Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
IUBMB Life ; 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822621

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that can actively participate in post-transcriptional regulation of genes. A number of studies have shown that miRNAs can serve as important regulators of cancer cell growth, differentiation, and apoptosis. They can also act as markers for the diagnosis and prognosis of certain cancers. To explore the potential prognosis-related miRNAs in liver cancer patients, to provide theoretical basis for early diagnosis and prognosis of liver cancer, as well as to provide a new direction for the targeted therapy of liver cancer. The miRNA expression profiles of liver cancer patients in the the Cancer Genome Atlas database were comprehensively analyzed and various prognostic-related miRNAs of liver cancer were screened out. The data was further subjected to survival analysis, prognostic analysis, gene ontology and kyoto encyclopedia of genes and genomes enrichment analysis, microenvironment analysis, and drug sensitivity analysis by R Language version 4.2.0. Finally, the screened miRNAs were further validated by different experiments. Thus, miNRAs involved in liver cancer diagnosis and prognosis were identified. MiRNA-3680-3p was found to be significantly different in 10 different cancers, including liver cancer, and was significantly associated with the microenvironment, survival, and prognosis of liver cancer patients. In addition, drug sensitivity analysis revealed that miRNA-3680-3p can provide a useful reference for drug selection in targeted therapy for liver cancer. MiRNA-3680-3p can serve as a biomarker for the diagnosis and prognosis of liver cancer patients and down-regulation of miRNA-3680-3p could significantly inhibit both the proliferation and migration of liver cancer cells.

2.
Pharm Dev Technol ; 29(6): 541-550, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769920

ABSTRACT

Currently, chemotherapy is one of the most practiced approaches for the treatment of cancers. However, existing chemotherapeutic drugs have poor aqueous solubility, poor selectivity, higher systematic toxicity, and poor target accumulation. In this study, we designed and synthesized a boronic acid/ester-based pH-responsive nano-valve that specifically targets the microenvironment in cancer cells. The nano-valve comprises phenylboronic acid-coated mesoporous silica nanoparticles (B-MSN) loaded with polyphenolic compound Rosmarinic acid (ROS-B-MSN). The nano-valve was further coated with lignin (LIG) to achieve our desired LIG-ROS-BMSN nano-valve for targeted chemotherapy against Hep-G2 and NCI-H460 cell lines. The structure and properties of NPs were characterized by Fourier-transformed infrared spectroscopy (FTIR), Scanning Electron Microscopy (SEM) in combination with EDX, and Dynamic light scattering (DLS). The outcomes revealed that the designed LIG-ROS-BMSN were in the nanorange (144.1 ± 0.70 nm), had negative Zeta potential (-15.7 ± 0.46 mV) and had a nearly spherical morphology. In vitro, drug release investigations showed a controlled pH-dependent release profile under mild acidic conditions that could enhance the targeted chemotherapeutic response against cancer in mild acidic environments. The obtained LIG-ROS-BMSN nano valve achieved significantly lower IC50 values of (1.70 ± 0.01 µg/mL and 3.25 ± 0.14 µg/mL) against Hep-G2 and NCI-H460 cell lines as compared to ROS alone, which was (14.0 ± 0.7 µg/mL and 29.10 ± 0.25 µg/mL), respectively. The cellular morphology before and after treatment was further confirmed via inverted microscopy. The outcomes of the current study imply that our designed LIG-ROS-BMSN nanovalve is a potential carrier for cancer chemotherapeutics.


Subject(s)
Boronic Acids , Cinnamates , Depsides , Drug Liberation , Liver Neoplasms , Lung Neoplasms , Nanoparticles , Rosmarinic Acid , Silicon Dioxide , Depsides/administration & dosage , Depsides/pharmacology , Depsides/chemistry , Cinnamates/administration & dosage , Cinnamates/pharmacology , Cinnamates/chemistry , Humans , Nanoparticles/chemistry , Boronic Acids/chemistry , Silicon Dioxide/chemistry , Hydrogen-Ion Concentration , Liver Neoplasms/drug therapy , Cell Line, Tumor , Lung Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Hep G2 Cells , Porosity , Drug Delivery Systems/methods , Cell Survival/drug effects , Drug Carriers/chemistry
3.
Int J Biochem Cell Biol ; 168: 106516, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38219975

ABSTRACT

In view of the tumor-inhibiting effect of α-santalol in various cancers and the role of E2F transcription factor 1 (E2F1) as an important target for anticancer research, this study investigates the relation between α-santalol and E2F1, as well as the effect of α-santalol on liver cancer progression and the corresponding mechanism. Concretely, liver cancer cells were treated with different concentrations of α-santalol. The IC50 value of α-santalol was determined using Probit regression analysis. Then, transcription factors that are targeted by α-santalol and differentially expressed in liver cancer were screened out. The clinicopathological impact of E2F1 and its targets were evaluated and predicted. The expressions of E2F1 and high-mobility group box 2 (HMGB2) and their correlation in the liver cancer tissues were analyzed by bioinformatics. The effects of E2F1 and HMGB2 on the biological characteristics of liver cancer cells were examined through loss/gain-of-function and molecular assays. With the extension of treatment time, the inhibitory effects of 10 µmol/L and 20 µmol/L α-santalol on cancer cell survival rate were enhanced (P < 0.001). E2F1 and HMGB2 were highly expressed and positively correlated in liver cancer tissues (P < 0.05). High E2F1 expression was correlated with large tumors and high TNM stages (P < 0.05). E2F1 knockdown promoted the effects of α-santalol on dose-dependently inhibiting viability, colony formation, invasion and migration (P < 0.05). Moreover, E2F1 knockdown reduced the IC50 value and HMGB2 level, while HMGB2 overexpression produced opposite effects. HMGB2 overexpression and E2F1 knockdown mutually counteracted their effects on the IC50 value and on the viability and apoptosis of α-santalol-treated liver cancer cells (P < 0.01). Collectively, blocking the E2F1/HMGB2 pathway enhances the intervention effects of α-santalol on the proliferation, migration and invasion of liver cancer cells.


Subject(s)
HMGB2 Protein , Liver Neoplasms , Polycyclic Sesquiterpenes , Humans , Cell Line, Tumor , HMGB2 Protein/genetics , Cell Proliferation , Liver Neoplasms/drug therapy , Transcription Factors/metabolism , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic
4.
Curr Protein Pept Sci ; 25(1): 71-82, 2024.
Article in English | MEDLINE | ID: mdl-37642183

ABSTRACT

BACKGROUND: Recently, the importance of the interactions between liver cancer cells and fibroblasts has been increasingly recognized; however, many details remain to be explored. METHODS: In this work, we first studied their intercellular interactions using conditioned medium from mouse embryonic fibroblasts (MEFs), then through a previously established coculture model. RESULTS: Culturing in a conditioned medium from MEFs could significantly increase the growth, migration, and invasion of liver cancer cells. The coculture model further demonstrated that a positive feedback loop was formed between transforming growth factor-ß (TGF-ß) from HepG2 cells and mHGF (mouse hepatocyte growth factor) from MEFs during coculture. In this feedback loop, c-Met expression in HepG2 cells was significantly increased, and its downstream signaling pathways, such as Src/FAK, PI3K/AKT, and RAF/MEK/ERK, were activated. Moreover, the proportion of activated MEFs was also increased. More importantly, the growth-promoting effects caused by the interaction of these two cell types were validated in vitro by a 3D spheroid growth assay and in vivo by a xenograft mouse model. CONCLUSION: Collectively, these findings provide valuable insights into the interactions between fibroblasts and liver cancer cells, which may have therapeutic implications for the treatment of liver cancer.


Subject(s)
Liver Neoplasms , Transforming Growth Factor beta , Humans , Animals , Mice , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Heterografts , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Fibroblasts/metabolism , Disease Models, Animal , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism
5.
Genomics ; 116(1): 110764, 2024 01.
Article in English | MEDLINE | ID: mdl-38113974

ABSTRACT

Sorafenib is currently the first-line treatment for patients with advanced liver cancer, but its therapeutic efficacy declines significantly after a few months of treatment. Therefore, it is of great importance to investigate the regulatory mechanisms of sorafenib sensitivity in liver cancer cells. In this study, we provided initial evidence demonstrating that circPHKB, a novel circRNA markedly overexpressed in sorafenib-treated liver cancer cells, attenuated the sensitivity of liver cancer cells to sorafenib. Mechanically, circPHKB sequestered miR-1234-3p, resulting in the up-regulation of cytochrome P450 family 2 subfamily W member 1 (CYP2W1), thereby reducing the killing effect of sorafenib on liver cancer cells. Moreover, knockdown of circPHKB sensitized liver cancer cells to sorafenib in vivo. The findings reveal a novel circPHKB/miR-1234-3p/CYP2W1 pathway that decreases the sensitivity of liver cancer cells to sorafenib, suggesting that circPHKB and the axis may serve as promising targets to improve the therapeutic efficacy of sorafenib against liver cancer.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , MicroRNAs/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Up-Regulation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Proliferation , Drug Resistance, Neoplasm , Cytochrome P450 Family 2/genetics
6.
Cells ; 12(22)2023 11 17.
Article in English | MEDLINE | ID: mdl-37998378

ABSTRACT

We investigated the possibility of using Raman spectroscopy assisted by artificial intelligence methods to identify liver cancer cells and distinguish them from their Non-Tumor counterpart. To this aim, primary liver cells (40 Tumor and 40 Non-Tumor cells) obtained from resected hepatocellular carcinoma (HCC) tumor tissue and the adjacent non-tumor area (negative control) were analyzed by Raman micro-spectroscopy. Preliminarily, the cells were analyzed morphologically and spectrally. Then, three machine learning approaches, including multivariate models and neural networks, were simultaneously investigated and successfully used to analyze the cells' Raman data. The results clearly demonstrate the effectiveness of artificial intelligence (AI)-assisted Raman spectroscopy for Tumor cell classification and prediction with an accuracy of nearly 90% of correct predictions on a single spectrum.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Artificial Intelligence , Spectrum Analysis, Raman/methods
7.
Noncoding RNA Res ; 8(4): 675-685, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37860266

ABSTRACT

Researches indicate miR-3200 is closely related to tumorigenesis, However, the role of miR-3200 in human hepatocarcinogenesis is still unclear. In this study, we clearly demonstrate that miR-3200 accelerates the growth of liver cancer cells in vivo and in vitro. Obviously, these findings are noteworthy that miR-3200 affects the transcriptional regulation for several genes, including DSP,BABAM2, Rab7A,SQSTM1,PRKAG2,CDK1,ABCE1,BECN1,PTEN,UPRT. And miR-3200 affects the transcriptional ability of several genes, such as, upregulating CADPS, DSP,FBXO32, PPCA,SGK1, PATXN7L1, PLK2,ITGB5,FZD3,HOXC8,HSPA1A,C-Myc,CyclnD1,CyclinE,PCNA and down -regulating SUV39H1, MYO1G, OLFML3, CBX5, PPDE2A, HOXA7, RAD54L, CDC45,SHMT7,MAD2L1,P27,IQGAP3,PTEN,P57,SCAMP3,etc...On the other hand, it is obvious that miR-3200 affects the translational ability of several genes, such as, upregulating GNS,UPRT,EIFAD,YOS1,SGK1,K-Ras,PKM2,C-myc,Pim1,CyclinD1,mTOR,erbB-2,CyclinE,PCNA,RRAS,ARAF,RAPH1,etc.. and down-regulating KDM2A, AATF, TMM17B, RAB8B, MYO1G,P21WAF1/Cip1,GADD45,PTEN,P27,P18,P57,SERBP1,RPL34,UFD1,Bax,ANXA6,GSK3ß. Strikingly, miR-3200 affects some signaling pathway in liver cancer, including carbon metabolism signaling pathway, DNA replication pathway, FoxO signaling pathway, Hippo signaling pathway, serine and threonine metabolism signaling pathway, mTOR signaling pathway, Fatty acid biosynthesis signaling pathway, carcinogenesis-receptor activation signaling pathway, autophagy signaling pathway. Furthermore, our results suggest that miR-3200 enhances expression of RAB7A, and then Rab7A regulates the carcinogenic function of miR-3200 by increasing telomere remodeling in human liver cancer. These results are of great significance for the prevention and treatment of human liver cancer.

8.
Artif Cells Nanomed Biotechnol ; 51(1): 361-370, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37524306

ABSTRACT

BACKGROUND: Curcumin has been used in the treatment of several diseases; however, its low pharmacologic profile reduces its therapeutic use. Towards improving its biological activity, nanoformulations have emerged. Thus, we aimed to determine whether curcumin nanoparticles (Cur-NPs) coated with PEG/chitosan improve the treatment of liver cancer (LC) cells and underpin the molecular mechanisms underlying their anti-cancer activity. METHODS: Cur-NPs were synthesised in the form of Cur-PLGA-PEG/chitosan NPs. The effect of Cur-NPs was assessed in HepG2 and Huh 7 LC cells and THLE-2 normal liver cells. RESULTS: The size of synthesised Cur-NPS was determined in the standard range of 141.2 ± 47.5 nm. Compared to THLE-2 cells, LC cells treated with Cur-NPs exerted cytotoxicity at 6.25 µg/mL after 48h. Treatment of HepG-2 cells with 2.5 µg/mL of Cur-NPs inhibited cell migration and this inhibition was augmented at 10 µg/mL (p < 0.001). Treatment of chicken embryo with 5 µg/mL Cur-NPs reduced angiogenesis (p < 0.001) of 4-day-old embryos. The nanoformulation upregulated Bax and p53 and downregulated Bcl-2 in a concentration-dependent manner and subsequently induce apoptosis in HepG-2 cells. CONCLUSION: Treatment of LC cells with Cur-NPs decreased cell proliferation, migration, and angiogenesis, and induced cell death by promoting the proapoptotic pathway.


Curcumin nanoparticles (Cur-NPs) increase the anticancer efficiency of Curcumin against liver cancer cells.Cur-NPs induce apoptotic cell death of Liver cancer cells.Cur-NPs have ant-angiogenesis and metastasis effect.


Subject(s)
Chitosan , Curcumin , Liver Neoplasms , Nanoparticles , Chick Embryo , Animals , Curcumin/pharmacology , Curcumin/therapeutic use , Cell Line, Tumor , Chitosan/pharmacology , Apoptosis , Liver Neoplasms/drug therapy
9.
J Biomater Sci Polym Ed ; 34(14): 1891-1910, 2023 10.
Article in English | MEDLINE | ID: mdl-37000910

ABSTRACT

Curcumin is one of the most important polyphenolic nutrients in pharmaceutical industries. Unfortunately, its poor solubility and bioavailability have hampered its clinical application. To improve curcumin solubility and bioavailability, a natural nanocarrier made from protein-polysaccharide conjugate has been developed. Following antisolvent precipitation method, zein (Z) nanoparticles coated with dextran sulphate (DS) have been fabricated as curcumin (C) nanocarrier (DSZCNPs). The physicochemical properties of the nanoconjugate were measured using different techniques. Morphologically, DSZCNPs appeared spherical and monodispersed in scanning electron microscope (SEM) and transmission electron microscope (TEM) images. Curcumin encapsulation efficiency was ≈ 96%. DSZCNPs size was 180 nm and the polydispersity index value (PDI) 0.28. Zeta potential for DSZCNPs was -28.5 mV. DSZCNPs showed stability either for shelf storage (100 days) or at different pHs. Furthermore, DSZCNPs protected zein nanoparticles degradation in gastric environment and achieved controlled curcumin release in intestinal environment. DSZCNPs greatly enhanced the antioxidant activity of curcumin as demonstrated by DPPH assay. DSZCNPs had significant results in the reduction of colony forming unit (CFU%) against the tested microbes when compared with free curcumin. Also, the anticancer activity of DSZCNPs and free curcumin against hepatocellular carcinoma cells (HepG2) were assessed by MTT assay. IC50 for DSZCNPs was 13 µg/ml compared to 50 µg/ml for free curcumin indicating the therapeutic impact of DSZCNPs over free curcumin.Based on the above results, the developed zein-dextran nanocomplex exhibited high stability and improved the efficacy and bioactivity of curcumin suggesting its potential utility as nanovehicle for the hydrophobic drug curcumin.


Subject(s)
Curcumin , Zein , Nanoconjugates , Curcumin/pharmacology , Curcumin/chemistry , Zein/chemistry , Dextrans , Particle Size
10.
Int J Mol Sci ; 24(4)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36835343

ABSTRACT

Mangosteen peel, a waste produced during mangosteen processing, has been reported to be rich in xanthone and anthocyanin, both of which possess vital biological activities such as anti-cancer properties. The objectives of this study were to analyze various xanthones and anthocyanins in mangosteen peel by UPLC-MS/MS for the subsequent preparation of both xanthone and anthocyanin nanoemulsions to study their inhibition effects on liver cancer cells HepG2. Results showed that methanol was the optimal solvent for the extraction of xanthones and anthocyanins, with a total amount of 68,543.39 and 2909.57 µg/g, respectively. A total of seven xanthones, including garcinone C (513.06 µg/g), garcinone D (469.82 µg/g), γ-mangostin (11,100.72 µg/g), 8-desoxygartanin (1490.61 µg/g), gartanin (2398.96 µg/g), α-mangostin (51,062.21 µg/g) and ß-mangostin (1508.01 µg/g), as well as two anthocyanins including cyanidin-3-sophoroside (2889.95 µg/g) and cyanidin-3-glucoside (19.72 µg/g), were present in mangosteen peel. The xanthone nanoemulsion was prepared by mixing an appropriate portion of soybean oil, CITREM, Tween 80 and deionized water, while the anthocyanin nanoemulsion composed of soybean oil, ethanol, PEG400, lecithin, Tween 80, glycerol and deionized water was prepared as well. The mean particle size of the xanthone extract and nanoemulsion were, respectively, 22.1 and 14.0 nm as determined by DLS, while the zeta potential was -87.7 and -61.5 mV. Comparatively, xanthone nanoemulsion was more effective than xanthone extract in inhibiting the growth of HepG2 cells, with the IC50 being 5.78 µg/mL for the former and 6.23 µg/mL for the latter. However, the anthocyanin nanoemulsion failed to inhibit growth of HepG2 cells. Cell cycle analysis revealed that the proportion of the sub-G1 phase followed a dose-dependent increase, while that of the G0/G1 phase showed a dose-dependent decline for both xanthone extracts and nanoemulsions, with the cell cycle being possibly arrested at the S phase. The proportion of late apoptosis cells also followed a dose-dependent rise for both xanthone extracts and nanoemulsions, with the latter resulting in a much higher proportion at the same dose. Similarly, the activities of caspase-3, caspase-8 and caspase-9 followed a dose-dependent increase for both xanthone extracts and nanoemulsions, with the latter exhibiting a higher activity at the same dose. Collectively, xanthone nanoemulsion was more effective than xanthone extract in inhibiting the growth of HepG2 cells. Further research is needed to study the anti-tumor effect in vivo.


Subject(s)
Garcinia mangostana , Liver Neoplasms , Xanthones , Humans , Anthocyanins , Tandem Mass Spectrometry , Soybean Oil , Chromatography, Liquid , Polysorbates , Xanthones/pharmacology , Cell Line, Tumor , Plant Extracts/pharmacology , Water
11.
Molecules ; 28(4)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36838549

ABSTRACT

Astragalus alcohol soluble polysaccharide (AASP) could present superior water solubility and antitumor activity with high concentration. Selenium nanoparticles (SeNPs) have received growing attention in various fields, but their unstable property increases the application difficulties. In the present study, functionalized nano-composites (AASP-SeNPs) were synthesized through SeNPs using AASP (average molecular weight of 2.1 × 103 Da) as a surface modifier, and the preliminary structural characteristics and inhibitory mechanism on liver cancer (HepG2) cells were investigated. Results showed that AASP-SeNPs prepared under a sodium selenite/AASP mass ratio of 1/20 (w/w) were uniformly spherical with a mean grain size of 49.80 nm and exhibited superior dispersivity and stability in water solution. Moreover, the composites could dose-dependently inhibit HepG2 cell proliferation and induce apoptosis through effectively regulating mitochondria-relevant indicators including ΔΨm depletion stimulation, intracellular ROS accumulation, Bax/Bcl-2 ratio improvement, and Cytochrome c liberation promotion. These results provide scientific references for future applications in functional food and drug industries.


Subject(s)
Liver Neoplasms , Nanoparticles , Selenium , Humans , Nanoparticles/chemistry , Polysaccharides , Selenium/chemistry , Hep G2 Cells
12.
Front Pharmacol ; 13: 1039376, 2022.
Article in English | MEDLINE | ID: mdl-36506551

ABSTRACT

The aim of this study was to investigate the anticancer mechanisms of white genius mushroom (WGM). WGM is a popular edible mushroom in Taiwan and has been demonstrated to mediate potent antiproliferation effects against human Hep3B liver cancer cells in our previous study. According to next generation sequencing technology and KEGG pathway enrichment analysis, mTOR and MAPK signaling pathways were markedly changed during treatment with WGM extracts in Hep3B cells. Therefore, this study examined the effects of WGM extracts on the expression of mTOR and MAPK signaling pathway-related proteins, such as PI3K, Akt, mTOR, Ras, Raf, MEK, ERK, p38 and JNK in Hep3B cells. According to the results of immunoblotting, we demonstrated that the protein expression of the members of PI3K/Akt/mTOR and MAPK signaling pathways were involved in WGM extracts-induced cell death. Furthermore, the inhibitors of PI3K/Akt/mTOR and MAPK signaling pathways such as rapamycin, MK2206, LY3214996 and SB202190, blocked the induction of cell death and vacuoles formation induced by WGM extracts. This study also demonstrated that WGM extracts is able to inhibit Hep3B cell migration and colony formation in a dose-dependent manner. In addition to being a very popular food, WGM should be a pharmacologically safe natural agent for cancer treatment. Therefore, WGM might be designed to develop into a dietary chemopreventive agent for the cancer treatment.

13.
Front Pharmacol ; 13: 888863, 2022.
Article in English | MEDLINE | ID: mdl-35784738

ABSTRACT

White strain of Hypsizygus marmoreus is named as white genius mushroom (WGM) and is a popular food in Taiwan. We have confirmed the cytotoxicity of WGM extracts on human Hep3B liver cancer cells. A total of 8711 significantly differential genes were identified through large-scale transcriptome sequencing. According to the KEGG pathway enrichment analysis, autophagy, mitophagy and apoptosis pathways were identified as significant in WGM extracts-treated cells. WGM extracts induced a dose-dependent generation of reactive oxygen species (ROS) and membrane-enclosed vacuoles in Hep3B cells. The inhibition of ROS by the ROS scavengers blocked the induction of cell death and vacuoles formation. We suggested that the cell death and membrane-enclosed vacuoles induced by WGM extracts are dependent on ROS production in Hep3B cells. (2E,6E)-3,7,11,15,19,23,27,31,35-Nonamethylhexatriaconta-2,6,34-triene-1,11,15,19,23,27,31-heptol and (18:2) lysophosphatidylcholine were identified in WGM extracts. In addition to being a very popular edible mushrooms, WGM may be developed into a dietary supplement or dietary chemopreventive agent for the cancer treatment.

14.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 37(4): 415-418, 2021 Jul.
Article in Chinese | MEDLINE | ID: mdl-34374263

ABSTRACT

Objective: To investigate the effects and molecular mechanisms of shikonin on liver cancer SMMC-772 cells. Methods: SMMC-7721 cells were treated with shikonin at the concentrations of 0, 5, 20, 80 and 320 ng/ml for 0, 24, 48 and 72 h respectively. The proliferative activity of the cells was detected by CCK8 assay. The nuclear type changes of cells was observed after hoechst 33342 staining. Flow cytometry was used to analyze cell apoptosis and death rate. The expressions of proteins in cells were determined by Western blot, and the tumor inhibitive effects were observed through anti-tumor experiment on the BALB/c mice. Results: In vitro experiments, shikonin could inhibit the proliferation of SMMC-7721 cells and induce their apoptosis(P<0.01), up-regulate the expression of p53 gene, down-regulate the phosphorylation levels of AKT and PI3K protein. In vivo study also confirmed that shikonin could significantly inhibit the growth of tumor in tumor-bearing mice(P<0.01)in dose-dependent and time-dependent manners. Conclusion: Shikonin can inhibit the proliferation activitity and induce apoptosis of SMMC-7721 cells by affecting the PI3K/AKT signal pathway and has potential anti-liver cancer functions.


Subject(s)
Liver Neoplasms , Phosphatidylinositol 3-Kinases , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Naphthoquinones , Proto-Oncogene Proteins c-akt
15.
Bioengineered ; 12(1): 4581-4592, 2021 12.
Article in English | MEDLINE | ID: mdl-34323647

ABSTRACT

Malignant tumors are a serious threat to human health. Surgical resection is the most effective treatment for liver cancer. However, liver cancer is mostly found at an advanced stage, is difficult to remove by surgery, and has a very high recurrence rate after surgery. The current liver cancer treatment drugs have serious side effects, and the treatment effect is not ideal, far from meeting the clinical needs. Based on this, this paper studies the effect of propofol on the proliferation and apoptosis of liver cancer cells through the TGF-B1/Smad2 signaling pathway, and explores the proliferation, adhesion and apoptosis of cancer cells in patients with propofol. This paper uses a comparative experiment. With medical imaging method, 80 rats with liver cancer in the same period were cultured. High-precision microscope and radiolocation method were used to observe and record the whole process of propofol regulating Smad2 signal pathway. The results show that propofol can effectively inhibit the proliferation of cancer cells in patients with liver cancer. Propofol can increase the activity and content of transforming growth factor-ß1 by 12% and 20%, respectively, and then inhibit the proliferation rate of liver cancer cells by 10% through the Smad2 signaling pathway, and exponentially increase the apoptotic number of liver cancer cells. This shows that propofol has a significant inhibitory effect on the cycle of liver cancer cells. Under the action of propofol, the life cycle of liver cancer cells is shortened, which provides a certain theoretical basis for the treatment of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Propofol/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/metabolism , Mice, Inbred C57BL , Rats , Smad2 Protein/metabolism , THP-1 Cells
16.
Spectrochim Acta A Mol Biomol Spectrosc ; 262: 120098, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34252742

ABSTRACT

As the main existing form of SO2 derivatives, bisulfite showed closely relationship to many diseases. In this work, a new fluorescent probe (SDPP-DM) based on thienyl-substituted diketopyrrolopyrrole (SDPP) was designed and synthesized for the detection of endogenous bisulfite. The probe displayed obvious color changes from green to pink towards bisulfite due to the reduced conjugated length caused by the addition to the α,ß-unsaturated double bond of its structure, and the change of the fluorescence intensity of SDPP-DM (I/I0) was about 16 folds. In addition, SDPP-DM was prepared a test strip for bisulfite identified by naked eye through color and fluorescence changes. Besides, SDPP-DM was successfully applied to imaging and discriminating different endogenous bisulfite levels in normal and cancer cells of liver. More importantly, the ROS generation and cell viability tests showed the phototoxicity of SDPP-DM triggered by bisulfite, indicating the specific phototoxicity of SDPP-DM towards liver cancer cells than normal liver cells.


Subject(s)
Fluorescent Dyes , Liver Neoplasms , Humans , Ketones , Pyrroles , Sulfites/toxicity
17.
Biosci Biotechnol Biochem ; 85(7): 1711-1715, 2021 Jun 24.
Article in English | MEDLINE | ID: mdl-33988672

ABSTRACT

Neoplastic cells secrete autocrine motility factor (AMF) to stimulate the motility of cancer cells. In this study, AMF secreted from HT-29 colorectal cancer cells selectively suppressed liver cancer cells by downregulating pAKT and ß-catenin. In addition, HT-29 AMF significantly augmented the activity of methyl jasmonate against liver cancer cells and is a promising alternative for liver cancer therapy.


Subject(s)
Acetates/pharmacology , Cell Proliferation/drug effects , Cyclopentanes/pharmacology , Glucose-6-Phosphate Isomerase/pharmacology , Liver Neoplasms/pathology , Oxylipins/pharmacology , Acetates/administration & dosage , Cyclopentanes/administration & dosage , Down-Regulation/drug effects , Glucose-6-Phosphate Isomerase/administration & dosage , HT29 Cells , Humans , Oxylipins/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism
18.
Exp Ther Med ; 21(6): 559, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33850531

ABSTRACT

Long non-coding RNA (lncRNA) ADAM metallopeptidase with thrombospondin type 1 motif 9 antisense RNA 2 (ADAMTS9-AS2) is involved in various types of cancer, such as ovarian cancer, lung cancer and clear cell renal cell carcinoma. However, the roles of ADAMTS9-AS2 in liver cancer are not completely understood. The present study aimed to determine the functional role of ADAMTS9-AS2 in human liver cancer and investigate the potential underlying molecular mechanisms. The expression levels of ADAMTS9-AS2 and ADAMTS9 were determined following ADAMTS9-AS2 overexpression and knockdown. The results indicated that ADAMTS9-AS2 overexpression and knockdown increased and decreased ADAMTS9 mRNA and protein expression levels, respectively, indicating that alterations in ADAMTS9 expression corresponded with ADAMTS9-AS2 expression. Subsequently, the effects of ADAMTS9-AS2 on liver cancer cell proliferation, migration and invasion were analyzed by performing Cell Counting Kit-8, wound healing and Transwell assays, respectively. The results demonstrated that ADAMTS9-AS2 inhibited liver cancer cell proliferation, migration and invasion. Finally, the effect of ADAMTS9 on PI3K/AKT/mTOR signaling pathway-associated proteins [AKT, phosphorylated-AKT, phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit ß (PIK3CB), mTOR and phosphorylated-mTOR], several key autophagy-related proteins [light chain 3-I/II (LC3-I/II), beclin 1 (BECN1) and sequestosome 1 (SQSTM1)] and apoptosis-related proteins (Bax and Bcl-2) was detected via western blotting. The results suggested that ADAMTS9-AS2 downregulated the phosphorylation of AKT and mTOR, the protein expression level of PIK3CB, as well as the expression levels of autophagy protein SQSTM1 and antiapoptotic protein Bcl-2. By contrast, ADAMTS9-AS2 upregulated the expression levels of autophagy proteins LC3-II and BECN1, and the proapoptotic protein Bax. Collectively, ADAMTS9-AS2 inhibited liver cancer cell proliferation, migration and invasion via inhibiting the PI3K/AKT/mTOR signaling pathway. The present study provided a novel insight into the role of ADAMTS9-AS2 in liver cancer.

19.
Technol Cancer Res Treat ; 20: 1533033820957021, 2021.
Article in English | MEDLINE | ID: mdl-33827350

ABSTRACT

Although the treatment of liver cancer has made great progress, the mechanism of its occurrence is not completely clear. miR-155 plays an important regulatory role in tumorigenesis and development, including survival, proliferation, migration and invasion. However, the role and regulatory mechanism of miR-155 in liver cancer has rarely been reported. We analyzed miR-155 expression in liver cancer tissue samples and cell lines by qRT-PCR. The expression of miR-155 was measured by qRT-PCR before and after miR-155-mimic and sh-miR-155 transfection. CCK-8 and clonogenic assays were used to detect the proliferation of liver cancer cells. Cell scratch and invasion assays were used to detect migration and invasion. RNA-seq was used to detect the difference in RNA expression in liver cancer cells. SRPK1 expression was detected in liver cancer cells before and after transfection by qRT-PCR and western blotting. We observed that miR-155 was downregulated in liver cancer tissues compared with normal tissues. Furthermore, we demonstrated that liver cancer cell proliferation, migration and invasion are markedly suppressed by miR-155. Importantly, we also demonstrated that SRPK1 is directly regulated by miR-155 during the process of liver cancer cell proliferation and metastasis. Finally, the overexpression of miR-155 inhibits malignant biological behavior of human liver cancer cells. We report the abnormal expression of the miR-155 cluster in liver cancer cells, which inhibits cancer cell proliferation and metastasis. In addition, we identified SRPK1 as a target gene of miR-155 during the process of liver cancer cell proliferation and metastasis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Protein Serine-Threonine Kinases/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation , Female , Gene Expression , Hep G2 Cells , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis/genetics , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, RNA , Transcriptome , Transfection , Tumor Stem Cell Assay
20.
Food Res Int ; 142: 110187, 2021 04.
Article in English | MEDLINE | ID: mdl-33773660

ABSTRACT

This study investigated the effects of ultrasound assisted-subcritical water (U-SW), subcritical water (SW), ultrasound (U) and hot water (HW) pretreatments and acid hydrolysis (AH) and alkaline hydrolysis (AlkH) on the phenolic composition, antioxidant potential and cytotoxicity of Tartary buckwheat hull extracts. The Folin Ciocalteu assay and HPLC-MS were used to characterize and quantify phenolics of the extracts. The ABTS, FRAP and TEAC assays were used to measure antioxidant activity and the MTT assay was used to measure cytotoxicity of the extracts in HepG2 human liver cancer cells. Results showed that U-SW gave the best AH yield of phenolics (128.45), followed by SW (85.82) and U (64.70), compared to the control, HW (35.82 mgg-1). The same trend was observed for phenols extracted using AlkH. U-SW had the highest antioxidant activity, followed by SW and U regardless of hydrolytic method used. Cytotoxicity followed a similar trend with U-SW and SW being the most cytotoxic to liver cancer cells, followed by U, with the least being HW. The findings suggested that plant materials such as Tartary buckwheat hulls can be pretreated with U-SW, SW and U prior to hydrolytic recovery of bound polyphenols. Also, AH was more efficient than AlkH for phenol extraction, and gave extracts with higher antioxidant activity and cytotoxicity in HepG2 liver carcinoma cells. This application allows for beneficial usage of agricultural biomass and help diversify income sources and products for industry.


Subject(s)
Fagopyrum , Antioxidants/pharmacology , Humans , Hydrolysis , Phenols , Polyphenols/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL