Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 50(2): e9, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34718755

ABSTRACT

Epigenetic therapy has significant potential for cancer treatment. However, few small potent molecules have been identified against DNA or RNA modification regulatory proteins. Current approaches for activity detection of DNA/RNA methyltransferases and demethylases are time-consuming and labor-intensive, making it difficult to subject them to high-throughput screening. Here, we developed a fluorescence polarization-based 'High-Throughput Methyl Reading' (HTMR) assay to implement large-scale compound screening for DNA/RNA methyltransferases and demethylases-DNMTs, TETs, ALKBH5 and METTL3/METTL14. This assay is simple to perform in a mix-and-read manner by adding the methyl-binding proteins MBD1 or YTHDF1. The proteins can be used to distinguish FAM-labelled substrates or product oligonucleotides with different methylation statuses catalyzed by enzymes. Therefore, the extent of the enzymatic reactions can be coupled with the variation of FP binding signals. Furthermore, this assay can be effectively used to conduct a cofactor competition study. Based on the assay, we identified two natural products as candidate compounds for DNMT1 and ALKBH5. In summary, this study outlines a powerful homogeneous approach for high-throughput screening and evaluating enzymatic activity for DNA/RNA methyltransferases and demethylases that is cheap, easy, quick, and highly sensitive.


Subject(s)
DNA Modification Methylases/metabolism , Drug Discovery/methods , High-Throughput Screening Assays/methods , Methyltransferases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Carrier Proteins/metabolism , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/standards , Humans , Methyltransferases/antagonists & inhibitors , Nucleotides/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , RNA/metabolism
2.
Article in English | MEDLINE | ID: mdl-37103434

ABSTRACT

A Gram-stain-negative, aerobic, rod-shaped, non-gliding bacterial strain, designated as MT39T, was isolated from a deep-sea sediment sample collected from the Mariana Trench. Strain MT39T grew optimally at 35°C and pH 7.0, and could tolerate up to 10% (w/v) NaCl. The strain was positive for catalase and negative for oxidase. The genome of strain MT39T was 4 033 307 bp, with a 41.1 mol % genomic G+C content and 3514 coding sequences. Phylogenetic analysis based on 16S rRNA gene sequences placed strain MT39T within the genus Salinimicrobium, showing the highest 16S rRNA gene sequence similarity to Salinimicrobium terrea CGMCC 1.6308T (98.1%). The average nucleotide identity and in silico DNA-DNA hybridization values between strain MT39T and the type strains of seven Salinimicrobium species were all less than the threshold values to discriminate bacterial species, indicating that strain MT39T is affiliated with a novel species within the genus. The major cellular fatty acids of strain MT39T were iso-C15 : 0, anteiso-C15 : 0 and iso-C17 : 0 3-OH. Polar lipids of strain MT39T included phosphatidylethanolamine, one unidentified aminolipid and four unidentified lipids. Menaquinone-6 was the only respiratory quinone in strain MT39T. On the basis of the polyphasic data present in this study, strain MT39T represents a novel species of the genus Salinimicrobium, for which the name Salinimicrobium profundisediminis sp. nov. is proposed, with type strain being MT39T (=MCCC 1K07832T=KCTC 92381T).


Subject(s)
Fatty Acids , Flavobacteriaceae , Fatty Acids/chemistry , Geologic Sediments/microbiology , Seawater/microbiology , Phospholipids/chemistry , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Base Composition , Bacterial Typing Techniques , DNA, Bacterial/genetics , Vitamin K 2/chemistry
3.
Bioorg Chem ; 139: 106726, 2023 10.
Article in English | MEDLINE | ID: mdl-37451145

ABSTRACT

O-GlcNAcylation is a specific type of post-translational glycosylation modification, which is regulated by two enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Aberrant overexpression of OGT is associated with the development of many solid tumors. In this study, we have developed and optimized a sensitive Homogeneous Time-Resolved Fluorescence (HTRF) assay then identified a novel OGT inhibitor CDDO (also called Bardoxolone) through a high-throughput screening (HTS) based on HTRF assay. Further characterization suggested that CDDO is an effective OGT inhibitor with an IC50 value of 6.56 ± 1.69 µM. CPMG-NMR analysis confirmed that CDDO is a direct binder of OGT with a binding affinity (Kd) of approximately 1.7 µM determined by the MST analysis. Moreover, HDX-MS analysis indicated that CDDO binds to the TPR domain and N-Terminal domain of OGT, which was further confirmed by the enzymatic competition experiments as the binding of CDDO to OGT was not affected by the catalytic site binding inhibitor OSMI-4. Our docking modeling analysis further predicted the possible interactions between CDDO and OGT, providing informative molecular basis for further optimization of the inhibitor in the future. Together, our results suggested CDDO is a new inhibitor of OGT with a distinct binding pocket from the reported OGT inhibitors. Our work paved a new direction for developing OGT inhibitors driven by novel mechanisms.


Subject(s)
High-Throughput Screening Assays , Protein Processing, Post-Translational , Glycosylation
4.
Bioorg Chem ; 134: 106453, 2023 05.
Article in English | MEDLINE | ID: mdl-36898211

ABSTRACT

Chromatin remodeling regulates many basic cellular processes, such as gene transcription, DNA repair, and programmed cell death. As the largest member of nucleosome remodeling factor (NURF), BPTF plays a vital role in the occurrence and development of cancer. Currently, BPTF bromodomain inhibitors are still in development. In this study, by conducting homogenous time-resolved fluorescence resonance energy transfer (HTRF) assay, we identified a potential, novel BPTF inhibitor scaffold Sanguinarine chloride with the IC50 value of 344.2 ± 25.1 nM. Biochemical analysis revealed that compound Sanguinarine chloride exhibited high binding affinity to the BPTF bromodomain. Molecular docking predicted the binding mode of Sanguinarine chloride and elucidated the activities of its derivatives. Moreover, Sanguinarine chloride showed a potent anti-proliferative effect in MIAPaCa-2 cells and inhibited the expression of BPTF target gene c-Myc. Taken together, Sanguinarine chloride provides a qualified chemical tool for developing potent BPTF bromodomain inhibitors.


Subject(s)
Neoplasms , Transcription Factors , Humans , Transcription Factors/metabolism , Molecular Docking Simulation , Chromatin Assembly and Disassembly
5.
Bioorg Chem ; 129: 106128, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36113266

ABSTRACT

A series of novel 2-aminopyridine derivatives 1-26 have been designed and synthesized by structural modifications on a lead USP7 inhibitor, GNE6640. All the compounds were evaluated for their USP7 inhibitory activities. The results showed that most of the compounds have good USP7 inhibitory activities at the concentration of 50 µM. Among them, compounds 7, 14 and 21 are the most potential ones from each category with the IC50 values of 7.6 ± 0.1 µM, 17.0 ± 0.2 µM and 11.6 ± 0.5 µM, respectively. Compounds 7 and 21 expressed significant binding interactions with USP7 by surface plasmon resonance (SPR)-based binding assay, but both of them presented moderate antiproliferative activities against HCT116 cells. They could effectively promote MDM2 degradation, p53 stabilization and p21 gene expression in the western blot analysis.


Subject(s)
Aminopyridines , Antineoplastic Agents , Humans , Ubiquitin-Specific Peptidase 7/metabolism , Aminopyridines/pharmacology , HCT116 Cells , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemistry , Cell Proliferation , Cell Line, Tumor
6.
Bioorg Chem ; 127: 106016, 2022 10.
Article in English | MEDLINE | ID: mdl-35841671

ABSTRACT

A series of novel benzimidazole-iminosugars linked a (substuituted) phenyl group on benzene ring of benzimidazole 5(a-p) and 6(a-p) have been rationally designed and conveniently synthesized through Suzuki coupling reaction in high yields. All compounds have been evaluated for their inhibitory activities against ß-glucosidase (almond). Six compounds 5d, 6d, 6e, 6i, 6n, and 6p showed more significant inhibitory activities with IC50 values in the range of 0.03-0.08 µM, almost 10-fold improved than that of the parent analogue 4, and much higher than that of the positive control castanospermine. The additional phenyl ring and the electron donating groups on it would be beneficial for the activity. Compounds 6d, 6n, and 4 had been chosen to be tested for their inhibition types against ß-glucosidase. Interestingly, three compounds have different inhibition types although they had very similar structure. Their Ki values were calculated to be 0.02 ± 0.01 µM, 0.02 ± 0.01 µM, and 0.66 ± 0.14 µM, respectively. The equilibrium dissociation constant (KD) for 6d, 6n, and 4 and ß-glucosidase was 0.04 µM, 0.03 µM and 0.45 µM by the ITC-based assay, respectively. Molecular docking work suggests that such benzimidazole-iminosugars derivatives might bind to the active site of ß-glucosidase mainly through hydrogen bonds, the additional phenyl ring towards the solvent-exposed region played an important effect on their inhibitory activity against ß-glucosidase.


Subject(s)
Benzimidazoles , beta-Glucosidase , Benzimidazoles/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , alpha-Glucosidases/metabolism , beta-Glucosidase/metabolism
7.
Microb Pathog ; 142: 104100, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32109570

ABSTRACT

In this study we generated and characterized a series of monoclonal antibodies (mAbs) against GII.6 norovirus (NoV) virus like particles (VLPs). Mice were immunized with purified GII.6 NoV VLPs and peptide-bovine serum albumin (BSA) conjugates with the peptide sequence (31 aa) derived from the trypsin cleavage region. An indirect enzyme-linked immunosorbent assay was used to identify positive cell clones during cloning and subcloning, and an in vitro VLP-histo-blood group antigens (HBGAs) binding blockade assay was used to identify mAbs with blocking ability. A total of seven mAbs comprising five (1F7, 1F11, 2B6, 2C4, and 2E10) reactive with major capsid proteins (VP1) and two (1E5 and 2B2) reactive with both VP1 proteins and the peptide were identified. mAb 1F7, 1F11, and 2B6 were identified as blocking antibodies. Sandwich ELISA indicated that all these mAbs recognized soluble GII.6 NoV VLPs. Cross-reactivities with GI.7, GII.3, and GII.4 NoV VLPs were observed in indirect and sandwich ELISA. Western blot analysis indicated that all non-blocking mAbs recognized denatured GII.6 VP1 proteins and blocking mAbs only recognized non-denatured proteins. The in vitro VLP-HBGA binding blockade assay indicated that the three blocking antibodies exhibited blocking effects against GII.6 NoV VLPs, but not GI.7, GII.3, and GII.4 NoV VLPs. Epitope mapping and HBGA blocking assay indicated that mAbs targeting the predicted surface-exposed loop region did not have blocking effects, suggesting a possible important role of this region in regulating NoV-HBGA interactions. This is the first report regarding the characterization of mAbs with blocking ability against GII.6 NoV VLPs. These mAbs might be useful in facilitating our understanding of this group of viruses.

8.
Virus Genes ; 55(3): 280-289, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30725444

ABSTRACT

Noroviruses are leading cause of acute gastroenteritis worldwide. In our previous study, we established an in vitro histo-blood group antigens (HBGAs) binding blockade assay against GII.3 Norovirus virus like particles (VLPs) with trypsin digestion. In this study, we characterized the blocking antibody binding site and epitope type (linear or conformational) by using hyperimmune sera produced against different antigens. VP1 from Jingzhou402 (GII.3, JZ402) strain was expressed by using pGEX-6p-1 expression vector and the insoluble proteins were purified for immunization in rabbit. Previously characterized chimeric VP1-assembled VLPs (GII.4-VP1/GII.3-P2) were used to immunize guinea pig. Peptides reactive with hyperimmune serum against VLPs derived from the VP1 of JZ402 strain were conjugated with BSA and used to immunize rabbits. Hyperimmune sera against above antigens and JZ402 and JZ403 strain-derived VLPs were used to compare their HBGAs blocking effects. Rabbit anti-GST-VP1 and BSA-peptide conjugated hyperimmune sera demonstrated no blocking effects against the binding of GII.3 and GII.4 NoV VLPs to salivary HBGAs. Guinea pig anti-GII.4-VP1/GII.3-P2 hyperimmune serum blocked the binding of trypsin cleaved GII.3 VLPs to salivary HBGAs with no or very weak blocking effects against the binding of GII.4 VLPs to salivary HBGAs. Our data indicated that HBGAs blocking antibodies primarily bound the P2 domain of GII.3 NoV VP1 and their binding epitopes were most probably conformation-dependent.


Subject(s)
Blood Group Antigens/genetics , Epitopes/genetics , Gastroenteritis/genetics , Norovirus/genetics , Animals , Antibodies/genetics , Antibodies/immunology , Binding Sites/immunology , Blood Group Antigens/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Epitopes/immunology , Gastroenteritis/immunology , Gastroenteritis/virology , Genotype , Guinea Pigs , Humans , Norovirus/immunology , Protein Binding , Rabbits , Signal Transduction/genetics
9.
J Med Virol ; 90(4): 671-676, 2018 04.
Article in English | MEDLINE | ID: mdl-29236287

ABSTRACT

Noroviruses (NoVs) are increasingly recognized as the leading cause of acute non-bacterial gastroenteritis worldwide. To screen for NoV-specific monoclonal antibodies (mAbs) with wide spectrum binding activities that could be used for the development of NoV-related detection reagents, we immunized mice with a combination of virus like particles (VLPs) derived from eight different genotypes (two from genogroup I and six from genogroup II), of which two (GI.7 and GII.2) were newly produced VLPs. Indirect enzyme-linked immunosorbent assay (ELISA) confirmed that two mAbs (8D8 and 10B11) bound to all eight major capsid proteins (VP1) with varied binding abilities. Epitope mapping using short peptides covering the N-terminal half of GII.3 VP1 indicated that the binding site of mAb 8D8 was located between amino acid 31 and 60. Multiple amino acid sequence alignment of VP1 suggested that this site harbors conservative sequences across all genogroups. Indirect and sandwich ELISA indicated that mAb 8D8 was unable bind intact VLPs. In summary, we successfully produced GI.7 and GII.2 VLPs using recombinant baculovirus expression system and a cross-reactive mAb by immunizing mice with eight different VLPs that might be useful in the studying and detecting NoVs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Norovirus/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Baculoviridae/genetics , Binding Sites , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Genetic Vectors , Genotype , Mice, Inbred BALB C , Norovirus/genetics , Protein Binding , Virosomes/genetics , Virosomes/immunology
10.
Arch Virol ; 163(12): 3265-3273, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30143876

ABSTRACT

GII.3 and GII.6 noroviruses (NoVs) are similar in several aspects, including the presence of a short sequence insertion in the P2 domain of the major capsid protein (VP1) and trypsin susceptibility of VP1-containing virus-like particles (VLPs). In this study, we generated two constructs with the S or P domains of VP1 from GII.3 and GII.6 NoV strains exchanged (GII.3S/GII.6P and GII.6S/GII.3P), and the resultant chimeric capsid proteins were expressed from recombinant baculoviruses. The assembly of VLPs was confirmed by electron microscopy, and the susceptibility of assembled VLPs to trypsin digestion was analyzed by SDS-PAGE. Salivary histo-blood group antigen (HBGA) binding and binding blockade assays were performed to determine the binding characteristics of chimeric VP1-containing VLPs with and without trypsin digestion. Our results indicated that both expressed GII.3S/GII.6P and GII.6S/GII.3P chimeric proteins successfully assembled into VLPs. Trypsin digestion of VLPs assembled from both chimeric proteins led to the generation of two fragments with molecular sizes similar to those of wild-type VP1-containing VLPs. An in vitro salivary HBGA binding assay demonstrated that VLPs assembled from both chimeric proteins exhibited enhanced binding after trypsin cleavage. An HBGA binding blockade assay indicated that the binding of GII.3S/GII.6P and GII.6S/GII.3P VLPs against salivary HBGAs could only be blocked by GII.3 and GII.6 NoV VLP-specific hyperimmune sera, respectively. For GII.6 and GII.3S/GII.6P VLPs, a difference in binding enhancement after trypsin cleavage was observed. Our results demonstrate that the S domains of GII.3 and GII.6 NoV VP1 are interchangeable and that the S domain affects the binding of the P domain to HBGAs.


Subject(s)
Blood Group Antigens/metabolism , Caliciviridae Infections/metabolism , Caliciviridae Infections/virology , Capsid Proteins/metabolism , Norovirus/metabolism , Caliciviridae Infections/genetics , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Gastroenteritis/genetics , Gastroenteritis/metabolism , Gastroenteritis/virology , Genotype , Humans , Norovirus/chemistry , Norovirus/genetics , Norovirus/ultrastructure , Protein Binding , Protein Domains , Trypsin/chemistry
11.
Future Oncol ; 14(8): 719-726, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29336610

ABSTRACT

AIM: Though lutein can inhibit cancer cell proliferation via alleviating oxidative injury, the molecular mechanisms of lutein involvement in the NrF2/antioxidant response element (ARE) and NF-κB pathways remain poorly understood. MATERIALS & METHODS: MTT, flow cytometry, quantitative real-time PCR (qRT-PCR) and western blot assays were performed. RESULTS: After treatment with lutein, breast cancer cell proliferation was significantly decreased in a dose-dependent manner. Lutein induced nuclear translocation and protein expression of NrF2, improved the expression of cellular antioxidant enzymes and attenuated reactive oxygen species levels. Moreover, lutein treatment decreased NF-κB signaling pathway related NF-κB p65 protein expression. CONCLUSION: The effect of lutein antiproliferation was mediated by activation of the NrF2/ARE pathway, and blocking of the NF-κB signaling pathway.


Subject(s)
Breast Neoplasms/drug therapy , Lutein/pharmacology , NF-E2-Related Factor 2/genetics , Transcription Factor RelA/genetics , Antioxidant Response Elements/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
12.
Arch Virol ; 162(12): 3863-3868, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28866835

ABSTRACT

In this study, we successfully produced VLPs derived from full-length or chimeric VP1 of a documented GII.6 strain. Trypsin digestion of purified VLPs led to total cleavage of VP1, while the integrity of assembled VLPs was not affected. In vitro VLP-histo-blood group antigen (HBGA) binding and binding blockade assays indicated that trypsin digestion enhanced the binding of GII.6 VLPs to salivary HBGAs and that this binding could only be blocked by serum produced against a homologous strain. The data regarding the assembly, morphology and binding patterns of GII.6 NoV VLPs presented here might be useful for further study of GII.6 NoVs.


Subject(s)
Capsid Proteins/genetics , Norovirus/genetics , Virosomes/genetics , Virosomes/metabolism , Blood Group Antigens/metabolism , Humans , Protein Multimerization , Virosomes/ultrastructure , Virus Assembly , Virus Attachment
13.
Virus Genes ; 52(5): 613-9, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27234312

ABSTRACT

Genogroup II, genotype 3 noroviruses (GII.3 NoVs) are secondary to GII.4 NoVs in causing acute non-bacterial gastroenteritis worldwide. In our previous study, we found that virus-like particles (VLPs) derived from a GII.3 NoV strain exhibited no binding activity to any salivary and synthetic histo-blood group antigens (HBGAs) tested. In this study, the nucleotide sequence encoding the major capsid protein of another documented GII.3 NoV strain was codon-optimized and synthesized, and the major capsid protein was expressed using recombinant baculovirus virus expression system. The assembly of VLPs was verified by electron microscopy, and the binding profiles of the assembled VLPs to salivary HBGAs were determined, and in vitro VLP-salivary HBGAs binding blockade assay was used to test the cross-blocking effects of hyperimmune sera produced against different genotypes (GI.2, GII.3, and GII.4). The expression of the major capsid proteins led to the successful assembly of VLPs, and in vitro VLP-salivary HBGAs binding assay indicated that the assembled VLPs bound to salivary HBGAs from blood type A, B, AB, and O individuals, with the highest binding capacity to type A salivary HBGAs. In vitro VLP-salivary HBGAs binding blockade assay demonstrated the absence of blocking activities for hyperimmune sera produced against GI.2and GII.4 VLPs and the presence of blocking activity for that against GII.3 VLPs. Our results suggest the absence of cross-blocking activities among different genotypes and the presence of blocking activities between GII.3 NoVs from different clusters, which might have implications for the design of multivalent NoV vaccines.


Subject(s)
Capsid Proteins/genetics , Norovirus/genetics , Baculoviridae/genetics , Base Sequence/genetics , Blood Group Antigens/genetics , Genotype , Humans , Receptors, Virus/genetics , Salivary Glands/virology , Virus Attachment
14.
Hepatogastroenterology ; 61(133): 1459-64, 2014.
Article in English | MEDLINE | ID: mdl-25436325

ABSTRACT

BACKGROUND/AIMS: SALL4 [encoding the Sal-Like 4 (Drosophila) protein, also known as spalt-like transcription factor 4] plays crucial roles in the development of different cancers, as well as vital embryogenic roles. However, knowledge of its relationship to gastric cancer is limited, and its association with prognosis in gastric cancer has not yet been reported. METHODOLOGY: comprehensive bioinformatics analyses including mRNA expression, gene occurrence, protein/gene interaction, pathway enrichment and biological processes annotation were performed. RESULTS: we indicated that the expression of SALL4 was up-regulated at least 2.2-fold in 103 cases of gastric cancers, compared with its expression in 65 normal controls. Further analysis indicated that SALL4 was co-occurred and interacted with 23 proteins/genes which all were associated with a poor gastric cancer prognosis. It also associated with 14 biological processes and Wnt signaling pathway, which have been proven to be closely related to gastric cancer and its prognosis. CONCLUSIONS: for the first time, we have shown that the up-regulation of SALL4 in gastric cancers should serve as a worthwhile biological marker of poor prognosis in gastric cancer therapies, via the strong interactions with a number of proteins, genes, biological processes and pathways which all were associated with poor prognosis in gastric and other cancer.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Carcinoma/pathology , Case-Control Studies , Computational Biology , Databases, Genetic , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Prognosis , RNA, Messenger/analysis , Stomach Neoplasms/pathology , Up-Regulation
15.
Arch Gerontol Geriatr ; 126: 105540, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38964091

ABSTRACT

OBJECT: The aim was to assess the effect of folic acid supplementation on cognitive function and inflammatory cytokines in elderly patients with mild cognitive impairment. METHODS: From its inception until February 2024, four databases including Web of Science were searched. Two researchers independently screened the literature, assessed the quality, extracted data, and conducted a meta-analysis using RevMan. RESULTS: The systematic review included seven studies (with a total of 1102 participants, mean age 65-80 years), seven of which were appropriate for meta-analysis. Although a small number of studies found relatively large heterogeneity, the majority of studies showed significant benefit from folic acid supplementation, including the FSIQ (823 individuals, standardized mean difference [SMD] = 8.36, 95 % confidence interval [CI] = 0.79 - 1.08), Arithmetic (823 individuals, SMD = 0.17, 95 % CI = -0.03-0.31), Information, SMD = 1.73, 95 % CI 0.41-3.05), Digit Span (823 individuals, SMD = 0.17, 95 % CI = -0.03 - 0.31), Block Design (823 individuals, SMD = 0.26, 95 % CI 0.03-0.49), Picture Completion (823 individuals, SMD = 0.27, 95 % CI = -0.15 - 0.69) and Picture Arrangement (823 individuals, SMD = -0.12, 95 % CI = -0.26 - 0.01). Finally, folic acid supplementation had a significant effect on the reduction of most inflammatory cytokines, blood biomarkers of Alzheimer's disease, and Hcy. CONCLUSIONS: Folic acid supplementation seems to have a positive impact on cognitive function in older adults with mild cognitive impairment, but further evidence of its effectiveness in improving inflammatory cytokines is needed from high-quality studies.

16.
Eur J Med Chem ; 265: 116044, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38145603

ABSTRACT

Ferroptosis is a form of non-apoptotic cell death, regulated by phospholipid hydroperoxide glutathione peroxidase 4 (GPX4), a selenoprotein with a selenocysteine residue (sec) in the active site. GPX4 is a promising target for cancer cells in therapy-resistant conditions via ferroptosis, which can reduce the level of lipid reactive oxygen species (ROS). So far, all existing GPX4 inhibitors covalently bind to GPX4 via a reactive alkyl chloride moiety or masked nitrile-oxide electrophiles with poor selectivity and pharmacokinetic properties and most were obtained by cell phenotype-based screening. Lacking of effective high-throughput screening methods for GPX4 protein limits the discovery of GPX4 inhibitors. Here, we report a fluorescence polarization (FP)-based high throughput screening (HTS) assay for GPX4-U46C-C10A-C66A in vitro, and found Metamizole sodium from our in-house compound library inhibits GPX4-U46C-C10A-C66A enzyme activity. Structure-activity relationships (SAR) demonstrated the importance of sulfonyl group on interaction between Metamizole sodium and GPX4-U46C-C10A-C66A. Our FP assay could be an effective tool for discovery of GPX4 inhibitors and Metamizole sodium was a potential inhibitor for GPX4 in vitro.


Subject(s)
Dipyrone , High-Throughput Screening Assays , Phospholipid Hydroperoxide Glutathione Peroxidase , Selenocysteine/metabolism , Structure-Activity Relationship , Glutathione Peroxidase/metabolism
17.
Signal Transduct Target Ther ; 9(1): 96, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38653754

ABSTRACT

The translocation of YAP from the cytoplasm to the nucleus is critical for its activation and plays a key role in tumor progression. However, the precise molecular mechanisms governing the nuclear import of YAP are not fully understood. In this study, we have uncovered a crucial role of SOX9 in the activation of YAP. SOX9 promotes the nuclear translocation of YAP by direct interaction. Importantly, we have identified that the binding between Asp-125 of SOX9 and Arg-124 of YAP is essential for SOX9-YAP interaction and subsequent nuclear entry of YAP. Additionally, we have discovered a novel asymmetrical dimethylation of YAP at Arg-124 (YAP-R124me2a) catalyzed by PRMT1. YAP-R124me2a enhances the interaction between YAP and SOX9 and is associated with poor prognosis in multiple cancers. Furthermore, we disrupted the interaction between SOX9 and YAP using a competitive peptide, S-A1, which mimics an α-helix of SOX9 containing Asp-125. S-A1 significantly inhibits YAP nuclear translocation and effectively suppresses tumor growth. This study provides the first evidence of SOX9 as a pivotal regulator driving YAP nuclear translocation and presents a potential therapeutic strategy for YAP-driven human cancers by targeting SOX9-YAP interaction.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Nucleus , SOX9 Transcription Factor , Transcription Factors , YAP-Signaling Proteins , Humans , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Cell Nucleus/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Active Transport, Cell Nucleus/genetics , Mice , Cell Line, Tumor , Animals , Repressor Proteins/genetics , Repressor Proteins/metabolism
18.
Cell Rep ; 42(7): 112798, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37453063

ABSTRACT

In castration-resistant prostate cancer (CRPC), clinical response to androgen receptor (AR) antagonists is limited mainly due to AR-variants expression and restored AR signaling. The metabolite spermine is most abundant in prostate and it decreases as prostate cancer progresses, but its functions remain poorly understood. Here, we show spermine inhibits full-length androgen receptor (AR-FL) and androgen receptor splice variant 7 (AR-V7) signaling and suppresses CRPC cell proliferation by directly binding and inhibiting protein arginine methyltransferase PRMT1. Spermine reduces H4R3me2a modification at the AR locus and suppresses AR binding as well as H3K27ac modification levels at AR target genes. Spermine supplementation restrains CRPC growth in vivo. PRMT1 inhibition also suppresses AR-FL and AR-V7 signaling and reduces CRPC growth. Collectively, we demonstrate spermine as an anticancer metabolite by inhibiting PRMT1 to transcriptionally inhibit AR-FL and AR-V7 signaling in CRPC, and we indicate spermine and PRMT1 inhibition as powerful strategies overcoming limitations of current AR-based therapies in CRPC.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Receptors, Androgen/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Spermine/pharmacology , Signal Transduction , Androgen Receptor Antagonists/therapeutic use , Cell Line, Tumor , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism
19.
Medicine (Baltimore) ; 101(41): e31061, 2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36254025

ABSTRACT

RATIONALE: Uterine metastasis from breast cancer is extremely rare. Asymptomatic patients with cervical metastases from breast cancer are rarer and more likely to be missed. We present an asymptomatic patient with breast cancer metastasized to the uterus and share opinions on diagnosing and treating for this kind of cases. PATIENT CONCERNS: We present the case of a 64-year-old woman who was diagnosed with both breast cancer and uterine fibroids after examination. She had no symptoms of gynecological disease during breast cancer treatment. A positron emission tomography/computed tomography (PET/CT) scan was performed during reexamination, revealing multiple metastases of the bone throughout the body and an abnormal hypermetabolic mass in the uterus. It was later confirmed as uterine metastasis by pathology. DIAGNOSIS: A diagnosis of metastatic breast invasive lobular carcinoma was established after a uterine curettage. INTERVENTIONS AND OUTCOMES: Treatment of the uterine metastasis included systemic chemotherapy, total abdominal hysterectomy and bilateral salpingo-oophorectomy (TAH and BSO), postoperative radiotherapy, and postoperative chemotherapy. The patient eventually refused further treatment for personal reasons and died at home. LESSONS: Breast cancer metastases to the uterus are very rare and further research is needed for their diagnosis and treatment. During reexamination of breast cancer patients, clinicians must be alert to metastasis to gynecologic organs. This is particularly important in hormone receptor-positive patients with asymptomatic distant metastasis.


Subject(s)
Breast Neoplasms , Carcinoma, Lobular , Leiomyoma , Uterine Neoplasms , Breast Neoplasms/pathology , Carcinoma, Lobular/pathology , Female , Humans , Middle Aged , Positron Emission Tomography Computed Tomography , Uterine Neoplasms/pathology , Uterus/pathology
20.
Acta Pharm Sin B ; 12(11): 4180-4192, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36386479

ABSTRACT

Glycolytic metabolism enzymes have been implicated in the immunometabolism field through changes in metabolic status. PGK1 is a catalytic enzyme in the glycolytic pathway. Here, we set up a high-throughput screen platform to identify PGK1 inhibitors. DC-PGKI is an ATP-competitive inhibitor of PGK1 with an affinity of K d = 99.08 nmol/L. DC-PGKI stabilizes PGK1 in vitro and in vivo, and suppresses both glycolytic activity and the kinase function of PGK1. In addition, DC-PGKI unveils that PGK1 regulates production of IL-1ß and IL-6 in LPS-stimulated macrophages. Mechanistically, inhibition of PGK1 with DC-PGKI results in NRF2 (nuclear factor-erythroid factor 2-related factor 2, NFE2L2) accumulation, then NRF2 translocates to the nucleus and binds to the proximity region of Il-1ß and Il-6 genes, and inhibits LPS-induced expression of these genes. DC-PGKI ameliorates colitis in the dextran sulfate sodium (DSS)-induced colitis mouse model. These data support PGK1 as a regulator of macrophages and suggest potential utility of PGK1 inhibitors in the treatment of inflammatory bowel disease.

SELECTION OF CITATIONS
SEARCH DETAIL