Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
FEBS Lett ; 595(14): 1949-1961, 2021 07.
Article in English | MEDLINE | ID: mdl-34056710

ABSTRACT

In embryonic stem (ES) cells, oxidative stress control is crucial for genomic stability, self-renewal, and cell differentiation. Heme oxygenase-1 (HO-1) is a key player of the antioxidant system and is also involved in stem cell differentiation and pluripotency acquisition. We found that the HO-1 gene is expressed in ES cells and induced after promoting differentiation. Moreover, downregulation of the pluripotency transcription factor (TF) OCT4 increased HO-1 mRNA levels in ES cells, and analysis of ChIP-seq public data revealed that this TF binds to the HO-1 gene locus in pluripotent cells. Finally, ectopic expression of OCT4 in heterologous systems repressed a reporter carrying the HO-1 gene promoter and the endogenous gene. Hence, this work highlights the connection between pluripotency and redox homeostasis.


Subject(s)
Gene Expression Regulation , Heme Oxygenase-1/genetics , Membrane Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heme Oxygenase-1/metabolism , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , NIH 3T3 Cells , Nanog Homeobox Protein/antagonists & inhibitors , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Promoter Regions, Genetic , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transcription, Genetic
2.
Int J Biol Macromol ; 180: 608-624, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33662423

ABSTRACT

Gastric cancer (GC) is a leading cause of death worldwide. GC is the third-most common cause of cancer-related death after lung and colorectal cancer. It is also the fifth-most commonly diagnosed cancer. Accumulating evidence has revealed the role of signaling networks in GC progression. Identification of these molecular pathways can provide new insight into therapeutic approaches for GC. Several molecular factors involved in GC can play both onco-suppressor and oncogene roles. Sex-determining region Y (Sry)-box-containing (SOX) family members are transcription factors with a well-known role in cancer. SOX proteins can bind to DNA to regulate cellular pathways via a highly conserved domain known as high mobility group (HMG). In the present review, the roles of SOX proteins in the progression and/or inhibition of GC are discussed. The dual role of SOX proteins as tumor-promoting and tumor-suppressing factors is highlighted. SOX members can affect upstream mediators (microRNAs, long non-coding RNAs and NF-κB) and down-stream mediators (FAK, HIF-1α, CDX2 and PTEN) in GC. The possible role of anti-tumor compounds to target SOX pathway members in GC therapy is described. Moreover, SOX proteins may be used as diagnostic or prognostic biomarkers in GC.


Subject(s)
Gene Expression Regulation, Neoplastic , SOX Transcription Factors/genetics , Signal Transduction/genetics , Stomach Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Disease Progression , Humans , Molecular Targeted Therapy/methods , SOX Transcription Factors/antagonists & inhibitors , SOX Transcription Factors/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
3.
Signal Transduct Target Ther ; 5(1): 135, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728033

ABSTRACT

SOX2 is a well-characterized pluripotent factor that is essential for stem cell self-renewal, reprogramming, and homeostasis. The cellular levels of SOX2 are precisely regulated by a complicated network at the levels of transcription, post-transcription, and post-translation. In many types of human cancer, SOX2 is dysregulated due to gene amplification and protein overexpression. SOX2 overexpression is associated with poor survival of cancer patients. Mechanistically, SOX2 promotes proliferation, survival, invasion/metastasis, cancer stemness, and drug resistance. SOX2 is, therefore, an attractive anticancer target. However, little progress has been made in the efforts to discover SOX2 inhibitors, largely due to undruggable nature of SOX2 as a transcription factor. In this review, we first briefly introduced SOX2 as a transcription factor, its domain structure, normal physiological functions, and its involvement in human cancers. We next discussed its role in embryonic development and stem cell-renewal. We then mainly focused on three aspects of SOX2: (a) the regulatory mechanisms of SOX2, including how SOX2 level is regulated, and how SOX2 cross-talks with multiple signaling pathways to control growth and survival; (b) the role of SOX2 in tumorigenesis and drug resistance; and (c) current drug discovery efforts on targeting SOX2, and the future perspectives to discover specific SOX2 inhibitors for effective cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Drug Discovery , Neoplasm Proteins , Neoplasms , SOXB1 Transcription Factors , Animals , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/metabolism
4.
Int J Mol Sci ; 21(12)2020 Jun 20.
Article in English | MEDLINE | ID: mdl-32575737

ABSTRACT

PURPOSE: Human corneal endothelial cells (hCECs) pump out water from the stroma and maintain the clarity of the cornea. The sex-determining region Y-box 2 (SOX2) participates in differentiation during the development of the anterior segment of the eye and is found in the periphery of wounded corneas. This study was performed to investigate the effect of SOX2 repression on hCECs. METHODS: Cultured hCECs were transfected by siRNA for SOX2. The wound healing rate and cell viability were measured. The cell proliferation-associated protein level was evaluated by Western blotting and RT-PCR. The energy production and mitochondrial function were measured, and cell shape and WNT signaling were assessed. RESULTS: Upon transfecting the cultured cells with siRNA for SOX2, the SOX2 level was reduced by 80%. The wound healing rate and viability were also reduced. Additionally, CDK1, cyclin D1, SIRT1, and ATP5B levels were reduced, and CDKN2A and pAMPK levels were increased. Mitochondrial oxidative stress and mitochondrial viability decreased, and the cell shape became elongated. Furthermore, SMAD1, SNAI1, WNT3A, and ß-catenin levels were increased. CONCLUSION: SOX2 repression disrupts the normal metabolism of hCECs through modulating WNT signaling and mitochondrial functions.


Subject(s)
Endothelium, Corneal/cytology , Mitochondria/metabolism , RNA, Small Interfering/pharmacology , SOXB1 Transcription Factors/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Corneal/drug effects , Endothelium, Corneal/metabolism , Energy Metabolism , Humans , Mitochondria/drug effects , Oxidative Stress , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/metabolism , Transfection , Wnt Signaling Pathway/drug effects
5.
In Vivo ; 34(3): 1085-1094, 2020.
Article in English | MEDLINE | ID: mdl-32354896

ABSTRACT

BACKGROUND/AIM: Cervical cancer is one of the leading causes of cancer death in women worldwide. BRM270 (BRMLife) has therapeutic potential for cancer treatment owing to its ability to inhibit cell proliferation, and expression of cluster of differentiation (CD) 133 in CD133+ cancer cells. This study was designed to evaluate the therapeutic effects of plant extract formulation BRM270 against cervical cancer progression. MATERIALS AND METHODS: The expression of sex-determining region Y-box 2 (SOX2) was tested in four different cervical cancer cell lines, HeLA, SiHa, Caski and C33A. SOX2-expressing SiHa and C33A cell lines were selected for further experiments on the in vitro and in vivo effects of BRM270 on cervical cancer progression using western blotting, flow cytometry, sphere-formation assay, magnetic-activated cell sorting of CD133+ cervical cancer cells, and xenografts in female athymic BALB/c nude mice. RESULTS: In the present study, in cervical cancer stem cells (CSCs), we found that BRM270 inhibited expression of SOX2, which is associated with cervical cancer initiation and metastasis. BRM270 also inhibited CD133 expression and induced apoptosis of CSCs and suppressed CD133+ CSC proliferation and sphere formation in vitro as well as SiHa and C33A cell xenograft tumor growth in vivo. This was accompanied by down-regulation of markers of epithelial-to-mesenchymal transition. CONCLUSION: BRM270 might be an effective agent for cervical cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Chinese Herbal/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Mice , Neoplastic Stem Cells/pathology , Uterine Cervical Neoplasms/genetics , Xenograft Model Antitumor Assays
6.
Anticancer Agents Med Chem ; 20(10): 1221-1232, 2020.
Article in English | MEDLINE | ID: mdl-32116204

ABSTRACT

BACKGROUND: Targeting DNA mismatch repair-deficient/KRAS-mutant Colorectal Cancer Stem Cells (CRCSCs) with chemical compounds remains challenging. Modulating stemness factors Bmi-1, Sox-2, Oct-4 and Nanog in CRCSCs which are direct downstream targets of carcinogenesis pathways may lead to the reactivation of caspase-3 and apoptosis in these cells. Omega-3 DHA modulates different signaling pathways involved in carcinogenesis. However, little is known, whether in vitro concentrations of DHA equal to human plasma levels are able to modulate pluripotency genes expression, caspase-3 reactivation and apoptosis in DNA mismatch repair-deficient/KRAS-mutant CRC stem-like cells. METHODS: DNA mismatch repair-deficient/KRAS-mutant CRC stem-like cells (LS174T cells) were treated with DHA, after which, cell number and proliferation-rate, Bmi-1, Sox-2, Nanog and Oct-4 expression, caspase-3 activation and apoptosis were evaluated with different cellular and molecular techniques. RESULTS: DHA changed the morphology of cells to apoptotic forms and disrupted cell connections. After 48h treatment with 50- to 200µM DHA, cell numbers and proliferation-rates were measured to be 86%-35% and 93.6%-45.7% respectively. Treatment with 200 µM DHA dramatically decreased the expression of Bmi-1, Sox- 2, Oct-4 and Nanog by 69%, 70%, 97.5% and 53% respectively. Concurrently, DHA induced caspase-3 activation by 1.8-4.7-fold increases compared to untreated cells. An increase in the number of apoptotic cells ranging from 9.3%-38.4% was also observed with increasing DHA concentrations. CONCLUSIONS: DHA decreases the high expression level of pluripotency network genes suggesting Bmi-1, Sox-2, Oct-4 and Nanog as promising molecular targets of DHA. DHA reactivates caspase-3 and apoptosis in DNA mismatch repair-deficient/KRAS-mutant CRC stem-like cells, representing the high potential of this safe compound for therapeutic application in CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Colorectal Neoplasms/drug therapy , Fatty Acids, Omega-3/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Mismatch Repair/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fatty Acids, Omega-3/chemistry , Humans , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Molecular Structure , Mutation , Nanog Homeobox Protein/antagonists & inhibitors , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Theranostics ; 10(7): 3151-3163, 2020.
Article in English | MEDLINE | ID: mdl-32194860

ABSTRACT

Up to one-third of diffuse large B cell lymphoma (DLBCL) patients eventually develop resistance to R-CHOP regimen, while the remaining therapeutic options are limited. Thus, understanding the underlying mechanisms and developing therapeutic approaches are urgently needed. Methods: We generated two germinal center B cell-like (GCB) and activated B cell-like (ABC) subtype R-CHO resistant DLBCL cell lines, of which the tumor-initiating capacity was evaluated by serial-transplantation and stemness-associated features including CD34 and CD133 expression, side population and ALDH1 activity were detected by flow cytometry or immunoblotting. Expression profiles of these resistant cells were characterized by RNA sequencing. The susceptibility of resistant cells to different treatments was evaluated by in vitro CytoTox-glo assay and in tumor-bearing mice. The expression levels of SOX2, phos-AKT, CDK6 and FGFR1/2 were detected in 12 R-CHOP-resistant DLBCL clinical specimens by IHC. Results: The stem-like CSC proportion significantly increased in both resistant DLBCL subtypes. SOX2 expression level remarkably elevated in both resistant cell lines due to its phosphorylation by activated PI3K/AKT signaling, thus preventing ubiquitin-mediated degradation. Further, multiple factors, including BCR, integrins, chemokines and FGFR1/2 signaling, regulated PI3K/AKT activation. CDK6 in GCB subtype and FGFR1/2 in ABC subtype were SOX2 targets, whose inhibition potently re-sensitized resistant cells to R-CHOP treatment. More importantly, addition of PI3K inhibitor to R-CHOP completely suppressed the tumor growth of R-CHO-resistant DLBCL cells, most likely by converting CSCs to chemo-sensitive differentiated cells. Conclusions: The PI3K/AKT/SOX2 axis plays a critical role in R-CHOP resistance development and the pro-differentiation therapy against CSCs proposed in this study warrants further study in clinical trials for the treatment of resistant DLBCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , SOXB1 Transcription Factors/antagonists & inhibitors , Aminopyridines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Benzimidazoles/pharmacology , Cell Differentiation/drug effects , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoquinolines/pharmacology , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Mice, SCID , Neoplasm Proteins/analysis , Neoplastic Stem Cells/drug effects , Phosphorylation , Piperazines/pharmacology , Prednisone/administration & dosage , Protein Processing, Post-Translational/drug effects , Purines/pharmacology , Pyrazoles/pharmacology , Rituximab/administration & dosage , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Transcription Factors/analysis , Ubiquitination , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
8.
Dev Biol ; 461(2): 160-171, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32059837

ABSTRACT

In amniotes, unlike primary neurulation in the anterior body, secondary neurulation (SN) proceeds along with axial elongation by the mesenchymal-to-epithelial transition of SN precursors in the tail bud. It has been under debate whether the SN is generated by neuromesodermal common progenitor cells (NMPs) or neural restricted lineage. Our direct cell labeling and serial transplantations identify uni-fated (neural) precursors in the early tail bud. The uni-fated SN precursor territory is further divided into two subpopulations, neural-differentiating and self-renewing cells, which are regulated by high- and low levels of Sox2, respectively. Unexpectedly, uni-fated SN precursors change their fate at later stages to produce both SN and mesoderm. Thus, chicken embryos adopt a previously unappreciated prolonged phase with uni-fated SN stem cells in the early tail bud, which is absent or very limited in mouse embryos.


Subject(s)
Cell Self Renewal/physiology , Chickens/genetics , Neural Stem Cells/cytology , Neural Tube/embryology , Neurulation/physiology , SOXB1 Transcription Factors/physiology , Tail/embryology , Animals , Cell Lineage , Chick Embryo , Genes, Reporter , Mesoderm/cytology , Neural Tube/cytology , Neurulation/genetics , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics , Tail/cytology
9.
Mol Ther ; 28(3): 901-913, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31991109

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a predominant cancer type in developing countries such as China, where ESCC accounts for approximately 90% of esophageal malignancies. Lacking effective and targeted therapy contributes to the poor 5-year survival rate. Recent studies showed that about 30% of ESCC cases have high levels of SOX2. Herein, we aim to target this transcription factor with aptamer. We established a peptide aptamer library and then performed an unbiased screening to identify several peptide aptamers including P42 that can bind and inhibit SOX2 downstream target genes. We further found that P42 overexpression or incubation with a synthetic peptide 42 inhibited the proliferation, migration, and invasion of ESCC cells. Moreover, peptide 42 treatment inhibited the growth and metastasis of ESCC xenografts in mouse and zebrafish. Further analysis revealed that P42 overexpression led to alternations in the levels of proteins that are important for the proliferation and migration of ESCC cells. Taken together, our study identified the peptide 42 as a key inhibitor of SOX2 function, reducing the proliferation and migration of ESCC cells in vitro and in vivo, and thereby offering a potential therapy against ESCC.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Peptide/pharmacology , SOXB1 Transcription Factors/antagonists & inhibitors , Animals , Aptamers, Peptide/chemistry , Aptamers, Peptide/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/mortality , Humans , Mice , Molecular Targeted Therapy , Prognosis , Protein Binding , SELEX Aptamer Technique , SOXB1 Transcription Factors/metabolism , Xenograft Model Antitumor Assays , Zebrafish
10.
J Mol Cell Biol ; 12(2): 85-98, 2020 02 20.
Article in English | MEDLINE | ID: mdl-30517668

ABSTRACT

Cancer stem cells (CSCs), a minor subpopulation of tumor bulks with self-renewal and seeding capacity to generate new tumors, posit a significant challenge to develop effective and long-lasting anti-cancer therapies. The emergence of drug resistance appears upon failure of chemo-/radiation therapy to eradicate the CSCs, thereby leading to CSC-mediated clinical relapse. Accumulating evidence suggests that transcription factor SOX2, a master regulator of embryonic and induced pluripotent stem cells, drives cancer stemness, fuels tumor initiation, and contributes to tumor aggressiveness through major drug resistance mechanisms like epithelial-to-mesenchymal transition, ATP-binding cassette drug transporters, anti-apoptotic and/or pro-survival signaling, lineage plasticity, and evasion of immune surveillance. Gaining a better insight and comprehensive interrogation into the mechanistic basis of SOX2-mediated generation of CSCs and treatment failure might therefore lead to new therapeutic targets involving CSC-specific anti-cancer strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , SOXB1 Transcription Factors/genetics , Signal Transduction , Tumor Escape
11.
Cell Death Differ ; 27(2): 632-645, 2020 02.
Article in English | MEDLINE | ID: mdl-31243344

ABSTRACT

Sex-determining region Y-box 2 (SOX2), a well-known stemness biomarker, is highly expressed in a variety of cancers, including human highly invasive bladder cancer (BC). However, the role of SOX2 may vary in different kinds of malignancy. In the present study, we discovered that ChlA-F, a novel conformation derivative of isolate Cheliensisin A (Chel A), remarkably inhibits the invasive ability of human invasive BC cells through downregulation of SOX2 protein expression. We found that ChlA-F treatment dramatically decreases SOX2 protein expression in human high-grade invasive BC cells. Ectopic expression of SOX2 reversed ChlA-F inhibition of cell invasion ability in human bladder cancer cells, suggesting that SOX2 is a major target of ChlA-F during its inhibition of human BC invasion. Mechanistic studies revealed that ChlA-F downregulates SOX2 at both the protein degradation and protein translation levels. Further studies revealed that ChlA-F treatment induces HuR protein expression and that the increased HuR interacts with USP8 mRNA, resulting in elevation of USP8 mRNA stability and protein expression. Elevated USP8 subsequently acts as an E3 ligase to promote SOX2 ubiquitination and protein degradation. We also found that ChlA-F treatment substantially increases c-Jun phosphorylation at Ser63 and Ser73, initiating miR-200c transcription. The increased miR-200c directly binds to the 3'-UTR of SOX2 mRNA to suppress SOX2 protein translation. These results present novel mechanistic insight into understanding SOX2 inhibition upon ChlA-F treatment and provide important information for further exploration of ChlA-F as a new therapeutic compound for the treatment of highly invasive/metastatic human BC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Lactones/pharmacology , SOXB1 Transcription Factors/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Down-Regulation/drug effects , Humans , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tumor Cells, Cultured , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
12.
Bioorg Med Chem ; 28(3): 115248, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31879179

ABSTRACT

Metastasis, a deadly feature of cancer, compromises the prognosis and accounts for mortality in the majority of cancer patients. SOX2, a well-known pluripotency transcription factor, plays a central role in cell fate determination and has an overlapping role as a regulatory factor in tumorigenesis and metastasis. The demand is increasing for clinically useful strategies for artificial control of SOX2 expression and its complex transcription machinery in cancer cells. N-Methylpyrrole (Py) and N-methylimidazole (Im) polyamides are small programmable designer ligands that can be pre-programmed to selectively recognize DNA sequence and control endogenous gene expression. Herein, we evaluated the anticancer activity of a designer ligand (SOX2i). SOX2i remarkably altered the expression of SOX2 at the mRNA and protein level in human cancer cell lines such as SW620 (colorectal adenocarcinoma), MKN45 (gastric adenocarcinoma), MCF7 (breast carcinoma), U2OS (osteosarcoma) and other cancer cell lines of different origin and type. Genome-wide transcriptome analysis and cell-based assays showed SOX2 to be a downregulated upstream regulator that alters cell proliferation, cell cycle progression, metabolism and apoptotic pathway. Studies in the mouse model confirmed the anti-metastatic property of SOX2i. SOX2i inhibited the expression of genes associated with EMT and stemness. Moreover, Wnt-canonical signaling was found to be downregulated in the SOX2i-treated group. Our proof-of-concept study supports the potential of DNA-based programmable small molecules for controlling the key regulatory factors associated with tumorigenesis and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Nylons/pharmacology , Pyrroles/pharmacology , SOXB1 Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemistry , Mice , Molecular Structure , Nylons/chemical synthesis , Nylons/chemistry , Pyrroles/chemistry , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Biomed Pharmacother ; 121: 109602, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31707349

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers in China, accompanied by an extremely high mortality rate. Chlorogenic acid (CGA) is a small-molecule compound, that has been shown to have a wide range of biological activities, including antitumor. However, the efficacy and molecular mechanism of CGA on ESCC remains unknown. In this study, we confirmed the inhibition of proliferation by CGA in ESCC cells, as well as the reduction of ESCC xenograft volume by CGA in vivo. In addition, CGA also suppressed both the migration and invasion of ESCC cells in vitro. In a carcinogen-induced murine model of ESCC, hyperplasia of the esophagus was slowed by CGA, while mice suffering from ESCC that were treated with CGA had longer survival times than mice in the control group. The measurement of pluripotency factors (BMI1, SOX2, OCT4 and Nanog) that are related to poor prognosis revealed reduced expression of both BMI1 and SOX2, but not of OCT4 or Nanog, in ESCC cells, in both a dose- and time-dependent manner. Together, our initial findings demonstrate that CGA suppresses ESCC progression, downregulates the expression of BMI1 and SOX2, and provide an anti-tumor candidate for ESCC therapy.


Subject(s)
Chlorogenic Acid/therapeutic use , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Growth Inhibitors/therapeutic use , Polycomb Repressive Complex 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Animals , Cell Line , Cell Line, Tumor , Chlorogenic Acid/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Growth Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics
15.
Biochem Biophys Res Commun ; 520(3): 586-593, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31623830

ABSTRACT

Accepted as crucial participators in human malignancies, long noncoding RNAs (lncRNAs) have been proven to exert significant function on the complicated processes of cancer progression. Although existing investigations have revealed the oncogenic role of lncRNA SOX2 overlapping transcript (SOX2-OT) in different kinds of cancers, such as osteosarcoma and cholangiocarcinoma, the potential role of it in prostate cancer (PC) is poorly understood. This study was the first attempt to decipher the underlying regulatory mechanism of SOX2-OT in PC. According to the data from this study, SOX2-OT expression was conspicuously elevated in PC tissues and cells. Silenced SOX2-OT could repress PC cell proliferation and migration. Besides, mechanism assays manifested that SOX2-OT bound with miR-369-3p and negatively correlated with miR-369-3p in PC. Additionally, miR-369-3p was confirmed to elicit suppressive impact on PC progression. What's more, cofilin 2 (CFL2) was testified to be a downstream target gene of miR-369-3p. Final rescue tests uncovered that CFL2 upregulation or miR-369-3p inhibition could largely restore SOX2-OT knockdown-mediated function on PC progression. To sum up, SOX2-OT accelerates cell proliferation and migration by targeting miR-369-3p/CFL2 axis in PC.


Subject(s)
Cofilin 2/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Long Noncoding/genetics , SOXB1 Transcription Factors/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cofilin 2/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/metabolism
16.
BMJ Case Rep ; 12(7)2019 Jul 16.
Article in English | MEDLINE | ID: mdl-31315840

ABSTRACT

Paraneoplastic syndromes (PS) are a rare presentation of cancer, most commonly associated with small cell lung cancer (SCLC), breast cancer and haematologic malignancies. The diagnosis of PS is challenging because it could affect multiple organ systems and it may present before the tumour is visible by imaging. We report a malignant tumour diagnosed in a male patient who referred long-term paraesthesia and proximal muscle strength loss. After ruling out common causes of polyneuropathy, the anti-SOX1 antibody gave light to the diagnosis. A pulmonary opacity in the upper right lobe was observed in the chest X-ray and a pulmonary tumour was later confirmed by CT scan. The biopsy of the cervical lymphadenopathy determined an SCLC, which caused a PS called Lambert-Eaton myasthenic syndrome (LEMS). Our case raises awareness of a rare PS presentation, which can be diagnosed by specific antibodies, allowing early diagnosis and treatment of lung cancer.


Subject(s)
Lambert-Eaton Myasthenic Syndrome/blood , Paraneoplastic Syndromes/diagnosis , Paresthesia/etiology , SOXB1 Transcription Factors/antagonists & inhibitors , Small Cell Lung Carcinoma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autoantibodies/blood , Diagnosis, Differential , Electromyography/methods , Humans , Lambert-Eaton Myasthenic Syndrome/complications , Lambert-Eaton Myasthenic Syndrome/diagnosis , Male , Paraneoplastic Syndromes/physiopathology , SOXB1 Transcription Factors/blood , Sentinel Lymph Node Biopsy/methods , Small Cell Lung Carcinoma/complications , Small Cell Lung Carcinoma/diagnostic imaging , Small Cell Lung Carcinoma/drug therapy , Tomography, X-Ray Computed/methods , Treatment Outcome
17.
Nutrients ; 11(7)2019 Jul 19.
Article in English | MEDLINE | ID: mdl-31331073

ABSTRACT

The lysine-specific histone demethylase 1A (LSD1) also known as lysine (K)-specific demethylase 1A (KDM1A) is a central epigenetic regulator of metabolic reprogramming in obesity-associated diseases, neurological disorders, and cancer. Here, we evaluated the ability of oleacein, a biophenol secoiridoid naturally present in extra virgin olive oil (EVOO), to target LSD1. Molecular docking and dynamic simulation approaches revealed that oleacein could target the binding site of the LSD1 cofactor flavin adenosine dinucleotide with high affinity and at low concentrations. At higher concentrations, oleacein was predicted to target the interaction of LSD1 with histone H3 and the LSD1 co-repressor (RCOR1/CoREST), likely disturbing the anchorage of LSD1 to chromatin. AlphaScreen-based in vitro assays confirmed the ability of oleacein to act as a direct inhibitor of recombinant LSD1, with an IC50 as low as 2.5 µmol/L. Further, oleacein fully suppressed the expression of the transcription factor SOX2 (SEX determining Region Y-box 2) in cancer stem-like and induced pluripotent stem (iPS) cells, which specifically occurs under the control of an LSD1-targeted distal enhancer. Conversely, oleacein failed to modify ectopic SOX2 overexpression driven by a constitutive promoter. Overall, our findings provide the first evidence that EVOO contains a naturally occurring phenolic inhibitor of LSD1, and support the use of oleacein as a template to design new secoiridoid-based LSD1 inhibitors.


Subject(s)
Aldehydes/pharmacology , Histone Demethylases/antagonists & inhibitors , Olive Oil/chemistry , Phenols/pharmacology , Aldehydes/analysis , Binding Sites/drug effects , Breast Neoplasms , Cell Line, Tumor , Co-Repressor Proteins/drug effects , Gene Expression/drug effects , Histone Demethylases/chemistry , Histone Demethylases/metabolism , Histones/metabolism , Humans , MCF-7 Cells , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Neoplastic Stem Cells/metabolism , Phenols/analysis , Recombinant Proteins/drug effects , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics
18.
Cancer Lett ; 454: 108-119, 2019 07 10.
Article in English | MEDLINE | ID: mdl-30981761

ABSTRACT

Treatment of oral squamous cell carcinoma (OSCC) remains a challenge because of the lack of effective early treatment strategies and high incidence of relapse. Here, we showed that combined 4SC-202 (a novel selective class I HDAC inhibitor) and INK128 (a selective mTORC1/C2 inhibitor) treatment exhibited synergistic effects on inhibiting cell growth, sphere-forming ability, subcutaneous tumor formation and ALDH1+ cancer stem cells (CSCs) in OSCC. The initiation of OSCC was significantly inhibited by combined treatment in 4NQO-induced rat model. In addition, upregulated SOX2 was associated with advanced and metastatic tumors in OSCC patients and was responsible for the drug-resistance property of OSCC cells. The inhibitory effect of combined treatment on cell viability and ALDH1+ CSCs were attenuated by SOX2 verexpression. Furthermore, combined treatment can effectively overcome chemoresistance and inhibit the growth of recurrent OSCC in vitro and in vivo. Mechanistically, 4SC-202 and INK128 repressed SOX2 expression through miR-429/miR-1181-mediated mRNA degradation and preventing cap-dependent mRNA translation, respectively. These results suggest that combined class I histone deacetylase and mTORC1/C2 inhibition suppresses the carcinogenesis and recurrence of OSCC by repressing SOX2.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mouth Neoplasms/drug therapy , SOXB1 Transcription Factors/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Benzamides/administration & dosage , Benzamides/pharmacology , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacology , Carcinogenesis , Drug Synergism , Female , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred BALB C , Mouth Neoplasms/metabolism , Neoplasm Recurrence, Local , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Random Allocation , Rats , Squamous Cell Carcinoma of Head and Neck/metabolism , Xenograft Model Antitumor Assays
19.
Nucleic Acids Res ; 45(16): 9219-9228, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934500

ABSTRACT

Targeted differentiation of human induced pluripotent stem cells (hiPSCs) using only chemicals would have value-added clinical potential in the regeneration of complex cell types including cardiomyocytes. Despite the availability of several chemical inhibitors targeting proteins involved in signaling pathways, no bioactive synthetic DNA-binding inhibitors, targeting key cell fate-controlling genes such as SOX2, are yet available. Here, we demonstrate a novel DNA-based chemical approach to guide the differentiation of hiPSCs using pyrrole-imidazole polyamides (PIPs), which are sequence-selective DNA-binding synthetic molecules. Harnessing knowledge about key transcriptional changes during the induction of cardiomyocyte, we developed a DNA-binding inhibitor termed PIP-S2, targeting the 5'-CTTTGTT-3' and demonstrated that inhibition of SOX2-DNA interaction by PIP-S2 triggers the mesoderm induction in hiPSCs. Genome-wide gene expression analyses revealed that PIP-S2 induced mesoderm by targeted alterations in SOX2-associated gene regulatory networks. Also, employment of PIP-S2 along with a Wnt/ß-catenin inhibitor successfully generated spontaneously contracting cardiomyocytes, validating our concept that DNA-binding inhibitors could drive the directed differentiation of hiPSCs. Because PIPs can be fine-tuned to target specific DNA sequences, our DNA-based approach could be expanded to target and regulate key transcription factors specifically associated with desired cell types.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/drug effects , Mesoderm/cytology , Myocytes, Cardiac/cytology , Nylons/pharmacology , Pyrroles/pharmacology , SOXB1 Transcription Factors/antagonists & inhibitors , Base Sequence , Binding Sites , Cell Line , Consensus Sequence , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mesoderm/drug effects , Mesoderm/metabolism , Myocytes, Cardiac/metabolism , Nylons/chemistry , Pyrroles/chemistry , SOXB1 Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects
20.
Cell Biol Int ; 41(12): 1335-1344, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28833847

ABSTRACT

Transcriptional master regulators like Sox2 and Oct4, which are expressed in various human tumors, have been shown to cause tumor growth promotion as well as epithelial dysplasia by means of interfering with progenitor cell differentiation. In order to investigate the potential of Sox2-Oct4 transcription factor decoy (TFD) strategy for differentiation therapy, mouse embryonic stem cells (mESCs) were used in this study as a model of cancer stem cells (CSCs). Sox2-Oct4 complex decoy ODNs (cd-ODNs) were designed according to their elements in the promoter region of Sox2 gene. DNA-protein interactions between decoy ODNs and their corresponding proteins were examined by electrophoretic mobility shift assay (EMSA). Then, decoy and scrambled ODNs were transfected into mESCs with lipofectamine under 2 inhibitors (2i) conditions. Fluorescence and confocal microscopy, cell viability, cell cycle and apoptosis analysis, alkaline phosphatase, embryoid body formation assay, and real-time PCR were used to conduct further investigations. EMSA data showed that Sox2-Oct4 decoy ODNs bound specifically to their recombinant proteins. The results revealed that the synthesized complex decoy can concomitantly target Sox2 and Oct4, which subsequently represses the stemness properties of mESCs compared to controls through decreasing cell viability, arresting cell cycle in G0 /G1 phases, inducing apoptosis, and modulating differentiation in mESCs despite the presence of 2i/LIF in cell culture. While cd-ODN strategy seems to offer great promise for cancer therapy, further studies are still required to put this powerful investigative tool in practice for a wide range of human cancers.


Subject(s)
Mouse Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/antagonists & inhibitors , Oligodeoxyribonucleotides/pharmacology , SOXB1 Transcription Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Electrophoretic Mobility Shift Assay/methods , Gene Expression Regulation/drug effects , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oligodeoxyribonucleotides/genetics , Promoter Regions, Genetic/drug effects , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL