Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Materials (Basel) ; 17(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38893752

ABSTRACT

Antibiotic residues have been found to have potentially harmful effects on ecological and human health. Carbon nitride-based photocatalysts have widely focused on antibiotic photocatalytic degradation. Herein, we prepared Fe-modified g-C3N4 nanorod bunches (FCNBs) using chemical vapor co-deposition. Specifically, through the process of calcination, a blend of urea and chlorophyllin sodium iron salt underwent an intriguing transformation, resulting in the integration of Fe into the framework of the g-C3N4 nanorod cluster. The resulting photocatalyst exhibited remarkable stability and superior dispersibility. The prepared FCNBs had a unique structure, which was beneficial for increasing light absorption. Furthermore, the Fe species formed a chemical coordination with the g-C3N4 matrix, thereby altering the electronic structure of the matrix. This modification facilitated charge transfer, prolonged the carrier lifetime, and enhanced light absorption, all of which significantly increased the photocatalytic activity. The oxytetracycline degradation efficiency of FCNBs was 82.5%, and they demonstrated outstanding stability in cycle trials. This work introduces a promising photocatalyst for the degradation of antibiotics.

2.
Genomics ; 115(5): 110704, 2023 09.
Article in English | MEDLINE | ID: mdl-37678441

ABSTRACT

RNA N6-methyladenosine (m6A) modifications influence gastrointestinal stromal tumors (GISTs) development, but the detailed molecular mechanisms have not been fully studied. Here, microRNA-675 was found to be aberrantly elevated in cancerous tissues and cells of GISTs, compared to the corresponding normal counterparts, and GISTs patients with high-expressed microRNA-675 have worse outcomes. Additional experiments confirmed that silencing of microRNA-675 hindered cell division, mobility and tumorigenesis in vitro and in vivo, whereas triggered apoptotic cell death in GISTs cells. Furthermore, microRNA-675-ablation increased the expression levels of myosin phosphatase targeting protein 1 (MYPT1) to inactivate the tumor-initiating RhoA/NF2/YAP1 signal pathway, and downregulation of MYPT1 recovered the malignant phenotypes in microRNA-675-silenced GISTs cells. In addition, we evidenced that METTL3-mediated m6A modifications were essential for sustaining the stability of microRNA-675, and silencing of METTL3 restrained tumorigenesis of GISTs cells by regulating the microRNA-675/MYPT1 axis. To summarize, theMETTL3/m6A/microRNA-675/MYPT1 axis could be used as novel biomarkers for the diagnosis and treatment of GISTs.


Subject(s)
Gastrointestinal Stromal Tumors , MicroRNAs , Humans , Myosin-Light-Chain Phosphatase/genetics , Gastrointestinal Stromal Tumors/genetics , Methyltransferases/genetics , Carcinogenesis/genetics , MicroRNAs/genetics
3.
Cell Biol Toxicol ; 39(5): 1995-2010, 2023 10.
Article in English | MEDLINE | ID: mdl-34822033

ABSTRACT

Cisplatin is the effective chemotherapeutic drug in colon cancer treatment, but its therapeutic efficacy is limited by intrinsic or acquired drug resistance and detrimental side effects. Therefore, improving the effect of cisplatin chemotherapy remains a great challenge. The previous study identified that USP39 was relevant to cisplatin resistance of lung cancer. However, the function and mechanisms of USP39 regulating the chemosensitivity of cisplatin in colorectal cancer remain unclear. In this study, we reveal that USP39 is associated with colon cancer cells sensitivity to cisplatin. Depletion of USP39 enhances the cisplatin-induced apoptosis in HCT116 cells. Conversely, overexpression of USP39 attenuates apoptosis in RKO cells. Furthermore, we demonstrate that USP39 depletion promotes apoptosis induced by cisplatin, which is related with the induction of oxidative stress and DNA damage response. Further studies show that USP39 regulates cisplatin-induced apoptosis dependent on p53. The underlying mechanism is demonstrated by knocking down USP39, that results in p53 upregulation, associated with its prolonged half-life. Collectively, our findings reveal that USP39 might be a negative factor of the p53 mediated cisplatin sensitivity of colon cancer, and suggest USP39 as a potential molecular target for cisplatin chemotherapy of colon cancer.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Lung Neoplasms , Humans , Cisplatin/pharmacology , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Lung Neoplasms/genetics , Apoptosis , Antineoplastic Agents/pharmacology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
4.
BMC Cancer ; 21(1): 460, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33902514

ABSTRACT

BACKGROUND: Gastric cancer is a fatal gastrointestinal cancer with high morbidity and poor prognosis. The dismal 5-year survival rate warrants reliable biomarkers to assess and improve the prognosis of gastric cancer. Distinguishing driver mutations that are required for the cancer phenotype from passenger mutations poses a formidable challenge for cancer genomics. METHODS: We integrated the multi-omics data of 293 primary gastric cancer patients from The Cancer Genome Atlas (TCGA) to identify key driver genes by establishing a prognostic model of the patients. Analyzing both copy number alteration and somatic mutation data helped us to comprehensively reveal molecular markers of genomic variation. Integrating the transcription level of genes provided a unique perspective for us to discover dysregulated factors in transcriptional regulation. RESULTS: We comprehensively identified 31 molecular markers of genomic variation. For instance, the copy number alteration of WASHC5 (also known as KIAA0196) frequently occurred in gastric cancer patients, which cannot be discovered using traditional methods based on significant mutations. Furthermore, we revealed that several dysregulation factors played a hub regulatory role in the process of biological metabolism based on dysregulation networks. Cancer hallmark and functional enrichment analysis showed that these key driver (KD) genes played a vital role in regulating programmed cell death. The drug response patterns and transcriptional signatures of KD genes reflected their clinical application value. CONCLUSIONS: These findings indicated that KD genes could serve as novel prognostic biomarkers for further research on the pathogenesis of gastric cancers. Our study elucidated a multidimensional and comprehensive genomic landscape and highlighted the molecular complexity of GC.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , Mutation , Proteins/genetics , Stomach Neoplasms/genetics , ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents/pharmacology , Apoptosis/genetics , DNA Copy Number Variations , Databases, Genetic , Gene Dosage , Genetic Markers , Genomics , Humans , Membrane Proteins/genetics , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/genetics , Prognosis , Proteasome Endopeptidase Complex/genetics , Stomach Neoplasms/drug therapy , Transcriptome
5.
Mol Carcinog ; 60(4): 265-278, 2021 04.
Article in English | MEDLINE | ID: mdl-33634905

ABSTRACT

Ubiquitin-specific protease 39 (USP39) is frequently overexpressed in a variety of cancers, and involved in the regulation of various biological processes, such as cell proliferation, cell cycle progression, apoptosis and pre-messenger RNA splicing. Nevertheless, the biological roles and mechanisms of USP39 in colon cancer remain largely unknown. In this study, we analyzed whether USP39 can be a molecular target for the treatment of colon cancer. Whilst overexpression of USP39 was detected in human colon cancer tissues and cell lines, USP39 knockdown was observed to inhibit the growth and subcutaneous tumor formation of colon cancer cells. Further analysis showed that USP39 knockdown can stabilize p21 by prolonging the half-life of p21 and by upregulating the promoter activity of p21. The RS domain and USP domain of USP39 were found to play an essential role. Additionally, our findings revealed that USP39 plays a regulatory role in the proliferation of colon cancer cells by the p53/p21/CDC2/cyclin B1 axis in a p21-dependent manner. Taken together, this study provided the theoretical basis that may facilitate the development of USP39 as a novel potential target of colon cancer therapy.


Subject(s)
Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclin-Dependent Kinase Inhibitor p21/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Promoter Regions, Genetic , Protein Domains , Protein Stability , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases/chemistry , Up-Regulation
6.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255748

ABSTRACT

Ubiquitin-specific protease 39 (USP39), a member of the deubiquitinating enzyme family, has been reported to participate in cytokinesis and metastasis. Previous studies determined that USP39 functions as an oncogenic factor in various types of cancer. Here, we reported that USP39 is frequently overexpressed in human lung cancer tissues and non-small-cell lung cancer (NSCLC) cell lines. USP39 knockdown inhibited the proliferation and colony formation of A549 and HCC827 cells and decreased tumorigenic potential in nude mice. Specifically, knocking down USP39 resulted in cell cycle arrest at G2/M and subsequent apoptosis through the activation of the p53 pathway, including upregulation of p21, cleaved-cas3, cleaved-cas9 and downregulation of CDC2 and CycinB1. Moreover, USP39 knockdown significantly inhibited migration and invasion of A549 and HCC827 cells, also via activation of the p53 pathway, and downregulation of MMP2 and MMP9. Importantly, we verified these results in metastasis models in vivo. Collectively, these results not only establish that USP39 functions as an oncogene in lung cancer, but reveal that USP39 has an essential role in regulating cell proliferation and metastasis via activation of the p53 pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Proteases/genetics , A549 Cells , Animals , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Heterografts , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Mice , Neoplasm Metastasis , Signal Transduction/genetics , Transcriptional Activation/genetics
7.
Int Immunopharmacol ; 81: 106240, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32044657

ABSTRACT

Morroniside and loganin are iridoid glycosides extracted from Cornus officinalis, a plant species widely used in traditional Chinese medicine. However, the anti-inflammatory effects of morroniside and loganin in colitis are barely understood. The aim of the present study was to explore the effects of morroniside and loganin on the dextran sodium sulfate (DSS)-induced murine model of colitis and an LPS-induced colorectal cancer (CRC) cell inflammation model, and to clarify the underlying mechanisms. We found that morroniside and loganin were able to ameliorate clinical features, including disease activity index (DAI), histological inflammation score and periodic acid-Schiff staining (PAS). In the mouse model, morroniside and loganin treatment increased expression of tight junction proteins (TJs) and decreased pro-inflammatory cytokine production. Moreover, our findings showed that the expression of p-STAT3 and p-p65 were suppressed compared to the disease group. In in vitro experiments, treatment with morroniside and loganin had no obvious effects on proliferative activity in HCT116 cells and HIEC-6 cells. Expression of pro-inflammatory cytokines was inhibited by morroniside and loganin treatment in comparison with the LPS-treated group. Taken together, morroniside and loganin have beneficial effects on colitis in vivo and are anti-inflammatory in vitro. Possible mechanisms of the anti-inflammatory response may include blockade of the STAT3/NF-κB pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Colitis/drug therapy , Glycosides/therapeutic use , Iridoid Glycosides/therapeutic use , Iridoids/therapeutic use , Animals , Cell Line , Colitis/chemically induced , Cornus/immunology , Dextran Sulfate , Disease Models, Animal , Humans , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...