Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
3.
Front Oncol ; 13: 1191646, 2023.
Article in English | MEDLINE | ID: mdl-37675235

ABSTRACT

Systemic chemotherapies are the primary treatment options for patients with unresectable and metastatic intrahepatic cholangiocarcinoma (ICC), but the effectiveness of current systemic therapies is limited. The development of targeted-therapy has changed the treatment landscape of ICC, and comprehensive genome sequencing of advanced cholangiocarcinoma patients could be beneficial to identify potential targets to guide individualized treatment. Herein, we reported an unresectable and metastatic ICC patient who detected EML4-ALK rearrangement in peripheral blood, which was later confirmed on tissue-based testing, and achieved partial response (PR) after first-line treatment with ensartinib. This case suggests that the liquid biopsy is of clinical value for unresectable or metastatic ICC, and the discovery of rare molecular targets provides new therapeutically approaches for advanced ICC patients.

5.
J Cell Mol Med ; 27(15): 2215-2227, 2023 08.
Article in English | MEDLINE | ID: mdl-37307404

ABSTRACT

Melatonin, a lipophilic hormone released from the pineal gland, has oncostatic effects on various types of cancers. However, its cancer treatment potential needs to be improved by deciphering its corresponding mechanisms of action and optimising therapeutic strategy. In the present study, melatonin inhibited gastric cancer cell migration and soft agar colony formation. Magnetic-activated cell sorting was applied to isolate CD133+ cancer stem cells. Gene expression analysis showed that melatonin lowered the upregulation of LC3-II expression in CD133+ cells compared to CD133- cells. Several long non-coding RNAs and many components in the canonical Wnt signalling pathway were altered in melatonin-treated cells. In addition, knockdown of long non-coding RNA H19 enhanced the expression of pro-apoptotic genes, Bax and Bak, induced by melatonin treatment. Combinatorial treatment with melatonin and cisplatin was investigated to improve the applicability of melatonin as an anticancer therapy. Combinatorial treatment increased the apoptosis rate and induced G0/G1 cell cycle arrest. Melatonin can regulate migration and stemness in gastric cancer cells by modifying many signalling pathways. Combinatorial treatment with melatonin and cisplatin has the potential to improve the therapeutic efficacy of both.


Subject(s)
Melatonin , Stomach Neoplasms , Humans , Cisplatin/pharmacology , Cisplatin/therapeutic use , Melatonin/pharmacology , Melatonin/therapeutic use , Stomach Neoplasms/pathology , Cell Line, Tumor , Signal Transduction , Apoptosis , Cell Proliferation
6.
Pharmacol Res ; 194: 106753, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37011775

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.

9.
Front Immunol ; 13: 1051998, 2022.
Article in English | MEDLINE | ID: mdl-36439106

ABSTRACT

With cancer incidence rates continuing to increase and occurrence of resistance in drug treatment, there is a pressing demand to find safer and more effective anticancer strategy for cancer patients. Natural products, have the advantage of low toxicity and multiple action targets, are always used in the treatment of cancer prevention in early stage and cancer supplement in late stage. Tumor microenvironment is necessary for cancer cells to survive and progression, and immune activation is a vital means for the tumor microenvironment to eliminate cancer cells. A number of studies have found that various natural products could target and regulate immune cells such as T cells, macrophages, mast cells as well as inflammatory cytokines in the tumor microenvironment. Natural products tuning the tumor microenvironment via various mechanisms to activate the immune response have immeasurable potential for cancer immunotherapy. In this review, it highlights the research findings related to natural products regulating immune responses against cancer, especially reveals the possibility of utilizing natural products to remodel the tumor microenvironment to overcome drug resistance.


Subject(s)
Biological Products , Neoplasms , Humans , Tumor Microenvironment , Biological Products/pharmacology , Biological Products/therapeutic use , Immunotherapy , Drug Resistance
11.
Cell Death Dis ; 13(5): 487, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35597793

ABSTRACT

Epigenetic alteration is a pivotal factor in tumor metastasis. PHD finger protein 13 (PHF13) is a recently identified epigenetic reader of H3K4me2/3 that functions as a transcriptional co-regulator. In this study, we demonstrate that PHF13 is required for pancreatic-cancer-cell growth and metastasis. Integrative analysis of transcriptome and epigenetic profiles provide further mechanistic insights into the epigenetic regulation of genes associated with cell metastasis during the epithelial-to-mesenchymal transition (EMT) induced by transforming growth factor ß (TGFß). Our data suggest PHF13 depletion impairs activation of TGFß stimulated genes and correlates with a loss of active epigenetic marks (H3K4me3 and H3K27ac) at these genomic regions. These observations argue for a dependency of TGFß target activation on PHF13. Furthermore, PHF13-dependent chromatin regions are enriched in broad H3K4me3 domains and super-enhancers, which control genes critical to cancer-cell migration and invasion, such as SNAI1 and SOX9. Overall, our data indicate a functional and mechanistic correlation between PHF13 and EMT.


Subject(s)
DNA-Binding Proteins , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Neoplasms , Transcription Factors , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Humans , Neoplasms/pathology , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism
12.
Front Oncol ; 12: 859195, 2022.
Article in English | MEDLINE | ID: mdl-35359416

ABSTRACT

B-cell receptor-associated protein 31 (BAP31) has been shown to overexpress in a wide range type of cancers. The present study aims to investigate the role of BAP31 on migration in lung cancer. Results showed that the migration of BAP31 knockdown cells was weaken than the control cells. Applying TGFß to treat BAP31 knockdown cells could reduce cell migration. The enhancement on proliferation by TGFß treatment was downregulated after BAP31 knockdown. The cell death and G0/G1 phase arrest was increased in the cells with TGFß and BAP31 siRNA treatment when compared with TGFß treatment alone. Gene expression analysis showed that Bax/Bcl2, MLKL and LC3 was upregulated in the cells with combinatorial treatment of TGFß and BAP31 siRNA. In addition, BAP31 was shown to regulate multiple signaling pathways, especially for Wnt signaling. It found that BAP31 knockdown cells treated with TGFß decreased ß-catenin cytosolic expression and nuclear localization. Wnt signaling activator BIO could restore the downregulation of proliferation by BAP31 knockdown. This finding suggested that BAP31 regulated cancer cell migration is possibly involved with cell death mechanisms and Wnt signaling.

13.
J Cell Mol Med ; 26(8): 2205-2217, 2022 04.
Article in English | MEDLINE | ID: mdl-35166018

ABSTRACT

The phytoalexin resveratrol exhibits anti-tumour activity in many types of cancer. In this study, we showed that resveratrol suppressed the survival of gastric tumour cells both in vivo and in vitro. Resveratrol promoted apoptosis, autophagy and endoplasmic reticulum (ER) stress in a dose-dependent manner. RNA-seq analysis showed that multiple cell death signalling pathways were activated after resveratrol treatment, while the use of ER stress activators (tunicamycin and thapsigargin) in combinatorial with resveratrol led to further inhibition of cancer cell survival. Results also showed that resveratrol altered the expression of several long non-coding RNAs (lncRNAs), including MEG3, PTTG3P, GAS5, BISPR, MALAT1 and H19. Knockdown of H19 in resveratrol-treated cells further enhanced the effects of resveratrol on apoptosis, ER stress and cell cycle S-phase arrest. Furthermore, the migratory ability of resveratrol-treated cells was dramatically decreased after H19 knockdown. In conclusion, resveratrol inhibited cancer cell survival, while knockdown of lncRNA H19 resulted in increased sensitivity to resveratrol therapy.


Subject(s)
Neoplasms , RNA, Long Noncoding , Resveratrol , Apoptosis , Cell Line, Tumor , Cell Proliferation/genetics , Endoplasmic Reticulum Stress/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Resveratrol/pharmacology
14.
J Clin Lab Anal ; 36(1): e24107, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34871464

ABSTRACT

BACKGROUND: Metabolic disturbance is closely correlated with intrahepatic cholangiocarcinoma (IHCC), and we aimed to identify metabolic gene marker for the prognosis of IHCC. METHODS: We obtained expression and clinical data from 141 patients with IHCC from public databases. Prognostic metabolic genes were selected using univariate Cox regression analysis. Unsupervised cluster analysis was applied to identify IHCC subtypes, and CIBERSORT was used for immune infiltration analysis of different subtypes. Then, the metabolic gene signature was screened using multivariate Cox regression analysis and the LASSO algorithm. The prognostic potential and regulatory network of the metabolic gene signature were further investigated. RESULTS: We screened 228 prognosis-related metabolic genes. Based on their expression levels, IHCC samples were divided into two subtypes, which showed significant differences in survival and immune cell infiltration. After LASSO analysis, eight metabolic genes including CYP19A1, SCD5, ACOT8, SRD5A3, MOGAT2, PFKFB3, PPARGC1B, and RPL17 were identified as the optimal genes for the prognosis signature. The prognostic model had excellent predictive abilities, with areas under the receiver-operating characteristic curves over 0.8. A nomogram model was also established based on two independent prognostic clinical factors (pathologic stage and prognostic model), and the generated calibration curves and c-indexes determined its excellent accuracy and discriminative ability to predict 1- and 5-year survival status (c-indexes>0.7). Finally, we found that miR-26a-5p, miR-27a-3p, and miR-27b-3p were the upstream regulators that mediate the involvement of gene signatures in metabolic pathways. CONCLUSION: We developed eight metabolic gene signatures to predict IHCC prognosis and proposed potential upstream regulatory axes of gene signatures.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Gene Regulatory Networks/genetics , MicroRNAs , Transcriptome/genetics , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Female , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Nomograms , Prognosis
16.
Front Cell Dev Biol ; 9: 733751, 2021.
Article in English | MEDLINE | ID: mdl-34568341

ABSTRACT

Cell death induction has become popular as a novel cancer treatment. Ferroptosis, a newly discovered form of cell death, features regulated, iron-dependent accumulation of lipid hydroperoxides. Since this word "ferroptosis" was coined, numerous studies have examined the complex relationship between ferroptosis and cancer. Here, starting from the intrinsic hallmarks of cancer and cell death, we discuss the theoretical basis of cell death induction as a cancer treatment. We review various aspects of the relationship between ferroptosis and cancer, including the genetic basis, epigenetic modification, cancer stem cells, and the tumor microenvironment, to provide information and support for further research on ferroptosis. We also note that exosomes can be applied in ferroptosis-based therapy. These extracellular vesicles can deliver different molecules to modulate cancer cells and cell death pathways. Using exosomes to control ferroptosis occurring in targeted cells is promising for cancer therapy.

17.
J Cell Mol Med ; 25(15): 7135-7145, 2021 08.
Article in English | MEDLINE | ID: mdl-34164906

ABSTRACT

In recent years, the repurposing of conventional and chemotherapeutic drugs is recognized as an alternative strategy for health care. The main purpose of this study is to strengthen the application of non-oncological drug metformin on breast cancer treatment in the perspective of epigenetics. In the present study, metformin was found to inhibit cell proliferation, promote apoptosis and induce cell cycle arrest in breast cancer cells at a dose-dependent manner. In addition, metformin treatment elevated acH3K9 abundance and decreased acH3K18 level. The expression of lncRNA MALAT1, HOTAIR, DICER1-AS1, LINC01121 and TUG1 was up-regulated by metformin treatment. In metformin-treated cells, MALAT1 knock-down increased the Bax/Bcl2 ratio and enhanced p21 but decreased cyclin B1 expression. The expression of Beclin1, VDAC1, LC3-II, CHOP and Bip was promoted in the cells received combinatorial treatment of metformin and MALAT1 knock-down. The reduced phosphorylation of c-Myc was further decreased in the metformin-treated cells in combination with MALAT1 knock-down than metformin treatment alone. Taken together, these results provide a promising repurposed strategy for metformin on cancer treatment by modulating epigenetic modifiers.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , RNA, Long Noncoding/metabolism , Apoptosis/drug effects , Beclin-1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Female , Humans , MCF-7 Cells , Microtubule-Associated Proteins/metabolism , RNA, Long Noncoding/genetics , Transcription Factor CHOP/metabolism , Voltage-Dependent Anion Channel 1/metabolism
18.
J Cell Mol Med ; 25(3): 1480-1492, 2021 02.
Article in English | MEDLINE | ID: mdl-33369155

ABSTRACT

Melatonin exhibits antitumour activities in the treatment of many human cancers. In the present study, we aimed to improve the therapeutic potential of melatonin in gastric cancer. Our results confirmed that melatonin dose-dependently suppressed the proliferation and necrosis, and increased G0/G1 phase arrest, apoptosis, autophagy and endoplasmic reticulum (ER) stress. The Ras-Raf-MAPK signalling pathway was activated in cells after melatonin treatment. RNA-seq was performed and GSEA analysis further confirmed that many down-regulated genes in melatonin-treated cells were associated with proliferation. However, GSEA analysis also indicated that many pathways related to metastasis were increased after melatonin treatment. Subsequently, combinatorial treatment was conducted to further investigate the therapeutic outcomes of melatonin. A combination of melatonin and thapsigargin increased the apoptotic rate and G0/G1 cell cycle arrest when compared to treatment with melatonin alone. Melatonin in combination with thapsigargin triggered the increased expression of Bip, LC3-II, phospho-Erk1/2 and phospho-p38 MAPK. In addition, STF-083010, an IRE1a inhibitor, further exacerbated the decrease in survival rate induced by combinatorial treatment with melatonin and thapsigargin. Collectively, melatonin was effective in gastric cancer treatment by modifying ER stress.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Melatonin/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Profiling , Humans
19.
Front Pharmacol ; 12: 804615, 2021.
Article in English | MEDLINE | ID: mdl-35153759

ABSTRACT

Magnesium, an essential mineral micronutrient, plays a role in the activation of various transporters and enzymes. The present study aimed to investigate the possibility of applying magnesium to enhance the efficacy of cisplatin which is still ranked as one of the major chemotherapeutic drugs for bladder cancer patients. Results showed that the survival rate and colony formation of bladder cancer cells were reduced by combinatorial treatment with cisplatin and magnesium chloride (MgCl2). The proportion of apoptotic cells was also increased in UC3 bladder cancer cells treated with a combination of cisplatin and MgCl2. Most importantly, a marked decrease in nuclear ß-catenin was observed in cells that received cisplatin treatment. In addition, the nuclear ß-catenin in cisplatin treated cells was further down-regulated by supplementing MgCl2. 6-bromoindirubin-3'-oxime (BIO), an inhibitor of glycogen synthase kinase-3 (GSK-3) that activates the Wnt/ß-catenin signaling pathway by modulating ß-catenin activity, was thus applied to further exploit the role of this signaling pathway in magnesium aided cancer treatment. The survival rate of bladder cancer cells was decreased by BIO treatment at concentrations of 1.0, 2.5 and 5.0 µM accompanied by increased ß-catenin expression. However, the expression of ß-catenin in MgCl2-treated cells was lower than in untreated cells under the same BIO concentration. The expression of cleaved caspase-3, cleaved caspase-9 and microtubule-associated protein 1 light chain 3- II (LC3-II) was highest in cells treated with MgCl2 and 5.0 µM BIO among the examined groups. Our findings reveal that magnesium could contribute to cisplatin-based chemotherapy by moderately regulating the Wnt/ß-catenin signaling pathway.

20.
Methods Mol Biol ; 2239: 1-18, 2021.
Article in English | MEDLINE | ID: mdl-33226609

ABSTRACT

Porcine cloning technology can be used to produce progenies genetically identical to the donor cells from high-quality breeding pigs. In addition, genetically modified pigs have been produced by somatic cell nuclear transfer using genetically modified porcine fetal fibroblasts. The method of preparing genetically modified pigs is critical for establishing pig models for human diseases, and for generating donor animals for future xenotransplantation. This chapter describes detailed procedures for generating cloned pigs using fetal fibroblasts as nuclear donors.


Subject(s)
Cloning, Organism/methods , Embryo Transfer/methods , Fetus/cytology , Fibroblasts/cytology , Nuclear Transfer Techniques , Oocytes/cytology , Animals , Animals, Genetically Modified , Cells, Cultured , Cloning, Organism/instrumentation , Cryopreservation/methods , Female , Pregnancy , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...