Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.514
Filter
Add more filters

Publication year range
1.
Cell ; 173(4): 864-878.e29, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29681454

ABSTRACT

Diversity in the genetic lesions that cause cancer is extreme. In consequence, a pressing challenge is the development of drugs that target patient-specific disease mechanisms. To address this challenge, we employed a chemistry-first discovery paradigm for de novo identification of druggable targets linked to robust patient selection hypotheses. In particular, a 200,000 compound diversity-oriented chemical library was profiled across a heavily annotated test-bed of >100 cellular models representative of the diverse and characteristic somatic lesions for lung cancer. This approach led to the delineation of 171 chemical-genetic associations, shedding light on the targetability of mechanistic vulnerabilities corresponding to a range of oncogenotypes present in patient populations lacking effective therapy. Chemically addressable addictions to ciliogenesis in TTC21B mutants and GLUT8-dependent serine biosynthesis in KRAS/KEAP1 double mutants are prominent examples. These observations indicate a wealth of actionable opportunities within the complex molecular etiology of cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Lung Neoplasms/pathology , Small Molecule Libraries/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cytochrome P450 Family 4/deficiency , Cytochrome P450 Family 4/genetics , Drug Discovery , G1 Phase Cell Cycle Checkpoints/drug effects , Glucocorticoids/pharmacology , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
2.
Mol Cell ; 81(5): 1084-1099.e6, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33450211

ABSTRACT

Cells have evolved an elaborate DNA repair network to ensure complete and accurate DNA replication. Defects in these repair machineries can fuel genome instability and drive carcinogenesis while creating vulnerabilities that may be exploited in therapy. Here, we use nascent chromatin capture (NCC) proteomics to characterize the repair of replication-associated DNA double-strand breaks (DSBs) triggered by topoisomerase 1 (TOP1) inhibitors. We reveal profound changes in the fork proteome, including the chromatin environment and nuclear membrane interactions, and identify three classes of repair factors according to their enrichment at broken and/or stalled forks. ATM inhibition dramatically rewired the broken fork proteome, revealing that ataxia telangiectasia mutated (ATM) signalling stimulates DNA end resection, recruits PLK1, and concomitantly suppresses the canonical DSB ubiquitination response by preventing accumulation of RNF168 and BRCA1-A. This work and collection of replication fork proteomes provide a new framework to understand how cells orchestrate homologous recombination repair of replication-associated DSBs.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/genetics , DNA Replication , DNA Topoisomerases, Type I/genetics , DNA/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Recombinational DNA Repair , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Camptothecin/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation , HeLa Cells , Humans , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , Proto-Oncogene Proteins/metabolism , Pyridines/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Topoisomerase I Inhibitors/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Polo-Like Kinase 1
3.
FASEB J ; 35(10): e21923, 2021 10.
Article in English | MEDLINE | ID: mdl-34551143

ABSTRACT

Our recent studies have shown that haspin, a protein kinase imperative for mitosis, is engaged in the interphase progression of HeLa and U2OS cancer cells. In this investigation, we employed the Fucci reporter system and time-lapse imaging to examine the impact of haspin gene silencing on cell cycle progressions at a single-cell level. We found that the loss of haspin induced multiple cell cycle defects. Specifically, the S/G2 duration was greatly prolonged by haspin gene depletion or inhibition in synchronous HeLa cells. Haspin gene depletion in asynchronous HeLa and U2OS cells led to a similarly protracted S/G2 phase, followed by mitotic cell death or postmitotic G1 arrest. In addition, haspin deficiency resulted in robust induction of the p21CIP1/WAF1 checkpoint protein, a target of the p53 activation. Also, co-depleting haspin with either p21 or p53 could rescue U2OS cells from postmitotic G1 arrest and partially restore their proliferation. These results substantiate the haspin's capacity to regulate interphase and mitotic progression, offering a broader antiproliferative potential of haspin loss in cancer cells.


Subject(s)
Cell Cycle , Cell Proliferation , Intracellular Signaling Peptides and Proteins/deficiency , Neoplasms/pathology , Protein Serine-Threonine Kinases/deficiency , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Fluorescent Dyes , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase/drug effects , Humans , Interphase/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Mitosis/drug effects , Neoplasms/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , S Phase/drug effects , Tubercidin/analogs & derivatives , Tubercidin/pharmacology , Tumor Suppressor Protein p53/genetics , Ubiquitination , Up-Regulation/drug effects
4.
Mol Cell Proteomics ; 19(9): 1450-1467, 2020 09.
Article in English | MEDLINE | ID: mdl-32554797

ABSTRACT

Insulin receptor substrate 2 (IRS2) is an essential adaptor that mediates signaling downstream of the insulin receptor and other receptor tyrosine kinases. Transduction through IRS2-dependent pathways is important for coordinating metabolic homeostasis, and dysregulation of IRS2 causes systemic insulin signaling defects. Despite the importance of maintaining proper IRS2 abundance, little is known about what factors mediate its protein stability. We conducted an unbiased proteomic screen to uncover novel substrates of the Anaphase Promoting Complex/Cyclosome (APC/C), a ubiquitin ligase that controls the abundance of key cell cycle regulators. We found that IRS2 levels are regulated by APC/C activity and that IRS2 is a direct APC/C target in G1 Consistent with the APC/C's role in degrading cell cycle regulators, quantitative proteomic analysis of IRS2-null cells revealed a deficiency in proteins involved in cell cycle progression. We further show that cells lacking IRS2 display a weakened spindle assembly checkpoint in cells treated with microtubule inhibitors. Together, these findings reveal a new pathway for IRS2 turnover and indicate that IRS2 is a component of the cell cycle control system in addition to acting as an essential metabolic regulator.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Insulin Receptor Substrate Proteins/metabolism , M Phase Cell Cycle Checkpoints , Amino Acid Motifs , Anaphase-Promoting Complex-Cyclosome/drug effects , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line , Chromatography, Liquid , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knockout Techniques , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , M Phase Cell Cycle Checkpoints/drug effects , Mice , Microtubules/drug effects , Microtubules/metabolism , Phosphorylation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Stability , Proteomics , Pyridines/pharmacology , Tandem Mass Spectrometry , Time-Lapse Imaging , Ubiquitination/drug effects , Ubiquitination/genetics
5.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163589

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and is one of the leading causes of cancer-related deaths worldwide. Regorafenib, a multi-kinase inhibitor, is used as a second-line treatment for advanced HCC. Here, we aimed to investigate the mechanism of the antitumor effect of regorafenib on HCC and evaluate altered microRNA (miRNA) expression. Cell proliferation was examined in six HCC cell lines (HuH-7, HepG2, HLF, PLC/PRF/5, Hep3B, and Li-7) using the Cell Counting Kit-8 assay. Xenografted mouse models were used to assess the effects of regorafenib in vivo. Cell cycle analysis, western blotting analysis, and miRNA expression analysis were performed to identify the antitumor inhibitory potential of regorafenib on HCC cells. Regorafenib suppressed proliferation in HuH-7 cell and induced G0/G1 cell cycle arrest and cyclin D1 downregulation in regorafenib-sensitive cells. During miRNA analysis, miRNA molecules associated with the antitumor effect of regorafenib were found. Regorafenib suppresses cell proliferation and tumor growth in HCC by decreasing cyclin D1 via alterations in intracellular and exosomal miRNAs in HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/drug therapy , MicroRNAs/biosynthesis , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , RNA, Neoplasm/biosynthesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , RNA, Neoplasm/genetics , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/genetics
6.
J Cell Mol Med ; 25(5): 2645-2654, 2021 03.
Article in English | MEDLINE | ID: mdl-33523587

ABSTRACT

Anwulignan is a monomer compound derived from Schisandra sphenanthera lignans. It has been reported to possess a spectrum of pharmacological activities, including anti-bacterial, anti-inflammatory, anticancer and hepatoprotective properties. However, its anticancer capacity and molecular mechanism(s) against non-small cell lung cancer (NSCLC) have not been fully elucidated. Anwulignan significantly inhibited cell growth and increased G1-phase cell cycle arrest in NSCLC cells. Anwulignan strongly attenuates the JAK1/STAT3 signalling pathway by directly targeting JAK1 protein kinase activity in vitro. The anticancer activity by Anwulignan is dependent upon the JAK1 protein expression. Remarkably, Anwulignan strongly inhibited tumour growth in vivo. In conclusion, Anwulignan is a novel JAK1 inhibitor that may have therapeutic implications for NSCLC management.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Schisandra/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Carcinoma, Non-Small-Cell Lung , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Lung Neoplasms , Mice , Protein Kinase Inhibitors/chemistry , Xenograft Model Antitumor Assays
7.
J Cell Physiol ; 236(7): 4954-4965, 2021 07.
Article in English | MEDLINE | ID: mdl-33305380

ABSTRACT

Integrin α2ß1 plays an important role in cellular migration and metastasis processes associated with prostate cancer. The aim of this study was to assess whether selective inhibition of integrin α2ß1 is an effective strategy to target metastatic prostate cancer cells. In this regard, we examined the effects of the inhibitor BTT-3033, which selectively interferes with the connection between integrin a2b1 and its ligand, on migration, epithelial-mesenchymal transition (EMT), cell cycle arrest, apoptosis, and specific intracellular signaling pathways using LNcap-FGC and DU-145 prostate cancer cell lines. Western blot analysis and immunocytochemistry assays showed that inhibition of integrin a2b1 inhibits EMT, through the increased expression of E-cadherin and decreased expression of N-cadherin and vimentin. Scratch wound healing assays revealed a direct effect on integrin α2ß1 in the migration capacity of cells. In addition, treatment with BTT-3033 induced a reduction in cell viability and proliferation, as assessed by MTT and BrdU assays. In addition, the results show that BTT-3033 inhibits cell proliferation by inducing G1 cell cycle arrest. Moreover, inhibition of integrin α2ß1 induces apoptosis through the activation of ROS, Bax protein upregulation, caspase-3 activation, and depletion of ΔΨm.  Molecular signaling studies showed that integrin α2ß1 was a positive regulator of MKK7 phosphorylation. In conclusion, our results reveal a critical role for integrin a2b1 in the proliferation of prostate cancer cells, as demonstrated by EMT inhibition, cell cycle arrest, and apoptosis induction in response to treatment with its specific inhibitor BT-3033.


Subject(s)
Apoptosis/physiology , Epithelial-Mesenchymal Transition/physiology , G1 Phase Cell Cycle Checkpoints/drug effects , Integrin alpha2beta1/antagonists & inhibitors , Prostatic Neoplasms/pathology , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Humans , Integrin alpha2beta1/metabolism , MAP Kinase Kinase 7/metabolism , Male , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Phosphorylation , Prostate/pathology , Vimentin/biosynthesis
8.
J Cell Physiol ; 236(3): 2214-2225, 2021 03.
Article in English | MEDLINE | ID: mdl-32783256

ABSTRACT

Retinoblastoma is the most common intraocular cancer with metastatic potential affecting infants and children. Although chemotherapy is available for retinoblastoma, side effects and drug resistance are frequent. Rpl41, encoding ribosomal protein L41 (RPL41), has been identified as a tumor suppressor gene, and its targeted degradation of activating transcription factor 4 (ATF4) produces an antitumor effect. The goal of the present study is to provide experimental evidence for the clinical application of a small peptide regimen in combination with chemotherapy for the treatment of retinoblastoma and to investigate the mechanism of their combined cytotoxicity. It was observed that treatment with the RPL41 peptide alone decreased the viability, migration, and invasion of retinoblastoma Y79 and Weri-Rb1 cells, in addition to promoting cell apoptosis and cell cycle arrest. Furthermore, RPL41 protein levels showed a significantly decreased trend in retinoblastoma specimens, whereas ATF4 protein levels tended to be increased. Mechanistically, ATF4 degradation as a result of RPL41 peptide treatment was observed in retinoblastoma Y79 and Weri-Rb1 cells. Most important, low-dose administration of the RPL41 peptide significantly enhanced the antitumor effect of carboplatin, and further analysis confirmed their synergistic effect as anti-retinoblastoma therapy, indicating that RPL41 sensitized Y79 and Weri-Rb1 retinoblastoma cells to carboplatin. Thus, our data provide a preclinical rationale for the exploration of the RPL41 peptide as a potential adjuvant to carboplatin treatment in retinoblastoma.


