Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Toxicol Appl Pharmacol ; 484: 116884, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38442791

ABSTRACT

BACKGROUND: The global increase in the aging population has led to a higher incidence of osteoporosis among the elderly. OBJECTIVE: This study aimed to evaluate the protective properties of pinoresinol diglucoside (PDG), an active constituent of Eucommia ulmoides, against dexamethasone-induced osteoporosis and chondrodysplasia. METHODS: A zebrafish model of osteoporosis was established by exposing larval zebrafish to dexamethasone. The impact of PDG on bone mineralization was assessed through alizarin red and calcein staining. Alkaline phosphatase activity was quantified to evaluate osteoblast function. The influence of PDG on chondrogenesis was estimated using alcian blue staining. Fluorescence imaging and motor behavior analysis were employed to assess the protective effect of PDG on the structure and function of dexamethasone-induced skeletal teratogenesis. qPCR determined the expression of osteogenesis and Wnt signaling-related genes. Molecular docking was used to assess the potential interactions between PDG and Wnt receptors. RESULTS: PDG significantly increased bone mineralization and corrected spinal curvature and cartilage malformations in the zebrafish model. Furthermore, PDG enhanced swimming abilities compared to the model group. PDG mitigated dexamethasone-induced skeletal abnormalities in zebrafish by upregulating Wnt signaling, showing potential interaction with Wnt receptors FZD2 and FZD5. CONCLUSION: PDG mitigates dexamethasone-induced osteoporosis and chondrodysplasia by promoting bone formation and activating Wnt signaling.


Subject(s)
Lignans , Osteoporosis , Zebrafish , Humans , Animals , Aged , Molecular Docking Simulation , Osteogenesis , Dexamethasone/pharmacology , Osteoporosis/chemically induced , Osteoporosis/prevention & control , Receptors, Wnt , Cell Differentiation
2.
Mol Cancer Ther ; 23(5): 721-732, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38295302

ABSTRACT

In this study, we explored the therapeutic potential of everolimus, an mTOR inhibitor, in a patient-derived xenograft (PDX) of rhabdomyosarcoma, the most prevalent malignant pediatric sarcoma. In addition, rhabdoid tumor cell line A-204 and Ewings sarcoma cell line A-673 were cultured to assess the in vitro effect of everolimus. Furthermore, the cell-derived xenograft (CDX) of A-673 was established and treated with everolimus in vivo. IHC and Western blotting were performed to detect the expressions of pertinent proteins. Results showed that everolimus intervention had limited inhibitory effect on PDX tumor growth compared with cyclophosphamide. Nevertheless, everolimus treatment significantly influenced the phosphorylation levels of S6 kinase beta 1 (S6K1) and eIF4E-binding protein 1 (p-4E-BP1), resulting in the inhibition of angiogenesis in vitro and in vivo. Interestingly, everolimus led to an upregulation in the level of IL17A in sarcoma cells. Notably, when secukinumab, a mAb of IL17A, was combined with everolimus, it synergistically enhanced the inhibitory effect of everolimus on sarcoma cell proliferation in vitro and on the growth of PDX or CDX xenograft tumors in vivo. Importantly, this combination therapy did not affect the mTOR signaling. These results indicate that everolimus exerts an antipediatric sarcoma effect by inhibiting mTOR signal. However, everolimus induces sarcoma cells to produce IL17A, which promotes tumor cell survival and counteracts its antipediatric sarcoma effect. The combination of secukinumab effectively eliminates the effects of IL17A, thereby improving the therapeutic efficacy of everolimus in the context of pediatric sarcomas.


Subject(s)
Antibodies, Monoclonal, Humanized , Cell Proliferation , Everolimus , Interleukin-17 , Xenograft Model Antitumor Assays , Everolimus/pharmacology , Humans , Animals , Mice , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Interleukin-17/metabolism , Interleukin-17/antagonists & inhibitors , Cell Proliferation/drug effects , Cell Line, Tumor , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Sarcoma/drug therapy , Sarcoma/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Synergism
3.
Cancer Chemother Pharmacol ; 93(4): 381-391, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38148335

ABSTRACT

BACKGROUND: Burkitt's lymphoma, one of the most common subtypes of pediatric malignant lymphoma, is notorious for its swift onset, aggressive proliferation, pronounced invasiveness, and marked malignancy. The therapeutic landscape for Burkitt's lymphoma currently falls short of providing universally effective and tolerable solutions. Andrographolide, a primary active component of Andrographis paniculata, is renowned for its properties of heat-clearing, detoxification, inflammation reduction, and pain relief. It is predominantly used in treating bacterial and viral infections of the upper respiratory tract, as well as dysentery. Various reports highlight the antitumor effects of andrographolide. Yet, its specific impact and the underlying mechanism of action on Burkitt's lymphoma remain an uncharted area of research. METHOD: We employed network pharmacology to pinpoint the targets of andrographolide's action on Burkitt's lymphoma and the associated pathways. We then evaluated the impact of andrographolide on Burkitt's lymphoma using both in vitro and in vivo patient-derived xenograft (PDX) models. Concurrently, we confirmed the molecular targets of andrographolide in Burkitt's lymphoma through immunofluorescence assays. RESULT: Utilizing network pharmacology, we identified 15 relevant targets, 60 interrelationships between these targets, and numerous associated signaling pathways for andrographolide's action on Burkitt's lymphoma. In vitro efficacy tests using High-throughput Drug Sensitivity Testing and in vivo PDX model evaluations revealed that andrographolide effectively curtailed the growth of Burkitt's lymphoma. Moreover, we observed a increased in the expression of JUN (c-Jun) and CASP3 (Caspase 3) proteins in Burkitt's lymphoma cells treated with andrographolide. CONCLUSION: Andrographolide inhibits the growth of Burkitt's lymphoma by inhibiting JUN and CASP3 proteins.


Subject(s)
Burkitt Lymphoma , Diterpenes , Humans , Child , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Caspase 3
4.
J Environ Sci (China) ; 139: 460-472, 2024 May.
Article in English | MEDLINE | ID: mdl-38105069

ABSTRACT

As an increasingly used alternative to perfluorooctanoic acid (PFOA), hexafluoropropylene oxide trimer acid (HFPO-TA) has been widely detected in global water environments. However, little is known regarding its toxic effects on cardiovascular development. Here, zebrafish embryos were treated with egg water containing 0, 60, 120, or 240 mg/L HFPO-TA. Results showed that HFPO-TA treatment led to a significant reduction in both larval survival percentage and heart rate. Furthermore, HFPO-TA exposure caused severe pericardial edema and elongation of the sinus venous to bulbus arteriosus distance (SV-BA) in Tg (myl7: GFP) transgenic larvae, disrupting the expression of genes involved in heart development and thus causing abnormal heart looping. Obvious sprouting angiogenesis was observed in the 120 and 240 mg/L exposed Tg (fli: GFP) transgenic larvae. HFPO-TA treatment also impacted the mRNA levels of genes involved in the vascular endothelial growth factor (VEGF) pathway and embryonic vascular development. HFPO-TA exposure significantly decreased erythrocyte number in Tg (gata1: DsRed) transgenic embryos and influenced gene expression associated with the heme metabolism pathway. HFPO-TA also induced oxidative stress and altered the transcriptional levels of genes related to cell cycle and apoptosis, inhibiting cell proliferation while promoting apoptosis. Therefore, HFPO-TA exposure may induce abnormal development of the cardiovascular and hematopoietic systems in zebrafish embryos, suggesting it may not be a suitable or safe alternative for PFOA.


Subject(s)
Fluorocarbons , Zebrafish , Animals , Vascular Endothelial Growth Factor A/genetics , Fluorocarbons/toxicity , Water
5.
J Hazard Mater ; 459: 132175, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37517235

ABSTRACT

The increasing use of cosmetics has raised widespread concerns regarding their ingredients. Cysteamine hydrochloride (CSH) is a newly identified allergenic component in cosmetics, and therefore its potential toxicity needs further elucidation. Here, we investigated the in vivo toxicity of CSH during ocular development utilizing a zebrafish model. CSH exposure was linked to smaller eyes, increased vasculature of the fundus and decreased vessel diameter in zebrafish larvae. Moreover, CSH exposure accelerated the process of vascular sprouting and enhanced the proliferation of ocular vascular endothelial cells. Diminished behavior in response to visual stimuli and ocular structural damage in zebrafish larvae after CSH treatment were confirmed by analysis of the photo-visual motor response and pathological examination, respectively. Through transcriptional assays, transgenic fluorescence photography and molecular docking analysis, we determined that CSH inhibited Notch receptor transcription, leading to an aberrant proliferation of ocular vascular endothelial cells mediated by Vegf signaling activation. This process disrupted ocular homeostasis, and induced an inflammatory response with neutrophil accumulation, in addition to the generation of high levels of reactive oxygen species, which in turn promoted the occurrence of apoptotic cells in the eye and ultimately impaired ocular structure and visual function during zebrafish development.


Subject(s)
Cysteamine , Zebrafish , Animals , Cysteamine/toxicity , Endothelial Cells , Molecular Docking Simulation , Inflammation/chemically induced
6.
Transl Cancer Res ; 12(4): 1006-1020, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37180669

ABSTRACT

Background: Some studies have shown that daucosterol has potential anti-tumor activity, but its therapeutic effect on multiple myeloma (MM) has not been reported. This study aimed to evaluate the therapeutic effect daucosterol against MM and explore its possible mechanism through network pharmacology. Methods: We collected daucosterol and approved drugs for MM, and their potential target profiles were obtained. We used 2 major methods to collect the gene sets related to the physiological process of MM. Based on the protein-protein interaction (PPI) network in the STRING database, the correlation between the therapeutic targets of daucosterol and MM-related genes was calculated by using the random walk with restart (RWR) algorithm to systematically evaluate the therapeutic potential of daucosterol for MM. On this basis, through intersection analysis, the potential targets of daucosterol in treating MM were identified, and the signaling pathways were mined. Furthermore, the key targets were identified. Finally, the regulatory relationship between the predicted daucosterol and potential targets was verified by molecular docking method, and the interaction mode between daucosterol and key targets was analyzed. Results: A total of 13 approved drugs reported to treat MM were retrieved from the DrugBank database. A total of 35 potential targets of daucosterol were obtained, including 8 known targets and 27 newly predicted targets. In the PPI network, the target of daucosterol was significantly correlated with MM-related genes, indicating that it has therapeutic potential for MM. A total of 18 therapeutic targets for MM were obtained, which were significantly enriched in the FoxO signaling pathway, prostate cancer, the PI3K-Akt signaling pathway, insulin resistance, the AMPK signaling pathway, and pathways related to the regulation of TP53. The core targets were HSP90AA1, MDM2, GSK3B, AKT3, PRKAA1, and PRKAB1. Molecular docking suggested that daucosterol had potential direct regulatory effects on 13 of the 18 predicted targets. Conclusions: This study highlights the use of daucosterol as a promising therapeutic drug for MM treatment. These data provide new insights into the potential mechanism of daucosterol in the treatment of MM, which may provide references for subsequent research and even the clinical treatment.

7.
Infect Agent Cancer ; 18(1): 22, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37081537

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) does not respond well to current treatment options like sorafenib, and there is an urgent need for developing therapeutical strategies for HBV + HCC. Brassicasterol has previously shown anti-cancer and anti-viral activities, however, its value against HBV + HCC remains to be explored. METHODS: The inhibitory effect of brassicasterol and sorafenib was evaluated on HBV + HCC cell lines and xenograft mouse model. The cytotoxicity of brassicasterol on normal liver cells were measured by LDH assay. AKT agonist was used to identify the targeted signaling pathway by brassicasterol. RESULTS: Brassicasterol induced HBV + HCC cell death in a both dose-dependent and time-dependent manner, and such inhibition was more potent than sorafenib. Brassicasterol did not show apparent cytotoxicity to normal liver cells. Xenograft mouse model further confirmed the inhibitory effect of brassicasterol on the growth of HBV + HCC. Furthermore, signaling pathway analysis showed that brassicasterol-treated HBV + HCC cells had decreased level of phosphor-AKT expression while the addition of AKT agonist could counteract the inhibitory effect of brassicasterol on HCC, indicating that brassicasterol suppressed AKT pathway to exhibit anti-cancer activity in HBV + HCC cells. In addition, brassicasterol showed similar levels of inhibition on HBV- and HBV + HCC cells. CONCLUSION: Brassicasterol possesses anti-cancer activity against HCC through the downregulation of AKT pathway and such activity is independent of HBV infection.

8.
Front Surg ; 9: 987075, 2022.
Article in English | MEDLINE | ID: mdl-36157427

ABSTRACT

Objective: The prognostic effect of delayed treatment on stage IA1 non-small cell lung cancer (NSCLC) patients is still unclear. This study aimed to explore the association between the waiting time before treatment and the prognosis in stage IA1 NSCLC patients. Methods: Eligible patients diagnosed with pathological stage IA1 NSCLC were included in this study. The clinical endpoints were overall survival (OS) and cancer-specific survival (CSS). The Kaplan-Meier method, the Log-rank test, univariable, and multivariable Cox regression analyses were used in this study. Propensity score matching was used to reduce the bias of data distribution. Results: There were eligible 957 patients in the study. The length of waiting time before treatment stratified the survival in patients [<3 months vs. ≥3-months, unadjusted hazard ratio (HR) = 0.481, P = 0.007; <2 months vs. ≥2-months, unadjusted HR = 0.564, P = 0.006; <1 month vs. ≥1-month, unadjusted HR = 0.537, P = 0.001]. The 5-year CSS rates were 95.0% and 77.0% in patients of waiting time within 3 months and over 3 months, respectively. After adjusting for other confounders, the waiting time was identified as an independent prognostic factor. Conclusions: A long waiting time before treatment may decrease the survival of stage IA1 NSCLC patients. We propose that the waiting time for those patients preferably is less than one month and should not exceed two months.

9.
Ecotoxicol Environ Saf ; 241: 113752, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35709675

ABSTRACT

Trifloxystrobin-tebuconazole (TFS-TBZ) is a novel, broad-spectrum fungicide that has been frequently detected in both the environment and agricultural products. However, its adverse effects on aquatic organisms remain unknown. In this study, the adverse effects of ecologically relevant TFS-TBZ concentrations (i.e., 75.0, 112.5, and 150.0 µg/L) on the heart and development of zebrafish were investigated. TFS-TBZ was found to substantially hinder development, inhibit growth, and cause significant abnormity at higher concentrations. Moreover, TFS-TBZ caused severe pericardial edema, heart loop failure, cardiac linearization, and ultra-slow heartbeat, implying that TFS-TBZ might induce congenital heart disease. TFS-TBZ inhibited Notch signaling and increased the intracellular generation of reactive oxygen species, resulting in decreased myocardial cell proliferation and increased apoptosis. The use of sodium valproate and Gadofullerene illustrated the relevance of the Notch signaling system and oxidative stress. Finally, TFS-TBZ exposure conveys severe developmental toxicity to the zebrafish heart. The underlying mechanism is regulation notch mediated-oxidative stress generation, implying that TFS-TBZ may be potentially hazardous to aquatic organisms in the environment.


Subject(s)
Oxidative Stress , Zebrafish , Acetates , Animals , Embryo, Nonmammalian , Imines , Strobilurins/toxicity , Triazoles
10.
Chemosphere ; 305: 135453, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35752317

ABSTRACT

Zeolite imidazolate framework-8 (ZIF-8) is a nanomaterial of metal-organic frameworks (MOFs), which have various applications in drug delivery and water pollution remediation. However, little is known about its developmental neurotoxicity in aquatic organisms, especially on the low-level exposure. In the present study, we investigated the toxic effects of ZIF-8 NPs on the neuron development, behavioral traits, oxidative stress and gene expression in zebrafish embryos. Firstly, our results showed that ZIF-8 induced significantly embryonic malformations and abnormal development of nervous system in zebrafish embryos with a concentration-dependent manner. Meanwhile, the locomotor behavior was obviously inhibited while the anxiety behavior was greatly increased after ZIF-8 exposure. Secondly, the levels of ROS and antioxidant enzyme activities (CAT, SOD and MDA) together with AChE and ATPase were substantially increased in the ZIF-8 exposed groups. At the molecular level, ZIF-8 NPs could down-regulate the expression profiles of neural development-related genes (gap43, synapsin 2a and neurogenin 1) and PD-like related genes (dj-1, dynactin and parkin), but up-regulate the expression levels of neuro-inflammatory genes (nox-1, glip1a and glip1b) in larval zebrafish. In addition, we further explored the molecular mechanism of neurotoxicity induced by ZIF-8 with pharmacological experiments. The results showed that specific inhibition of ROS-mediated oxidative stress by the astaxanthin could reverse the expression patterns of ATPase, AChE and neurodevelopmental genes. Moreover, astaxanthin can partially rescue the ZIF-8-modulated locomotor behavior. Taken together, our results demonstrated that ZIF-8 had the potential to cause neurotoxicity in zebrafish embryos. These informations presented in this study will help to elucidate the molecular mechanisms of ZIF-8 nanoparticles exposure in zebrafish, which providing a scientific evaluation of its safety to aquatic ecosystems.


Subject(s)
Nanoparticles , Water Pollutants, Chemical , Zeolites , Adenosine Triphosphatases/metabolism , Animals , Antioxidants/metabolism , Ecosystem , Embryo, Nonmammalian , Oxidative Stress , Reactive Oxygen Species/metabolism , Water Pollutants, Chemical/metabolism , Zebrafish/metabolism , Zeolites/toxicity
11.
Infect Agent Cancer ; 16(1): 20, 2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33757557

