Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Endocrinol (Lausanne) ; 13: 894883, 2022.
Article in English | MEDLINE | ID: mdl-36147572

ABSTRACT

Background: Aged women appear to be at a higher risk of developing memory impairment than men. Whether menopausal hormone therapy (MHT) could improve memory in postmenopausal women remains unclear. We thus conducted a meta-analysis to investigate the potential effect of MHT on memory, especially verbal memory, in postmenopausal women. Methods: PubMed, Cochrane, Embase, Chinese Biomedical Literature Database, and web of ClinicalTrials.gov were systematically searched for randomized controlled trials comparing MHT versus placebo in postmenopausal women. Our primary outcome of interest is memory function. Results: We included 10 studies with 2,818 participants in the final analysis. There was no significant differences in immediate recall (weighted mean difference [WMD] 0.34, 95% confidence interval [CI]: -0.73, 1.40), delayed recall (WMD 0.99, 95% CI: -0.51, 2.48), short-delay (WMD -0.00, 95% CI: -0.37, 0.37), and long-delay (WMD -0.19, 95% CI: -0.69, 0.31) recall between WMT and placebo. WMT was associated with a lower digit span forward (mean reduction -0.20, 95% CI: -0.36, -0.03). In women within 5 years of menopause, MHT did not differ in immediate (0.45, 95% CI: -0.75, 1.65) or delayed recall (1.03, 95% CI: -0.93, 3.00), and digit span forward (-0.11, 95% CI: -0.72, 0.50), when compared with placebo. Conclusion: This meta-analysis suggested that MHT had no effect on verbal memory in postmenopausal women, and may impair some domains of short-term memory. Current available evidence does not support MHT for improving memory in women less than 60 years, even in recently menopausal women. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO, identifier CRD42021233255.


Subject(s)
Hormone Replacement Therapy , Menopause , Aged , Female , Humans , Memory
2.
J Chem Phys ; 153(14): 144501, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33086840

ABSTRACT

The thermal properties of ß-Ga2O3 can significantly affect the performance and reliability of high-power electronic devices. To date, due to the absence of a reliable interatomic potential, first-principles calculations based on density functional theory (DFT) have been routinely used to probe the thermal properties of ß-Ga2O3. DFT calculations can only tackle small-scale systems due to the huge computational cost, while the thermal transport processes are usually associated with large time and length scales. In this work, we develop a machine learning based Gaussian approximation potential (GAP) for accurately describing the lattice dynamics of perfect crystalline ß-Ga2O3 and accelerating atomic-scale simulations. The GAP model shows excellent convergence, which can faithfully reproduce the DFT potential energy surface at a training data size of 32 000 local atomic environments. The GAP model is then used to predict ground-state lattice parameters, coefficients of thermal expansion, heat capacity, phonon dispersions at 0 K, and anisotropic thermal conductivity of ß-Ga2O3, which are all in excellent agreement with either the DFT results or experiments. The accurate predictions of phonon dispersions and thermal conductivities demonstrate that the GAP model can well describe the harmonic and anharmonic interactions of phonons. Additionally, the successful application of our GAP model to the phonon density of states of a 2500-atom ß-Ga2O3 structure at elevated temperature indicates the strength of machine learning potentials to tackle large-scale atomic systems in long molecular simulations, which would be almost impossible to generate with DFT-based molecular simulations at present.

3.
J Biochem Mol Toxicol ; 34(5): e22460, 2020 May.
Article in English | MEDLINE | ID: mdl-32022984

ABSTRACT

Gypenosides have anticancer activity against many cancers. Gypenoside LI is a gypenoside monomer from Gynostemma pentaphyllum, its pharmacological functions in melanoma have not been reported. In this study, we found that gypenoside LI had a potent cytotoxic effect on melanoma cells. Gypenoside LI can induce intrinsic apoptosis along with S phase arrest. Furthermore, gypenoside LI inhibited the colony formation ability of melanoma through inhibition of the Wnt/ß-catenin signaling pathway. Interestingly, we also found that gypenoside LI can induce the upregulation of the tumor suppressor miR-128-3p during melanoma apoptosis. In contrast, gypenoside LI induced apoptosis, cell cycle arrest, and inhibition of the Wnt/ß-catenin signaling pathway, which were abolished by overexpression of the miR-128-3p inhibitor in A375 cells. Taken together, these results showed that gypenoside LI could inhibit human melanoma cells through inducing apoptosis, arresting cell cycle at the S phase and suppressing the Wnt/ß-catenin signaling pathway in a miR-128-3p dependent manner.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Gynostemma/chemistry , Melanoma/metabolism , MicroRNAs/metabolism , Plant Extracts/pharmacology , Skin Neoplasms/metabolism , Up-Regulation/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Humans , Melanoma/pathology , MicroRNAs/genetics , S Phase Cell Cycle Checkpoints/drug effects , Skin Neoplasms/pathology , Transfection , Wnt Signaling Pathway/drug effects
4.
Planta Med ; 85(5): 394-405, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30562828

ABSTRACT

Damulin B, a dammarane-type saponin from steamed Gynostemma pentaphyllum, exhibits the strongest activity against human lung carcinoma A549 cells among the isolated active saponins. In this study, the structure-activity relationship of a series of saponin compounds was discussed. The inhibitory effect of damulin B on human lung cancer A549 and H1299 cells was investigated from apoptosis, cell cycle, and migration aspects. In vitro, human lung cancer cells were more susceptible to damulin B treatment than human normal fibroblasts. Damulin B exhibited a strong cytotoxic effect, as evidenced by the increase of apoptosis rate, reduction of mitochondrial membrane potential (MMP), generation of reactive oxygen species, and G0/G1 phase arrest. Furthermore, damulin B activated the following: both intrinsic and extrinsic apoptosis pathways along with early G1 phase arrest via the upregulation of the Bax, Bid, tBid, cleaved caspase-8, and p53 expression levels; downregulation of the procaspase-8/-9, CDK4, CDK6, and cyclin D1 expression levels; and more release of cytochrome c in the cytoplasm. In addition, antimigratory activities and suppressive effects on metastasis-related factors, such as MMP-2 and MMP-9, accompanied by the upregulation of IL-24 were revealed. Altogether, the results proved that damulin B could inhibit human lung cancer cells by inducing apoptosis, blocking the cell cycle at early G0/G1 phase and suppressing the migration. Hence, damulin B has potential therapeutic efficacy against lung cancer.


Subject(s)
Gynostemma/chemistry , Lung Neoplasms/drug therapy , Saponins/pharmacology , Triterpenes/pharmacology , A549 Cells , Cell Cycle/drug effects , G1 Phase , Humans , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species , Saponins/chemistry , Signal Transduction , Structure-Activity Relationship , Triterpenes/chemistry , Dammaranes
6.
J Ethnopharmacol ; 219: 161-172, 2018 Jun 12.
Article in English | MEDLINE | ID: mdl-29545210

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Gypenosides are major constituents in Gynostemma pentaphyllum (Thunb.) Makino. Previous studies have shown that gypenosides isolated from G. pentaphyllum possess inhibitory effect on the growth of cancer cells, especially A549 cells, with structure-activity relationship (SAR). However, the underlying mechanism of gypenoside-induced A549 cell death remains to be clarified. AIM OF THE STUDY: To further investigate SAR and the underlying mechanism of gypenosides in A549 cells. MATERIALS AND METHODS: Gypenosides were isolated from G. pentaphyllum using chromatography methods and identified using MS and NMR data. The cytotoxicity was determined with CCK-8 assay. The effects of gypenosides on apoptosis, cell cycle and migration were investigated through cell morphology observation, flow cytometry analysis and key proteins detection. RESULTS: Three gypenosides, 2α,3ß,12ß,20(S)-tetrahydroxydammar-24-ene-3-O-ß-D-glucopyranoside-20-O-ß-D-glucopyranoside, gypenoside L and gypenoside LI were isolated from G. pentaphyllum. Gypenoside stereoisomers, gypenoside L (S configuration at C20) and gypenoside LI (R configuration at C20) showed stronger activity against A549 cells. Furthermore, both induced A549 cell apoptosis through intrinsic and extrinsic pathways evidenced by reducing mitochondrial membrane potential (MMP), generating reactive oxygen species (ROS), releasing more cytochrome c and down-regulating procaspase 8. However, gypenoside L blocked A549 cells in G0/G1, while gypenoside LI induced G2/M arrest, which was further verified by different expression of CDK1, CDK2 and CDK4. In addition, both inhibited A549 cell migration, which was evidenced by down-regulation of MMP-2/9 as well as scratch wound assay and transwell assay. CONCLUSION: C20 of gypenoside played an important role in A549 cell cytotoxicity and gypenoside stereoisomers could be used as potential multi-target chemopreventive agents for cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Growth Inhibitors/pharmacology , Gynostemma , Lung Neoplasms , Plant Extracts/pharmacology , A549 Cells , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Movement/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Growth Inhibitors/chemistry , Growth Inhibitors/isolation & purification , Gynostemma/chemistry , Hep G2 Cells , Humans , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Stereoisomerism
7.
Biomed Environ Sci ; 29(1): 80-4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26822515

ABSTRACT

The molecular mechanism of DNA damage induced by hydroquinone (HQ) remains unclear. Poly(ADP-ribose) polymerase-1 (PARP-1) usually works as a DNA damage sensor, and hence, it is possible that PARP-1 is involved in the DNA damage response induced by HQ. In TK6 cells treated with HQ, PARP activity as well as the expression of apoptosis antagonizing transcription factor (AATF), PARP-1, and phosphorylated H2AX (γ-H2AX) were maximum at 0.5 h, 6 h, 3 h, and 3 h, respectively. To explore the detailed mechanisms underlying the prompt DNA repair reaction, the above indicators were investigated in PARP-1-silenced cells. PARP activity and expression of AATF and PARP-1 decreased to 36%, 32%, and 33%, respectively, in the cells; however, γ-H2AX expression increased to 265%. Co-immunoprecipitation (co-IP) assays were employed to determine whether PARP-1 and AATF formed protein complexes. The interaction between these proteins together with the results from IP assays and confocal microscopy indicated that poly(ADP-ribosyl)ation (PARylation) regulated AATF expression. In conclusion, PARP-1 was involved in the DNA damage repair induced by HQ via increasing the accumulation of AATF through PARylation.


Subject(s)
Antioxidants/toxicity , Apoptosis Regulatory Proteins/metabolism , DNA Damage/drug effects , Hydroquinones/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Repressor Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Cell Line , Gene Expression Regulation/drug effects , Gene Silencing , Histones/genetics , Histones/metabolism , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Transport , Repressor Proteins/genetics
8.
World J Gastroenterol ; 21(29): 8858-67, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26269675

ABSTRACT

AIM: To investigate pim-3 expression in hepatic stellate cells (HSCs) stimulated by lipopolysaccharide (LPS), and its protective effect on HSCs. METHODS: Rat HSC-T6 cells were stimulated by LPS. The effect of LPS on proliferation and apoptosis of HSC-T6 cells was investigated by methyl thiazoyltetrazolium (MTT) assay and flow cytometry after annexin V-fluorescein isothiocyanate/propidium iodide double staining. pim-3 mRNA and protein were detected by reverse transcriptase polymerase chain reaction and Western blotting at 48 h when HSC-T6 cells were stimulated with 1 µg/mL LPS for 0, 3, 6, 12, 24 and 48 h. The cells without stimulation served as controls. To study the effect of pim-3 kinase on HSC-T6 cells, si-pim3 (siRNA against pim-3) was transfected into HSC-T6 cells. HSC-T6 cells were subjected to different treatments, including LPS, si-pim3, or si-pim3 plus LPS, and control cells were untreated. Protein expression of pim-3 was detected at 48 h after treatment, and cell proliferation at 24 and 48 h by MTT assay. Apoptosis was detected by flow cytometry, and confirmed with caspase-3 activity assay. RESULTS: LPS promoted HSC-T6 cell proliferation and protected against apoptosis. Significantly delayed upregulation of pim-3 expression induced by LPS occurred at 24 and 48 h for mRNA expression (pim-3/ß-actin RNA, 24 or 48 h vs 0 h, 0.81 ± 0.20 or 0.78 ± 0.21 vs 0.42 ± 0.13, P < 0.05), and occurred at 12 h and peaked at 24 and 48 h for protein expression (pim-3/GAPDH protein, 12, or 24 or 48 h vs 0 h, 0.68 ± 0.12, 1.47 ± 0.25 or 1.51 ± 0.23 vs 0.34 ± 0.04, P < 0.01). pim-3 protein was ablated by si-pim3 and upregulated by LPS in HSC-T6 cells at 48 h after treatment (pim-3/GAPDH: si-pim3, si-pim3 plus LPS or LPS vs control, 0.11 ± 0.05, 0.12 ± 0.05 or 1.08 ± 0.02 vs 0.39 ± 0.03, P < 0.01). Ablation of pim-3 by si-pim3 in HSC-T6 cells partly abolished proliferation (OD at 24 h, si-pim3 group or si-pim3 plus LPS vs control, 0.2987 ± 0.050 or 0.4063 ± 0.051 vs 0.5267 ± 0.030, P < 0.01; at 48 h 0.4634 ± 0.056 or 0.5433 ± 0.031 vs 0.8435 ± 0.028, P < 0.01; si-pim3 group vs si-pim3 plus LPS, P < 0.01 at 24 h and P < 0.05 at 48 h), and overexpression of pim-3 in the LPS group increased cell proliferation (OD: LPS vs control, at 24 h, 0.7435 ± 0.028 vs 0.5267 ± 0.030, P < 0.01; at 48 h, 1.2136 ± 0.048 vs 0.8435 ± 0.028, P < 0.01). Ablation of pim3 with si-pim3 in HSC-T6 cells aggravated apoptosis (si-pim3 or si-pim3 plus LPS vs control, 42.3% ± 1.1% or 40.6% ± 1.3% vs 16.8% ± 3.3%, P < 0.01; si-pim3 vs si-pim3 plus LPS, P > 0.05), and overexpression of pim-3 in the LPS group attenuated apoptosis (LPS vs control, 7.32% ± 2.1% vs 16.8% ± 3.3%, P < 0.05). These results were confirmed by caspase-3 activity assay. CONCLUSION: Overexpression of pim-3 plays a protective role in LPS-stimulated HSC-T6 cells.


Subject(s)
Hepatic Stellate Cells/drug effects , Lipopolysaccharides/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic , Hepatic Stellate Cells/enzymology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/metabolism , Rats , Time Factors , Transfection , Up-Regulation
9.
Biomed Environ Sci ; 22(1): 14-21, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19462682

ABSTRACT

OBJECTIVE: To screen miRNA profiles of malignantly transformed human bronchial epithelial cells, 16HBE-T, induced by anti-benzo[a]pyrene-trans-7,8-diol-9,10-epoxide (anti-BPDE), and to analyze putative miR-10a targets in 16HBE-T. METHODS: A novel microarray platform was employed to screen miRNA profiles of 16HBE-T cells transformed by anti-BPDE. Microarray data for miR-10a and miR-320 were validated using quantitative real time polymerase chain reaction (QRT-PCR). The expression of a putative target for miR-10a, HOXA1, was analyzed by reverse transcription polymerase chain reaction (RT-PCR) and QRT-PCR. RESULTS: In comparison with the vehicle-treated cells (16HBE-N), 16HBE-T exhibited differential expression of 54 miRNAs, in which, 45 were over-expressed and 9 were down-regulated. The five most highly expressed miRNAs were miR-494, miR-320, miR-498, miR-129, and miR-106a. The lowest expressed miRNAs were miR-10a, miR-493-5p, and miR-363*. Three members of miR-17-92 cluster, miR-17-5p, miR-20a, and miR-92, showed significantly higher abundance in 16BHE-T as miR-21, miR-141, miR-27a, miR-27b, miR-16 and miRNAs of the let-7 family. The putative target for miR-10a, HOXA1 mRNA was up-regulated 3-9-fold in 16HBE-T, as compared with 16HBE-N. CONCLUSION: The findings of the study provide information on differentially expressed miRNA in malignant 16HBE-T, and also suggest a potential role of these miRNAs in cell transformation induced by anti-BPDE. HOXA1 is similarly up-regulated, suggesting that miR-10a is associated with the process of HOXA 1-mediated transformation.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Carcinogens/toxicity , Cell Transformation, Neoplastic/chemically induced , MicroRNAs/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...