Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
J Biol Chem ; 299(9): 105083, 2023 09.
Article in English | MEDLINE | ID: mdl-37495110

ABSTRACT

c-Myc is a critical regulator of cell proliferation and growth. Elevated levels of c-Myc cause transcriptional amplification, leading to various types of cancers. Small molecules that specifically inhibit c-Myc-dependent regulation are potentially invaluable for anticancer therapy. Because c-Myc does not have enzymatic activity or targetable pockets, researchers have attempted to obtain small molecules that inhibit c-Myc cofactors, activate c-Myc repressors, or target epigenetic modifications to regulate the chromatin of c-Myc-addicted cancer without any clinical success. In this study, we screened for c-Myc inhibitors using a cell-dependent assay system in which the expression of c-Myc and its transcriptional activity can be inferred from monomeric Keima and enhanced GFP fluorescence, respectively. We identified one mitochondrial inhibitor, antimycin A, as a hit compound. The compound enhanced the c-Myc phosphorylation of threonine-58, consequently increasing the proteasome-mediated c-Myc degradation. The mechanistic analysis of antimycin A revealed that it enhanced the degradation of c-Myc protein through the activation of glycogen synthetic kinase 3 by reactive oxygen species (ROS) from damaged mitochondria. Furthermore, we found that the inhibition of cell growth by antimycin A was caused by both ROS-dependent and ROS-independent pathways. Interestingly, ROS-dependent growth inhibition occurred only in the presence of c-Myc, which may reflect the representative features of cancer cells. Consistently, the antimycin A sensitivity of cells was correlated to the endogenous c-Myc levels in various cancer cells. Overall, our study provides an effective strategy for identifying c-Myc inhibitors and proposes a novel concept for utilizing ROS inducers for cancer therapy.


Subject(s)
Antimycin A , Proteolysis , Proto-Oncogene Proteins c-myc , Antimycin A/pharmacology , Cell Line, Tumor , High-Throughput Screening Assays , Phosphorylation , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Reactive Oxygen Species/metabolism , Threonine/metabolism , Proteolysis/drug effects , Transcription, Genetic/drug effects , Antineoplastic Agents/pharmacology , HCT116 Cells , HeLa Cells , Cell Survival/drug effects , Humans
2.
J Biol Chem ; 298(12): 102635, 2022 12.
Article in English | MEDLINE | ID: mdl-36273581

ABSTRACT

Cancer cells intrinsically proliferate in an autonomous manner; however, the expansion of cancer cell areas in a tissue is known to be regulated by surrounding nontransformed cells. Whether these nontransformed cells can be targeted to control the spread of cancer cells is not understood. In this study, we established a system to evaluate the cancer-inhibitory activity of surrounding nontransformed cells and screened chemical compounds that could induce this activity. Our findings revealed that lonidamine (LND) and domperidone (DPD) inhibited expansion of oncogenic foci of KRASG12D-expressing transformed cells, whereas they did not inhibit the proliferation of monocultured KRASG12D-expressing cells. Live imaging revealed that LND and DPD suppressed the movement of nontransformed cells away from the attaching cancer cells. Moreover, we determined that LND and DPD promoted stress fiber formation, and the dominant-negative mutant of a small GTPase RhoA relieved the suppression of focus expansion, suggesting that RhoA-mediated stress fiber formation is involved in the inhibition of the movement of nontransformed cells and focus expansion. In conclusion, we suggest that elucidation of the mechanism of action of LND and DPD may lead to the development of a new type of drug that could induce the anticancer activity of surrounding nontransformed cells.


