéreuses en culture
10.
Nutrients ; 12(8)2020 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-32824273

RÉSUMÉ

Sinensetin (SIN) has been reported to exhibit anti-inflammatory and anti-cancer activity. However, the cellular and molecular mechanism by which SIN promotes hepatocellular carcinoma (HCC) cell death remains unclear. In the present study, we investigated the induction of cell death by SIN and its underlying mechanism in HepG2 cells, an HCC cell line. We found that SIN significantly induced cell death in HepG2 cells, whereas the proliferation rate of Thle2, human liver epithelial cells, was unaffected by SIN. SIN-treated HepG2 cells were not affected by apoptotic cell death; instead, autophagic cell death was induced through the p53-mediated AMPK/mTOR signaling pathway. Inhibition of p53 degradation led to both autophagy and apoptosis in HepG2 cells. p53 translocation led to SIN-induced autophagy, whereas p53 translocation inhibited SIN-induced apoptosis. However, SIN showed apoptosis in the p53-mutant Hep3B cell line. Molecular docking simulation of the p53 core domain showed effective binding with SIN, which was found significant compared with the known p53 activator, RITA. Collectively, these data suggest that SIN may be a potential anti-cancer agent targeting autophagic cell death in human liver cancer.


Sujet(s)
Antinéoplasiques/pharmacologie , Mort cellulaire par autophagie/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/métabolisme , Flavonoïdes/pharmacologie , Tumeurs du foie/métabolisme , AMP-Activated Protein Kinases/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/anatomopathologie , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules HepG2 , Humains , Tumeurs du foie/anatomopathologie , Simulation de docking moléculaire/méthodes , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme
11.
Oncol Rep ; 44(3): 939-958, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32705238

RÉSUMÉ

Scutellarein (SCU), a flavone that belongs to the flavonoid family and abundantly present in Scutellaria baicalensis a flowering plant in the family Lamiaceae, has been reported to exhibit anticancer effects in several cancer cell lines including gastric cancer (GC). Although our previous study documented the mechanisms of Scutellarein­induced cytotoxic effects, the literature shows that the proteomic changes that are associated with the cellular response to SCU have been poorly understood. To avoid adverse side­effects and significant toxicity of chemotherapy in patients who react poorly, biomarkers anticipating therapeutic responses are imperative. In the present study, we utilized a comparative proteomic analysis to identify proteins associated with Scutellarein (SCU)­induced cell death in GC cells (AGS and SNU484), by integrating two­dimensional gel electrophoresis (2­DE), mass spectrometry (MS), and bioinformatics to analyze the proteins. Proteomic analysis between SCU­treated and DMSO (control) samples successfully identified 41 (AGS) and 31 (SNU484) proteins by MALDI­TOF/MS analysis and protein database search. Comparative proteomics analysis between AGS and SNU484 cells treated with SCU revealed a total of 7 protein identities commonly expressed and western blot analysis validated a subset of identified critical proteins, which were consistent with those of the 2­DE outcome. Molecular docking studies also confirmed the binding affinity of SCU towards these critical proteins. Phosphatidylinositol 4,5­bisphosphate 3­kinase catalytic subunit ß isoform (PIK3CB) protein expression was accompanied by a distinct group of cellular functions, including cell growth, and proliferation. Cancerous inhibitor of protein phosphatase 2A (CIP2A), is one of the oncogenic molecules that have been shown to promote tumor growth and resistance to apoptosis and senescence­inducing therapies. In the present study, both PIK3CB and CIP2A proteins were downregulated in SCU­treated cells, which boosts our previous results of SCU to induce apoptosis and inhibits GC cell growth by regulating these critical proteins. The comparative proteomic analysis has yielded candidate biomarkers of response to SCU treatment in GC cell models and further validation of these biomarkers will help the future clinical development of SCU as a novel therapeutic drug.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Apigénine/pharmacologie , Marqueurs biologiques tumoraux/génétique , Tumeurs de l'estomac/traitement médicamenteux , Antinéoplasiques d'origine végétale/usage thérapeutique , Apigénine/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Autoantigènes/analyse , Autoantigènes/génétique , Autoantigènes/métabolisme , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Phosphatidylinositol 3-kinases de classe I/analyse , Phosphatidylinositol 3-kinases de classe I/génétique , Phosphatidylinositol 3-kinases de classe I/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Protéines et peptides de signalisation intracellulaire/analyse , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines membranaires/analyse , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Simulation de docking moléculaire , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Protéomique , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie
12.
Am J Chin Med ; 48(3): 679-702, 2020.
Article de Anglais | MEDLINE | ID: mdl-32329644

RÉSUMÉ

Autophagy is a process of active programmed cell death, where a dying cell induces autophagosomes and subsequently regulated by degradative machinery. The aim of this study was to investigate the mechanism behind induction of autophagic cell death by Naringin flavonoid in AGS cancer cells. Growth inhibition of AGS cells showed downregulation of PI3K/Akt/mTOR signaling by Naringin treatment. Transmission electron microscopy observation showed swollen mitochondria and lysosome near peri-nuclear zone fused with autophagic vacuoles. Rapamycin pre-treatment with Naringin showed significant decrease in mTOR phosphorylation and increase in LC3B activation in AGS cells. Decrease in mTOR phosphorylation is associated with lysosomal function activation was observed by time-dependent treatment of Naringin. Induction of lysosomal membrane permeabilization (LMP) was observed by LAMP1 activation leading lysosomal cell death by releasing Cathepsin D from lysosomal lumen to cytosol. Naringin treated AGS cells showed up-regulating BH3 domain Bad, down-regulating Bcl-xL, and Bad phosphorylation and significant mitochondrial fluorescence intensity expression. Significant localization of mitochondria and LC3B activation was examined by person coefficient correlation. Activation of ERK1/2-p38 MAPKs and production of intracellular ROS has been observed over Naringin treatment. It has also been elucidated that pre-treatment with NAC inhibited mitochondria-LC3B colocalization, where ROS acted as upstream of ERK1/2-p38 MAPKs activation. Lysosomal cell death involvement has been evaluated by BAF A1 pre-treatment, inhibiting LAMP1, Cathepsin D, ROS, and blocking autophagolysosome in AGS cell death. Taken together, these findings show that, Naringin induced autophagy cell death involves LMP mediated lysosomal damage and BH3 protein Bad activation in AGS cancer cells.


Sujet(s)
Autophagie/effets des médicaments et des substances chimiques , Flavanones/pharmacologie , Lysosomes/anatomopathologie , Tumeurs de l'estomac/anatomopathologie , Perméabilité des membranes cellulaires/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Humains , Lysosomes/métabolisme , Protéines associées aux microtubules/métabolisme , Protéines de transport de la membrane mitochondriale/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Cellules cancéreuses en culture
13.
Article de Anglais | MEDLINE | ID: mdl-31781269

RÉSUMÉ

The use of adipose-derived stem cells (ADSCs) to enhance wound healing and tissue regeneration is progressively being accepted. Proteomic profiling of cultured ADSCs by mass spectrometry (MS) is a valuable tool to determine the identity of the proteins involved in multiple pathways, which make these ADSCs unique. In the current study, Nano-LC-MS/MS analysis was implemented on the membrane-free stem cell component (MFSCC), and the MS analysis revealed the presence of 252 proteins, that are involved in several biological functions, like metabolic process, biological regulation, developmental process, cell proliferation, and many more. Furthermore, bioinformatic analyses of the identified proteins in MFSCC found them to be involved in versatile pathways, like integrin pathway and wound healing response-related pathways. In addition, we also investigated the anti-inflammatory effects of MFSCC on lipopolysaccharide (LPS) stimulated mouse macrophage (RAW264.7) cells. The cell cytotoxicity of MFSCC was measured using MTT and LDH assays, the production of nitric oxide (NO) was measured by the Griess assay, and the protein expression levels of inducible nitric oxide (iNOS) and cyclooxygenase (COX-2) were examined by western blot analysis. The results showed that MFSCC concentrations ranging from 0.1 to 3 µg/mL did not show any significant cytotoxicity in LPS-induced RAW264.7 cells. Treatment with MFSCC of LPS-stimulated RAW264.7 cells significantly suppressed the production of NO and the expression of iNOS and COX-2 proteins related to inflammation. The present findings lead to a better understanding of the therapeutic potential of MFSCC and strongly promote it for the future clinical development of novel non-cell-based stem cell therapeutics.

14.
Int J Mol Sci ; 20(19)2019 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-31575035

RÉSUMÉ

Membrane-free stem cell components (MFSCC) from basal adipose tissue-derived stem cells (ADSCs) are unknown for the treatment strategies in osteoarthritis (OA). OA has been considered to be associated with inflammatory damage and cartilage degradation. In this study, we intended to investigate the molecular mechanism of the anti-inflammation and cartilage protection effect of MFSCC in vitro (rat primary chondrocytes) and in vivo (rat OA model). The MFSCC treatment significantly inhibited interleukin-1α (IL-1α) stimulated inflammation and cartilage degradation. The MFSCC considerably reduced the levels of inflammatory factors such as iNOS, COX-2, NO, and PGE2 and was suppressed NF-κB and MAPKs signaling pathways in IL-1α-stimulated rat chondrocytes. Additionally, biomarkers of OA such as MMP-9, COMP, and CTX-II decreased in the monosodium iodoacetate (MIA)-induced rat OA model by MFSCC treatment. In conclusion, the MFSCC was established to suppress IL-1α induced inflammation and cartilage degradation in vitro and in vivo. These findings provide new insight for understanding OA therapy using membrane-free stem cell approaches.


Sujet(s)
Cartilage hyalin/métabolisme , Interleukine-1 alpha/métabolisme , Arthrose/étiologie , Arthrose/métabolisme , Cellules souches/métabolisme , Animaux , Marqueurs biologiques , Chondrocytes/métabolisme , Cyclooxygenase 2/génétique , Cyclooxygenase 2/métabolisme , Cytokines/métabolisme , Dinoprostone/métabolisme , Modèles animaux de maladie humaine , Expression des gènes , Mitogen-Activated Protein Kinases/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Monoxyde d'azote/métabolisme , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Arthrose/anatomopathologie , Rats
15.
Sci Rep ; 9(1): 10029, 2019 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-31296948

RÉSUMÉ

The first 3-D direct observation of clusters of Nd oxide inside silicate glasses was achieved using atom probe tomography. Three-dimensional elemental maps of major chemical elements in glasses such as Si, Al, Zn and O showed no evidence of regions that had concentrations higher than the average values, whereas the Nd aggregated into regions of high concentration. Elemental maps of Nd and Pb recorded from the glasses containing PbS QDs showed highly-concentrated areas of both elements at the same locations; this result indicates that PbS QDs formation started in association with the Nd clusters.

16.
Int J Mol Med ; 43(1): 393-403, 2019 Jan.
Article de Anglais | MEDLINE | ID: mdl-30387810

RÉSUMÉ

To date, Korean hinoki cypress (Chamaecyparis obtusa), has been widely used for household and commercial purposes. Although the medicinal efficacy of hinoki cypress essential oil has been observed, that of the essential oil­derived terpenes, which exhibit a mechanism that acts against lung inflammation, remains to be fully elucidated. The present study investigated the anti­inflammatory effect of hinoki cypress leaf extracted essential oil on lipopolysaccharide (LPS)­stimulated WI38 fibroblast cells by inhibiting the nuclear factor κ­light­chain­enhancer of activated B cells (NF­κB) pathway, which exhibited lung tissue protection through the olfactory administration of essential oil in Sprague­Dawley rats. GC/MS analysis derived 24 terpenes from the essential oil. The morphological observations revealed that, upon LPS stimulation of WI38 fibroblast cells, inflammation was induced, whereas the condition of the cells reverted to normal in the essential oil extract pre­treated group. The results of western blot analysis revealed the inhibition of inducible nitric oxide synthase, activation of cyclooxygnase­2, and the degradation of cytosolic p65 and inhibitor of NF­κB­α in the LPS­stimulated group. Additionally, confocal imaging of nuclei revealed the translocation of phosphorylated p65, which was recovered in the cytosol in the phytoncide essential oil pre­treated group. Histopathological observation revealed that the alveolar capacity was enhanced in the essential oil olfactory administered rat group, compared with that in the normal rat group. These findings suggest that terpenes in essential oil from the Chamaecyparis obtusa leaf have therapeutic potential against respiratory inflammation­related disease.