ABSTRACT

BACKGROUND: HBV promotes cell survival by upregulating the expression of the cellular inhibitor of apoptosis protein 2 (cIAP2), however whether it is involved in HBV-induced sorafenib resistance in liver cancer remains unclear. METHODS: cIAP2 overexpression and knockdown was adopted to assess the involvement of cIAP2 in HBV-induced sorafenib resistance. Anti-HBV drug lamivudine and Akt inhibitor were used to investigate the impact of HBV replication on cIAP2 expression and sorafenib resistance. Xenotransplantation mouse model was used to confirm the data on cell lines in vitro. RESULTS: Liver cancer cell line HepG2.215 showed increased cIAP2 expression and enhanced resistance to sorafenib. Upon sorafenib treatment, overexpression of cIAP2 in HepG2 lead to decreased cleaved caspase 3 level and increased cell viability, while knockdown of cIAP2 in HepG2.215 resulted in increased level of cleaved caspase 3 and decreased cell viability, suggesting the involvement of cIAP2 in HBV-induced sorafenib resistance. Furthermore, anti-HBV treatment reduced cIAP2 expression and partially restored sorafenib sensitivity in HepG2.215 cells. Xenotransplantation mouse model further confirmed that co-treatment with lamivudine and sorafenib could reduce sorafenib-resistant HepG2.215 tumor cell growth. CONCLUSION: cIAP2 is involved in HBV-induced sorafenib resistance in liver cancer and anti-HBV treatments reduce cIAP2 expression and partially restore sorafenib sensibility.

12.
Cell Immunol ; 363: 104342, 2021 05.
Article in English | MEDLINE | ID: mdl-33765541

ABSTRACT

BACKGROUND: Chimeric antigen receptor T cells (CAR-T) against B-cell maturation antigen (BCMA) has been used to treat multiple myeloma (MM). CAR-T cells co-expressing a truncated human EGFR (tEGFR) has been proposed for in vivo cell ablation. METHODS: We designed and tested a novel anti-BCMA CAR. We transduced T cells with retroviral vectors encoding CAR and tEGFR. The anti-BCMA-CAR-transduced T cells were evaluated for the functions including cytokine production, proliferation, cytotoxicity, and in vivo tumor eradication of BCMA. Cetuximab was used for in vivo cell ablation. RESULTS: The CAR-T cells could specifically recognize BCMA, and anti-BCMA CAR-T cells could exhibit interferon-γ and cytotoxicity specifically produced by BCMA and eradicate tumor in vivo. Cetuximab could mediate antibody-dependent cellular cytotoxicity and in vivo elimination. CONCLUSIONS: We confirm that BCMA is a suitable target for CAR- T cells and tEGFR is a effective tool for cellular ablation.


Subject(s)
B-Cell Maturation Antigen/immunology , ErbB Receptors/genetics , Immunotherapy, Adoptive/methods , Adult , Animals , B-Cell Maturation Antigen/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Female , HEK293 Cells , Heterografts , Humans , K562 Cells , Male , Mice , Mice, Inbred NOD , Middle Aged , Neoplasms/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Transgenes , Xenograft Model Antitumor Assays
13.
BMC Cancer ; 21(1): 125, 2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33549034

ABSTRACT

BACKGROUND: Side population (SP) cells, which have similar features to those of cancer stem cells, show resistance to dexamethasone (Dex) treatment. Thus, new drugs that can be used in combination with Dex to reduce the population of SP cells in multiple myeloma (MM) are required. Diallyl thiosulfinate (DATS, allicin), a natural organosulfur compound derived from garlic, has been shown to inhibit the proliferation of SP cells in MM cell lines. Therefore, we investigated the effect of a combination of DATS and Dex (DAT + Dex) on MM SP cells. METHODS: SP cells were sorted from MM RPMI-8226 and NCI-H929 cell lines using Hoechst 33342-labeled fluorescence-activated cell sorting. The growth of SP cells was evaluated using the cell counting kit-8 assay. Cell cycle and apoptosis assays were conducted using a BD Calibur flow cytometer. miRNA expression was measured using quantitative reverse transcription-polymerase chain reaction. Phosphoinositide 3-kinase (PI3K), phosphorylated AKT (p-AKT), AKT, p-mechanistic target of rapamycin (mTOR), and mTOR levels were measured using western blot analysis. RESULTS: Our results showed that the combination of DATS+Dex inhibited sphere formation, colony formation, and proliferation of MM SP cells by inducing apoptosis and cell cycle arrest in the G1/S phase. In addition, the combination of DATS+Dex promoted miR-127-3p expression and inhibited PI3K, p-AKT, and p-mTOR expression in SP cells. Knockdown of miR-127-3p expression weakened the effect of DATS+Dex on cell proliferation, colony formation, apoptosis, and cell cycle of MM SP cells. Additionally, knockdown of miR-127-3p activated the PI3K/AKT/mTOR signaling pathway in MM SP cells cotreated with DATS+Dex. CONCLUSION: We demonstrated that cotreatment with DATS+Dex reduced cell proliferation, promoted apoptosis, and caused cell cycle arrest of MM SP cells by promoting miR-127-3p expression and deactivating the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Dexamethasone/pharmacology , Disulfides/pharmacology , MicroRNAs/drug effects , Multiple Myeloma/drug therapy , Phosphatidylinositol 3-Kinase/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Side-Population Cells/drug effects , Sulfinic Acids/pharmacology , Aldehyde Dehydrogenase 1 Family/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Databases, Genetic , Drug Resistance, Neoplasm , Drug Synergism , G1 Phase Cell Cycle Checkpoints , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplastic Stem Cells/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , S Phase Cell Cycle Checkpoints , Sex-Determining Region Y Protein/metabolism , Side-Population Cells/metabolism , Side-Population Cells/pathology , Signal Transduction/drug effects , Spheroids, Cellular/pathology , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
14.
Medicine (Baltimore) ; 99(27): e21025, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32629725

ABSTRACT

BACKGROUND: Given the huge burden of atrial fibrillation (AF) and AF-related stroke in Asia, stroke prevention represents an urgent issue in this region. We herein performed a network meta-analysis to examine the role of non-vitamin K antagonist oral anticoagulants (NOACs) in Asian patients with AF. METHODS: A systematic search of the publications was conducted in PubMed and Embase databases for eligible studies until July 2019. The odds ratios (ORs) and 95% confidence intervals (CIs) were regarded as the effect estimates. The surface under the cumulative ranking area (SUCRA) for the ranking probabilities was calculated. RESULTS: A total of 17 studies were included. For comparisons of NOACs vs warfarin, dabigatran (OR = 0.77, 95% CI 0.68-0.86), rivaroxaban (OR = 0.72, 95% CI 0.65-0.81), apixaban (OR = 0.56, 95% CI 0.49-0.65), but not edoxaban reduced the risk of stroke or systemic embolism, wheres dabigatran (OR = 0.56, 95% CI 0.41-0.76), rivaroxaban (OR = 0.66, 95% CI 0.50-0.86), apixaban (OR = 0.49, 95% CI 0.36-0.66), and edoxaban (OR = 0.34, 95% CI 0.24-0.49) decreased the risk of major bleeding. In reducing the risk of stroke or systemic embolism, apixaban and rivaroxaban ranked the best and second best (SUCRA 0.2% and 31.4%, respectively), followed by dabigatran (50.2%), edoxaban (75.2%), and warfarin (93.0%). In reducing the risk of major bleeding, edoxaban, and apixaban ranked the best and second best (1.5% and 30.8%, respectively), followed by dabigatran (48.4%), rivaroxaban (69.2%), and warfarin (100%). CONCLUSION: NOACs were at least as effective as warfarin, but more safer in Asians with AF. Apixaban was superior to other NOACs for reducing stroke or systemic embolism, while edoxaban showed a better safety profile than other NOACs.


Subject(s)
Anticoagulants/therapeutic use , Atrial Fibrillation/drug therapy , Stroke/prevention & control , Warfarin/therapeutic use , Administration, Oral , Aged , Antithrombins/therapeutic use , Asia/epidemiology , Asian People/ethnology , Cost of Illness , Dabigatran/therapeutic use , Embolism/prevention & control , Factor Xa Inhibitors/therapeutic use , Female , Hemorrhage/prevention & control , Humans , Male , Network Meta-Analysis , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Pyridones/therapeutic use , Rivaroxaban/therapeutic use , Safety , Stroke/epidemiology , Thiazoles/therapeutic use
15.
Medicine (Baltimore) ; 99(25): e20782, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32569222

ABSTRACT

BACKGROUND: The HAS-BLED, HEMORR2HAGES, ATRIA, and ORBIT scores are used to predict bleeding risk in anticoagulated patients with atrial fibrillation (AF). Recently, these scores have been validated in various studies. Therefore, we aimed to compare the occurrence of major bleeding across different risk categories between HAS-BLED and any of HEMORR2HAGES, ATRIA, or ORBIT scores. METHODS: A systemic literature search of PubMed and Embase databases was conducted to screen the relevant studies. We calculated and pooled the odds ratios (ORs) and 95% confidence intervals (CIs) for a comparative analysis of the occurrence of major bleeding. RESULTS: Nine studies fulfilled the inclusion criteria in this meta-analysis. Compared with HEMORR2HAGES, there were 87% and 39% reduced rates of major bleeding in the HAS-BLED "low-risk" and "moderate-risk" groups, respectively. Compared with ATRIA, there was an 89% decreased rate of major bleeding in the HAS-BLED "low-risk" group. Compared with ORBIT, there were 84% and 44% reduced rates of major bleeding in the HAS-BLED "low-risk" and "moderate-risk" groups, respectively. Patients with HAS-BLED scores ≥3 showed an approximately 3-fold greater risk of major bleeding compared with patients with scores <3 (OR=3.00, CI: 1.21-7.43). CONCLUSIONS: Compared with any of HEMORR2HAGES, ATRIA, or ORBIT scores, the HAS-BLED score distributed more major bleeding events into the "low" or "moderate" risk categories.


Subject(s)
Anticoagulants/adverse effects , Atrial Fibrillation/complications , Hemorrhage/chemically induced , Risk Assessment/methods , Anticoagulants/therapeutic use , Atrial Fibrillation/drug therapy , Humans
16.
Medicine (Baltimore) ; 99(18): e19542, 2020 May.
Article in English | MEDLINE | ID: mdl-32358343

ABSTRACT

BACKGROUND: Data of non-vitamin K antagonist oral anticoagulants (NOACs) in current management of atrial fibrillation (AF) are predominantly derived from North American and European regions. However, the effects of NOACs for stroke prevention in Latin America remain unclear. Therefore, we aimed to compare the efficacy and safety of NOACs with warfarin in Latin American patients with AF. METHODS: The PubMed and Embase databases were systematically searched until July 12, 2019 for applicable randomized clinical trials. The risk ratios (RRs) and 95% confidence intervals (CIs) were pooled using a random-effects model. RESULTS: Four trials involving 8943 Latin American patients were included in this meta-analysis. In anticoagulated patients with AF, Latin American patients had higher rates of stroke or systemic embolism and all-cause death compared with non-Latin American subjects. Compared with warfarin use, the use of NOACs was significantly associated with reduced risks of stroke or systemic embolism, major bleeding, intracranial bleeding, and any bleeding in Latin American patients. There were no significant differences in the risks of ischemic stroke, all-cause death, and gastrointestinal bleeding between Latin and non-Latin American groups. All the interactions between Latin and non-Latin American groups about efficacy and safety outcomes of NOACs compared with warfarin were non-significant (all Pinteraction > .05). CONCLUSIONS: Our meta-analysis suggested that the use of NOACs was at least non-inferior to warfarin use for stroke prevention in Latin American patients with AF.


Subject(s)
Anticoagulants/therapeutic use , Atrial Fibrillation/drug therapy , Hispanic or Latino/statistics & numerical data , Stroke/prevention & control , Warfarin/therapeutic use , Administration, Oral , Adult , Atrial Fibrillation/complications , Atrial Fibrillation/ethnology , Embolism/ethnology , Embolism/prevention & control , Female , Hemorrhage/chemically induced , Hemorrhage/ethnology , Humans , Latin America , Male , Middle Aged , Randomized Controlled Trials as Topic , Stroke/ethnology , Treatment Outcome
17.
Oncol Lett ; 19(3): 2043-2052, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32194701

ABSTRACT

Activation of antiapoptotic genes has been indicated as one of the factors that contributes to hepatitis B virus (HBV) infection-induced liver cancer. The cellular inhibitor of apoptosis protein 2 (cIAP2), a member of the IAP family, is upregulated in various types of cancer and serves as a potential treatment target. However, to the best of our knowledge, the importance of cIAP2 in HBV-induced liver cancer has not been investigated. In the present study, cIAP2 expression in liver cells in response to HBV infection and the underlying mechanism involved was investigated. Western blot analysis of clinical liver samples showed that higher cIAP2 expression was detected in HBV-positive non-cancerous tissue compared with that in HBV-negative non-cancerous tissue, and the expression was further increased in HBV-positive liver cancer tissue. Reverse transcription-quantitative PCR and western blot experiments performed on two liver cell lines also confirmed that cIAP2 expression was increased upon HBV infection at both the mRNA and protein levels. Promoter analysis revealed that HBV could activate cIAP2 promoter in an infection dose-dependent manner, and this activation involved a NF-κB-binding site in the cIAP2 promoter. Further analysis demonstrated that HBV enhanced NF-κB phosphorylation and nuclear translocation via the PI3K/AKT signaling pathway, leading to the binding and activation of cIAP2 promoter. The present data demonstrates that HBV-infection induces cIAP2 expression in the liver by activation of the PI3K/AKT/NF-κB signaling pathway through promoting the binding of NF-κB to cIAP2 promoter, which may lead to carcinogenesis. The findings from the present study provide more information for understanding HBV-induced liver cancer and also offer a potential target for treatment or diagnosis of this disease.

18.
Heart Fail Rev ; 25(6): 973-983, 2020 11.
Article in English | MEDLINE | ID: mdl-31713086

ABSTRACT

We performed this meta-analysis to compare the efficacy and safety of reduced-dose non-vitamin K antagonist oral anticoagulants (NOACs) versus warfarin in patients with atrial fibrillation (AF). The PubMed and Embase databases were systematically searched until July 2019 for eligible studies that comparing the effect between any reduced-dose NOAC and warfarin in patients with AF. Risk ratios (RRs) and 95% confidence intervals (CIs) were pooled by using a random-effects model. A total of 14 observational cohorts were included. Compared with warfarin use, the use of reduced-dose NOACs was associated with decreased risks of stroke or systemic embolism (RR, 0.83; 95% CI 0.74-0.93), ischemic stroke (RR, 0.87; 95% CI 0.77-0.98), major bleeding (RR, 0.71; 95% CI 0.60-0.84), intracranial hemorrhage (RR, 0.51; 95% CI 0.44-0.60), and gastrointestinal bleeding (RR, 0.72; 95% CI 0.54-0.94), but not all cause death (RR, 0.84; 95% CI 0.67-1.06). In the subgroup analyses, all NOAC users had lower or similar rates of thromboembolic and bleeding events; and the reductions in stroke or systemic embolism, all-cause death, major bleeding, and gastrointestinal bleeding were more prominent in Asians than non-Asians. In conclusion, current published data suggest that the use of reduced-dose NOACs is non-inferior to warfarin in patients with AF (in particular Asians).


Subject(s)
Atrial Fibrillation/drug therapy , Embolism/prevention & control , Warfarin/administration & dosage , Administration, Oral , Anticoagulants/administration & dosage , Atrial Fibrillation/complications , Dose-Response Relationship, Drug , Embolism/etiology , Humans
19.
Biomed Pharmacother ; 117: 109099, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31200255

ABSTRACT

Inflammation and coagulation are two important processes implicated in venous thromboembolism (VTE). 15-epi-lipoxin A4 (15-epi-LXA4) is the epimer of LXA4, a small lipid molecule, is known to play a key role in the resolution of inflammation. This study aimed to demonstrate whether 15-epi-LXA4 could suppress the inflammatory factor tumor necrosis factor-alpha (TNF-α)-induced upregulation of tissue factor (TF), an important regulator of the blood coagulation cascade, and evaluated the possible underlying mechanisms. We found that 15-epi-LXA4 not only reduced the up-regulation of mRNA and antigens, but also lowered the activity of TF (elevated by TNF-α) in primary culture of human umbilical vein endothelial cells (pc-HUVECs). In addition, 15-epi-LXA4 suppressed the activation of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway, induced by TNF-α, in pc-HUVECs. 15-epi-LXA4 also inhibited the binding of NF-κB on the TF promoter, which is otherwise enhanced by TNF-α. The role of 15-epi-LXA4 in regulating TNF-α-induced effects was enhanced by the PI3K inhibitor and prevented by the PI3K activator. In conclusion, 15-epi-LXA4 lowered the TNF-α-induced high TF expression and activity by suppressing PI3K/AKT signaling activation, thereby reducing the binding capacity of NF-κB on the TF promoter in pc-HUVECs.


Subject(s)
Lipoxins/pharmacology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/drug therapy , Inflammation/metabolism
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(9): 1234-1239, 2017 Sep.
Article in Chinese | MEDLINE | ID: mdl-29089083

ABSTRACT

Objective To observe the effect of metadherin/astrocyte elevated gene-1 (MTDH/AEG-1) silencing on the proliferation and metastasis of SGC7901 human gastric cancer cells. Methods The eukaryotic shRNA expression vectors targeting MTDH gene were constructed and transfected into SGC7901 cells. Effects of gene silencing were confirmed by Western blotting. MTT assay was used to detect the effect of MTDH on the proliferation of SGC7901 cells, and TranswellTM assay was employed to detect the effect of MTDH on the migration of SGC7901 cells. Results MTDH-shRNA was constructed and transfected into SGC-7901 cells successfully. After transfection, the level of MTDH protein expression was reduced significantly. Knockdown of MTDH in SGC7901 cells, the abilities of cell proliferation and migration decreased obviously. Conclusion Knockdown of MTDH gene effectively inhibits the proliferation and metastasis of SGC7901 cells.


Subject(s)
Cell Adhesion Molecules/physiology , Stomach Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Membrane Proteins , Neoplasm Invasiveness , RNA, Small Interfering/genetics , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...