Subject(s)
Antineoplastic Agents , Domperidone , Indazoles , Neoplasms , Domperidone/pharmacology , Indazoles/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Mice , Epithelial Cells , Mammary Glands, Animal/cytology , Drug Screening Assays, Antitumor
3.
J Transl Med ; 21(1): 361, 2023 06 03.
Article in English | MEDLINE | ID: mdl-37268950

ABSTRACT

BACKGROUND: Restoration of salivary gland function in Sjogren's syndrome (SS) is still a challenge. Dental pulp stem cells (DPSCs) derived exosomes had shown anti-inflammatory, anti-oxidative, immunomodulatory, and tissue function restorative abilities. However, the salivary gland function restoration potential of DPSCs-derived exosomes (DPSC-Exos) during SS has not been investigated yet. METHODS: DPSC-Exos was isolated by ultracentrifugation methods and characterized. Salivary gland epithelial cells (SGEC) were treated with interferon-gamma (IFN-γ) to mimic SS in vitro and cultured with or without DPSC-Exos. SGEC survival and aquaporin 5 (AQP5) expression were analyzed. mRNA sequencing and bioinformatics analysis were performed in IFN-γ vs. DPSC-Exos+ IFN-γ treated SGEC. Non-obese diabetic (NOD)/ltj female mice (SS model), were intravenously administered with DPSC-Exos, and salivary gland functions and SS pathogenicity were analyzed. Furthermore, the mRNA sequencing and bioinformatics predicted mechanism of the therapeutic effect of DPSC-Exos was further investigated both in vitro and in vivo using RT-qPCR, Western blot, immunohistochemistry, immunofluorescence, flowcytometry analysis. RESULTS: DPSC-Exos partially rescued IFN-γ triggered SGEC death. IFN-γ inhibited AQP5 expression in SGEC and DPSC-Exos reversed this effect. Transcriptome analysis showed GPER was the upregulated DEG in DPSC-Exos-treated SGEC with a positive correlation with salivary secretion-related DEGs. Pathway enrichment analysis revealed that DEGs were mainly attributed to estrogen 16 alpha-hydroxylase activity, extracellular exosome function, cAMP signaling, salivary secretion, and estrogen signaling. Intravenous injection of DPSC-Exos in NOD/ltj mice alleviated the SS syndrome as indicated by the increased salivary flow rate, attenuated glandular inflammation, and increased AQP5 expression. GPER was also upregulated in the salivary gland of DPSC-Exos-treated NOD/ltj mice compared with the PBS-treated NOD/ltj mice. IFN-γ+DPSC-Exos-treated SGEC showed higher expression of AQP5, p-PKA, cAMP, and intracellular Ca2+ levels compared with IFN-γ-treated SGEC. These effects were reversed by the inhibition of GPER. CONCLUSIONS: Our results showed that DPSC-Exos revitalize salivary gland epithelial cell function during SS via the GPER-mediated cAMP/PKA/CREB pathway suggesting the possible therapeutic potential of DPSC-Exos in SS-treatment.


Subject(s)
Dental Pulp , Exosomes , Salivary Glands , Sjogren's Syndrome , Humans , Animals , Mice , Dental Pulp/cytology , Cells, Cultured , Exosomes/metabolism , Female , Mice, Inbred NOD , Interferon-gamma/pharmacology , Salivary Glands/cytology , Epithelial Cells/metabolism , Sjogren's Syndrome/therapy
4.
Biol Proced Online ; 24(1): 26, 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36575389

ABSTRACT

BACKGROUND: Sjogren's syndrome (SS) is an autoimmune disorder characterized by the destruction of exocrine glands, resulting in dry mouth and eyes. Currently, there is no effective treatment for SS, and the mechanisms associated with inadequate salivary secretion are poorly understood. METHODS: In this study, we used NOD mice model to monitor changes in mice's salivary secretion and water consumption. Tissue morphology of the submandibular glands was examined by H&E staining, and Immunohistochemical detected the expression of AQP5 (an essential protein in salivary secretion). Global gene expression profiling was performed on submandibular gland tissue of extracted NOD mice model using RNA-seq. Subsequently, a series of bioinformatics analyses of transcriptome sequencing was performed, including differentially expressed genes (DEGs) identification, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, PPI network construction, hub gene identification, and the validity of diagnostic indicators using the dataset GSE40611. Finally, IFN-γ was used to treat the cells, the submandibular gland tissue of NOD mice model was extracted, and RT-qPCR was applied to verify the expression of hub genes. RESULTS: We found that NOD mice model had reduced salivary secretion and increased water consumption. H&E staining suggests acinar destruction and basement membrane changes in glandular tissue. Immunohistochemistry detects a decrease in AQP5 immunostaining within acinar. In transcriptome sequencing, 42 overlapping DEGs were identified, and hub genes (REN, A2M, SNCA, KLK3, TTR, and AZGP1) were identified as initiating targets for insulin signaling. In addition, insulin signaling and cAMP signaling are potential pathways for regulating salivary secretion and constructing a regulatory relationship between target-cAMP signaling-salivary secretion. CONCLUSION: The new potential targets and signal axes for regulating salivary secretion provide a strategy for SS therapy in a clinical setting.

5.
Cancer Cell Int ; 22(1): 52, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35101047

ABSTRACT

BACKGROUND: Triple-receptor negative breast cancer (TNBC) is an aggressive breast tumor subtype that generally has a poor prognosis. This study aimed to investigate the role and regulatory mechanisms of Zinc finger MIZ-type containing 2 (ZMIZ2) in relation to TNBC. METHODS: Based on data from The Cancer Genome Atlas (TCGA), the expression of ZMIZ2 in different subtypes and its correlation with androgen receptor (AR) were analyzed, and a regulatory mechanism network was constructed. The expression and prognostic value of ZMIZ2 in clinical TNBC tissue samples were also investigated. Furthermore, in vitro studies were conducted to investigate the effects of ZMIZ2 knockdown on the malignant behaviors of TNBC cells and target gene expression. RESULTS: Based on TCGA data, ZMIZ2 was found to be significantly upregulated in TNBC tissues and its expression was negatively correlated with AR expression. Key relationships, such as the ZMIZ2-CCL5, ZMIZ2/AR-MCM3, ZMIZ2/AR-E2F4, and the ZMIZ2/AR-DHX38 were identified, which were enriched in NOD-like receptor signaling pathway/toll-like receptor signaling pathway, DNA replication, cell cycle, and spliceosome, respectively. Moreover, ZMIZ2 was upregulated in clinical breast cancer tissues and its high expression was correlated with the poor prognosis of TNBC patients. Furthermore, ZMIZ2 expression was increased in breast cancer cells, and a knockdown of ZMIZ2 inhibited MDA-MB-231 cell proliferation, migration, and invasion, induced cell cycle arrest in the G1 phase, and promoted cell apoptosis. Furthermore, ZMIZ2 knockdown inhibited the mRNA and protein expression of CCL5, MCM3, E2F4, and DHX38. CONCLUSION: Our findings reveal that ZMIZ2 is upregulated in TNBC tissues and is associated with its poor prognosis. ZMIZ2 may promote TNBC progression by promoting the expression of its target genes and affecting the corresponding pathways. Consequently, ZMIZ2 may serve as a promising target for future TNBC treatments.

6.
Bioorg Med Chem ; 66: 116830, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35594648

ABSTRACT

The identification, structure-activity relationships (SARs), and biological effects of new antimalarials consisting of a 2,3,4,9-tetrahydro-1H-ß-carboline core, a coumarin ring, and an oxyalkanoyl linker are described. A cell-based phenotypic approach was employed in this search for novel antimalarial drugs with unique modes of action. Our screening campaign of the RIKEN compound library succeeded in the identification of the known tetrahydro-ß-carboline derivative (4e) as a hit compound showing significant in vitro activity. SAR studies on this chemical series led to the discovery of compound 4h having a (R)-methyl group on the oxyacetyl linker with potent inhibition of parasite growth (IC50 = 2.0 nM). Compound 4h was also found to exhibit significant in vivo antimalarial effects in mouse models. Furthermore, molecular modeling studies on 4e, 4h, and its diastereomer (4j) suggested that the (R)-methyl group of 4h forces the preferential adoption of a specific conformer which is considered to be an active conformer.


Subject(s)
Antimalarials , Animals , Antimalarials/pharmacology , Carbolines/chemistry , Carbolines/pharmacology , Coumarins/pharmacology , Mice , Structure-Activity Relationship
7.
Appl Microbiol Biotechnol ; 106(19-20): 6567-6581, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36112204

ABSTRACT

Leptospirosis is a zoonotic disease caused by pathogenic Leptospira spp., with global implications primarily in tropical countries. However, the mechanisms of leptospiral pathogenesis are still not fully known and not all virulence factors (VFs) have been identified. Budding yeast, Saccharomyces cerevisiae is a popular eukaryotic model which has been used to identify bacterial VFs that target the conserved eukaryotic cellular processes. In this study, we screened for putative VFs of L. interrogans, one of the dominant species causing leptospirosis, by expressing candidate VFs in budding yeast and examining their impact on yeast growth in a high-throughput format. From an initial selection of 288 L. interrogans ORFs, we screened 226 candidate VFs in a yeast growth inhibition assay and identified nine putative VFs in four categories (adhesion, enzymatic, host structure interaction, and immunogenicity). Notably, LIC10280 was highly toxic even when expressed at low copies. We also observed specific subcellular localization for several putative VFs. This study shows that there are still potential L. interrogans VFs that await discovery. KEY POINTS: • High-throughput cloning and expression of leptospiral proteins in yeast. • Heterologous expression of nine leptospiral proteins inhibited yeast growth. • An uncharacterized protein LIC10280 maybe a putative VF for further validation.


Subject(s)
Leptospira interrogans , Leptospira , Leptospirosis , Fungal Proteins/metabolism , Humans , Leptospira interrogans/genetics , Leptospira interrogans/metabolism , Leptospirosis/metabolism , Leptospirosis/microbiology , Leptospirosis/pathology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
8.
J Neurochem ; 157(6): 1992-2007, 2021 06.
Article in English | MEDLINE | ID: mdl-32799401

ABSTRACT

Aggregation of amyloid-ß peptide 1-42 (Aß42) initiates the onset of Alzheimer's disease (AD), and all the drugs designed to attenuate AD have failed in clinical trials. Emodin reduces levels of ß-amyloid, tau aggregation, oxidative stress, and inflammatory response, demonstrating AD therapeutic potential, whereas its effect on the accumulation of the amyloid-ß protein is not well understood. In this work, we investigated emodin activity on Aß aggregation using a range of biochemical, biophysical, and cell-based approaches. We provide evidence to suggest that emodin blocks Aß42 fibrillogenesis and Aß-induced cytotoxicity, displaying a greater effect than that of curcumin. Through adopting three short peptides (Aß1-16, Aß17-33, and Aß28-42), it was proven that emodin interacts with the Leu17-Gly33 sequence. Furthermore, our findings indicated that Val18 and Phe19 in Aß42 are the target residues with which emodin interacts according amino acid mutation experiments. When fed to 8-month-old B6C3-Tg mice for 2 months, high-dose emodin ameliorates cognitive impairment by 60%-70%. Pathological results revealed that levels of Aß deposition in the brains of AD mice treated with a high dose of emodin decreased by 50%-70%. Therefore, our study indicates that emodin may represent a promising drug for AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , Cognitive Dysfunction/drug therapy , Emodin/therapeutic use , Peptide Fragments/toxicity , Protein Aggregates/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Emodin/pharmacology , Female , Male , Mice , Mice, Transgenic , Peptide Fragments/antagonists & inhibitors , Protein Aggregates/physiology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
9.
J Biol Chem ; 294(9): 2988-2996, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30610116

ABSTRACT

Carcinoma-associated fibroblasts are fibroblasts activated by surrounding cancer cells. Carcinoma-associated fibroblasts exhibit enhanced cell migration, which plays an important role in cancer metastasis. Previously, we demonstrated enhanced migration of NIH3T3 fibroblasts when they were cultured in the presence of MCF7 breast cancer cells. Human fibroblasts displayed a similar phenomenon even when they were co-cultured with cancer cells other than MCF7 cells. In this study, we screened ∼16,000 compounds from the RIKEN Natural Products Depository chemical library for inhibitors of enhanced NIH3T3 cell migration in the presence of MCF7. We identified NPD8733 as an inhibitor of cancer cell-enhanced fibroblast migration. This inhibition was observed not only in a wound-healing co-culture assay but also in a Transwell migration assay. Using NPD8733 and a structurally similar but inactive derivative, NPD8126, on immobilized beads, we found that NPD8733, but not NPD8126, specifically binds to valosin-containing protein (VCP)/p97, a member of the ATPase-associated with diverse cellular activities (AAA+) protein family. Using VCP truncation variants, we found that NPD8733 binds to the D1 domain of VCP. Because VCP's D1 domain is important for its function, we concluded that NPD8733 may act on VCP by binding to this domain. siRNA-mediated silencing of VCP in NIH3T3 fibroblasts, but not in MCF7 cells, reduced the migration of the co-cultured NIH3T3 fibroblasts. These results indicate that MCF7 activates the migration of NIH3T3 cells through VCP and that NPD8733 binds VCP and thereby inhibits its activity.


Subject(s)
Cell Movement/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Valosin Containing Protein/metabolism , Animals , Coculture Techniques , Drug Evaluation, Preclinical , Humans , Ligands , MCF-7 Cells , Mice , NIH 3T3 Cells , Protein Domains , Valosin Containing Protein/chemistry
10.
Biosci Biotechnol Biochem ; 84(12): 2484-2490, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32867616

ABSTRACT

Discovery of small-molecule inducers of unique phenotypic changes combined with subsequent target identification often provides new insights into cellular functions. Here, we applied integrated profiling based on cellular morphological and proteomic changes to compound screening. We identified an indane derivative, NPD9055, which is mechanistically distinct from reference compounds with known modes of action. Employing a chemical proteomics approach, we then showed that NPD9055 binds subunits of heterotrimeric G-protein Gi. An in vitro [35S]GTPγS-binding assay revealed that NPD9055 inhibited GDP/GTP exchange on a Gαi subunit induced by a G-protein-coupled receptor agonist, but not on another G-protein from the Gαs family. In intact HeLa cells, NPD9055 induced an increase in intracellular Ca2+ levels and ERK/MAPK phosphorylation, both of which are regulated by Gßγ, following its dissociation from Gαi. Our observations suggest that NPD9055 targets Gαi and thus regulates Gßγ-dependent cellular processes, most likely by causing the dissociation of Gßγ from Gαi.


Subject(s)
Drug Discovery , Heterotrimeric GTP-Binding Proteins/metabolism , Phenotype , Proteomics , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Humans
11.
Cell Commun Signal ; 17(1): 153, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31752909

ABSTRACT

BACKGROUND: Abnormal reactivation of androgen receptor (AR) signaling in castration-resistant prostate cancer (CRPC) mainly results from overexpression and down-regulation of AR. Sumoylation of AR can influence its function. However, regulation of AR sumoylation by SUMO E3 ligases PIASs to modify AR distribution and stability are not well understood. METHODS: We assessed the potential effect of SUMO3 modification on AR intracellular localization by immunostaining in AR-negative prostate cancer DU145 cells, and detected the effect of PIAS1/SUMO3 overexpression on AR sumoylation related degradation. Then we characterized AR sumoylation sites involved modified by SUMO3, and the key residue of PIAS1 involved in itself sumoylation and further mediated AR sumoylation (sumo3-conjugated), translocation and degradation. Finally we detected the recognition of PIAS1 (sumoylation ligase) to MDM2, a ubiquin ligase mediated AR degradation. RESULTS: We demonstrate that SUMO E3 ligase PIAS1, along with SUMO3, mediates AR cytosolic translocation and subsequent degradation via a ubiquitin-proteasome pathway. Although AR sumoylation occurs prior to ubiquitination, the SUMO-acceptor lysine 386 on AR, together with ubiquitin-acceptor lysine 845, contribute to PIAS1/SUMO3-induced AR nuclear export, ubiquitination and subsequent degradation. Moreover, PIAS1 itself is modified by SUMO3 overexpression, and mutation of SUMO-acceptor lysine 117 on PIAS1 can impair AR cytoplasmic distribution, demonstrating the essential role of sumoylated PIAS1 in AR translocation. We further determine that sumoylated PIAS1 interacts with AR lysine 386 and 845 to form a binary complex. Consistent with the effect on AR distribution, SUMO3 modification of PIAS1 is also required for AR ubiquitination and degradation by recruiting ubiquitin E3 ligase MDM2. CONCLUSION: Taken together, SUMO3 modification of PIAS1 modulates AR cellular distribution and stability. Our study provided the evidence the crosstalk between AR sumoylation and ubquitination mediated by PIAS1 and SUMO3.


Subject(s)
Protein Inhibitors of Activated STAT/metabolism , Receptors, Androgen/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism , Humans , Protein Stability , Signal Transduction , Sumoylation , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Bioorg Med Chem ; 26(8): 1453-1461, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29170028

ABSTRACT

Reactive oxygen species (ROS) play an essential role in the survival and progression of cancer. Moderate oxidative stress drives proliferation, whereas high levels of ROS induce cytotoxicity. Compared to cancer cells, healthy cells often exhibit lower levels of oxidative stress. Elevation of cellular ROS levels by small molecules could therefore induce cancer-specific cytotoxicity. We have employed high-throughput phenotypic screening to identify inducers of ROS accumulation. We found 4,5-dihalo-2-methylpyridazin-3-one (DHMP) and 2,3,4,5(6)-tetrachloro-6(5)-methylpyridine (TCMP) moieties to strongly deplete GSH, to cause ROS accumulation and to induce cell death. Small molecules containing these fragments will most likely share the same properties and should therefore be carefully considered in the development of bioactive molecules.


Subject(s)
Antineoplastic Agents/pharmacology , Glutathione/metabolism , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , High-Throughput Screening Assays , Humans , Molecular Structure , Oxidative Stress/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured
13.
Exp Cell Res ; 358(2): 279-289, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28684116

ABSTRACT

CD147 is a multifunctional trans-membrane glycoprotein, which is highly expressed in many cancers. However, the mechanism by which CD147 modulates cell proliferation is not fully understood. The aim of this study is to investigate the role of CD147 in cell proliferation associated with the TGF-ß/Smad4 signaling pathway. Here, we used cell viability and clone formation assays in LNCaP prostate cancer cells to demonstrate that CD147 promotes cell proliferation. The luciferase assay and western blotting show that silencing CD147 using shRNA enhances transcription and expression of p21WAF1. Using immunofluorescence and nuclear-cytoplasmic separation, we show that this is primarily attributed to transport of Smad4 from the cytoplasm to nucleus. Other assays (GST pull-down, co-immunoprecipitation and immunofluorescence) demonstrate that Smad4 is a new interaction partner of CD147, with the Smad4 MH2 domain and CD147 intracellular domain (CD147-ICD) being involved in the interaction. Furthermore, we report that a phosphoserine (pSer) in CD147 (pSer252) is responsible for this interaction and inhibition of the Smad4/p21WAF1 signal that promotes cell proliferation. Our results provide a novel molecular mechanism for CD147-induced cell proliferation associated with Smad4 signal inhibition.


Subject(s)
Basigin/genetics , Cell Proliferation/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Smad4 Protein/metabolism , Basigin/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism
14.
Plant Cell Physiol ; 58(1): 95-105, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28011868

ABSTRACT

Plants have a remarkable ability to perceive and respond to various wavelengths of light and initiate regulation of different cascades of light signaling and molecular components. While the perception of red light and the mechanisms of its signaling involving phytochromes are largely known, knowledge of the mechanisms of blue light signaling is still limited. Chemical genetics involves the use of diverse small active or synthetic molecules to evaluate biological processes. By combining chemicals and analyzing the effects they have on plant morphology, we identified a chemical, 3-bromo-7-nitroindazole (3B7N), that promotes hypocotyl elongation of wild-type Arabidopsis only under continuous blue light. Further evaluation with loss-of-function mutants confirmed that 3B7N inhibits photomorphogenesis through cryptochrome-mediated light signaling. Microarray analysis demonstrated that the effect of 3B7N treatment on gene expression in cry1cry2 is considerably smaller than that in the wild type, indicating that 3B7N specifically interrupts cryptochrome function in the control of seedling development in a light-dependent manner. We demonstrated that 3B7N directly binds to CRY1 protein using an in vitro binding assay. These results suggest that 3B7N is a novel chemical that directly inhibits plant cryptochrome function by physical binding. The application of 3B7N can be used on other plants to study further the blue light mechanism and the genetic control of cryptochromes in the growth and development of plant species.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Cryptochromes/genetics , Indazoles/pharmacology , Light , Seedlings/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Cryptochromes/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/radiation effects , Gene Expression Regulation, Plant/drug effects , Gene Expression Regulation, Plant/radiation effects , Hypocotyl/genetics , Hypocotyl/metabolism , Immunoblotting , Indazoles/chemistry , Indazoles/metabolism , Light Signal Transduction/drug effects , Light Signal Transduction/genetics , Light Signal Transduction/radiation effects , Molecular Structure , Morphogenesis/drug effects , Morphogenesis/genetics , Morphogenesis/radiation effects , Mutation , Oligonucleotide Array Sequence Analysis , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Seedlings/growth & development , Seedlings/metabolism
15.
Angew Chem Int Ed Engl ; 56(28): 8153-8157, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28544137

ABSTRACT

Autophagy is a critical regulator of cellular homeostasis and metabolism. Interference with this process is considered a new approach for the treatment of disease, in particular cancer and neurological disorders. Therefore, novel small-molecule autophagy modulators are in high demand. We describe the discovery of autophinib, a potent autophagy inhibitor with a novel chemotype. Autophinib was identified by means of a phenotypic assay monitoring the formation of autophagy-induced puncta, indicating accumulation of the lipidated cytosolic protein LC3 on the autophagosomal membrane. Target identification and validation revealed that autophinib inhibits autophagy induced by starvation or rapamycin by targeting the lipid kinase VPS34.


Subject(s)
Autophagy/drug effects , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Autophagosomes/drug effects , Drug Discovery , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Sirolimus/pharmacology , Structure-Activity Relationship
16.
Biochem Biophys Res Commun ; 477(4): 737-742, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27363336

ABSTRACT

Cancer stem cells (CSCs) have robust systems to maintain cancer stemness and drug resistance. Thus, targeting such robust systems instead of focusing on individual signaling pathways should be the approach allowing the identification of selective CSC inhibitors. Here, we used the alkaline phosphatase (ALP) assay to identify inhibitors for cancer stemness in induced cancer stem-like (iCSCL) cells. We screened several compounds from natural product chemical library and evaluated hit compounds for their efficacy on cancer stemness in iCSCL tumorspheres. We identified artesunate, an antimalarial drug, as a selective inhibitor of cancer stemness. Artesunate induced mitochondrial dysfunction that selectively inhibited cancer stemness of iCSCL cells, indicating an essential role of mitochondrial metabolism in cancer stemness.


Subject(s)
Artemisinins/administration & dosage , High-Throughput Screening Assays/methods , Mitochondria/drug effects , Mitochondria/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Artesunate , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Humans , Mitochondrial Proteins/metabolism , Neoplastic Stem Cells/ultrastructure
17.
Biochem Biophys Res Commun ; 474(3): 528-533, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27120460

ABSTRACT

Peptidyl prolyl cis/trans isomerization by Pin1 regulates various oncogenic signals during cancer progression, and its inhibition through multiple approaches has established Pin1 as a therapeutic target. However, lack of simplified screening systems has limited the discovery of potent Pin1 inhibitors. We utilized phosphorylation-dependent binding of Pin1 to its specific substrate to develop a screening system for Pin1 inhibitors. Using this system, we screened a chemical library, and identified a novel selenium derivative as Pin1 inhibitor. Based on structure-activity guided chemical synthesis, we developed more potent Pin1 inhibitors that inhibited cancer cell proliferation.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Selenium Compounds/chemistry , Selenium Compounds/therapeutic use , Binding Sites , Cell Line, Tumor , Humans , Neoplasms, Experimental/pathology , Protein Binding , Protein Interaction Mapping/methods , Treatment Outcome
18.
Bioorg Med Chem ; 24(15): 3246-54, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27017542

ABSTRACT

Protein-protein interaction is one of the key events in the signal transduction pathway. The interaction changes the conformations, activities, localization and stabilities of the proteins, and transduces the signal to the next step. Frequently, this interaction occurs upon the protein phosphorylation. When upstream signals are stimulated, protein kinase(s) is/are activated and phosphorylate(s) their substrates, and induce the phosphorylation dependent protein-protein interaction. For this interaction, several domains in proteins are known to specifically recognize the phosphorylated residues of target proteins. These specific domains for interaction are important in the progression of the diseases caused by disordered signal transduction such as cancer. Thus small molecules that modulate this interaction are attractive lead compounds for the treatment of such diseases. In this review, we focused on three examples of phosphorylation dependent protein-protein interaction modules (14-3-3, polo box domain of Plk1 and F-box proteins in SCF ubiquitin ligases) and summarize small molecules that modulate their interaction. We also introduce our original screening system to identify such small molecules.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Interaction Maps/drug effects , Small Molecule Libraries/pharmacology , Animals , Humans , Phosphorylation/drug effects , Protein Binding , Signal Transduction
19.
Biosci Biotechnol Biochem ; 79(4): 633-5, 2015.
Article in English | MEDLINE | ID: mdl-25471083

ABSTRACT

Quinomycin A and its derivatives were identified as potent antimalarial (Plasmodium falciparum) agents in a screen of the RIKEN NPDepo chemical library. IC50 values of quinomycin A and UK-63,598 were approximately 100 times lower than that of the antimalarial drug chloroquine. This activity was mitigated by the addition of plasmid DNA, suggesting that these compounds act against parasites by intercalating into their DNA.


Subject(s)
Antimalarials/pharmacology , DNA, Protozoan/antagonists & inhibitors , Echinomycin/pharmacology , Intercalating Agents/pharmacology , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Chloroquine/pharmacology , DNA, Protozoan/chemistry , Drug Discovery , Echinomycin/chemistry , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Inhibitory Concentration 50 , Intercalating Agents/chemistry , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Plasmids/chemistry , Plasmids/pharmacology , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167121, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38471652

ABSTRACT

BACKGROUND: Sjögren's syndrome (SS) is a chronic autoimmune disease that predominantly affects exocrine glands. Previous studies have demonstrated that upregulated interferon-gamma (IFN-γ) in SS triggers ferroptosis in salivary gland epithelial cells (SGECs), resulting in impaired salivary gland secretion. However, the immune cells responsible for secreting IFN-γ remain unclear. Therefore, this study conducted bioinformatics analysis and molecular validation to identify the origin of IFN-γ in SS salivary gland. METHODS: The 'limma' package in R software was utilized to identify differentially expressed genes (DEGs) in the human SS dataset. Subsequently, the identified DEGs were compared with the ferroptosis database and screened through Cytoscape to determine candidate genes. The cellular localization and expression patterns of candidate genes were further confirmed in the salivary gland single-cell RNA sequence (scRNA-seq) data set from healthy control and SS mice. Furthermore, in vitro and in vivo studies were performed to analyze the effect of CD4 T-secreted IFN-γ on SGECs' ferroptosis and functions. RESULTS: Upregulated TLR4, IFNG, and IL33 were screened as candidates ferroptosis ferroptosis-inducing genes in SS salivary glands. The association of IFNG and IL33 with CD4 T cells was established through immune infiltration analysis. The expression of IFN-γ on CD4 T cells was robustly higher compared with that of IL33 as evidenced by scRNA-seq and immunofluorescence co-localization. Subsequent experiments conducted on candidate genes consistently demonstrated the potent ability of IFN-γ to induce SGECs' ferroptosis and inhibit AQP5 expression. CONCLUSIONS: Our findings indicate that CD4 T cell-secreted IFN-γ in SS induces SGECs' ferroptosis and inhibits AQP5 expression.


Subject(s)
Ferroptosis , Sjogren's Syndrome , Humans , Animals , Mice , Interferon-gamma/metabolism , CD4-Positive T-Lymphocytes , Interleukin-33/metabolism , Salivary Glands , Epithelial Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL