Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Infect Drug Resist ; 17: 1099-1105, 2024.
Article En | MEDLINE | ID: mdl-38590553

Purpose: To explore the clinical characteristics, diagnosis, and treatment of family outbreak of psittacosis and to improve the success rate of treatment. Patients and Methods: The clinical characteristics, diagnosis, treatment, and outcome of family outbreak of psittacosis, which consists three patients, diagnosed by clinical analysis and metagenomic next-generation sequencing (mNGS) in our hospital were analyzed retrospectively. Results: We report on three instances of clustered atypical pneumonia, which were caused by Chlamydia psittaci during the COVID-19 pandemic. All patients exhibited symptoms of fever and cough, while one patient also experienced gastrointestinal symptoms such as nausea, vomiting, and diarrhea. Laboratory tests indicated no significant increase in leukocytes and neutrophils, but a mild increase in C-reactive protein was observed in all three patients. Chest computed tomography (CT) scans revealed a consolidation shadow in a unilateral lung lobe in all three patients. Both patients were treated with empirical moxifloxacin, yielding unsatisfactory outcomes. mNGS was conducted on sputum samples from one adult patient, revealing the presence of Chlamydia psittaci. Additional doxycycline was prescribed immediately, and then the patients' temperatures were stabilized, and the lesion in chest CT was absorbed. The pediatric patient exhibited less severe symptoms compared to the adult patients and exhibited a favorable response to azithromycin administration. Conclusion: This study reports a cluster of a family outbreak of atypical pneumonia caused by C. psittaci in China. The occurrence of a family outbreak during the COVID-19 pandemic may be attributed to familial aggregation resulting from the epidemic. The three cases reported in this study did not experience severe complications, which can be attributed to the prompt medical intervention and swift diagnosis. This finding implies the need to enhance patients' awareness and vigilance towards their health. Additionally, mNGS emerges as a valuable technique for accurately identifying pathogens causing pulmonary infections.

2.
Curr Med Chem ; 31(19): 2821-2837, 2024.
Article En | MEDLINE | ID: mdl-38351696

INTRODUCTION: Senescence of activated hepatic stellate cells (HSC) reduces extracellular matrix expression to reverse liver fibrosis. Ferroptosis is closely related to cellular senescence, but its regulatory mechanisms need to be further investigated. The iron ions weakly bound to ferritin in the cell are called labile iron pool (LIP), and together with ferritin, they maintain cellular iron homeostasis and regulate the cell's sensitivity to ferroptosis. METHODS: We used lipopolysaccharide (LPS) to construct a pathological model group and divided the hepatic stellate cells into a blank group, a model group, and a curcumol 12.5 mg/L group, a curcumol 25 mg/L group, and a curcumol 50 mg/L group. HIF-1α-NCOA4- FTH1 signalling axis, ferroptosis and cellular senescence were detected by various cellular molecular biology experiments. RESULT: We found that curcumol could induce hepatic stellate cell senescence by promoting iron death in hepatic stellate cells. Curcumol induced massive deposition of iron ions in hepatic stellate cells by activating the HIF-1α-NCOA4-FTH1 signalling axis, which further led to iron overload and lipid peroxidation-induced ferroptosis. Interestingly, our knockdown of HIF-1α rescued curcumol-induced LIP and iron deposition in hepatic stellate cells, suggesting that HIF-1α is a key target of curcumol in regulating iron metabolism and ferroptosis. We were able to rescue curcumol-induced hepatic stellate cell senescence when we reduced LIP and iron ion deposition using iron chelators. CONCLUSION: Overall, curcumol induces ferroptosis and cellular senescence by increasing HIF-1α expression and increasing NCOA4 interaction with FTH1, leading to massive deposition of LIP and iron ions, which may be the molecular biological mechanism of its anti-liver fibrosis.


Cellular Senescence , Ferroptosis , Hepatic Stellate Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Liver Cirrhosis , Sesquiterpenes , Signal Transduction , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Ferroptosis/drug effects , Cellular Senescence/drug effects , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Signal Transduction/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Nuclear Receptor Coactivators/metabolism , Lipopolysaccharides/pharmacology , Cells, Cultured , Ferritins , Oxidoreductases
3.
Eur Radiol ; 34(2): 1053-1064, 2024 Feb.
Article En | MEDLINE | ID: mdl-37581663

OBJECTIVES: To explore the performance of low-dose computed tomography (LDCT) with deep learning reconstruction (DLR) for the improvement of image quality and assessment of lung parenchyma. METHODS: Sixty patients underwent chest regular-dose CT (RDCT) followed by LDCT during the same examination. RDCT images were reconstructed with hybrid iterative reconstruction (HIR) and LDCT images were reconstructed with HIR and DLR, both using lung algorithm. Radiation exposure was recorded. Image noise, signal-to-noise ratio, and subjective image quality of normal and abnormal CT features were evaluated and compared using the Kruskal-Wallis test with Bonferroni correction. RESULTS: The effective radiation dose of LDCT was significantly lower than that of RDCT (0.29 ± 0.03 vs 2.05 ± 0.65 mSv, p < 0.001). The mean image noise ± standard deviation was 33.9 ± 4.7, 39.6 ± 4.3, and 31.1 ± 3.2 HU in RDCT, LDCT HIR-Strong, and LDCT DLR-Strong, respectively (p < 0.001). The overall image quality of LDCT DLR-Strong was significantly better than that of LDCT HIR-Strong (p < 0.001) and comparable to that of RDCT (p > 0.05). LDCT DLR-Strong was comparable to RDCT in evaluating solid nodules, increased attenuation, linear opacity, and airway lesions (all p > 0.05). The visualization of subsolid nodules and decreased attenuation was better with DLR than with HIR in LDCT but inferior to RDCT (all p < 0.05). CONCLUSION: LDCT DLR can effectively reduce image noise and improve image quality. LDCT DLR provides good performance for evaluating pulmonary lesions, except for subsolid nodules and decreased lung attenuation, compared to RDCT-HIR. CLINICAL RELEVANCE STATEMENT: The study prospectively evaluated the contribution of DLR applied to chest low-dose CT for image quality improvement and lung parenchyma assessment. DLR can be used to reduce radiation dose and keep image quality for several indications. KEY POINTS: • DLR enables LDCT maintaining image quality even with very low radiation doses. • Chest LDCT with DLR can be used to evaluate lung parenchymal lesions except for subsolid nodules and decreased lung attenuation. • Diagnosis of pulmonary emphysema or subsolid nodules may require higher radiation doses.


Deep Learning , Humans , Quality Improvement , Radiation Dosage , Lung/diagnostic imaging , Tomography, X-Ray Computed/methods , Algorithms , Radiographic Image Interpretation, Computer-Assisted/methods
4.
FASEB J ; 36(12): e22665, 2022 12.
Article En | MEDLINE | ID: mdl-36398583

To explore the effect of curcumol on autophagy and ferroptosis of hepatic stellate cells, and to clarify the molecular mechanism of its anti-hepatic fibrosis. In the present study, we report that curcumol promotes the death of activated HSCs and reduces the deposition of extracellular matrix. Interestingly, curcumol treatment can trigger ferroptosis to eliminate activated HSCs characterized by iron overload, lipid ROS accumulation, glutathione depletion, and lipid peroxidation. Curcumol promotes HSC autophagy, which may be the key mechanism for its induction of ferroptosis. It is worth noting that the upregulation of nuclear receptor coactivator 4 (NCOA4) may play a key molecular mechanism. NCOA4 mediates the release of iron ions and induces the occurrence of ferroptosis. Overall, curcumol promotes autophagy in hepatic stellate cells, mediates the degradation of NCOA4 and FTH1 complexes, releases iron ions, leads to iron overload, and induces ferroptosis, which may be an important mechanism for its anti-hepatic fibrosis effect.


Ferroptosis , Iron Overload , Humans , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Autophagy , Iron Overload/metabolism , Iron/metabolism
5.
Biomed Pharmacother ; 152: 113204, 2022 Aug.
Article En | MEDLINE | ID: mdl-35653891

OBJECTIVE: Liver fibrosis is a reversible pathological process, and its prevention and treatment hold great significance for patients with chronic liver disease. This study combined 16S rRNA analysis of gut microbiota and serum metabolomics to explore the mechanism of curcumol's effect on liver fibrosis in mice. The results clarified the relationship between the gut microbiota and metabolites in the process of liver fibrosis. MATERIALS AND METHODS: In this study, we randomly divided mice into a control group, a model group, and a curcumol treatment group to analyze the pathological changes in the liver tissue as well as the activities of the toll-like receptor 4 (TLR4)/nuclear factory kappa B (NF-κB) signaling pathway and inflammatory factors, such as tumor necrosis factor (TNF), interleukin 6 (IL-6), and IL-8. The gut microbiota were analyzed by 16 S rRNA sequencing, and serum metabolites were examined by liquid chromatography-mass spectrometry (LC-MS) metabolomic analysis. RESULTS: Molecular biological testing found that curcumol could significantly improve the pathological changes of the liver tissue and inhibit the occurrence of liver inflammation. Intestinal flora testing found that curcumol could significantly change the abundances of Veillonellaceae, Prerotella_oulorum, and Alistipes_finegoldii. Metabolomics analysis found that curcumol's antihepatic fibrosis effect may be related to its regulation of arachidonic acid metabolism. Correlation analysis suggested that curcumol regulated the abundances of Bacteroidota and Bacteroides and participated in the metabolism of Prostaglandin B2. CONCLUSIONS: When liver fibrosis occurs, the intestinal flora and metabolic network are altered. The effect of curcumol on liver fibrosis may be related to its regulation of intestinal flora and the resulting interference with metabolic pathways, thereby reducing liver inflammation.


Gastrointestinal Microbiome , Animals , Humans , Inflammation , Liver Cirrhosis/drug therapy , Liver Cirrhosis/prevention & control , Metabolomics/methods , Mice , RNA, Ribosomal, 16S , Sesquiterpenes
6.
Cancer Lett ; 536: 215651, 2022 06 28.
Article En | MEDLINE | ID: mdl-35315340

Copper is involved in various biochemical and physiological processes. The absorbed copper ions are transported to the intracellular destination via copper chaperones, such as ATOX1. Previous studies have demonstrated that neoplastic cells have a high demand for copper; however, its role in cancer cells has not been fully elucidated. Here, we reveal that the high level of copper contributes to drug resistance and repair of damaged DNA in cancer cells at least partially via ATOX1-induced expression of MDC1, a crucial protein involved in double-strand DNA damage repair. Specifically, ATOX1 enters into nuclear to target MDC1 promoter after treatments of various genotoxic agents, thus promoting the transcription of MDC1 in a copper-dependent manner. Therefore, knockout or blockage of ATOX1 conferred sensitivity to Gemcitabine in transplanted tumor mouse models. Together, our findings gain new insight into the role of copper in DNA damage repair and provide a novel strategy for clinical cancer therapy of drug-resistance cancers.


Cation Transport Proteins , Copper , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/pharmacology , Copper Transport Proteins , DNA Damage , Drug Resistance , Humans , Mice , Molecular Chaperones/genetics
7.
Drug Chem Toxicol ; 45(1): 33-43, 2022 Jan.
Article En | MEDLINE | ID: mdl-35100937

1,4-naphthoquinone and its derivatives have attracted widespread attention due to their multiple biological activities, such as induction of cancer cell apoptosis; however, most of these compounds have high cytotoxicity. In this study, in order to reduce their toxicity and increase their potential anti-tumor effects, we synthesized a novel 1,4-naphthoquinone derivative named 2-(naphthalene-2-thio)-5,8-dimethoxy-1,4-naphthoquinone (NTDMNQ), and investigated its apoptotic effects and underlying mechanism. Our results showed that NTDMNQ inhibited the viability of HepG2, Hep3B, and Huh7 human hepatocellular carcinoma (HCC) cells. It also increased the accumulation of cells in the G0/G1 phase of the cell cycle by increasing the expression levels of p-p53, p21 and p27, while decreasing the levels of Cyclin D1, Cyclin E, Cyclin-dependent kinase 2 (CDK2), CDK4, and CDK6. Inhibition of reactive oxygen species (ROS) by the ROS scavenger N-acetyl-L-cysteine (NAC) decreased apoptosis in NTDMNQ-treated cells. Western blot analysis showed that NTDMNQ increased the phosphorylation of p38 and c-Jun N-terminal kinase (JNK), and decreased the phosphorylation of extracellular signal-regulated kinase (ERK), AKT, and signal transducer and activator of transcription-3 (STAT3); these effects were blocked by NAC. Both the JNK inhibitor (SP600125) and p38 inhibitor (SB203580) reversed the phosphorylation of STAT3, and the ERK inhibitor (FR180204) and AKT inhibitor (LY294002) reduced the expression of STAT3. Taken together, these findings suggest that NTDMNQ induces apoptosis via ROS-mediated MAPK, AKT and STAT3 signaling pathways in HepG2 cells, and may be a potent anticancer agent.


Carcinoma, Hepatocellular , Liver Neoplasms , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Naphthalenes , Naphthoquinones , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor , Signal Transduction
8.
Front Neurol ; 12: 632063, 2021.
Article En | MEDLINE | ID: mdl-34552546

Background: Revascularization surgery sometimes can achieve recanalization in patients with internal carotid artery occlusion (ICAO). High-resolution vessel wall magnetic resonance imaging (HRVWI) is a feasible technique to give detailed characteristics of the vessel wall, which may help to identify patients that carry higher success rates and more suitable for revascularization surgery. Objective: To examine the association between HRVWI characteristics of ICAO and the success rate of revascularization surgery in ICAO patients. Methods: We conducted a retrospective analysis of 31 ICAO recanalization patients enrolled from October 2017 to May 2019. The clinical data of patients and lesions were collected and analyzed. Results: A total of 31 ICAO patients were enrolled in this study. No significant differences were found between recanalization success and recanalization failure groups with regard to occlusion length, distal end of the occluded segment, and the treatment applied. The ipsilateral-to-contralateral diameter ratios (I/C ratios) of C1 or C2 and the diameter of C7 were positively related to recanalization success. A two-factor predictive model was constructed, and the I/C ratio of C2 < 0.86 and the diameter of C7 < 1.75mm were separately assigned 1 point. The ICAO patients who scored 0, 1, or 2 points had a risk of 5.6% (1/18), 55.6% (5/9), or 100% (4/4) to fail in the recanalization. Conclusions: The I/C ratios of C1 or C2 and the diameter of C7 are predictive factors of a revascularization surgery success in ICAO patients. A risk stratification model involving C2 and C7 was constructed for future clinical applications.

9.
J Ethnopharmacol ; 280: 114480, 2021 Nov 15.
Article En | MEDLINE | ID: mdl-34358654

ETHNOPHARMACOLOGICAL RELEVANCE: Curcuma is a common Chinese herbal medicine that is used in the clinical treatment of chronic liver disease. Studies have found that curcumol is the main active ingredient of curcuma and has good hepatoprotective and anti-inflammatory effects. However, there are few reports on the molecular mechanism underlying the anti-liver fibrosis effect of curcumol. AIM: To explore the effect of curcumol on liver angiogenesis, and to reveal the mechanism of curcumol against liver fibrosis. MATERIALS AND METHODS: We used liver collagenase perfusion combined with Percoll density gradient sedimentation to separate primary liver sinusoidal endothelial cells, and then applied a leptin-activated cell pathological model. The cells were divided into four treatment groups as follows: blank group, model group, curcumol group, and solafini group. MTT was used to detect the cell proliferation rate in each group, and RT-PCR and western blotting were used to detect the expressions of VEGF, AKT, eNOS, CD31, and vWF. A fluorescent probe was used to detect NO expression, and scanning electron microscopy was used to observe changes in the cell fenestration structure. Angiogenesis assays were used to observe blood vessel formation in each group. RESULTS: The results of the MTT test found that the proliferation rate of each group was higher. The results of the molecular biology tests found that curcumol inhibited the activity of the VEGF/AKT/eNOS pathway, thereby increasing fenestration of sinusoidal endothelial cells and inhibiting liver angiogenesis. These differences were statistically significant compared with the model group. CONCLUSIONS: Curcumol inhibits the activity of the VEGF/AKT/eNOS signaling pathway, regulates the structure of hepatic sinusoidal endothelial cells, and inhibits liver angiogenesis, which together may explain its anti-liver fibrosis mechanism.


Angiogenesis Inhibitors/pharmacology , Liver Cirrhosis/drug therapy , Neovascularization, Pathologic/drug therapy , Sesquiterpenes/pharmacology , Animals , Endothelial Cells/drug effects , Liver Cirrhosis/pathology , Male , Neovascularization, Pathologic/pathology , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
10.
Metallomics ; 13(7)2021 07 02.
Article En | MEDLINE | ID: mdl-34114637

Copper is one of the indispensable trace metal elements in organisms, but excess copper means cytotoxicity. Cells protect themselves by storing excess copper in copper-binding proteins. Metallothioneins (MTs) are a group of low-molecular-weight, cysteine-rich proteins, which are well known for sensing and binding the overcharged Zn(Ⅱ), Cd(Ⅱ), and Cu(Ⅰ) in cells. However, there are only few reports on MTs that can specifically respond to intracellular copper ions in mammals in real-time. Here, we screened copper-response MTs in pancreatic cancer cells through data-mining, RNA-seq, and qPCR analysis. We found that MT1E, MT1F, and MT1X mRNA were significantly upregulated after exogenous copper ion induction. By constructing the stable cell lines with MT1E, MT1F, or MT1X promoter-driven EGFP as reporters, we found that only PMT1F-EGFP could specifically and stably report the intracellular Cu(Ⅰ) changes in multiple cell lines including Panc-1, 8988T, 293T, HepG2, and normal hepatic cells, indicating that PMT1F-EGFP is an ideal in vivo Cu(Ⅰ) reporter. Using the PMT1F-EGFP reporter, we found that MEK inhibitors (U0126) and Astragaloside IV could significantly increase intracellular copper ions. According to these results, PMT1F-EGFP reporter can sense intracellular copper change and can be used to screen copper-target drugs and study copper-related cellular physiology and pathology.


Copper/metabolism , Green Fluorescent Proteins/metabolism , Metallothionein/metabolism , Pancreatic Neoplasms/pathology , Apoptosis , Cell Proliferation , Green Fluorescent Proteins/genetics , Humans , Metallothionein/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Tumor Cells, Cultured
11.
Ann Transl Med ; 9(2): 120, 2021 Jan.
Article En | MEDLINE | ID: mdl-33569422

BACKGROUND: Autosomal dominant polycystic liver disease (ADPLD) is characterized by multiple cysts in the liver without (or only occasional) renal cysts. At least seven genes are associated with high risk for developing ADPLD; however, clear genetic involvement is undetermined in more than 50% of ADPLD patients. METHODS: To identify additional ADPLD-associated genes, we collected 18 unrelated Chinese ADPLD cases, and performed whole exome sequencing on all the participants. After filtering the sequencing data against the human gene mutation database (HGMD) professional edition, we identified new mutations. We then sequenced this gene in family members of the patient. RESULTS: Among the 18 ADPLD cases analyzed by whole exome sequencing, we found 2 cases with a PRKCSH mutation (~11.1%), 2 cases with a PKD2 mutation (~11.1%), 1 case with both PKHD1 and PKD1 mutations (~5.6%), 1 case with GANAB mutation (~5.6%), 1 case with PKHD1 mutation (~5.6%), and 1 case with PKD1 mutations (~5.6%). We identified a new PKHD1 missense mutation in an ADPLD family, in which both patients showed innumerable small hepatic cysts, as reported previously. Additionally, we found that PRKCSH and SEC63 mutation frequencies were lower in the Chinese population compared with those in European and American populations. CONCLUSIONS: We report a family with ADPLD associated with a novel PKHD1 mutation (G1210R). The genetic profile of ADPLD in the Chinese population is different from that in European and American populations, suggesting that further genetic research on genetic mutation of ADPLD in the Chinese population is warranted.

12.
Article En | MEDLINE | ID: mdl-32849902

Two novel compounds, 2-(2-hydroxyethylthio)-5,8-dimethoxy-1,4-naphthoquinone (HEDMNQ) and 2-(6-hydroxyhexylthio)-5,8-dimethoxy-1,4-naphthoquinone (HHDMNQ), were synthesized to investigate the kill effects and mechanism of 1,4-naphthoquinone derivatives in lung cancer cells. The results of the CCK-8 assay showed that HEDMNQ and HHDMNQ had significant cytotoxic effects on A549, NCI-H23, and NCI-H460 NSCLC cells. Flow cytometry and western blot results indicated that HHDMNQ induced A549 cell cycle arrest at the G2/M phase by decreasing the expression levels of cyclin-dependent kinase 1/2 and cyclin B1. Fluorescence microscopy and flow cytometry results indicated that HHDMNQ could induce A549 cell apoptosis, and western blot analysis showed that HHDMNQ induced apoptosis through regulating the mitochondria pathway, as well as the MAPK, STAT3, and NF-κB signalling pathways. Flow cytometry results showed that intracellular reactive oxygen species (ROS) levels were increased after HHDMNQ treatment, and western blot showed that ROS could modulate the intrinsic pathway and MAPK, STAT3, and NF-κB signalling pathways. These effects were blocked by the ROS inhibitor N-acetyl-L-cysteine in A549 cells. Our findings suggest that compared with HEDMNQ, HHDMNQ had the stronger ability to inhibit the cell viability of lung cancer cells and induce apoptosis by regulating the ROS-mediated intrinsic pathway and MAPK/STAT3/NF-κB signalling pathways. Thus, HHDMNQ might be a potential antitumour compound for treating lung cancer.

13.
Article En | MEDLINE | ID: mdl-32595742

OBJECTIVE: To study the effect of curcumol on liver sinusoidal endothelial cells (LSECs) and to analyze the mechanism of antihepatic fibrosis. METHODS: The effects of drug intervention on cell proliferation rates were detected by MTT assay. The expression of NF-κB was detected by RT-PCR and WB. The NF-κB expression and entry into the nucleus were detected by immunofluorescence; scanning electron microscopy was used to observe the changes of LSECs fenestrae. RESULTS: MTT results showed that the interference of cell proliferation in each group was small. RT-PCR showed that the expression of NF-κB in the curcumol intervention group was significantly lower than that in the positive control group (P < 0.05). The WB detection found that, in the curcumol intervention group, the expression of pNF-κB in the NF-κB signaling pathway was significantly lower than that in the positive control group (P < 0.05). Scanning electron microscopy showed that the LSEC fenestrae were significantly improved compared with the positive control group. CONCLUSION: Curcumol may be one of the mechanisms of antihepatic fibrosis by inhibiting the activity of the NF-κB signaling pathway and increasing the fenestrae of LSECs.

14.
Naunyn Schmiedebergs Arch Pharmacol ; 393(10): 1987-1999, 2020 10.
Article En | MEDLINE | ID: mdl-31956937

Liquiritin (LIQ), a major constituent of Glycyrrhiza Radix, exhibits various pharmacological activities. In this study, to explore the potential anti-cancer effects and its underlying molecular mechanisms of LIQ in hepatocellular carcinoma (HCC) cells. LIQ significantly decreased viability and induced apoptosis in HepG2 cells by decreasing mitochondrial membrane potential and regulating Bcl-2 family proteins, cytochrome c, cle-caspase-3, and cle-PARP. The cell cycle analysis and western blot analysis revealed that LIQ induced G2/M phase arrest through increased expression of p21 and decreased levels of p27, cyclin B, and CDK1/2. The flow cytometry and western blot analysis also suggested that LIQ promoted the accumulation of ROS in HepG2 cells and up-regulated the phosphorylation expression levels of p38 kinase, c-Jun N-terminal kinase (JNK), and inhibitor of NF-κB (IκB-α); the phosphorylation levels of extracellular signal-regulated kinase (ERK), protein kinase B (AKT), signal transducer activator of transcription 3 (STAT3), and nuclear factor kappa B (NF-κB) were down-regulated. However, these effects were reversed by N-acetyl-L-cysteine (NAC), MAPK, and AKT inhibitors. The findings demonstrated that LIQ induced cell cycle arrest and apoptosis via the ROS-mediated MAPK/AKT/NF-κB signaling pathway in HepG2 cells, and the LIQ may serve as a potential therapeutic agent for the treatment of human HCC.


Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Flavanones/pharmacology , Glucosides/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Flavanones/therapeutic use , Glucosides/therapeutic use , Glycyrrhiza , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , MAP Kinase Signaling System/physiology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism
15.
Artif Cells Nanomed Biotechnol ; 48(1): 84-95, 2020 Dec.
Article En | MEDLINE | ID: mdl-31852250

Cytisine is a natural product isolated from plants and is a member of the quinolizidine alkaloid family. This study aims to investigate the effect of cytisine in human lung cancer. Cell viability was determined using the CCK-8 assay, and the results showed that cytisine inhibited the growth of lung cancer cell lines. The apoptotic effects were evaluated using flow cytometry, and the results showed that cytisine induced mitochondrial-dependent apoptosis through loss of the mitochondrial membrane potential; increased expression of BAD, cleaved caspase-3, and cleaved-PARP; and decreased expression levels of Bcl-2, pro-caspase-3, and pro-PARP. In addition, cytisine caused G2/M phase cell cycle arrest that was associated with inhibiting the AKT signalling pathway. During apoptosis, cytisine increased the phosphorylation levels of JNK, p38, and I-κB, and decreased the phosphorylation levels of ERK, STAT3, and NF-κB. Furthermore, cytisine treatment led to the generation of ROS, and the NAC attenuated cytisine-induced apoptosis. In vivo, cytisine administration significantly inhibited the lung cancer cell xenograft tumorigenesis. In conclusion, cytisine plays a critical role in suppressing the carcinogenesis of lung cancer cells through cell cycle arrest and induction of mitochondria-mediated apoptosis, suggesting that it may be a promising candidate for the treatment of human lung cancer.


Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Lung Neoplasms/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Azocines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinolizines/pharmacology , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
16.
Mol Med Rep ; 20(5): 4576-4586, 2019 Nov.
Article En | MEDLINE | ID: mdl-31702038

Quinalizarin has been demonstrated to exhibit potent antitumor activities in lung cancer and gastric cancer cells, but currently, little is known regarding its anticancer mechanisms in human breast cancer cells. The aim of the present study was to investigate the apoptotic effects of quinalizarin in MCF­7 cells and to analyze its molecular mechanisms. The MTT assay was used to evaluate the viability of human breast cancer cells that had been treated with quinalizarin and 5­fluorouracil. Flow cytometric analyses and western blotting were used to investigate the effects of quinalizarin on apoptosis and cycle arrest in MCF­7 cells with focus on reactive oxygen species (ROS) production. The results demonstrated that quinalizarin exhibited significant cytotoxic effects on human breast cancer cells in a dose­dependent manner. Accompanying ROS, quinalizarin induced MCF­7 cell mitochondrial­associated apoptosis by regulating mitochondrial­associated apoptosis, and caused cell cycle arrest at the G2/M phase in a time­dependent manner. Furthermore, quinalizarin can activate p38 kinase and JNK, and inhibit the extracellular signal­regulated kinase, signal transducer and activator of transcription 3 (STAT3) and NF­κB signaling pathways. These effects were blocked by mitogen­activated protein kinase (MAPK) inhibitor and N­acetyl­L­cysteine. The results from the present study suggested that quinalizarin induced G2/M phase cell cycle arrest and apoptosis in MCF­7 cells through ROS­mediated MAPK, STAT3 and NF­κB signaling pathways. Thus, quinalizarin may be useful for human breast cancer treatment, as well as the treatment of other cancer types.


Anthraquinones/pharmacology , Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , MCF-7 Cells
17.
Drug Dev Res ; 80(8): 1040-1050, 2019 12.
Article En | MEDLINE | ID: mdl-31432559

Quinalizarin, a bioactive and highly selective compound, is known to promote apoptosis in colon and lung cancer cells. However, studies evaluating quinalizarin-induced apoptosis in melanoma cells have not been conducted. In the present study, we investigated the underlying mechanisms of antimelanoma activity of quinalizarin in human melanoma A375 cells. The MTT assay and Trypan blue staining were used to evaluate the cell viability. The flow cytometry was used to detect cell cycle, apoptosis and reactive oxygen species (ROS). Western blot was used to detect the expression of cell cycle and apoptosis-related proteins, MAPK, and STAT3. The results revealed a significant dose and time dependent effect of quinalizarin on inhibiting proliferation in three kinds of human melanoma cells, and had no significant toxic effects on normal cells. Moreover, quinalizarin triggered G2/M phase cell arrest by modulating the protein expression levels of CDK 1/2, cyclin A, cyclin B, p21 and p27, and induced apoptosis by down-regulating the antiapoptotic protein Bcl-2 and upregulating the proapoptotic protein BAD, leading to the activation of caspase-3 and PARP in the caspase cascade in A375 cells. Quinalizarin treatment led to apoptosis of A375 cells via activation of MAPK and inhibition of STAT3 signaling pathways. In addition, quinalizarin increased the level of ROS, but ROS scavenger NAC inhibited quinalizarin-induced apoptosis by regulating MAPK and STAT3 signaling pathways. In summary, quinalizarin induces cell cycle arrest and apoptosis via ROS-mediated MAPK and STAT3 signaling pathways in human melanoma A375 cells, and quinalizarin may be used as a novel and effective antimelanoma therapeutic.


Anthraquinones/pharmacology , Melanoma/metabolism , Reactive Oxygen Species/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/drug therapy , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction/drug effects
18.
Mol Med Rep ; 20(3): 2571-2582, 2019 Sep.
Article En | MEDLINE | ID: mdl-31322207

1,4­Naphthoquinone derivatives have superior anticancer effects, but their use has been severely limited in clinical practice due to adverse side effects. To reduce the side effects and extend the anticancer effects of 1,4­naphthoquinone derivatives, 2­(butane­1­sulfinyl)­1,4­naphthoquinone (BQ) and 2­(octane­1­sulfinyl)­1,4­naphthoquinone (OQ) were synthesized, and their anticancer activities were investigated. The anti­proliferation effects, determined by MTT assays, showed that BQ and OQ significantly inhibited the viability of gastric cancer cells and had no significant cytotoxic effect on normal cell lines. The apoptotic effect was determined by flow cytometry, and the results showed that BQ and OQ induced cell apoptosis by regulating the mitochondrial pathway and cell cycle arrest at the G2/M phase via inhibition of the Akt signaling pathway in AGS cells. Furthermore, BQ and OQ significantly increased the levels of reactive oxygen species (ROS) and this effect was blocked by the ROS scavenger NAC in AGS cells. BQ and OQ induced apoptosis by upregulating the protein expression of p38 and JNK and downregulating the levels of ERK and STAT3. Furthermore, expression levels of these proteins were also blocked after NAC treatment. These results demonstrated that BQ and OQ induced apoptosis and cell cycle arrest at the G2/M phase in AGS cells by stimulating ROS generation, which caused subsequent activation of MAPK, Akt and STAT3 signaling pathways. Thus, BQ and OQ may serve as potential therapeutic agents for the treatment of human gastric cancer.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Naphthoquinones/pharmacology , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Naphthoquinones/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
19.
J Cell Mol Med ; 23(8): 4876-4882, 2019 08.
Article En | MEDLINE | ID: mdl-31210419

Despite aggressive multimodality treatment, the prognosis of glioma, especially malignant glioma, remains very poor. After decades of effort, anti-angiogenic therapy has become an important method of cancer treatment in addition to surgery, radiotherapy and chemotherapy. Although the performance of anti-angiogenic therapy in colorectal cancer is good, its performance in malignant glioma remains unsatisfactory. Several phase III clinical trials showed no overall survival benefits. To solve this problem, the division of patients into groups based on their molecular biomarkers is an important step. This paper provides current insights into anti-angiogenic drugs undergoing clinical trials and discusses the potential of molecular biomarkers to guide glioma diagnosis.


Angiogenesis Inhibitors/therapeutic use , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Glioma/drug therapy , Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Biomarkers, Tumor/blood , Brain Neoplasms/classification , Brain Neoplasms/enzymology , Brain Neoplasms/metabolism , Glioma/classification , Glioma/enzymology , Glioma/metabolism , Humans , Indoles/pharmacology , Indoles/therapeutic use , Molecular Targeted Therapy , Prognosis , Snake Venoms/pharmacology , Snake Venoms/therapeutic use
20.
J Chemother ; 31(4): 214-226, 2019 Jul.
Article En | MEDLINE | ID: mdl-31074342

The 1,4-naphthoquinones and their derivatives have garnered great interest due to their antitumor pharmacological properties in various cancers; however, their clinical application is limited by side effects. In this study, to reduce side effects and improve therapeutic efficacy, a novel 1,4-naphthoquinone derivative-2-(4-methoxyphenylthio)-5,8-dimethoxy-1,4-naphthoquinone (MPTDMNQ) was synthesized. We investigated the effects and underlying mechanisms of MPTDMNQ on cell viability, apoptosis, and reactive oxygen species (ROS) generation in human gastric cancer cells. Our results showed that MPTDMNQ decreased cell viability in nine human gastric cancer cell lines. MPTDMNQ significantly induced apoptosis accompanied by the accumulation of ROS in GC cells. However, pre-treatment with the ROS scavenger N-acetyl-L-cysteine (NAC) attenuated the MPTDMNQ-induced apoptosis. Moreover, MPTDMNQ decreased the phosphorylation levels of extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription 3 (STAT3); and increased the phosphorylation levels of c-Jun N-terminal kinase (JNK) and p38 kinase. However, phosphorylation was inhibited by NAC and a mitogen-activated protein kinase (MAPK) inhibitor. These findings showed that MPTDMNQ induced AGS cell apoptosis via ROS-mediated MAPK and STAT3 signaling pathways. Thus, MPTDMNQ may be a promising candidate for treating gastric cancer.


Apoptosis/drug effects , Mitogen-Activated Protein Kinases/metabolism , Naphthoquinones/pharmacology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Signal Transduction/drug effects , Stomach Neoplasms/metabolism
...