Sujet(s)
Chamaecyparis/composition chimique , Fibroblastes/anatomopathologie , Inflammation/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Huile essentielle/pharmacologie , Feuilles de plante/composition chimique , Agents protecteurs/pharmacologie , Respiration/effets des médicaments et des substances chimiques , Administration par inhalation , Animaux , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cyclooxygenase 2/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Humains , Lipopolysaccharides , Mâle , Modèles biologiques , Nitric oxide synthase type II/métabolisme , Huile essentielle/administration et posologie , Agents protecteurs/administration et posologie , Alvéoles pulmonaires/effets des médicaments et des substances chimiques , Alvéoles pulmonaires/anatomopathologie , Rat Sprague-Dawley , Terpènes/analyse
17.
Nutrients ; 10(11)2018 Oct 30.
Article de Anglais | MEDLINE | ID: mdl-30380781

RÉSUMÉ

Pectolinarigenin (PEC), a natural flavonoid that is present in citrus fruits, has been reported to exhibit antitumor effects in several cancers. Though the mechanism of PEC-induced cytotoxicity effects has been documented, the proteomic changes that are associated with the cellular response to this flavonoid are poorly understood in gastric cancer cells. In this study, a comparative proteomic analysis was performed to identify proteins associated with PEC-induced cell death in two human gastric cancer cell lines: AGS and MKN-28. Two-dimensional gel electrophoresis (2-DE) revealed a total of 29 and 56 protein spots with significant alteration were screened in AGS and MKN-28 cells respectively. In total, 13 (AGS) and 39 (MKN28) proteins were successfully identified by mass spectrometry from the differential spots and they are known to be involved in signal transduction, apoptosis, transcription and translation, cell structural organization, and metabolism, as is consistent with multiple effects of PEC on tumor cells. Notably, novel target proteins like Probable ATP-dependent RNA helicase DDX4 (DDX4) and E3 ubiquitin-protein ligase LRSAM1 (LRSAM1) along with the commonly differential expressed proteins on both the cell lines that are treated with PEC were confirmed by immunoblotting. The DDX4 accelerates cell cycle progression by abrogating the G2 checkpoint when overexpressed in cancer cells, while the aberrant expression of LRSAM1 may be involved in the cancer pathology. Thus, proteomic analysis provides vital information about target proteins that are important for PEC-induced cell death in gastric cancer cells.


Sujet(s)
Antinéoplasiques/pharmacologie , 4H-1-Benzopyran-4-ones/pharmacologie , Protéines tumorales/effets des médicaments et des substances chimiques , Tumeurs de l'estomac/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , DEAD-box RNA helicases/effets des médicaments et des substances chimiques , Électrophorèse bidimensionnelle sur gel , Analyse de profil d'expression de gènes , Humains , Protéomique , Transduction du signal/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/effets des médicaments et des substances chimiques
18.
Oncol Rep ; 40(6): 3249-3260, 2018 Dec.
Article de Anglais | MEDLINE | ID: mdl-30272339

RÉSUMÉ

Proteomic analysis serves as an important biological tool for identifying biological events. Novel biomarkers of a specific disease such as cancer may be identified using these promising techniques. The aim of the present study was to investigate the effect of tangeretin and to identify potential biomarkers in AGS gastric cancer cells using a proteomics approach. The results of the present study revealed that tangeretin inhibited AGS cell viability dose­dependently with a half­maximal inhibitory concentration of 100 µM. Two­dimensional gel electrophoresis was performed to determine the potential biomarker between control and tangeretin (100 µM)­treated AGS cells. A total of 16 proteins was identified from 36 significant protein spots using matrix­assisted laser­desorption/ionization time­of­flight­mass spectrometry using peptide fingerprinting. The bioinformatics tools Protein ANalysis THrough Evolutionary Relationships (PANTHER) and Database for Annotation, Visualization and Integrated Discovery (DAVID) were used to identify the functional properties and association of the proteins obtained. Using western blot analysis, the regulatory pattern of four selected proteins, protein kinase Cε, mitogen­activated protein kinase 4, phosphoinositide 4­kinase and poly(ADP­ribose) polymerase 14, were successfully verified in replicate sample sets. These selected proteins are primarily involved in apoptosis signaling, angiogenesis, cell cycle regulation, receptor kinase binding, intracellular cytoplasmic and nuclear alterations. Therefore, aim of the present study was to identify potential diagnostic biomarkers from the functional categories of altered protein expression in tangeretin­inhibited AGS gastric cancer cell viability.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Flavones/pharmacologie , Protéomique/méthodes , Tumeurs de l'estomac/métabolisme , 1-Phosphatidylinositol 4-kinase/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Électrophorèse bidimensionnelle sur gel , Extracellular Signal-Regulated MAP Kinases/métabolisme , Humains , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Protein kinase C-epsilon/métabolisme , Tumeurs de l'estomac/traitement médicamenteux
19.
Nutrients ; 10(8)2018 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-30096805

RÉSUMÉ

Pectolinarigenin (PEC), a natural flavonoid present in Cirsium chanroenicum and in some species of Citrus fruits, has various pharmacological benefits such as anti-inflammatory and anti-cancer activities. In the present study, we investigated the anti-cancer mechanism of PEC induced cell death caused by autophagy and apoptosis in AGS and MKN28 human gastric cancer cells. The PEC treatment significantly inhibited the AGS and MKN28 cell growth in a dose-dependent manner. Further, PEC significantly elevated sub-G1 phase in AGS cells and G2/M phase cell cycle arrest in both AGS and MKN28 cells. Apoptosis was confirmed by Annexin V and Hoechst 33342 fluorescent staining. Moreover, Immunoblotting results revealed that PEC treatment down-regulated the inhibitor of apoptosis protein (IAP) family protein XIAP that leads to the activation of caspase-3 thereby cleavage of PARP (poly-ADP-ribose polymerase) in both AGS and MKN28 cells in a dose-dependent manner. The autophagy-inducing effect was indicated by the increased formation of acidic vesicular organelles (AVOs) and increased protein levels of LC3-II conversion in both AGS and MKN28 cells. PEC shows the down regulation of PI3K/AKT/mTOR pathway which is a major regulator of autophagic and apoptotic cell death in cancer cells that leads to the down-regulation of p-4EBP1, p-p70S6K, and p-eIF4E in PEC treated cells when compared with the untreated cells. In conclusion, PEC treatment might have anti-cancer effect by down-regulation of PI3K/AKT/mTOR pathway leading to G2/M phase cell cycle arrest, autophagic and apoptotic cell death in human gastric cancer cells. Further studies of PEC treatment can support to develop as a potential alternative therapeutic agent for human gastric carcinoma.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , 4H-1-Benzopyran-4-ones/pharmacologie , Phosphatidylinositol 3-kinase/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Sérine-thréonine kinases TOR/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Protéines associées à l'autophagie/métabolisme , Lignée cellulaire tumorale , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de l'estomac/enzymologie , Tumeurs de l'estomac/anatomopathologie
20.
Oncotarget ; 9(5): 5993-6006, 2018 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-29464049

RÉSUMÉ

Gastric cancer is the fifth most common cancer and the third leading cause of cancer deaths worldwide. South Korea is in first place with 9,180 death alone attributed to gastric cancer in 2013. Plenty of literature suggests the evasion of apoptosis is implicated in neurodegeneration, autoimmune diseases, and tumors development due to dysregulation in the apoptotic mechanism. Reduced apoptosis or its resistance in cancer cells plays a significant role in carcinogenesis. It's imperative to understand apoptosis, which provides the basis for novel targeted therapies that can induce cancer cell death or sensitize them to cytotoxic agents by regulating key factors like IAPs, MDM2, p53, caspases and much more. Studies have demonstrated that Scutellarein have the ability to inhibit several cancer cells by inducing apoptosis with both: Scutellarein monomers as well as scutellarein containing flavonoids. MTT results revealed that scutellarein inhibited cell viability in both dose and time dependent manner. Flow cytometry and western blot analysis showed that scutellarein induces apoptosis in both AGS and SNU-484 human gastric cancer cells and G2/M phase cell cycle arrest in SNU-484 cells. This study demonstrated that the Scutellarein on AGS and SNU-484 cells significantly inhibits cell proliferation and induces apoptotic cell death via down regulating MDM2 and activated the tumor suppresser protein p53, subsequently down regulating the IAP family proteins (cIAP1, cIAP2, and XIAP) leading to caspase-dependent apoptosis in AGS and SNU-484 cells.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...