Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
2.
Sci Rep ; 14(1): 7489, 2024 03 29.
Article En | MEDLINE | ID: mdl-38553504

This study aims to evaluate the safety of Alprazolam by analyzing the FAERS database, provide data analysis for monitoring adverse drug reactions. This research encompasses adverse event (AE) reports related to Alprazolam from the first quarter of 2004 to the second quarter of 2023. Four signal mining and analysis methods were utilized, including Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Empirical Bayesian Geometric Mean (EBGM). Further exploration was conducted regarding patient characteristics and types of AEs. A total of 23,575 AE reports in which Alprazolam was the primary suspect drug were collected, identifying 347 Preferred Term (PT) signals and 27 System Organ Classes (SOCs). The number of AE reports increased annually, especially in 2015, 2018, 2019, and 2020. The main affected groups were females and the age range of 18 to 45. Psychiatric disorders, Nervous system disorders, and Gastrointestinal disorders were the most common the organ system in which the AEs occurred. There is a certain risk of drug abuse and suicide with Alprazolam. Most notably, several AEs not recorded in the Alprazolam leaflet appeared among the top 30 PTs in signal strength, including but not limited to Benzodiazepine drug level abnormal, Acquired amegakaryocytic thrombocytopenia, Cutaneous T-cell dyscrasia, and Coronary No-reflow Phenomenon. For the first time, AEs related to the cardiovascular system and platelet function were unveiled. The severe AE reports that resulted in "hospitalization" and "death" accounted for 30.96% and 21.86%. This study highlights the risks of suicide and misuse of Alprazolam. Other potential severe or fatal AEs, such as those related to the cardiovascular system, platelet function, and others, require further research to determine their precise mechanisms and risk factors.


Alprazolam , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Male , Alprazolam/adverse effects , Bayes Theorem , Benzodiazepines , Risk Factors , Drug-Related Side Effects and Adverse Reactions/epidemiology , Risk Assessment
3.
Front Immunol ; 13: 950884, 2022.
Article En | MEDLINE | ID: mdl-36238304

Hepatocellular carcinoma (HCC) responds poorly to standard chemotherapy or targeted therapy; hence, exploration for novel therapeutic targets is urgently needed. CEP192 protein is indispensable for centrosome amplification, which has been extensively characterized in both hematological malignancies and solid tumors. Here, we combined bioinformatics and experimental approaches to assess the potential of CEP192 as a prognostic and therapeutic target in HCC. CEP192 expression increased with tumor stage and was associated with poor clinicopathologic features, frequent recurrence, and higher mortality. Upon single-cell RNA sequencing, CEP192 was found to be involved in the proliferation and self-renewal of hepatic progenitor-like cells. This observation was further evidenced using CEP192 silencing, which prevented tumor cell proliferation and self-renewal by arresting cells in the G0/G1 phase of the cell cycle. Notably, CEP192 was highly correlated with multiple tumor-associated cytokine ligand-receptor axes, including IL11-IL11RA, IL6-IL6R, and IL13-IL13RA1, which could promote interactions between hepatic progenitor-like cells, PLVAP+ endothelial cells, tumor-associated macrophages, and CD4+ T cells. Consequently, CEP192 expression was closely associated with an immunosuppressive tumor microenvironment and low immunophenoscores, making it a potential predictor of response to immune checkpoint inhibitors. Taken together, our results unravel a novel onco-immunological role of CEP192 in establishing the immunosuppressive tumor microenvironment and provide a novel biomarker, as well as a potential target for therapeutic intervention of HCC.


Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Chromosomal Proteins, Non-Histone/metabolism , Endothelial Cells/metabolism , Humans , Immune Checkpoint Inhibitors , Interleukin-11 , Interleukin-13 , Interleukin-6 , Ligands , Liver Neoplasms/pathology , Prognosis , Tumor Microenvironment
4.
J Cancer ; 12(22): 6665-6675, 2021.
Article En | MEDLINE | ID: mdl-34659556

Background: The long-term survival rate of gastric cancer (GC) patients at advanced stages remains low worldwide. Circular RNAs (circRNAs) a newly studied type of non-coding RNA that play an important role in the pathogenesis and diagnosis of various diseases. In this research, we aimed to explore the functions of hsa_circRNA_101996 in GC cells and an animal model of GC. Methods: The expression of hsa_circRNA_101996, microRNA (miR)-143, and ten-eleven translocation (TET)-2 in GC tissues, the adjacent tissues, and cell lines were determined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Transwell assays were used to analyze the knockdown effects of hsa_circRNA_101996, miR-143, and overexpression of TET2 on cell proliferation, migration, and invasion abilities. Western blotting was used to analyze the expression of matrix metalloproteinases (MMP)2/MMP9. Binding interactions between, hsa_circRNA_101996 and miR-143 and between, miR-143 and TET2 were detected by Dual-luciferase reporter assays. Levels of protein expression were analyzed by Western blotting. Tumor models were established by subcutaneous injection of tumor cells in Bl6/Rag2/GammaC double knockout mice. Results: The result showed that hsa_circRNA_101996 expression was significantly upregulated in GC tissues compared to that in the adjacent tissues, and its level in cancer tissue was correlated with tumor size, lymphatic metastasis, and distant metastasis. Compared with the low hsa_circRNA_101996 expression group, the three-year survival rate of patients in the high hsa_circRNA_101996 expression group was significantly lower. The knockdown of hsa_circRNA_101996 dramatically suppressed the cell migration, invasion, and proliferation of GC cells by sponging to absorb miR-143 and elevated the expression of TET2. In vivo studies showed that the knockdown of hsa_circRNA_101996 delayed tumor growth. Furthermore, we revealed that TET2 regulates MMP2/MMP9 expression through the DNA demethylation pathway. Conclusion: Our findings indicate that hsa_circRNA_101996 promotes GC development by upregulating MMP2/MMP9 through miR-143/TET2 pathway, which may provide a novel target for GC.

5.
PPAR Res ; 2018: 6970407, 2018.
Article En | MEDLINE | ID: mdl-29686697

BACKGROUND AND AIMS: Accumulating evidence reveals that PPARγ plays a unique role in the regulation of hepatic fibrosis and hepatic stellate cells (HSCs) activation. This study was aimed at investigating the role of PPARγ in hypoxia-induced hepatic fibrogenesis and its possible mechanism. METHODS: Rats used for CCl4-induced hepatic fibrosis model were exposed to hypoxia for 8 hours each day. Rats exposed to hypoxia were treated with or without the PPARγ agonist rosiglitazone. Liver sections were stained with HE and Sirius red staining 8 weeks later. HSCs were exposed to hypoxic environment in the presence or absence of rosiglitazone, and expression of PPARγ and two fibrosis markers, α-SMA and desmin, were measured using western blot and immunofluorescence staining. Next, levels of PPARγ, α-SMA, and desmin as well as PKG and cGMP activity were detected using PI3K/AKT and a cGMP activator or inhibitor. RESULTS: Hypoxia promoted the induction and progress of hepatic fibrosis and HSCs activation. Meanwhile, rosiglitazone significantly antagonized the effects induced by hypoxia. Signaling by sGC/cGMP/PKG promoted the inhibitory effect of PPARγ on hypoxia-induced activation of HSCs. Moreover, PI3K/AKT signaling or PDE5 blocked the above response of PPARγ. CONCLUSION: sGC/cGMP/PKG and PI3K/AKT signals act on PPARγ synergistically to attenuate hypoxia-induced HSC activation.

6.
Environ Sci Pollut Res Int ; 24(1): 500-508, 2017 Jan.
Article En | MEDLINE | ID: mdl-27730508

Air pollution caused by particulate matter with aerodynamic diameters less than 2.5 µm (PM2.5) is a serious environmental problem. Plants can improve air quality by removing PM2.5 from the atmosphere. However, direct evidence of PM2.5 absorption and assimilation into plants has not yet been found. In this study, we demonstrate that 15NH4+ in PM2.5 was absorbed by poplar leaves in low and high PM2.5 treatment groups (namely, LPT and HPT). Then, 15N was subsequently transferred to other parts of the treated seedlings as shown by 15N tracing and simulated PM2.5 generation. 15N and total N contents were the highest in high pollution treatment (HPT), followed by that in low pollution treatment (LPT) and the control. Glutamate dehydrogenase (GDH) contributed more to NH4+ assimilation than glutamine synthetase and glutamate synthase in the leaves of treated seedlings. GDH aminating activity was induced upon NH4+ exposure whereas GDH deaminating activity was repressed in both LPT and HPT, suggesting that poplar seedlings can alleviate NH4+ toxicity by enhancing NH4+ assimilation. At the end of PM2.5 treatment period, the decreased amino acid content in the treated seedlings was attributed to the probably altered balance of amino acid metabolism. The decline in the net photosynthetic rate (Pn) was accompanied by the decrease in the stomatal conductance in poplar leaves with the extension of PM2.5 treatment time, indicating that stomatal limitation is a major reason for Pn reduction. This study may provide novel insights into the relationship between PM2.5 pollution and plants.


Air Pollutants/metabolism , Nitrogen/metabolism , Particulate Matter/metabolism , Populus/metabolism , Ammonium Compounds/metabolism , Nitrogen Isotopes/analysis , Particle Size , Plant Leaves/drug effects , Plant Leaves/enzymology , Plant Leaves/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Populus/drug effects , Populus/enzymology , Seedlings/drug effects , Seedlings/enzymology , Seedlings/metabolism
7.
Am J Transl Res ; 9(12): 5538-5547, 2017.
Article En | MEDLINE | ID: mdl-29312505

Cancer-derived mesenchymal stem cells (MSCs) seem to play an important role in mediating tumor angiogenesis. Recently, curcumin has been shown to display multiple therapeutic properties, including anticancer activity. In the present study, we have tried to explore the role of curcumin in regulating gastric cancer cells-derived mesenchymal stem cells (GC-MSCs) mediated angiogenesis. Our results showed that curcumin attenuated the high expression levels of fibroblast proteins (α-SMA & Vimentin) in GC-MSCs. Its treatment also inhibited GC-MSCs induced human umbilical vein endothelial cells (HUVEC) tube formation, migration and colony formation. Furthermore, it was noticed that curcumin abrogated NF-κB signaling activity and VEGF production in GC-MSCs. Next, to establish the link between regulation of NF-κB/VEGF signaling by curcumin, and its influence on GC-MSC-derived angiogenesis, we pretreated GC-MSCs with either NF-κB inhibitor PDTC or a neutralizing antibody against VEGF (NA-VEGF), and then collected conditioned media (CM). The HUVEC cells were then cultured in this conditioned media to test their ability to form tubes, migrate and form colonies. Our results demonstrated that NF-κB/VEGF signaling is important for GC-MSCs induced tube formation, migration and colony formation in HUVEC cells. Moreover, we also observed that NF-κB/VEGF signaling regulated VEGF expression of gastric cancer cells both in vitro and in vivo. Overall, our study indicated that curcumin may serve as a novel therapeutic target for GC-MSCs derived angiogenesis, by inhibiting NF-κB/VEGF signaling.

...