Subject(s)
Activating Transcription Factor 4/metabolism , Antineoplastic Agents/pharmacology , Proteolysis , Retinoblastoma/metabolism , Ribosomal Proteins/metabolism , Apoptosis/drug effects , Carboplatin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Neoplasm Invasiveness , Peptides/pharmacology , Proteolysis/drug effects , Retinoblastoma/pathology
9.
Biochem Biophys Res Commun ; 535: 73-79, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33341676

ABSTRACT

The Wnt/ß-catenin pathway is an attractive target for the treatment of acute myelogenous leukemia (AML), since aberrant activation of the Wnt/ß-catenin pathway contributes to carcinogenesis in various types of cancers including AML. Screening of an in-house compound library, constructed at Kyoto Pharmaceutical University, identified a novel compound designated "31" that was found to be an inhibitor of the Wnt/ß-catenin pathway. The compound inhibited T-cell factor (TCF) activity in a TCF firefly luciferase-reporter assay and suppressed the proliferation of several human AML cell lines in a dose-dependent manner. Compound 31 arrested the cell cycle of AML cells at the G1 stage and induced apoptosis. Decrease in protein and mRNA expression level of Wnt pathway-related molecules was confirmed by the analyses of western blotting and quantitative reverse transcription-polymerase chain reaction. In addition, compound 31 combined with idarubicin synergistically inhibited the proliferation of AML cells. In conclusion, these results strongly suggest that compound 31 has potential as a novel anti-AML agent targeting the Wnt/ß-catenin signaling pathway.


Subject(s)
Dipeptides/pharmacology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Wnt Signaling Pathway/drug effects , Antineoplastic Agents/analysis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dipeptides/chemistry , Drug Design , Drug Evaluation, Preclinical , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Idarubicin/pharmacology , Luciferases/metabolism
10.
Genes Cells ; 25(7): 450-465, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32277721

ABSTRACT

Androgens stimulate the proliferation of epithelial cells in the prostate by activating topoisomerase 2 (TOP2) and regulating the transcription of target genes. TOP2 resolves the entanglement of genomic DNA by transiently generating double-strand breaks (DSBs), where TOP2 homodimers covalently bind to 5' DSB ends, called TOP2-DNA cleavage complexes (TOP2ccs). When TOP2 fails to rejoin TOP2ccs generating stalled TOP2ccs, tyrosyl DNA phosphodiesterase-2 (TDP2) removes 5' TOP2 adducts from stalled TOP2ccs prior to the ligation of the DSBs by nonhomologous end joining (NHEJ), the dominant DSB repair pathway in G0 /G1 phases. We previously showed that estrogens frequently generate stalled TOP2ccs in G0 /G1 phases. Here, we show that physiological concentrations of androgens induce several DSBs in individual human prostate cancer cells during G1 phase, and loss of TDP2 causes a five times higher number of androgen-induced chromosome breaks in mitotic chromosome spreads. Intraperitoneally injected androgens induce several DSBs in individual epithelial cells of the prostate in TDP2-deficient mice, even at 20 hr postinjection. In conclusion, physiological concentrations of androgens have very strong genotoxicity, most likely by generating stalled TOP2ccs.


Subject(s)
Androgens/toxicity , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Genomic Instability/genetics , Phosphoric Diester Hydrolases/metabolism , Prostate/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromosome Breakage , DNA End-Joining Repair/drug effects , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , Epithelial Cells/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Genomic Instability/drug effects , Histones/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoric Diester Hydrolases/genetics , Prostate/drug effects , Prostatic Neoplasms/genetics , RNA, Small Interfering , Receptors, Androgen/metabolism
11.
Mol Carcinog ; 60(11): 769-783, 2021 11.
Article in English | MEDLINE | ID: mdl-34437731

ABSTRACT

Although CAMKK2 is overexpressed in several cancers, its role and relevant downstream signaling pathways in gastric cancer (GC) are poorly understood. Treatment of AGS GC cells with a CAMKK2 inhibitor, STO-609, resulted in decreased cell proliferation, cell migration, invasion, colony-forming ability, and G1/S-phase arrest. Quantitative phosphoproteomics in AGS cells with the CAMKK2 inhibitor led to the identification of 9603 unique phosphosites mapping to 3120 proteins. We observed decreased phosphorylation of 1101 phosphopeptides (1.5-fold) corresponding to 752 proteins upon CAMKK2 inhibition. Bioinformatics analysis of hypo-phosphorylated proteins revealed enrichment of MAPK1/MAPK3 signaling. Kinase enrichment analysis of hypo-phosphorylated proteins using the X2K Web tool identified ERK1, cyclin-dependant kinase 1 (CDK1), and CDK2 as downstream substrates of CAMKK2. Moreover, inhibition of CAMKK2 and MEK1 resulted in decreased phosphorylation of ERK1, CDK1, MCM2, and MCM3. Immunofluorescence results were in concordance with our mass spectroscopy data and Western blot analysis results. Taken together, our data reveal the essential role of CAMKK2 in the pathobiology of GC through the activation of the MEK/ERK1 signaling cascade.


Subject(s)
Benzimidazoles/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Naphthalimides/pharmacology , Proteomics/methods , Stomach Neoplasms/metabolism , CDC2 Protein Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromatography, Liquid , Cyclin-Dependent Kinase 2/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Tandem Mass Spectrometry
12.
Microvasc Res ; 135: 104136, 2021 05.
Article in English | MEDLINE | ID: mdl-33450295

ABSTRACT

Reversine, or 2-(4-morpholinoanilino)-6cyclohexylaminopurine, is a 2,6-disubstituted purine derivative. This small molecule exhibits tumor-suppressive activities through different molecular mechanisms. In this study, in vitro and in vivo angiogenic models were used to elucidate the effect of Reversine on angiogenesis in the tumor suppression. Firstly, we grafted osteosarcoma-derived MNNG/HOS cell aggregates onto chick embryonic chorioallantoic membrane (CAM) to examine the vascularization of these grafts following Reversine treatment. Following culture, it was determined that Reversine inhibited MNNG/HOS grafts growth, and decreased the density of blood vessels in the chick CAM. We then used CAM and chick embryonic yolk-sac membrane (YSM) to investigate the effects of Reversine on angiogenesis. The results revealed Reversine inhibited the proliferation of endothelial cells, where cells were mainly arrested at G1/S phase of the cell cycle. Scratch-wound assay with HUVECs revealed that Reversine suppressed cell migration in vitro. Furthermore, endothelial cells tube formation assay and chick aortic arch sprouting assay demonstrated Reversine inhibited the sprouting, migration of endothelial cells. Lastly, qPCR and western blot analyses showed BMP-associated Smad1/5/8 signaling expressions were up-regulated by Reversine treatment. Our results showed that Reversine could suppress tumor growth by inhibiting angiogenesis through BMP signaling, and suggests a potential use of Reversine as an anti-tumor therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bone Morphogenetic Proteins/metabolism , Bone Neoplasms/drug therapy , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Morpholines/pharmacology , Neovascularization, Physiologic/drug effects , Osteosarcoma/drug therapy , Purines/pharmacology , Smad Proteins/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Chick Embryo , G1 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Signal Transduction , Smad Proteins/genetics , Smad1 Protein/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
13.
Arch Biochem Biophys ; 709: 108965, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34129838

ABSTRACT

OBJECTIVE: MicroRNAs (miRNAs) have been demonstrated to be differently expressed in colorectal cancer (CRC) and were identified as biomarkers and therapeutic targets for CRC. We aimed to identify the effect of microRNA-424 (miR-424) on process of CRC. METHODS: Exosomes were obtained from bone marrow mesenchymal stem cells (BMSCs). MiR-424, transforming growth factor-ß receptor 3 (TGFBR3) vimentin, S100A4, p-Smad1 expression in tissues and cells was measured. After treated with miR-424 inhibitor or TGFBR3 overexpression plasmid, the migration, invasion, cell cycle distribution and apoptosis of Lovo cells and exosomes-transfected Lovo cells were determined. The subcutaneous tumor models were established and the tumor growth was observed. The target relation between miR-424 and TGFBR3 was confirmed. RESULTS: MiR-424 was upregulated while TGFBR3 was downregulated in CRC tissues. TGFBR3 was targeted by miR-424. Inhibited miR-424 or elevated TGFBR3 upregulated p-Smad1, indicating that TGFBR3 mediated the Smad1 pathway, thus regulating CRC progression. MiR-424 inhibition or TGFBR3 restoration also suppressed migration and invasion of CRC cells, arrested the CRC cells at G0/G1 phase, and promoted CRC cell apoptosis. Moreover, exosomal miR-424 from BMSCs promoted CRC development. CONCLUSION: Inhibited exosomal miR-424 from BMSCs inhibited malignant behaviors of CRC cells by targeting TGFBR3, thus suppressing the progression of CRC.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Carriers/chemistry , Exosomes/chemistry , Mesenchymal Stem Cells/cytology , MicroRNAs/antagonists & inhibitors , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Down-Regulation/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Middle Aged , RNA, Small Interfering/pharmacology , Up-Regulation/drug effects
14.
BMC Cancer ; 21(1): 1088, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34625047

ABSTRACT

BACKGROUND: The aminoisoquinoline FX-9 shows pro-apoptotic and antimitotic effects against lymphoblastic leukemia cells and prostate adenocarcinoma cells. In contrast, decreased cytotoxic effects against non-neoplastic blood cells, chondrocytes, and fibroblasts were observed. However, the actual FX-9 molecular mode of action is currently not fully understood. METHODS: In this study, microarray gene expression analysis comparing FX-9 exposed and unexposed prostate cancer cells (PC-3 representing castration-resistant prostate cancer), followed by pathway analysis and gene annotation to functional processes were performed. Immunocytochemistry staining was performed with selected targets. RESULTS: Expression analysis revealed 0.83% of 21,448 differential expressed genes (DEGs) after 6-h exposure of FX-9 and 0.68% DEGs after 12-h exposure thereof. Functional annotation showed that FX-9 primarily caused an activation of inflammatory response by non-canonical nuclear factor-kappa B (NF-κB) signaling. The 6-h samples showed activation of the cell cycle inhibitor CDKN1A which might be involved in the secondary response in 12-h samples. This secondary response predominantly consisted of cell cycle-related changes, with further activation of CDKN1A and inhibition of the transcription factor E2F1, including downstream target genes, resulting in G1-phase arrest. Matching our previous observations on cellular level senescence signaling pathways were also found enriched. To verify these results immunocytochemical staining of p21 Waf1/Cip1 (CDKN1A), E2F1 (E2F1), PAI-1 (SERPNE1), and NFkB2/NFkB p 100 (NFKB2) was performed. Increased expression of p21 Waf1/Cip1 and NFkB2/NFkB p 100 after 24-h exposure to FX-9 was shown. E2F1 and PAI-1 showed no increased expression. CONCLUSIONS: FX-9 induced G1-phase arrest of PC-3 cells through activation of the cell cycle inhibitor CDKN1A, which was initiated by an inflammatory response of noncanonical NF-κB signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Isoquinolines/pharmacology , NF-kappa B/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Antineoplastic Agents/therapeutic use , E2F1 Transcription Factor/antagonists & inhibitors , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression/drug effects , Gene Expression Profiling/methods , Humans , Isoquinolines/therapeutic use , Male , Middle Aged , PC-3 Cells , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , S Phase Cell Cycle Checkpoints , Time Factors , Tissue Array Analysis
15.
BMC Cancer ; 21(1): 1061, 2021 Sep 26.
Article in English | MEDLINE | ID: mdl-34565342

ABSTRACT

BACKGROUND: Neuroblastoma (NB) patients with MYCN amplification or overexpression respond poorly to current therapies and exhibit extremely poor clinical outcomes. PI3K-mTOR signaling-driven deregulation of protein synthesis is very common in NB and various other cancers that promote MYCN stabilization. In addition, both the MYCN and mTOR signaling axes can directly regulate a common translation pathway that leads to increased protein synthesis and cell proliferation. However, a strategy of concurrently targeting MYCN and mTOR signaling in NB remains unexplored. This study aimed to investigate the therapeutic potential of targeting dysregulated protein synthesis pathways by inhibiting the MYCN and mTOR pathways together in NB. METHODS: Using small molecule/pharmacologic approaches, we evaluated the effects of combined inhibition of MYCN transcription and mTOR signaling on NB cell growth/survival and associated molecular mechanism(s) in NB cell lines. We used two well-established BET (bromodomain extra-terminal) protein inhibitors (JQ1, OTX-015), and a clinically relevant mTOR inhibitor, temsirolimus, to target MYCN transcription and mTOR signaling, respectively. The single agent and combined efficacies of these inhibitors on NB cell growth, apoptosis, cell cycle and neurospheres were assessed using MTT, Annexin-V, propidium-iodide staining and sphere assays, respectively. Effects of inhibitors on global protein synthesis were quantified using a fluorescence-based (FamAzide)-based protein synthesis assay. Further, we investigated the specificities of these inhibitors in targeting the associated pathways/molecules using western blot analyses. RESULTS: Co-treatment of JQ1 or OTX-015 with temsirolimus synergistically suppressed NB cell growth/survival by inducing G1 cell cycle arrest and apoptosis with greatest efficacy in MYCN-amplified NB cells. Mechanistically, the co-treatment of JQ1 or OTX-015 with temsirolimus significantly downregulated the expression levels of phosphorylated 4EBP1/p70-S6K/eIF4E (mTOR components) and BRD4 (BET protein)/MYCN proteins. Further, this combination significantly inhibited global protein synthesis, compared to single agents. Our findings also demonstrated that both JQ1 and temsirolimus chemosensitized NB cells when tested in combination with cisplatin chemotherapy. CONCLUSIONS: Together, our findings demonstrate synergistic efficacy of JQ1 or OTX-015 and temsirolimus against MYCN-driven NB, by dual-inhibition of MYCN (targeting transcription) and mTOR (targeting translation). Additional preclinical evaluation is warranted to determine the clinical utility of targeted therapy for high-risk NB patients.


Subject(s)
Acetanilides/pharmacology , Azepines/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/drug therapy , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation , Drug Synergism , Eukaryotic Initiation Factor-4E/drug effects , Eukaryotic Initiation Factor-4E/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Sirolimus/pharmacology , Spheroids, Cellular/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
16.
Pharmacol Res ; 173: 105870, 2021 11.
Article in English | MEDLINE | ID: mdl-34500061

ABSTRACT

Colorectal cancer (CRC) is one of the most common causes of cancer-related death worldwide. Nannocystin ax (NAN), a 21-membered cyclodepsipeptide initially isolated from myxobacteria of the Nannocystis genus, was found to target the eukaryotic elongation factor 1A (eEF1A). The current study was designed to evaluate the anticancer effect and underlying mechanisms of NAN with in vitro and in vivo models. Results showed that NAN induced G1 phase cell cycle arrest and caspase-independent apoptosis in HCT116 and HT29 human CRC cells. NAN significantly downregulated cyclin D1 level in a short time, but NAN did not affect the transcription level and ubiquitin-dependent degradation of cyclin D1. Furthermore, NAN treatment directly targeted eEF1A and partially decreased the synthesis of new proteins, contributing to the downregulation of cyclin D1. Besides, NAN significantly suppressed tumor growth in the zebrafish xenograft model. In conclusion, NAN triggered G1 phase cell cycle arrest through cyclin D1 downregulation and eEF1A-targeted translation inhibition and promoted caspase-independent apoptosis in CRC cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Depsipeptides/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Depsipeptides/pharmacology , Down-Regulation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , HCT116 Cells , HT29 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Peptide Elongation Factor 1/genetics , Zebrafish
17.
Bioorg Med Chem Lett ; 40: 127965, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33744442

ABSTRACT

Small molecule inhibitors of the focal adhesion kinase are regarded as promising tools in our armamentarium for treating cancer. Here, we identified four 1,2,4-triazole derivatives that inhibit FAK kinase significantly and evaluated their therapeutic potential. Most tested compounds revealed potent antiproliferative activity in HepG2 and Hep3B liver cancer cells, in which 3c and 3d were the most potent (IC50 range; 2.88 ~ 4.83 µM). Compound 3d possessed significant FAK inhibitory activity with IC50 value of 18.10 nM better than the reference GSK-2256098 (IC50 = 22.14 nM). The preliminary mechanism investigation by Western blot analysis showed that both 3c and 3d repressed FAK phosphorylation comparable to GSK-2256098 in HepG2 cells. As a result of FAK inhibition, 3c and 3d inhibited the pro-survival pathways by decreasing the phosphorylation levels of PI3K, Akt, JNK, and STAT3 proteins. This effect led to apoptosis induction and cell cycle arrest. Taken together, these results indicate that 3d could serve as a potent preclinical candidate for the treatment of cancers.


Subject(s)
Acetanilides/pharmacology , Aminobenzoates/pharmacology , Antineoplastic Agents/pharmacology , Focal Adhesion Kinase 1/metabolism , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , Acetanilides/chemical synthesis , Aminobenzoates/chemical synthesis , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Focal Adhesion Kinase 1/chemistry , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , S Phase Cell Cycle Checkpoints/drug effects , Triazoles/chemical synthesis
18.
Inorg Chem ; 60(6): 3939-3951, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33657313

ABSTRACT

Palladacycles are versatile organometallic compounds that show potential for therapeutic use. Here are described the synthesis and characterization of mono- and dinuclear palladacycles bearing diphosphines. Their biological effect was investigated in A2780, an ovarian-derived cancer line, and in normal dermal fibroblasts. The compounds displayed selective cytotoxicity toward the A2780 cell line. Compound 3 decreased the cell viability through cell cycle retention in G0/G1, triggered apoptosis through the intrinsic pathway, and induced autophagy in A2780 cells. Compound 9 also induced cell cycle retention, apoptosis, and cellular detachment. Notably, compound 9 induced the production of intracellular reactive oxygen species (ROS). Our work demonstrated that compound 3 enters A2780 cells via active transport, which requires energy, while compound 9 enters A2780 cells mostly passively. The potential effect of palladacycles in angiogenesis was investigated for the first time in an in vivo chorioallantoic membrane model, showing that while compound 3 displayed an antiangiogenic effect crucial to fighting cancer progression, compound 9 promoted angiogenesis. These results show that palladacycles may be used in different clinical applications where pro- or antiangiogenic effects may be desirable.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Coordination Complexes/pharmacology , Organometallic Compounds/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chickens , Coordination Complexes/chemical synthesis , Embryo, Nonmammalian/drug effects , Female , Fibroblasts/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Organometallic Compounds/chemical synthesis , Ovarian Neoplasms/drug therapy , Palladium/chemistry , Reactive Oxygen Species/metabolism
19.
Bioorg Med Chem ; 37: 116107, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33735799

ABSTRACT

Triple-negative breast cancer (TNBC) is one of the most aggressive cancer with high mortality and recurrence rates. Hecogenin, a steroidal sapogenin, is reported as a potential anti-tumor agent against breast cancer. However, the moderate activity limits its further application in clinical. With the aim to identify novel analogues that are especially efficacious in therapy of TNBC, a series of novel hecogenin thiosemicarbazone and semicarbazone derivatives were designed, synthesized and biologically evaluated. Screening of cytotoxicity revealed that 4c could potently inhibit the proliferation of breast cancer cells (MCF-7 and MDA-MB-231 cells), lung cancer cells (A549) and colon cancer cells (HT-29) at low µM level. Importantly, further mechanism studies indicated the ability of 4c in inducing apoptosis of MDA-MB-231 cells by arresting the cell cycle. Moreover, 4c notably suppressed the migration and invasion of MDA-MB-231 cells compared to its parent hecogenin at the equal concentration.


Subject(s)
Antineoplastic Agents/pharmacology , Sapogenins/pharmacology , Thiosemicarbazones/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Sapogenins/chemical synthesis , Sapogenins/toxicity , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/toxicity
20.
Bioorg Med Chem ; 37: 116097, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33743356

ABSTRACT

Naproxen is a common non-steroidal anti-inflammatory drug, which is the most usually used propionic acid derivative for the treatment of many types of diseases. In this study, a series of novel (S)-Naproxen derivatives bearing hydrazide-hydrazone moiety were designed, synthesized, and evaluated for anticancer activity. The structures of these compounds were characterized by spectral (1H-13C NMR, FT-IR, and HR-MS analyses) methods. All synthesized compounds were screened for anticancer activity against two different human breast cancer cell lines (MDA-MB-231 and MCF-7). Among them, (S)-2-(6-methoxynaphthalen-2-yl)-N'-{(E)-[2-(trifluoromethoxy)phenyl]methylidene} propanehydrazide (3a) showed the most potent anticancer activity against both cancer cell lines with a good selectivity (IC50 = 22.42 and 59.81 µM, respectively). Furthermore, the molecular modeling of these compounds was studied on Vascular Endothelial Growth Factor Receptor 2. Inhibition of VEGFR-2 and apoptotic protein Bcl-2 was investigated in MDA-MB-231 cells treated with compound 3a by using Western Blotting. Apoptosis was also detected by staining with DAPI in fluorescence microscopy. Flow Cytometry analyses related to cell cycle phases showed that a dramatic increase in S and M phases was established compared to untreated control cells indicating the cancer cell cycle arrest. The anticancer activity of compound 3a was investigated in the Ehrlich acid tumor model, a well-validated in vivo ectopic breast cancer model, in mice. Our results showed that compound 3a had anticancer activity and decreased the tumor volume in both low (60 mg/kg) and high (120 mg/kg) doses in mice.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Hydrazones/therapeutic use , Naproxen/analogs & derivatives , Naproxen/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Hydrazones/chemical synthesis , Hydrazones/metabolism , Hydrazones/pharmacology , Mice, Inbred BALB C , Molecular Docking Simulation , Naproxen/metabolism , Naproxen/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL