Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.033
1.
J Exp Clin Cancer Res ; 43(1): 137, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711119

BACKGROUND: The C-terminal-binding protein 1/brefeldin A ADP-ribosylation substrate (CtBP1/BARS) acts both as an oncogenic transcriptional co-repressor and as a fission inducing protein required for membrane trafficking and Golgi complex partitioning during mitosis, hence for mitotic entry. CtBP1/BARS overexpression, in multiple cancers, has pro-tumorigenic functions regulating gene networks associated with "cancer hallmarks" and malignant behavior including: increased cell survival, proliferation, migration/invasion, epithelial-mesenchymal transition (EMT). Structurally, CtBP1/BARS belongs to the hydroxyacid-dehydrogenase family and possesses a NAD(H)-binding Rossmann fold, which, depending on ligands bound, controls the oligomerization of CtBP1/BARS and, in turn, its cellular functions. Here, we proposed to target the CtBP1/BARS Rossmann fold with small molecules as selective inhibitors of mitotic entry and pro-tumoral transcriptional activities. METHODS: Structured-based screening of drug databases at different development stages was applied to discover novel ligands targeting the Rossmann fold. Among these identified ligands, N-(3,4-dichlorophenyl)-4-{[(4-nitrophenyl)carbamoyl]amino}benzenesulfonamide, called Comp.11, was selected for further analysis. Fluorescence spectroscopy, isothermal calorimetry, computational modelling and site-directed mutagenesis were employed to define the binding of Comp.11 to the Rossmann fold. Effects of Comp.11 on the oligomerization state, protein partners binding and pro-tumoral activities were evaluated by size-exclusion chromatography, pull-down, membrane transport and mitotic entry assays, Flow cytometry, quantitative real-time PCR, motility/invasion, and colony assays in A375MM and B16F10 melanoma cell lines. Effects of Comp.11 on tumor growth in vivo were analyzed in mouse tumor model. RESULTS: We identify Comp.11 as a new, potent and selective inhibitor of CtBP1/BARS (but not CtBP2). Comp.11 directly binds to the CtBP1/BARS Rossmann fold affecting the oligomerization state of the protein (unlike other known CtBPs inhibitors), which, in turn, hinders interactions with relevant partners, resulting in the inhibition of both CtBP1/BARS cellular functions: i) membrane fission, with block of mitotic entry and cellular secretion; and ii) transcriptional pro-tumoral effects with significantly hampered proliferation, EMT, migration/invasion, and colony-forming capabilities. The combination of these effects impairs melanoma tumor growth in mouse models.  CONCLUSIONS: This study identifies a potent and selective inhibitor of CtBP1/BARS active in cellular and melanoma animal models revealing new opportunities to study the role of CtBP1/BARS in tumor biology and to develop novel melanoma treatments.


Alcohol Oxidoreductases , DNA-Binding Proteins , Melanoma , Humans , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Alcohol Oxidoreductases/genetics , Animals , Mice , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Xenograft Model Antitumor Assays
2.
J Phys Chem B ; 128(16): 3795-3806, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38606592

The Hippo signaling pathway is a highly conserved signaling network that plays a central role in regulating cellular growth, proliferation, and organ size. This pathway consists of a kinase cascade that integrates various upstream signals to control the activation or inactivation of YAP/TAZ proteins. Phosphorylated YAP/TAZ is sequestered in the cytoplasm; however, when the Hippo pathway is deactivated, it translocates into the nucleus, where it associates with TEAD transcription factors. This partnership is instrumental in regulating the transcription of progrowth and antiapoptotic genes. Thus, in many cancers, aberrantly hyperactivated YAP/TAZ promotes oncogenesis by contributing to cancer cell proliferation, metastasis, and therapy resistance. Because YAP and TAZ exert their oncogenic effects by binding with TEAD, it is critical to understand this key interaction to develop cancer therapeutics. Previous research has indicated that TEAD undergoes autopalmitoylation at a conserved cysteine, and small molecules that inhibit TEAD palmitoylation disrupt effective YAP/TAZ binding. However, how exactly palmitoylation contributes to YAP/TAZ-TEAD interactions and how the TEAD palmitoylation inhibitors disrupt this interaction remains unknown. Utilizing molecular dynamics simulations, our investigation not only provides detailed atomistic insight into the YAP/TAZ-TEAD dynamics but also unveils that the inhibitor studied influences the binding of YAP and TAZ to TEAD in distinct manners. This discovery has significant implications for the design and deployment of future molecular interventions targeting this interaction.


Lipoylation , Molecular Dynamics Simulation , TEA Domain Transcription Factors , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Humans , Acyltransferases/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Allosteric Regulation/drug effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , Protein Binding , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , TEA Domain Transcription Factors/chemistry , TEA Domain Transcription Factors/metabolism , Trans-Activators/metabolism , Trans-Activators/chemistry , Trans-Activators/antagonists & inhibitors , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transcriptional Coactivator with PDZ-Binding Motif Proteins/chemistry , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , YAP-Signaling Proteins/chemistry , YAP-Signaling Proteins/metabolism
3.
Int J Mol Sci ; 23(23)2022 Dec 06.
Article En | MEDLINE | ID: mdl-36499741

The existence of a tight relationship between inflammation and epigenetics that in primary breast tumor cells can lead to tumor progression and the formation of bone metastases was investigated. It was highlighted how the induction of tumor progression and bone metastasis by Interleukin-1 beta, in a non-metastatic breast cancer cell line, MCF-7, was dependent on the de-methylating actions of ten-eleven translocation proteins (TETs). In fact, the inhibition of their activity by the Bobcat339 molecule, an inhibitor of TET enzymes, determined on the one hand, the modulation of the epithelial-mesenchymal transition process, and on the other hand, the reduction in the expression of markers of bone metastasis, indicating that the epigenetic action of TETs is a prerequisite for IL-1ß-dependent tumor progression and bone metastasis formation.


Bone Neoplasms , Breast Neoplasms , Inflammatory Breast Neoplasms , Female , Humans , Bone Neoplasms/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Interleukin-1beta/pharmacology , MCF-7 Cells , Dioxygenases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology
4.
Comput Math Methods Med ; 2022: 8801484, 2022.
Article En | MEDLINE | ID: mdl-35844444

Objective: The lack of certain trace elements such as selenium, molybdenum, magnesium or related nutrients in the soil, water quality and food in the disease area, which caused disturbance of myocardium metabolism and resulted in injury and necrosis. The aim of the study was to explore the mechanism of ibuprofen alleviating myocardial injury caused by acute pancreatitis (AP). Method: We have established AP cell model and rat model. HE staining is used for histological examination. ELISA is used to determine the levels of proinflammatory cytokines (TNF-α and IL-6) and markers of myocardial injury (LDH and CK-MB). qRT-PCR and Western blot are used to analyze the mRNA and protein levels of related genes. Results: The expression level of AIM2 was significantly increased in AP cells; downregulation of AIM2 alleviated inflammation and myocardial injury induced by AP cells; ibuprofen could inhibit the expression of AIM2 and alleviate inflammation and myocardial injury induced by AP cells. In vivo experiments have found that ibuprofen can inhibit the expression of AIM2 to alleviate myocardial injury in AP rat. Conclusion: Ibuprofen can alleviate myocardial injury caused by acute pancreatitis by inhibiting the expression of AIM2.


DNA-Binding Proteins , Ibuprofen , Pancreatitis , Acute Disease , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Ibuprofen/pharmacology , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Pancreatitis/genetics , Pancreatitis/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
5.
Dis Markers ; 2022: 9312971, 2022.
Article En | MEDLINE | ID: mdl-35769815

T-cell acute lymphoblastic leukemia (T-ALL) is a serious hematological tumor derived from early T-cell progenitors, which is extremely resistant to chemotherapy. Classically, doxorubicin (DOX) is an effective first-line drug for the treatment of T-ALL; however, DOX resistance limits its clinical effect. The DEK proto-oncogene (DEK) has been involved in neoplasms but remains unexplored in T-ALL. We silenced DEK on Jurkat cells and detected cell proliferation with cell counting and colony formation assay. Then, we detected DEK's drug sensitivity to DOX with CCK-8, cell cycle, and apoptosis with DOX treatment. Western blot analysis was performed to determine protein expression of apoptosis and cell cycle-related genes, including BCL2L1, caspase-3, and cyclin-dependent kinases (CDK). Finally, the tumorigenic ability of DEK was analyzed using a BALB/C nude mouse model. In this study, DEK was highly expressed in Jurkat cells. Inhibition of DEK can lead to decreased cell proliferation and proportion of S-phase cells in the cell cycle and more cell apoptosis, and the effect is more obvious after DOX treatment. Western blot results showed that DOX treatment leads to cell cycle arrest, reduction of cyclin-dependent kinase 6 (CDK6) protein, accumulation of CDKN1A protein, and DOX-induced apoptosis accompanied by reductions in protein levels of BCL2L1, as well as increases in protein level of caspase-3. Furthermore, DEK-silenced Jurkat cells generated a significantly smaller tumor mass in mice. Our study found that DEK is a novel, potential therapeutic target for overcoming DOX resistance in T-ALL.


DNA-Binding Proteins , Doxorubicin , Oncogene Proteins , Poly-ADP-Ribose Binding Proteins , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Drug Synergism , Humans , Jurkat Cells , Mice , Mice, Inbred BALB C , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
6.
Asian Pac J Cancer Prev ; 23(5): 1539-1545, 2022 May 01.
Article En | MEDLINE | ID: mdl-35633536

OBJECTIVE: Among sarcomas, Ewing sarcoma (EWS) is characterized as a highly malignant type of bone tumor caused by the fusion of EWS RNA Binding Protein-1 (EWSR1)/ Friend leukemia integration 1 (FLI1) genes. The product of fusion gene gives rise to EWSR1/FLI1 which activates the activity of Eyes absent homolog 3 (EYA3) which causes tumor growth and angiogenesis. EYA3 is now considered as a therapeutic drug target for EWS . The study was designed to gather potential inhibitors for the EYA3 target using medicinal compounds. METHODS: In this study, we have obtained a list of medicinal compounds from the NuBBE database and downloaded their structural information. Then insilico screening analysis of >2,000 medicinal compounds was performed with PyRX virtual drug screening software to discover potential inhibitors for the treatment of EWS. RESULTS: Our investigation revealed that Sorbifolin and 1,7-Dihydroxy-3-methylanthracene-9.10-dione show interactive affinity for EYA3 active residues. Moreover, these compounds have adequate toxicity, can induce cytotoxicity in EWS cells, and are capable of regulating the expression of genes activated by EWSR1/FLI1. CONCLUSION: Our study concluded that Sorbifolin and 1,7-Dihydroxy-3-methylanthracene-9.10-dione are promising drug candidates for the treatment of EWS and should be further subjected to invitro testing.


Antineoplastic Agents , Bone Neoplasms , Sarcoma, Ewing , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , Oncogene Proteins, Fusion/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Protein c-fli-1/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics
7.
Clin Transl Med ; 12(4): e742, 2022 04.
Article En | MEDLINE | ID: mdl-35384342

BACKGROUND: Metabolic diseases, including type 2 diabetes, have long been considered incurable, chronic conditions resulting from a variety of pathological conditions in obese patients. Growing evidence suggests the Wnt/ß-catenin pathway is a major pathway in adipose tissue remodelling, pancreatic ß-cell regeneration and energy expenditure through regulation of key metabolic target genes in various tissues. CXXC5-type zinc finger protein 5 (CXXC5) is identified negative feedback regulator of the Wnt/ß-catenin pathway that functions via Dishevelled (Dvl) binding. METHODS: Expression level of CXXC5 was characterised in clinical samples and diabetes-induced mice model. Diabetes-induced mice model was established by using high-fat diet (HFD). HFD-fed mice treated with KY19334, a small molecule inhibiting CXXC5-Dvl protein-protein interaction (PPI), was used to assess the role of CXXC5 in metabolic diseases. RESULTS: Here, we show that CXXC5 is overexpressed with suppression of Wnt/ß-catenin signalling in visceral adipose tissues of patients with obesity-related diabetes. Meanwhile, Cxxc5-/- mice fed an HFD exhibited resistance to metabolic dysregulation. KY19334 restores the lowered Wnt/ß-catenin signalling and reverses metabolic abnormalities as observed in HFD-fed Cxxc5-/- mice. Administration of KY19334 on HFD-fed mice had a long-lasting glucose-controlling effect through remodelling of adipocytes and regeneration of pancreatic ß-cells. CONCLUSION: Overall, the inhibition of CXXC5 function by small molecule-mediated interference of Dvl binding is a potential therapeutic strategy for the treatment of obesity-related diabetes.


DNA-Binding Proteins , Diabetes Mellitus, Type 2 , Transcription Factors , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Humans , Mice , Mice, Knockout , Obesity/complications , Obesity/drug therapy , Obesity/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Wnt Signaling Pathway
8.
Cell Death Dis ; 13(4): 350, 2022 04 15.
Article En | MEDLINE | ID: mdl-35428778

Patients with late-stage and human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC) continue to have a very poor prognosis. The development of more effective novel therapies that improve overall survival and overcome drug resistance is an urgent priority. Here we report that HNSCC tumors significantly overexpress NEDD8 and exhibit high sensitivity to the first-in-class NEDD8-activating enzyme (NAE) inhibitor pevonedistat. Additional studies established that disruption of NEDD8-mediated protein turnover with pevonedistat dramatically augmented cisplatin-induced DNA damage and apoptosis in HNSCC models. Further analysis revealed that the specific pevonedistat target CUL4A played an essential role in driving the synergy of the pevonedistat and cisplatin combination. Targeted inhibition of CUL4A resulted in significant downregulation in Damage Specific DNA binding protein 2 (DDB2), a DNA-damage recognition protein that promotes nucleotide excision repair and resistance to cisplatin. Silencing of CUL4A or DDB2 enhanced cisplatin-induced DNA damage and apoptosis in a manner similar to that of pevonedistat demonstrating that targeted inhibition of CUL4A may be a novel approach to augment cisplatin therapy. Administration of pevonedistat to mice bearing HNSCC tumors significantly decreased DDB2 expression in tumor cells, increased DNA damage and potently enhanced the activity of cisplatin to yield tumor regression and long-term survival of all animals. Our findings provide strong rationale for clinical investigation of CUL4A inhibition with pevonedistat as a novel strategy to augment the efficacy of cisplatin therapy for patients with HNSCC and identify loss of DDB2 as a key pharmacodynamic mediator controlling sensitivity to this regimen.


Cisplatin , Cullin Proteins , DNA-Binding Proteins , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Animals , Cell Line, Tumor , Cisplatin/pharmacology , Cullin Proteins/antagonists & inhibitors , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Synergism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
9.
Eur J Med Chem ; 236: 114311, 2022 Jun 05.
Article En | MEDLINE | ID: mdl-35385803

TRIM24 (tripartite motif-containing protein 24) and BRPF1 (bromodomain and PHD finger containing protein 1) are epigenetics "readers" and potential therapeutic targets for cancer and other diseases. Here we describe the structure-guided design of 1-(indolin-1-yl)ethan-1-ones as novel TRIM24/BRPF1 bromodomain inhibitors. The representative compound 20l (Y08624) is a new TRIM24/BRPF1 dual inhibitor, with IC50 values of 0.98 and 1.16 µM, respectively. Cellular activity of 20l was validated by viability assay in prostate cancer (PC) cell lines. In PC xenograft models, 20l suppressed tumor growth (50 mg/kg/day, TGI = 53%) without exhibiting noticeable toxicity. Compound 20l represents a versatile starting point for the development of more potent TRIM24/BRPF1 inhibitors.


Adaptor Proteins, Signal Transducing , Carrier Proteins , DNA-Binding Proteins , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Carrier Proteins/antagonists & inhibitors , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , Humans , Male , Prostatic Neoplasms/drug therapy , Protein Domains
10.
J Med Chem ; 65(5): 3943-3961, 2022 03 10.
Article En | MEDLINE | ID: mdl-35192363

Chromodomain helicase DNA-binding protein 1 like (CHD1L) is an oncogene implicated in tumor progression, multidrug resistance, and metastasis in many types of cancer. In this article, we described the optimization of the first lead CHD1L inhibitors (CHD1Li) through drug design and medicinal chemistry. More than 30 CHD1Li were synthesized and evaluated using a variety of colorectal cancer (CRC) tumor organoid models and functional assays. The results led to the prioritization of six lead CHD1Li analogues with improved potency, antitumor activity, and drug-like properties including metabolic stability and in vivo pharmacokinetics. Furthermore, lead CHD1Li 6.11 proved to be an orally bioavailable antitumor agent, significantly reducing the tumor volume of CRC xenografts generated from isolated quasi mesenchymal cells (M-phenotype), which possess enhanced tumorigenic properties. In conclusion, we reported the optimization of first-in-class inhibitors of oncogenic CHD1L as a novel therapeutic strategy with potential for the treatment of cancer.


Antineoplastic Agents , DNA Helicases , DNA-Binding Proteins , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Cell Line, Tumor , DNA Helicases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Drug Design , Humans , Oncogenes
11.
J Immunol ; 208(5): 1007-1020, 2022 03 01.
Article En | MEDLINE | ID: mdl-35181641

E-protein transcription factors limit group 2 innate lymphoid cell (ILC2) development while promoting T cell differentiation from common lymphoid progenitors. Inhibitors of DNA binding (ID) proteins block E-protein DNA binding in common lymphoid progenitors to allow ILC2 development. However, whether E-proteins influence ILC2 function upon maturity and activation remains unclear. Mice that overexpress ID1 under control of the thymus-restricted proximal Lck promoter (ID1tg/WT) have a large pool of primarily thymus-derived ILC2s in the periphery that develop in the absence of E-protein activity. We used these mice to investigate how the absence of E-protein activity affects ILC2 function and the genomic landscape in response to house dust mite (HDM) allergens. ID1tg/WT mice had increased KLRG1- ILC2s in the lung compared with wild-type (WT; ID1WT/WT) mice in response to HDM, but ID1tg/WT ILC2s had an impaired capacity to produce type 2 cytokines. Analysis of WT ILC2 accessible chromatin suggested that AP-1 and C/EBP transcription factors but not E-proteins were associated with ILC2 inflammatory gene programs. Instead, E-protein binding sites were enriched at functional genes in ILC2s during development that were later dynamically regulated in allergic lung inflammation, including genes that control ILC2 response to cytokines and interactions with T cells. Finally, ILC2s from ID1tg/WT compared with WT mice had fewer regions of open chromatin near functional genes that were enriched for AP-1 factor binding sites following HDM treatment. These data show that E-proteins shape the chromatin landscape during ILC2 development to dictate the functional capacity of mature ILC2s during allergic inflammation in the lung.


Antigens, Dermatophagoides/immunology , Asthma/immunology , Inhibitor of Differentiation Protein 1/metabolism , T-Lymphocytes/immunology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Allergens/immunology , Animals , Asthma/pathology , Cell Differentiation/immunology , Chromatin/metabolism , Cytokines/immunology , DNA-Binding Proteins/antagonists & inhibitors , Female , Lectins, C-Type/genetics , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pyroglyphidae/immunology , Receptors, Immunologic/genetics , Stem Cells/cytology , T-Lymphocytes/cytology , Transcription Factor AP-1/metabolism
12.
Mol Med Rep ; 25(4)2022 04.
Article En | MEDLINE | ID: mdl-35169856

Polycystic ovary syndrome is one of the most common endocrine and metabolic gynecological disorders, of which dysfunction of ovarian granulosa cells is a key contributing factor. The aim of the present study was to explore the role of ferrostatin­1 (Fer­1), a ferroptosis inhibitor, in a cell injury model established by homocysteine (Hcy)­induced ovarian granulosa KGN cell line and the potential underlying mechanism. Cell viability was measured using Cell Counting Kit­8 assay in the presence or absence of Hcy and Fer­1. Cell apoptosis was assessed using TUNEL staining and the expression levels of apoptosis­related proteins were measured using western blotting. To explore the effects of Fer­1 on oxidative stress in Hcy­treated ovarian granulosa cells, the levels of reactive oxygen species (ROS), malondialdehyde (MDA), lactate dehydrogenase (LDH) and glutathione (GSH) were measured using their corresponding kits. Furthermore, Fe2+ levels were assessed using Phen Green™ SK labeling and western blotting was performed to measure the protein expression levels of ferroptosis­associated proteins GPX4, SLC7A11, ASCL4 and DMT1. Subsequently, DNA methylation and ten­eleven translocation (TET) 1/2 demethylase levels were also detected to evaluate the extent of overall DNA methylation in ovarian granulosa cells after Hcy treatment. The TET1/2 inhibitor Bobcat339 hydrochloride was applied to treat ovarian granulosa cells before evaluating the possible effects of Fer­1 on TET1/2 and DNA methylation. Fer­1 was found to markedly elevate ovarian granulosa cell viability following Hcy treatment. The apoptosis rate in Fer­1­treated groups was also markedly decreased, which was accompanied by downregulated Bax and cleaved caspase­3 expression and upregulated Bcl­2 protein expression. In addition, Fer­1 treatment reduced the levels of ROS, MDA and LDH whilst enhancing the levels of GSH. Fe2+ levels were significantly decreased following Fer­1 treatment, which also elevated glutathione peroxidase 4 expression whilst reducing solute carrier family 7 member 11, achaete­scute family BHLH transcription factor 4 and divalent metal transporter 1 protein expression. Fer­1 significantly inhibited DNA methylation and enhanced TET1/2 levels, which were reversed by treatment with Bobcat339 hydrochloride. Subsequent experiments on cell viability, oxidative stress, Fe2+ content, ferroptosis­ and apoptosis­related proteins levels revealed that Bobcat339 hydrochloride reversed the effects of Fer­1 on ovarian granulosa Hcy­induced cell injury. These results suggest that Fer­1 may potentially protect ovarian granulosa cells against Hcy­induced injury by increasing TET levels and reducing DNA methylation.


Cyclohexylamines/pharmacology , DNA Methylation/drug effects , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Ferroptosis/drug effects , Granulosa Cells/drug effects , Mixed Function Oxygenases/metabolism , Phenylenediamines/pharmacology , Proto-Oncogene Proteins/metabolism , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Dioxygenases/antagonists & inhibitors , Female , Glutathione/metabolism , Homocysteine/toxicity , Humans , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors
13.
Nat Commun ; 13(1): 866, 2022 02 14.
Article En | MEDLINE | ID: mdl-35165282

Epstein-Barr virus (EBV) is reportedly the first identified human tumor virus, and is closely related to the occurrence and development of nasopharyngeal carcinoma (NPC), gastric carcinoma (GC), and several lymphomas. PD-L1 expression is elevated in EBV-positive NPC and GC tissues; however, the specific mechanisms underlying the EBV-dependent promotion of PD-L1 expression to induce immune escape warrant clarification. EBV encodes 44 mature miRNAs. In this study, we find that EBV-miR-BART11 and EBV-miR-BART17-3p upregulate the expression of PD-L1 in EBV-associated NPC and GC. Furthermore, EBV-miR-BART11 targets FOXP1, EBV-miR-BART17-3p targets PBRM1, and FOXP1 and PBRM1 bind to the enhancer region of PD-L1 to inhibit its expression. Therefore, EBV-miR-BART11 and EBV-miR-BART17-3p inhibit FOXP1 and PBRM1, respectively, and enhance the transcription of PD-L1 (CD274, http://www.ncbi.nlm.nih.gov/gene/29126 ), resulting in the promotion of tumor immune escape, which provides insights into potential targets for EBV-related tumor immunotherapy.


Herpesvirus 4, Human/genetics , MicroRNAs/genetics , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Neoplasms/immunology , Stomach Neoplasms/immunology , Tumor Escape/immunology , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Epstein-Barr Virus Infections/virology , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Herpesvirus 4, Human/immunology , Humans , Lymphoma/immunology , Lymphoma/virology , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/virology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Stomach Neoplasms/virology , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tumor Escape/genetics , Tumor Microenvironment/immunology
14.
PLoS One ; 17(2): e0262328, 2022.
Article En | MEDLINE | ID: mdl-35148314

Planar cell polarity (PCP) signaling regulates several polarization events during development of ommatidia in the Drosophila eye, including directing chirality by polarizing a cell fate choice and determining the direction and extent of ommatidial rotation. The pksple isoform of the PCP protein Prickle is known to participate in the R3/R4 cell fate decision, but the control of other polarization events and the potential contributions of the three Pk isoforms have not been clarified. Here, by characterizing expression and subcellular localization of individual isoforms together with re-analyzing isoform specific phenotypes, we show that the R3/R4 fate decision, its coordination with rotation direction, and completion of rotation to a final ±90° rotation angle are separable polarization decisions with distinct Pk isoform requirements and contributions. Both pksple and pkpk can enforce robust R3/R4 fate decisions, but only pksple can correctly orient them along the dorsal-ventral axis. In contrast, pksple and pkpk can fully and interchangeably sustain coordination of rotation direction and rotation to completion. We propose that expression dynamics and competitive interactions determine isoform participation in these processes. We propose that the selective requirement for pksple to orient the R3/R4 decision and their interchangeability for coordination and completion of rotation reflects their previously described differential interaction with the Fat/Dachsous system which is known to be required for orientation of R3/R4 decisions but not for coordination or completion of rotation.


DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Eye/metabolism , LIM Domain Proteins/metabolism , Animals , Brain/metabolism , CRISPR-Cas Systems/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Gene Editing , Genotype , LIM Domain Proteins/antagonists & inhibitors , LIM Domain Proteins/genetics , Membrane Proteins/metabolism , Phenotype , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/genetics
15.
J Med Chem ; 65(4): 3306-3331, 2022 02 24.
Article En | MEDLINE | ID: mdl-35133824

ATAD2 is an epigenetic bromodomain-containing target which is overexpressed in many cancers and has been suggested as a potential oncology target. While several small molecule inhibitors have been described in the literature, their cellular activity has proved to be underwhelming. In this work, we describe the identification of a novel series of ATAD2 inhibitors by high throughput screening, confirmation of the bromodomain region as the site of action, and the optimization campaign undertaken to improve the potency, selectivity, and permeability of the initial hit. The result is compound 5 (AZ13824374), a highly potent and selective ATAD2 inhibitor which shows cellular target engagement and antiproliferative activity in a range of breast cancer models.


ATPases Associated with Diverse Cellular Activities/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , DNA-Binding Proteins/antagonists & inhibitors , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Drug Screening Assays, Antitumor , Female , Humans , Models, Molecular , Small Molecule Libraries , Structure-Activity Relationship , Substrate Specificity , Tumor Stem Cell Assay
16.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article En | MEDLINE | ID: mdl-35193978

The architectural protein histone-like protein from Escherichia coli strain U93 (HU) is the most abundant bacterial DNA binding protein and highly conserved among bacteria and Apicomplexan parasites. It not only binds to double-stranded DNA (dsDNA) to maintain DNA stability but also, interacts with RNAs to regulate transcription and translation. Importantly, HU is essential to cell viability for many bacteria; hence, it is an important antibiotic target. Here, we report that Gp46 from bacteriophage SPO1 of Bacillus subtilis is an HU inhibitor whose expression prevents nucleoid segregation and causes filamentous morphology and growth defects in bacteria. We determined the solution structure of Gp46 and revealed a striking negatively charged surface. An NMR-derived structural model for the Gp46-HU complex shows that Gp46 occupies the DNA binding motif of the HU and therefore, occludes DNA binding, revealing a distinct strategy for HU inhibition. We identified the key residues responsible for the interaction that are conserved among HUs of bacteria and Apicomplexans, including clinically significant Mycobacterium tuberculosis, Acinetobacter baumannii, and Plasmodium falciparum, and confirm that Gp46 can also interact with these HUs. Our findings provide detailed insight into a mode of HU inhibition that provides a useful foundation for the development of antibacteria and antimalaria drugs.


Bacterial Proteins/antagonists & inhibitors , Bacteriophages/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Glycoproteins/metabolism , Viral Proteins/metabolism , Bacterial Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/metabolism , Protein Binding
17.
Inflamm Res ; 71(2): 243-253, 2022 Feb.
Article En | MEDLINE | ID: mdl-35059772

OBJECTIVE: Inflammation and proliferation of vascular smooth muscle cells (VSMCs), induced by angiotensin II (AngII) and other growth factors, play important roles in the pathogenesis of hypertension, restenosis, and atherosclerosis. Dihydroartemisinin (DHA) exhibits broad protective effects. However, the effects of DHA on AngII-induced inflammation and proliferation of VSMCs remain unknown. MATERIALS AND METHODS: AngII was used to construct VSMCs and vascular inflammation model in vitro and in vivo. The protective roles of DHA in inflammatory response and proliferation were evaluated through CCK-8, BrdU assay and immunofluorescence staining. The level of mRNA N6-methyladenosine was measured by m6A-RNA immunoprecipitation (MeRIP) assay. Western blot and quantitative real-time PCR were used to investigate the relationship between FTO and its potential downstream signaling molecules. RESULTS: In the present study, we found that DHA significantly suppressed AngII-induced proliferation of VSMCs and the expression of IL-6 and Ccl2 in a dose-dependent manner. Additionally, we confirmed that fat mass and obesity-associated (FTO) plays a critical role in AngII-induced VSMC proliferation and inflammation. FTO knockdown increased the methylation level of NR4A3 mRNA, whereas FTO, but not mutated FTO overexpression, reduced the methylation level of NR4A3 mRNA. These results suggest that DHA plays a protective role in AngII-induced VSMC proliferation and the associated inflammation by inhibiting the FTO/NR4A3 axis. CONCLUSION: Our findings provide new insight into the mechanisms of DHA and its critical role in the pathogenesis of hypertension-related vascular complications.


Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Angiotensin II/pharmacology , Artemisinins/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Inflammation/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, Steroid/antagonists & inhibitors , Receptors, Thyroid Hormone/antagonists & inhibitors , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/physiology , Animals , Cell Proliferation/drug effects , Cells, Cultured , DNA-Binding Proteins/physiology , Mice , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Nerve Tissue Proteins/physiology , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , Signal Transduction/drug effects
18.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article En | MEDLINE | ID: mdl-35054774

Single-stranded DNA (ssDNA)-binding protein (SSB) plays a crucial role in DNA replication, repair, and recombination as well as replication fork restarts. SSB is essential for cell survival and, thus, is an attractive target for potential antipathogen chemotherapy. Whether naturally occurring products can inhibit SSB remains unknown. In this study, the effect of the flavonols myricetin, quercetin, kaempferol, and galangin on the inhibition of Pseudomonas aeruginosa SSB (PaSSB) was investigated. Furthermore, SSB was identified as a novel quercetin-binding protein. Through an electrophoretic mobility shift analysis, myricetin could inhibit the ssDNA binding activity of PaSSB with an IC50 of 2.8 ± 0.4 µM. The effect of quercetin, kaempferol, and galangin was insignificant. To elucidate the flavonol inhibition specificity, the crystal structure of PaSSB complexed with the non-inhibitor quercetin was solved using the molecular replacement method at a resolution of 2.3 Å (PDB entry 7VUM) and compared with a structure with the inhibitor myricetin (PDB entry 5YUN). Although myricetin and quercetin bound PaSSB at a similar site, their binding poses were different. Compared with myricetin, the aromatic ring of quercetin shifted by a distance of 4.9 Å and an angle of 31o for hydrogen bonding to the side chain of Asn108 in PaSSB. In addition, myricetin occupied and interacted with the ssDNA binding sites Lys7 and Glu80 in PaSSB whereas quercetin did not. This result might explain why myricetin could, but quercetin could not, strongly inhibit PaSSB. This molecular evidence reveals the flavonol inhibition specificity and also extends the interactomes of the natural anticancer products myricetin and quercetin to include the OB-fold protein SSB.


DNA-Binding Proteins/antagonists & inhibitors , Flavonols/pharmacology , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/antagonists & inhibitors , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Flavonoids/pharmacology , Flavonols/chemistry , Kaempferols/pharmacology , Models, Molecular , Protein Conformation , Quercetin/chemistry , Quercetin/pharmacology
19.
J Clin Invest ; 132(4)2022 02 15.
Article En | MEDLINE | ID: mdl-35085104

Eltrombopag, an FDA-approved non-peptidyl thrombopoietin receptor agonist, is clinically used for the treatment of aplastic anemia, a disease characterized by hematopoietic stem cell failure and pancytopenia, to improve platelet counts and stem cell function. Eltrombopag treatment results in a durable trilineage hematopoietic expansion in patients. Some of the eltrombopag hematopoietic activity has been attributed to its off-target effects, including iron chelation properties. However, the mechanism of action for its full spectrum of clinical effects is still poorly understood. Here, we report that eltrombopag bound to the TET2 catalytic domain and inhibited its dioxygenase activity, which was independent of its role as an iron chelator. The DNA demethylating enzyme TET2, essential for hematopoietic stem cell differentiation and lineage commitment, is frequently mutated in myeloid malignancies. Eltrombopag treatment expanded TET2-proficient normal hematopoietic stem and progenitor cells, in part because of its ability to mimic loss of TET2 with simultaneous thrombopoietin receptor activation. On the contrary, TET inhibition in TET2 mutant malignant myeloid cells prevented neoplastic clonal evolution in vitro and in vivo. This mechanism of action may offer a restorative therapeutic index and provide a scientific rationale to treat selected patients with TET2 mutant-associated or TET deficiency-associated myeloid malignancies.


Anemia, Aplastic , Benzoates/pharmacology , Cell Proliferation , DNA-Binding Proteins , Dioxygenases , Hematopoietic Stem Cells/enzymology , Hydrazines/pharmacology , Pyrazoles/pharmacology , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/antagonists & inhibitors , Dioxygenases/genetics , Dioxygenases/metabolism , Humans , Mice , Mice, Knockout
20.
Hum Pathol ; 119: 41-50, 2022 01.
Article En | MEDLINE | ID: mdl-34666078

Gynecologic cancers are routinely screened for DNA mismatch repair (MMR) gene mutations using immunohistochemistry (IHC) and/or polymerase chain reaction (PCR) for microsatellite instability (MSI) to enable selection of immune checkpoint inhibitor therapy and screen for Lynch syndrome. The limited data that compare IHC and MSI in endometrial tumors have shown discordance rates of 5-10%. We reviewed MMR/MSI results in gynecologic cancers and used next-generation sequencing (NGS) to interrogate discrepancies. Of the 328 cases with both IHC and MSI results, 256 (78.0%) were microsatellite stable (MSS) with preserved MMR (pMMR), 64 (19.5%) cases were MSI-High (MSI-H) with MMR deficient (dMMR), 2 cases showed subclonal loss of MLH1 and PMS2 with MSI-H, and 6 cases were discordant. Overall, there was a 98.2% (322/328) IHC/MSI concordance. Discordant cases were retested and/or subject to NGS. Of the six discrepant cases, five showed dMMR with MSS and one showed pMMR with MSI-H. One dMMR/MSI-L case showed loss of PMS2 with a germline pathogenic mutation. The pMMR/MSI-H case was found to harbor pathogenic variants in MLH1 and MSH6. One of the two cases with subclonal populations demonstrated MSI-H in the dMMR area and MSS in the pMMR area. These results emphasize the importance of selecting the appropriate tumor tissue for both IHC and molecular testing and demonstrate that NGS can help resolve discrepant MMR and MSI results.


Biomarkers, Tumor , DNA Mismatch Repair , Genital Neoplasms, Female/diagnosis , High-Throughput Nucleotide Sequencing , Immunohistochemistry , Microsatellite Instability , Polymerase Chain Reaction , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , DNA Mutational Analysis , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Databases, Factual , Female , Genital Neoplasms, Female/enzymology , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Humans , Middle Aged , Mismatch Repair Endonuclease PMS2/analysis , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/analysis , MutL Protein Homolog 1/genetics , Mutation , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies
...