Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 52(5): 2711-2723, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38281192

ABSTRACT

Class IIa Histone deacetylases (HDACs), including HDAC4, 5, 7 and 9, play key roles in multiple important developmental and differentiation processes. Recent studies have shown that class IIa HDACs exert their transcriptional repressive function by interacting with tissue-specific transcription factors, such as members of the myocyte enhancer factor 2 (MEF2) family of transcription factors. However, the molecular mechanism is not well understood. In this study, we determined the crystal structure of an HDAC4-MEF2A-DNA complex. This complex adopts a dumbbell-shaped overall architecture, with a 2:4:2 stoichiometry of HDAC4, MEF2A and DNA molecules. In the complex, two HDAC4 molecules form a dimer through the interaction of their glutamine-rich domain (GRD) to form the stem of the 'dumbbell'; while two MEF2A dimers and their cognate DNA molecules are bridged by the HDAC4 dimer. Our structural observations were then validated using biochemical and mutagenesis assays. Further cell-based luciferase reporter gene assays revealed that the dimerization of HDAC4 is crucial in its ability to repress the transcriptional activities of MEF2 proteins. Taken together, our findings not only provide the structural basis for the assembly of the HDAC4-MEF2A-DNA complex but also shed light on the molecular mechanism of HDAC4-mediated long-range gene regulation.


Subject(s)
DNA , Histone Deacetylases , MEF2 Transcription Factors , Repressor Proteins , DNA/chemistry , DNA/metabolism , Gene Expression Regulation , Genes, Reporter , MEF2 Transcription Factors/chemistry , MEF2 Transcription Factors/metabolism , Myogenic Regulatory Factors/chemistry , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Humans , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism
2.
Mol Cell Proteomics ; 22(6): 100567, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37172717

ABSTRACT

Nasopharyngeal carcinoma (NPC), a malignant tumor distinctly characterized by ethnic and geographic distribution, is highly prevalent in Southern China and Southeast Asia. However, the molecular mechanisms of NPC have not been fully revealed at the proteomic level. In this study, 30 primary NPC samples and 22 normal nasopharyngeal epithelial tissues were collected for proteomics analysis, and a relatively complete proteomics landscape of NPC was depicted for the first time. By combining differential expression analysis, differential co-expression analysis, and network analysis, potential biomarkers and therapeutic targets were identified. Some identified targets were verified by biological experiments. We found that 17-AAG, a specific inhibitor of the identified target heat shock protein 90 (HSP90), could be a potential therapeutic drug for NPC. Finally, consensus clustering identified two NPC subtypes with specific molecular features. The subtypes and the related molecules were verified by an independent data set and may have different progression-free survival. The results of this study provide a comprehensive understanding of the proteomics molecular signatures of NPC and provide new perspectives and inspiration for prognostic determination and treatment of NPC.


Subject(s)
Carcinoma , Nasopharyngeal Neoplasms , Humans , Nasopharyngeal Carcinoma , Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Proteomics/methods , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
3.
Proc Natl Acad Sci U S A ; 119(22): e2120633119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35605119

ABSTRACT

Dysregulated epigenetic and transcriptional programming due to abnormalities of transcription factors (TFs) contributes to and sustains the oncogenicity of cancer cells. Here, we unveiled the role of zinc finger protein 280C (ZNF280C), a known DNA damage response protein, as a tumorigenic TF in colorectal cancer (CRC), required for colitis-associated carcinogenesis and Apc deficiency­driven intestinal tumorigenesis in mice. Consistently, ZNF280C silencing in human CRC cells inhibited proliferation, clonogenicity, migration, xenograft growth, and liver metastasis. As a C2H2 (Cys2-His2) zinc finger-containing TF, ZNF280C occupied genomic intervals with both transcriptionally active and repressive states and coincided with CCCTC-binding factor (CTCF) and cohesin binding. Notably, ZNF280C was crucial for the repression program of trimethylation of histone H3 at lysine 27 (H3K27me3)-marked genes and the maintenance of both focal and broad H3K27me3 levels. Mechanistically, ZNF280C counteracted CTCF/cohesin activities and condensed the chromatin environment at the cis elements of certain tumor suppressor genes marked by H3K27me3, at least partially through recruiting the epigenetic repressor structural maintenance of chromosomes flexible hinge domain-containing 1 (SMCHD1). In clinical relevance, ZNF280C was highly expressed in primary CRCs and distant metastases, and a higher ZNF280C level independently predicted worse prognosis of CRC patients. Thus, our study uncovered a contributor with good prognostic value to CRC pathogenesis and also elucidated the essence of DNA-binding TFs in orchestrating the epigenetic programming of gene regulation.


Subject(s)
Chromatin , Colorectal Neoplasms , Epigenetic Repression , CCCTC-Binding Factor/metabolism , Carcinogenesis/genetics , Chromatin/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA-Binding Proteins , Histones/genetics , Histones/metabolism , Humans , Prognosis , Transcription Factors , Zinc Fingers
4.
Bioinformatics ; 39(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36655793

ABSTRACT

MOTIVATION: Side effects of drugs could cause severe health problems and the failure of drug development. Drug-target interactions are the basis for side effect production and are important for side effect prediction. However, the information on the known targets of drugs is incomplete. Furthermore, there could be also some missing data in the existing side effect profile of drugs. As a result, new methods are needed to deal with the missing features and missing labels in the problem of side effect prediction. RESULTS: We propose a novel computational method based on transductive matrix co-completion and leverage the low-rank structure in the side effects and drug-target data. Positive-unlabelled learning is incorporated into the model to handle the impact of unobserved data. We also introduce graph regularization to integrate the drug chemical information for side effect prediction. We collect the data on side effects, drug targets, drug-associated proteins and drug chemical structures to train our model and test its performance for side effect prediction. The experiment results show that our method outperforms several other state-of-the-art methods under different scenarios. The case study and additional analysis illustrate that the proposed method could not only predict the side effects of drugs but also could infer the missing targets of drugs. AVAILABILITY AND IMPLEMENTATION: The data and the code for the proposed method are available at https://github.com/LiangXujun/GTMCC. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Drug-Related Side Effects and Adverse Reactions , Humans , Drug Development , Drug Interactions , Proteins/chemistry
5.
Rheumatology (Oxford) ; 63(1): 251-258, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-37184873

ABSTRACT

OBJECTIVES: Galectin-9, as immune checkpoint protein, plays a role in regulating autoimmunity and tumour immunity. Therefore, we explored the pathophysiological link between galectin-9 and malignancy in cancer-related DM (CRDM). METHODS: Serum galectin-9 were quantified via enzyme-linked immunosorbent assay, and its association with serological indices was evaluated using Spearman analysis. Receiver operating characteristic (ROC) analysis was utilized to determine the cut-off value of galectin-9. RESULTS: Serum levels of galectin-9 were significantly higher in DM patients [23.38 (13.85-32.57) ng/ml] than those in healthy controls (HCs) [6.81 (5.42-7.89) ng/ml, P < 0.0001], and were positively correlated with the cutaneous dermatomyositis disease area severity index activity (CDASI-A) scores (rs=0.3065, P = 0.0172). DM patients with new-onset and untreated cancer (new-CRDM) [31.58 (23.85-38.84) ng/ml] had higher levels of galectin-9 than those with stable and treated cancer (stable-CRDM) [17.49 (10.23-27.91) ng/ml, P = 0.0288], non-cancer-related DM (non-CRDM) [21.05 (11.97-28.02) ng/ml, P = 0.0258], and tumour patients without DM [7.46 (4.90-8.51) ng/ml, P < 0.0001]. Serum galectin-9 levels significantly decreased [27.79 (17.04-41.43) ng/ml vs 13.88 (5.15-20.37) ng/ml, P = 0.002] after anti-cancer treatment in CRDM patients. The combination of serum galectin-9 and anti-transcriptional intermediary factor 1-γ (anti-TIF1-γ) antibody (AUC = 0.889, 95% CI 0.803-0.977) showed the highest predictive value for the presence of cancer in DM. CONCLUSION: Increased galectin-9 levels were related to tumor progression in CRDM, and galectin-9 was downregulated upon cancer treatment. Monitoring serum galectin-9 levels and anti-TIF1-γ antibodies might be an attractive strategy to achieve tumour diagnosis and predict CRDM outcome.


Subject(s)
Dermatomyositis , Neoplasms , Humans , Dermatomyositis/complications , Neoplasms/complications , Galectins , Antibodies , Biomarkers , Autoantibodies
6.
Nucleic Acids Res ; 49(18): 10235-10249, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34551426

ABSTRACT

Forkhead box (FOX) proteins are an evolutionarily conserved family of transcription factors that play numerous regulatory roles in eukaryotes during developmental and adult life. Dysfunction of FOX proteins has been implicated in a variety of human diseases, including cancer, neurodevelopment disorders and genetic diseases. The FOX family members share a highly conserved DNA-binding domain (DBD), which is essential for DNA recognition, binding and function. Since the first FOX structure was resolved in 1993, >30 FOX structures have been reported to date. It is clear now that the structure and DNA recognition mechanisms vary among FOX members; however, a systematic review on this aspect is lacking. In this manuscript, we present an overview of the mechanisms by which FOX transcription factors bind DNA, including protein structures, DNA binding properties and disease-causing mutations. This review should enable a better understanding of FOX family transcription factors for basic researchers and clinicians.


Subject(s)
DNA/metabolism , Forkhead Transcription Factors/metabolism , Neurodevelopmental Disorders/metabolism , Humans , Molecular Structure , Protein Binding
7.
Nucleic Acids Res ; 49(6): 3573-3583, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33577686

ABSTRACT

Forkhead transcription factors bind a canonical consensus DNA motif, RYAAAYA (R = A/G, Y = C/T), as a monomer. However, the molecular mechanisms by which forkhead transcription factors bind DNA as a dimer are not well understood. In this study, we show that FOXO1 recognizes a palindromic DNA element DIV2, and mediates transcriptional regulation. The crystal structure of FOXO1/DIV2 reveals that the FOXO1 DNA binding domain (DBD) binds the DIV2 site as a homodimer. The wing1 region of FOXO1 mediates the dimerization, which enhances FOXO1 DNA binding affinity and complex stability. Further biochemical assays show that FOXO3, FOXM1 and FOXI1 also bind the DIV2 site as homodimer, while FOXC2 can only bind this site as a monomer. Our structural, biochemical and bioinformatics analyses not only provide a novel mechanism by which FOXO1 binds DNA as a homodimer, but also shed light on the target selection of forkhead transcription factors.


Subject(s)
DNA/metabolism , Forkhead Box Protein O1/chemistry , Forkhead Box Protein O1/metabolism , DNA/chemistry , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , Inverted Repeat Sequences , Models, Molecular , Protein Binding , Protein Multimerization , Transcription, Genetic
8.
Int J Mol Sci ; 24(5)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36902201

ABSTRACT

The major cause (more than 90%) of all cancer-related deaths is metastasis, thus its prediction can critically affect the survival rate. Metastases are currently predicted by lymph-node status, tumor size, histopathology and genetic testing; however, all these are not infallible, and obtaining results may require weeks. The identification of new potential prognostic factors will be an important source of risk information for the practicing oncologist, potentially leading to enhanced patient care through the proactive optimization of treatment strategies. Recently, the new mechanobiology-related techniques, independent of genetics, based on the mechanical invasiveness of cancer cells (microfluidic, gel indentation assays, migration assays etc.), demonstrated a high success rate for the detection of tumor cell metastasis propensity. However, they are still far away from clinical implementation due to complexity. Hence, the exploration of novel markers related to the mechanobiological properties of tumor cells may have a direct impact on the prognosis of metastasis. Our concise review deepens our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incites further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit. It may open a new clinical dimension that will improve cancer prognosis and increase the effectiveness of tumor therapies.


Subject(s)
Proteomics , Humans , Neoplasm Invasiveness
9.
Biochem Biophys Res Commun ; 595: 1-6, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35091108

ABSTRACT

Farnesoid X receptor (FXR) is a bile acid-related nuclear receptor and is considered a promising target to treat several liver disorders. Cilofexor is a selective FXR agonist and has already entered phase III trials in primary sclerosing cholangitis (PSC) patients. Pruritis caused by cilofexor treatment is dose dependent. The binding characteristics of cilofexor with FXR and its pruritogenic mechanism remain unclear. In our research, the affinity of cilofexor bound to FXR was detected using an isothermal titration calorimetry (ITC) assay. The binding mechanism between cilofexor and FXR-LBD is explained by the cocrystal structure of the FXR/cilofexor complex. Structural models indicate the possibility that cilofexor activates Mas-related G protein-coupled receptor X4 (MRGPRX4) or G protein-coupled bile acid receptor 1 (GPBAR1), leading to pruritus. In summary, our analyses provide a molecular mechanism of cilofexor binding to FXR and provide a possible explanation for the dose-dependent pruritis of cilofexor.


Subject(s)
Azetidines/chemistry , Isonicotinic Acids/chemistry , Molecular Docking Simulation , Protein Domains , Receptors, Cytoplasmic and Nuclear/chemistry , Azetidines/metabolism , Azetidines/pharmacology , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Binding Sites , Binding, Competitive , Calorimetry/methods , Crystallization , Humans , Hydrogen Bonding , Isonicotinic Acids/metabolism , Isonicotinic Acids/pharmacology , Isoxazoles/chemistry , Isoxazoles/metabolism , Isoxazoles/pharmacology , Ligands , Molecular Structure , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
10.
Biochem Biophys Res Commun ; 598: 15-19, 2022 04 02.
Article in English | MEDLINE | ID: mdl-35151199

ABSTRACT

Ponatinib is a multi-target tyrosine kinase inhibitor that targets ABL, SRC, FGFR, and so on. It was designed to overcome the resistance of BCR-ABL mutation to imatinib, especially the gatekeeper mutation ABLT315I. The molecular mechanism by which ponatinib overcomes mutations of BCR-ABL and some other targets has been explained, but little information is known about the characteristics of ponatinib binding to SRC. Here, we showed that ponatinib inhibited wild type SRC kinase but failed to inhibit SRC gatekeeper mutants in both biochemical and cellular assays. We determined the crystal structure of ponatinib in complex with the SRC kinase domain. In addition, by structural analysis, we provided a possible explanation for why ponatinib showed different effects on SRC and other kinases with gatekeeper mutations. The resistance mechanism of SRC gatekeeper mutations to ponatinib may provide meaningful information for designing inhibitors against SRC family kinases in the future.


Subject(s)
Imidazoles/chemistry , Imidazoles/pharmacology , Protein Kinase Inhibitors/chemistry , Pyridazines/chemistry , Pyridazines/pharmacology , src-Family Kinases/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Imidazoles/metabolism , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-hck/chemistry , Proto-Oncogene Proteins c-hck/metabolism , Pyridazines/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
11.
Biochem Biophys Res Commun ; 605: 9-15, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35306364

ABSTRACT

Fumarates (fumaric acid esters), primarily dimethyl fumarate (DMF) and monoethyl fumarate (MEF) and its salts, are orally administered systemic agents used for the treatment of psoriasis and multiple sclerosis. It is widely believed that the pharmaceutical activities of fumarates are exerted through the Keap1-Nrf2 pathway. Although it has been revealed that DMF and MEF differentially modify specific Keap1 cysteine residues and result in the differential activation of Nrf2, how the modification of DMF and MEF impacts the biochemical properties of Keap1 has not been well characterized. Here, we found that both DMF and MEF can only modify the BTB domain of Keap1 and that only C151 is accessible for covalent binding in vitro. Dynamic fluorescence scanning (DSF) assays showed that the modification of DMF to Keap1 BTB increased its thermal stability, while the modification of MEF dramatically decreased its thermal stability. Further crystal structures revealed no significant conformational variation between the DMF-modified and MEF-modified BTBs. Overall, our biochemical and structural study provides a better understanding of the covalent modification of fumarates to Keap1 and may suggest fundamentally different mechanisms adopted by fumarates in regulating the Keap1-Nrf2 pathway.


Subject(s)
Dimethyl Fumarate , NF-E2-Related Factor 2 , Dimethyl Fumarate/pharmacology , Fumarates/chemistry , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Protein Binding
12.
Cancer Immunol Immunother ; 71(7): 1757-1769, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34854950

ABSTRACT

PURPOSE: Pancreatic adenocarcinoma (PAAD) is one of the most common causes of death among solid tumors, and its pathogenesis remains to be clarified. This study aims to elucidate the value of immune/stromal-related genes in the prognosis of PAAD through comprehensive bioinformatics analysis based on the immune microenvironment and validated in Chinese pancreatic cancer patients. METHODS: Gene expression profiles of pancreatic cancer patients were obtained from TCGA database. Differentially expressed genes (DEGs) were identified based on the ESTIMATE algorithm. Gene co-expression networks were constructed using WGCNA. In the key module, survival analysis was used to reveal the prognostic value. Subsequently, we performed functional enrichment analysis to construct a protein-protein interaction (PPI) network. The relationship between tumor immune infiltration and hub genes was analyzed by TIMER and CIBERSORT. Finally, it was validated in the GEO database and in tissues of Chinese pancreatic cancer patients. RESULTS: In the TCGA pancreatic cancer cohort, a low immune/stromal score was associated with a good prognosis. After bioinformatic analysis, 57 genes were identified to be significantly associated with pancreatic cancer prognosis. Among them, up-regulation of four genes (COL6A3, PLAU, MMP11 and MMP14) indicated poor prognosis and was associated with multiple immune cell infiltration. IHC results showed that PLAU protein levels from Chinese pancreatic cancer tissues were significantly higher than those from adjacent non-tumor tissues and were also associated with tumor TNM stage and lymph node metastasis. CONCLUSION: In conclusion, this study demonstrates that PLAU may serve as a new diagnostic and therapeutic target, which is highly expressed in Chinese pancreatic cancer tissues and associated with lymph node metastasis.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prognosis , Tumor Microenvironment/genetics , Pancreatic Neoplasms
13.
Rheumatology (Oxford) ; 61(5): 2176-2184, 2022 05 05.
Article in English | MEDLINE | ID: mdl-34508564

ABSTRACT

OBJECTIVES: DM and clinically amyopathic DM (CADM) patients with positive expression of anti-transcription intermediary factor 1-γ (anti-TIF1-γ) antibody (Ab) are characterized by distinct clinicopathological features. We aimed to determine the role of cytokine/chemokine profiles in the classification of anti-TIF1-γ positive DM/CADM patients. METHODS: Serum levels of 24 cytokines/chemokines were measured in 27 anti-TIF1-γ positive DM/CADM patients by a Luminex 200 system. Principal components analysis and unsupervised hierarchical clustering were used to reduce variables and establish patient subgroups. Spearman's correlation coefficient was calculated between cytokine/chemokine levels and disease activity markers. RESULTS: Among anti-TIF1-γ positive DM/CADM patients, two distinct patient clusters were identified. The diagnosis of CADM was more common in cluster 1 than in cluster 2 (58.3% vs 6.7%, P = 0.008). Skin disease activity was higher in cluster 2 than in cluster 1 as measured by Cutaneous DM Disease Area and Severity Index-Activity [38.6 (10.4) vs 25.3 (10.0), P = 0.003]. Patients within cluster 2 exhibited significant muscle weakness (Medical Research Council scale ≤ 3, 33.3% vs 0.0%, P = 0.047), higher levels of anti-TIF1-γ Ab [92.4 (20.6) vs 66.9 (13.9), P = 0.001] and an increased malignancy rate (73.3% vs 25.0%, P = 0.021). Cluster 2 exhibited higher serum levels of CXCL10 [564.2 (258.8) vs 122.0 (97.8), P < 0.001], CCL2 [1136.6 (545.4) vs 441.6 (163.3), P < 0.001], galectin-9 [38879.6 (20009.3) vs 12612.4 (6640.0), P < 0.001], IL-18 [436.1 (188.9) vs 243.0 (114.5), P = 0.003], TNF-α [9.3 (3.8) vs 5.6 (2.4), P = 0.007] and TNFRI [1385.1 (338.2) vs 2605.6 (928.5), P < 0.001] than cluster 1. CONCLUSION: In anti-TIF1-γ positive DM/CADM, we identified a 'skin-predominant' cluster and a 'hyperinflammation' cluster based on the cytokine/chemokine profiles.Cytokine/chemokine profiles in anti-TIF1-γ positive DM/CADM can identify discrete clusters of patients with different disease patterns, organ involvements and clinical outcomes.


Subject(s)
Dermatomyositis , Neoplasms , Autoantibodies , Chemokines , Cytokines , Humans
14.
World J Surg Oncol ; 20(1): 118, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35413978

ABSTRACT

BACKGROUND: Colon adenocarcinoma (COAD) is one of the most common malignant tumors and is diagnosed at an advanced stage with a poor prognosis worldwide. Pyroptosis is involved in the initiation and progression of tumors. This research focused on constructing a pyroptosis-related ceRNA network to generate a reliable risk model for risk prediction and immune infiltration analysis of COAD. METHODS: Transcriptome data, miRNA-sequencing data, and clinical information were downloaded from the TCGA database. First, differentially expressed mRNAs (DEmRNAs), miRNAs (DEmiRNAs), and lncRNAs (DElncRNAs) were identified to construct a pyroptosis-related ceRNA network. Second, a pyroptosis-related lncRNA risk model was developed applying univariate Cox regression analysis and least absolute shrinkage and selection operator method (LASSO) regression analysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses were utilized to functionally annotate RNAs contained in the ceRNA network. In addition, Kaplan-Meier analysis, receiver operating characteristic (ROC) curves, univariate and multivariate Cox regression, and nomogram were applied to validate this risk model. Finally, the relationship of this risk model with immune cells and immune checkpoint blockade (ICB)-related genes was analyzed. RESULTS: A total of 5373 DEmRNAs, 1159 DElncRNAs, and 355 DEmiRNAs were identified. A pyroptosis-related ceRNA regulatory network containing 132 lncRNAs, 7 miRNAs, and 5 mRNAs was constructed, and a ceRNA-based pyroptosis-related risk model including 11 lncRNAs was built. The tumor tissues were classified into high- and low-risk groups according to the median risk score. Kaplan-Meier analysis showed that the high-risk group had a shorter survival time; ROC analysis, independent prognostic analysis, and nomogram further indicated the risk model was a significant independent prognostic factor what had an excellent ability to predict patients' risk. Moreover, immune infiltration analysis indicated that the risk model was related to immune infiltration cells (i.e., B cell naïve, T cell follicular helper, macrophage M1) and ICB-related genes (i.e., PD-1, CTLA4, HAVCR2). CONCLUSIONS: This pyroptosis-related lncRNA risk model possessed good prognostic value, and the ability to predict the outcome of ICB immunotherapy in COAD.


Subject(s)
Colonic Neoplasms , MicroRNAs , Pulmonary Disease, Chronic Obstructive , RNA, Long Noncoding , Biomarkers, Tumor/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Immunity , MicroRNAs/genetics , Prognosis , Pulmonary Disease, Chronic Obstructive/genetics , Pyroptosis , RNA, Long Noncoding/genetics , RNA, Messenger/genetics
15.
World J Surg Oncol ; 20(1): 347, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36258216

ABSTRACT

BACKGROUND: Gelsolin-like capping actin protein (CapG) modulates actin dynamics and actin-based motility with a debatable role in tumorigenic progression. The motility-associated functions and potential molecular mechanisms of CapG in nasopharyngeal carcinoma (NPC) remain unclear. METHODS: CapG expression was detected by immunohistochemistry in a cohort of NPC tissue specimens and by Western blotting assay in a variety of NPC cell lines. Loss of function and gain of function of CapG in scratch wound-healing and transwell assays were performed. Inactivation of Rac1 and ROCK with the specific small molecular inhibitors was applied to evaluate CapG's role in NPC cell motility. GTP-bound Rac1 and phosphorylated-myosin light chain 2 (p-MLC2) were measured in the ectopic CapG overexpressing cells. Finally, CapG-related gene set enrichment analysis was conducted to figure out the significant CapG-associated pathways in NPC. RESULTS: CapG disclosed increased level in the poorly differentiated NPC tissues and highly metastatic cells. Knockdown of CapG reduced NPC cell migration and invasion in vitro, while ectopic CapG overexpression showed the opposite effect. Ectopic overexpression of CapG compensated for the cell motility loss caused by simultaneous inactivation of ROCK and Rac1 or inactivation of ROCK alone. GTP-bound Rac1 weakened, and p-MLC2 increased in the CapG overexpressing cells. Bioinformatics analysis validated a positive correlation of CapG with Rho motility signaling, while Rac1 motility pathway showed no significant relationship. CONCLUSIONS: The present findings highlight the contribution of CapG to NPC cell motility independent of ROCK and Rac1. CapG promotes NPC cell motility at least partly through MLC2 phosphorylation and contradicts with Rac1 activation.


Subject(s)
Actins , Nasopharyngeal Neoplasms , Humans , Actins/metabolism , Nasopharyngeal Carcinoma/genetics , Gelsolin/analysis , Gelsolin/genetics , Gelsolin/metabolism , Cell Line, Tumor , Cell Movement/genetics , Nasopharyngeal Neoplasms/genetics , Guanosine Triphosphate , Gene Expression Regulation, Neoplastic , Microfilament Proteins/metabolism , Nuclear Proteins/genetics
16.
Biochem Biophys Res Commun ; 579: 1-7, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34571387

ABSTRACT

Chronic myeloid leukemia (CML) is a hematologic malignancy originating from BCR-ABL oncogene-transformed hematopoietic stem cells (HSCs) known as leukemia stem cells (LSCs). Therefore, targeting LSCs is of primary importance to eradicate CML. The present study demonstrates that picropodophyllin (PPP) effectively induces apoptosis and inhibits colony formation in CML stem/progenitor cells as well as quiescent CML progenitors resistant to imatinib therapy, while sparing normal hematopoietic cells in vitro. Administration of PPP in vivo markedly diminishes CML stem/progenitor cells in a transgenic mouse model of CML by inhibition of cell proliferation and enhancement of apoptosis in LSK cells, and significantly improves survival of CML mice. Furthermore, PPP treatment preferentially leads to transcriptional activation of p53 in CML but not normal CD34+ cells, upregulation of p53 protein in LSCs-enriched Sca-1+ cells from CML mice, and increased phosphorylation of p53 and upregulation of Bax protein in Ku812 cells. These results suggest that the inhibitory effects of PPP on CML stem/progenitor cells are associated with selective activation of p53 pathway and propose that PPP is a potent agent that selectively targets CML LSCs, and may be of value in the CML therapy.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Podophyllotoxin/analogs & derivatives , Stem Cells/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, CD34/biosynthesis , Apoptosis , Disease Models, Animal , Fetal Blood/metabolism , Fusion Proteins, bcr-abl/metabolism , Humans , In Vitro Techniques , K562 Cells , Mice , Mice, Transgenic , Microtubules/metabolism , Phosphorylation , Podophyllotoxin/chemistry , Signal Transduction , Transcriptional Activation
17.
Biochem Biophys Res Commun ; 534: 1047-1052, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33121679

ABSTRACT

Farnesoid X receptor (FXR) is considered as a potential target for the treatment of several liver disorders such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). Tropifexor is a highly potent and non-steroidal FXR agonist that has progressed into phase II clinical trials in patients with PBC. The clinical trials demonstrate that tropifexor improved serum markers of patients with liver diseases and lower side effect such as pruritus that might be implicated with TGR5 activation. However, the molecular mechanism of the potency and selectivity of tropifexor remains unclear. In this study, the binding affinity of FXR and tropifexor is measured by isothermal titration calorimetry (ITC) assays. The crystal structure of the FXR/tropifexor complex is determined at 2.7 Å resolution to explain the molecular mechanism of tropifexor bound to FXR-LBD. Structural comparison with other FXR/agonists structures reveals the conformational change in the FXR/tropifexor structure. Moreover the structural superposition of TGR5/tropifexor indicates that the steric hindrance between tropifexor and TGR5 might be a possible explanation for the impotency arises of tropifexor to TGR5. Overall, our analyses might provide an insight into the molecular mechanism of tropifexor binding to FXR-LBD and account for the high selectivity of tropifexor for FXR versus TGR5.


Subject(s)
Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Isoxazoles/chemistry , Isoxazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation/drug effects , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism
18.
Hepatology ; 72(5): 1682-1700, 2020 11.
Article in English | MEDLINE | ID: mdl-32080887

ABSTRACT

BACKGROUND AND AIMS: Forkhead box M1 (FOXM1) and nuclear factor kappa B (NF-ĸB) are oncogenic drivers in liver cancer that positively regulate each other. We showed that methionine adenosyltransferase 1A (MAT1A) is a tumor suppressor in the liver and inhibits NF-ĸB activity. Here, we examined the interplay between FOXM1/NF-κB and MAT1A in liver cancer. APPROACH AND RESULTS: We examined gene and protein expression, effects on promoter activities and binding of proteins to promoter regions, as well as effects of FOXM1 inhibitors T0901317 (T0) and forkhead domain inhibitory-6 (FDI-6) in vitro and in xenograft and syngeneic models of liver cancer. We found, in both hepatocellular carcinoma and cholangiocarcinoma, that an induction in FOXM1 and NF-κB expression is accompanied by a fall in MATα1 (protein encoded by MAT1A). The Cancer Genome Atlas data set confirmed the inverse correlation between FOXM1 and MAT1A. Interestingly, FOXM1 directly interacts with MATα1 and they negatively regulate each other. In contrast, FOXM1 positively regulates p50 and p65 expression through MATα1, given that the effect is lost in its absence. FOXM1, MATα1, and NF-κB all bind to the FOX binding sites in the FOXM1 and MAT1A promoters. However, binding of FOXM1 and NF-κB repressed MAT1A promoter activity, but activated the FOXM1 promoter. In contrast, binding of MATα1 repressed the FOXM1 promoter. MATα1 also binds and represses the NF-κB element in the presence of p65 or p50. Inhibiting FOXM1 with either T0 or FDI-6 inhibited liver cancer cell growth in vitro and in vivo. However, inhibiting FOXM1 had minimal effects in liver cancer cells that do not express MAT1A. CONCLUSIONS: We have found a crosstalk between FOXM1/NF-κB and MAT1A. Up-regulation in FOXM1 lowers MAT1A, but raises NF-κB, expression, and this is a feed-forward loop that enhances tumorigenesis.


Subject(s)
Forkhead Box Protein M1/metabolism , Liver Neoplasms/genetics , Methionine Adenosyltransferase/genetics , NF-kappa B/genetics , Tumor Suppressor Proteins/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Datasets as Topic , Feedback, Physiological/drug effects , Forkhead Box Protein M1/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Hepatocytes , Humans , Hydrocarbons, Fluorinated/administration & dosage , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Methionine Adenosyltransferase/metabolism , Mice , Mice, Knockout , Primary Cell Culture , Promoter Regions, Genetic/genetics , Pyridines/administration & dosage , S-Adenosylmethionine/metabolism , Sulfonamides/administration & dosage , Thiophenes/administration & dosage , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
19.
Bioorg Med Chem Lett ; 34: 127757, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33359446

ABSTRACT

Ibrutinib is a BTK-targeted irreversible inhibitor. In this study, we demonstrate that ibrutinib potently inhibits SRC activity in a non-covalent manner via mass spectrometry and crystallography. The S345C mutation renders SRC to bind covalently with ibrutinib, and restores the potency of ibrutinib against the gatekeeper mutant. The co-crystal structure of ibrutinib/SRC shows Ser345 of SRC did not form covalent bond with ibrutinib, leading to a decrease of potency and loss of the ability to overcome the gatekeeper mutation of SRC. The X-ray crystallographic studies also provide structural insight into why ibrutinib behaves differently against gatekeeper mutants of different kinases.


Subject(s)
Adenine/analogs & derivatives , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , Adenine/chemistry , Adenine/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Piperidines/chemistry , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , src-Family Kinases/metabolism
20.
Bioorg Med Chem ; 50: 116457, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34670167

ABSTRACT

An aromatic trifluoromethyl ketone moiety was characterized as a new warhead for covalently reversible kinase inhibitor design to target the non-catalytic cysteine residue. Potent and selective covalently reversible inhibitors of FGFR4 kinase were successfully designed and synthesized by utilizing this new warhead. The binding mode of a representative inhibitor was fully characterized by using multiple technologies including MALDI-TOF mass spectrometry, dialysis assay and X-ray crystallographic studies etc. This functional group was also successfully applied to discovery of a new JAK3 inhibitor, suggesting its potential application in designing other kinase inhibitors.


Subject(s)
Hydrocarbons, Fluorinated/pharmacology , Janus Kinase 3/antagonists & inhibitors , Ketones/pharmacology , Protein Kinase Inhibitors/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Janus Kinase 3/metabolism , Ketones/chemical synthesis , Ketones/chemistry , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL