Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
1.
Anticancer Res ; 43(3): 1131-1138, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36854532

ABSTRACT

BACKGROUND/AIM: Givinostat is a pan-histone deacetylase (HDAC) inhibitor that has demonstrated excellent tolerability as well as efficacy in patients with polycythemia vera. Accumulating in vitro and in vivo evidence suggests givinostat is also promising as a therapeutic agent targeting glioma stem cells (GSCs), the cancer stem cells of glioblastoma (GBM) considered responsible for its intractable nature. However, it remains to be shown how givinostat impacts the therapeutic effects of temozolomide, a DNA-alkylating agent and the key component of GBM treatment given not only during postoperative radiotherapy but also thereafter as maintenance chemotherapy. MATERIALS AND METHODS: The effects of givinostat and knockdown of O6-methylguanine-DNA methyltransferase (MGMT) or Sp1 on the mRNA and protein expression of relevant genes in human GSC lines were examined by RT-PCR and western blot analyses. The dye exclusion method was used to evaluate cell viability. RESULTS: Givinostat enhanced the cytotoxic activity of temozolomide in GSC lines expressing MGMT, in which the MGMT expression was shown to contribute to their temozolomide resistance. Givinostat inhibited MGMT expression in GSCs and, in parallel, the expression of Sp1, a transcription factor involved in the control of MGMT promoter activity. Knockdown experiments demonstrated Sp1 expression was indeed required for MGMT expression in GSCs. CONCLUSION: Givinostat, in addition to its own cytotoxic activity, sensitizes GSCs to temozolomide by inhibiting Sp1-dependent MGMT expression in GSCs. Combining givinostat with temozolomide could therefore be a rational therapeutic strategy to effectively eliminate GSCs and thus help overcome the therapy resistance of GBM.


Subject(s)
Glioblastoma , Glioma , Neoplastic Stem Cells , O(6)-Methylguanine-DNA Methyltransferase , Temozolomide , Humans , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/metabolism , Glioma/drug therapy , Glioma/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics
2.
Cancer Immunol Immunother ; 71(2): 311-325, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34159436

ABSTRACT

Recent studies have shown that tumor-derived exosomes participate in the communication between tumor cells and their microenvironment and mediate malignant biological behaviors including immune escape. In this study, we found that gastric cancer (GC) cell-derived exosomes could be effectively uptaken by Vγ9Vδ2 T cells, decrease the cell viability of Vγ9Vδ2 T cells, induce apoptosis, and reduce the production of cytotoxic cytokines IFN-γ and TNF-α. Furthermore, we demonstrated that exosomal miR-135b-5p was delivered into Vγ9Vδ2 T cells. Exosomal miR-135b-5p impaired the function of Vγ9Vδ2 T cells by targeting specificity protein 1 (SP1). More importantly, blocking the SP1 function by Plicamycin, an SP1 inhibitor, abolished the effect of stable miR-135b-5p knockdown GC cell-derived exosomes on Vγ9Vδ2 T cell function. Collectively, our results suggest that GC cell-derived exosomes impair the function of Vγ9Vδ2 T cells via miR-135b-5p/SP1 pathway, and targeting exosomal miR-135b-5p/SP1 axis may improve the efficiency of GC immunotherapy based on Vγ9Vδ2 T cells.


Subject(s)
Exosomes/genetics , MicroRNAs/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Stomach Neoplasms/pathology , T-Lymphocytes/immunology , Tumor Microenvironment , Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
3.
Int J Biol Sci ; 17(12): 3118-3132, 2021.
Article in English | MEDLINE | ID: mdl-34421354

ABSTRACT

In our previous study, we demonstrated that norcantharidin (NCTD) is a potential therapeutic agent for renal interstitial fibrosis (RIF). Recently, we found that lncRNA Gm26669 (Gm26669) contributed to the development of RIF and could be regulated by NCTD. However, the upstream mechanisms of Gm26669 and whether the anti-RIF effects of NCTD are related to its regulatory action on Gm26669 remain unclear. Our bioinformatics analysis indicated that special protein1 (Sp1), a transcription factor, may bind to the promoter of Gm26669. In the present study, we observed a significant increase in the nuclear translocation of Sp1 using both in vivo and in vitro models of RIF. Furthermore, the knockdown of Sp1 inhibited the expression of collagen type I (CoL-I) and fibronectin (Fn). Mechanistically, Sp1 promoted the expression levels of CoL-I and Fn by directly binding to the promoter of Gm26669 to elevate its expression level. Moreover, we found that NCTD alleviated RIF by inhibiting Gm26669 and the nuclear translocation of Sp1. Collectively, above results suggested that NCTD might prevent RIF via targeting the Sp1/Gm26669 axis, thus providing a new theoretical basis for the clinical application of NCTD in the treatment of RIF.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Kidney/pathology , Nephritis, Interstitial/prevention & control , RNA, Long Noncoding/antagonists & inhibitors , Sp1 Transcription Factor/antagonists & inhibitors , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Drug Delivery Systems , Epithelial Cells/metabolism , Fibrosis/prevention & control , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Kidney/metabolism , Kidney Tubules, Proximal/metabolism , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Nephritis, Interstitial/genetics , RNA, Long Noncoding/genetics , Real-Time Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Transfection , Transforming Growth Factor beta1/pharmacology
4.
Cancer Sci ; 112(9): 3846-3855, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34286904

ABSTRACT

Heterogeneous nuclear ribonucleoprotein L-like (HNRNPLL), a suppressor of colorectal cancer (CRC) metastasis, is transcriptionally downregulated when CRC cells undergo epithelial-mesenchymal transition (EMT). Here we show that decrease of MYB mediates the downregulation of HNRNPLL during EMT. The promoter activity was attributed to a region from -273 to -10 base pairs upstream of the transcription start site identified by 5'-RACE analysis, and the region contained potential binding sites for MYB and SP1. Luciferase reporter gene assays and knockdown or knockout experiments for genes encoding the MYB family proteins, MYB, MYBL1, and MYBL2, revealed that MYB was responsible for approximately half of the promoter activity. On the other hand, treatment with mithramycin A, an inhibitor for SP1 and SP3, suppressed the promoter activity and their additive contribution was confirmed by knockout experiments. The expression level of MYB was reduced on EMT while that of SP1 and SP3 was unchanged, suggesting that the downregulation of HNRNPLL during EMT was mediated by the decrease of MYB expression while SP1 and SP3 determine the basal transcription level of HNRNPLL. Histopathological analysis confirmed the accumulation of MYB-downregulated cancer cells at the invasion front of clinical CRC tissues. These results provide an insight into the molecular mechanism underlying CRC progression.


Subject(s)
Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Binding Sites , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Knockout Techniques , HT29 Cells , Humans , Neoplasm Metastasis , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myb/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic/genetics , Transfection
5.
Clin Immunol ; 229: 108775, 2021 08.
Article in English | MEDLINE | ID: mdl-34116211

ABSTRACT

The pathogenesis of recurrent tonsillitis is to be further investigated. B cell-derived interleukin (IL)-10 plays a critical role in immune regulation. Ras activation plays an important role in cancer and many immune disorders. This study aims to investigate the role of Ras activation in down regulating IL-10 expression in tonsillar B cells. Surgically removed tonsil tissues were collected from patients with recurrent acute tonsillar inflammation; B cells were isolated from the tonsillar tissues by flow cytometry sorting to be analyzed by the Ras-specific enzyme-linked immunosorbent assay and pertinent immunological approaches. We found that, compared to peripheral B cells (pBC), B cells isolated from the tonsillar tissues with recurrent inflammation (tBC) showed higher Ras activation, lower IL-10 expression and higher Bcl2L12 expression. Bcl2L12 formed a complex with GAP (GTPase activating protein) to prevent Ras from deactivating. The Ras activation triggered the MAPK/Sp1 pathway to promote the Bcl2L12 expression in B cells. Bcl2L12 prevented the IL-10 expression in tBCs, that was counteracted by inhibition of Ras or the Ras signal transduction pathway. In conclusion, Bcl2L12 interacts with Ras activation to compromise immune tolerance in the tonsils by inhibiting the IL-10 expression in tBCs. Inhibition of Bcl2L12 can restore the IL-10 expression in tBCs.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Interleukin-10/metabolism , Muscle Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , ras Proteins/metabolism , Adolescent , Adult , B-Lymphocytes/pathology , Child , Down-Regulation , Female , GTPase-Activating Proteins/metabolism , Gene Knockdown Techniques , Humans , Immune Tolerance , Interleukin-10/genetics , Male , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Recurrence , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Tonsillitis/immunology , Tonsillitis/metabolism , Tonsillitis/pathology , Up-Regulation , Young Adult
6.
Biochem Pharmacol ; 188: 114544, 2021 06.
Article in English | MEDLINE | ID: mdl-33831396

ABSTRACT

Although YM155 is reported to suppress survivin (also known as BIRC5) expression in cancer cells, its cytotoxic mechanism in human acute myeloid leukemia (AML) cells has not been clearly resolved. In this study, we analyzed the mechanistic pathways that modulate the sensitivity of human AML U937 and HL-60 cells to YM155. YM155 induced apoptosis in AML cells, which was characterized by p38 MAPK phosphorylation and downregulation of survivin and MCL1 expression. Phosphorylated p38 MAPK causes autophagy-mediated Sp1 degradation, thereby inhibiting the transcription of survivin and MCL1. The reduction of survivin and MCL1 levels further facilitated Sp1 protein degradation through autophagy. The restoration of Sp1, survivin, or MCL1 expression protected U937 and HL-60 cells from YM155-mediated cytotoxicity. U937 and HL-60 cells were continuously exposed to hydroquinone (HQ) to generate U937/HQ and HL-60/HQ cells, which showed increased SLC35F2 expression. The increase in SLC35F2 expression led to an increase in the sensitivity of U937/HQ cells to YM155-mediated cytotoxicity, whereas no such effect was observed in HL-60/HQ cells. Of note, myeloperoxidase (MPO) activity in HL-60 and HL-60/HQ cells enhanced YM155 cytotoxicity in these cells, and the enforced expression of MPO also increased the sensitivity of U937 cells to YM155. Taken together, we conclude that p38 MAPK-modulated autophagy inhibits Sp1-mediated survivin and MCL1 expression, which, in turn, leads to the death of U937 and HL-60 cells following YM155 treatment. In addition, our data indicate that SLC35F2 increases the sensitivity of U937 cells to YM155-mediated cytotoxicity, whereas MPO enhances YM155 cytotoxicity in U937 and HL-60 cells.


Subject(s)
Imidazoles/toxicity , Membrane Transport Proteins/biosynthesis , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Naphthoquinones/toxicity , Peroxidase/biosynthesis , Sp1 Transcription Factor/biosynthesis , Survivin/biosynthesis , Cell Survival/drug effects , Cell Survival/physiology , Cytotoxins/toxicity , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , Leukemia/genetics , Leukemia/metabolism , Membrane Transport Proteins/genetics , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Peroxidase/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Survivin/antagonists & inhibitors , Survivin/genetics , U937 Cells
7.
Retrovirology ; 18(1): 9, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33836787

ABSTRACT

BACKGROUND: Maturation inhibitors (MIs) potently block HIV-1 maturation by inhibiting the cleavage of the capsid protein and spacer peptide 1 (CA-SP1). Bevirimat (BVM), a highly efficacious first-in-class MI against HIV-1 subtype B isolates, elicited sub-optimal efficacy in clinical trials due to polymorphisms in the CA-SP1 region of the Gag protein (SP1:V7A). HIV-1 subtype C inherently contains this polymorphism thus conferring BVM resistance, however it displayed sensitivity to second generation BVM analogs. RESULTS: In this study, we have assessed the efficacy of three novel second-generation MIs (BVM analogs: CV-8611, CV-8612, CV-8613) against HIV-1 subtype B and C isolates. The BVM analogs were potent inhibitors of both HIV-1 subtype B (NL4-3) and subtype C (K3016) viruses. Serial passaging of the subtype C, K3016 virus strain in the presence of BVM analogs led to identification of two mutant viruses-Gag SP1:A1V and CA:I201V. While the SP1:A1V mutant was resistant to the MIs, the CA:I120V mutant displayed partial resistance and a MI-dependent phenotype. Further analysis of the activity of the BVM analogs against two additional HIV-1 subtype C strains, IndieC1 and ZM247 revealed that they had reduced sensitivity as compared to K3016. Sequence analysis of the three viruses identified two polymorphisms at SP1 residues 9 and 10 (K3016: N9, G10; IndieC1/ZM247: S9, T10). The N9S and S9N mutants had no change in MI-sensitivity. On the other hand, replacing glycine at residue 10 with threonine in K3016 reduced its MI sensitivity whereas introducing glycine at SP1 10 in place of threonine in IndieC1 and ZM247 significantly enhanced their MI sensitivity. Thus, the specific glycine residue 10 of SP1 in the HIV-1 subtype C viruses determined sensitivity towards BVM analogs. CONCLUSIONS: We have identified an association of a specific glycine at position 10 of Gag-SP1 with an MI susceptible phenotype of HIV-1 subtype C viruses. Our findings have highlighted that HIV-1 subtype C viruses, which were inherently resistant to BVM, may also be similarly predisposed to exhibit a significant degree of resistance to second-generation BVM analogs. Our work has strongly suggested that genetic differences between HIV-1 subtypes may produce variable MI sensitivity that needs to be considered in the development of novel, potent, broadly-active MIs.


Subject(s)
Anti-HIV Agents/pharmacology , Gene Expression Regulation, Viral/genetics , HIV-1/drug effects , HIV-1/genetics , Polymorphism, Genetic/drug effects , Sp1 Transcription Factor/antagonists & inhibitors , gag Gene Products, Human Immunodeficiency Virus/genetics , Cell Line , Drug Resistance, Viral/genetics , HEK293 Cells , Humans , Sp1 Transcription Factor/genetics , Succinates/pharmacology , Triterpenes/pharmacology , Virus Assembly/drug effects , Virus Replication/drug effects
8.
In Vitro Cell Dev Biol Anim ; 57(3): 315-323, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33580416

ABSTRACT

Specific protein 1 (SP1) might act as a critical transcription regulator in myocardial infarction (MI), but little evidence about its function in regulating cardiac apoptosis, a major cause of MI development, has been revealed. This study tried to investigate the role of SP1 in MI and its interaction with poly-ADP-ribose polymerase (PARP)-1 by using SP1 inhibitor, mithramycin A (mithA). Primary mouse cardiomyocytes and commercial mouse cardiomyocytes were subjected to mithA treatment under hypoxia conditions, while cell viability, Nix promoter activity, and its expression were detected correspondingly. PARP overexpression and knockdown were conducted, respectively, in mithA-treated and SP1-overexpressing cells. Co-immunoprecipitation was used to verify the interaction between PARP and SP1. For in vivo experiments, mithA administration was performed after the injections of adenovirus for PARP overexpression, and then, MI introduction was carried out. Infarct size and lactate dehydrogenase level were measured to assess MI injury. SP1 inhibitor mithA attenuated hypoxia-induced decrease of cell viability and Nix transcriptional activation, which could be inhibited by PARP overexpression. Knockdown of PARP prevented SP1-induced transcription of Nix and cell viability change, and PARP showed direct interaction with SP1. Furthermore, mithA administration reduced MI injuries, while PARP overexpression could suppress the improvement. The cardioprotective role of SP1 inhibitor mithA was demonstrated here expanding the role of SP1 in MI development involving hypoxia-induced cardiac apoptosis. Moreover, PARP acted as a transcriptional coactivator in Nix transcription involving its interaction with SP1.


Subject(s)
Cardiotonic Agents/pharmacology , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Plicamycin/analogs & derivatives , Poly(ADP-ribose) Polymerases/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Hypoxia/drug effects , Cell Survival/drug effects , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/drug effects , Plicamycin/pharmacology , Protein Binding/drug effects , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects
9.
Carcinogenesis ; 42(3): 344-356, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33146712

ABSTRACT

Recently, we identified that the atypical protein kinase C isoform ι (PKCι) enhances the expression of Yes-associated protein 1 (YAP1) to promote the tumorigenesis of pancreatic adenocarcinoma harboring mutant KRAS (mu-KRAS). To advance our understanding about underlying mechanisms, we analyze the transcription of YAP1 in pancreatic cancer cells and reveal that transcription factor specificity protein 1 (Sp1) is upregulated by PKCι and subsequently binds to multiple sites in YAP1 promoter to drive the transactivation of YAP1 in pancreatic cancer cells carrying mu-KRAS. The bioinformatics analysis further substantiates that the expression of PKCι, Sp1 and YAP1 is correlated and associated with the stages and prognosis of pancreatic tumors. Moreover, our apoptotic detection data demonstrate that combination of PKCι and Sp1 inhibitors at subtoxic doses displays synergistic effects on inducing apoptosis and reversing the immunosuppression of pancreatic cancer cells, establishing the combination of PKCι and Sp1 inhibitors as a promising novel therapeutic approach, or an adjuvant strategy to potentiate the antitumor effects of other immunotherapeutic agents in pancreatic cancer treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Isoenzymes/metabolism , Pancreatic Neoplasms/genetics , Protein Kinase C/metabolism , Sp1 Transcription Factor/genetics , Transcription Factors/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Computational Biology , Datasets as Topic , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Isoenzymes/antagonists & inhibitors , Mutation , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prognosis , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , RNA-Seq , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/immunology , Tumor Escape/drug effects , Tumor Escape/genetics , Up-Regulation/drug effects , Up-Regulation/immunology , YAP-Signaling Proteins
10.
Biomed Res Int ; 2020: 1358674, 2020.
Article in English | MEDLINE | ID: mdl-32851058

ABSTRACT

Phloretin is a flavonoid existed in various plants and has been reported to possess anticarcinogenic activity. However, the anticancer mechanism of phloretin in prostate cancer (PCa) remains unclear. Here, our in vitro and in vivo experimental data demonstrate that phloretin inhibits the phosphorylation and the activation of EGFR and then inhibits its downstream PI3K/AKT and MEK/ERK1/2 pathways in PCa cells. Inhibition of these two pathways further decreases expression of Sp1 by inhibiting Sp1 gene transcription, induces degradation of Sp1 protein by inhibiting GSK3ß phosphorylation, suppresses nucleolin-enhanced translation of Sp1 mRNA by inhibiting nucleolin phosphorylation, and directly inactivates transcription activity of Sp1. Inhibition of Sp1 subsequently decreases the expression of Sp3/4, VEGF, and Survivin and then upregulates apoptosis-related proteins and downregulates cell cycle-related proteins in PCa cells. Finally, phloretin treatment in PCa cells induces cell growth inhibition and apoptosis, suggesting that phloretin may be an effective therapy compound in the treatment of prostate cancer.


Subject(s)
Glycogen Synthase Kinase 3 beta/genetics , Phloretin/pharmacology , Phosphoproteins/genetics , Prostatic Neoplasms/drug therapy , RNA-Binding Proteins/genetics , Sp1 Transcription Factor/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Phosphatidylinositol 3-Kinases/genetics , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Survivin/genetics , Vascular Endothelial Growth Factor A/genetics , Nucleolin
11.
J Cell Mol Med ; 24(18): 10876-10888, 2020 09.
Article in English | MEDLINE | ID: mdl-32743904

ABSTRACT

Osteosarcoma (OS) is a malignant bone cancer lacking of effective treatment target when the metastasis occurred. This study investigated the implication of MicroRNA-326 in OS proliferation and metastasis to provide the clue for the treatment of metastatic OS. This study knocked down SP1 in MG63 and 143B cells and then performed Microarray assay to find the expression of miRNAs that were influenced by SP1. MTT, EdU, wound-healing and cell invasion assays were performed to evaluated cell proliferation and invasion. OS metastasis to lung was detected in a nude mice model. ChIP assay and DAPA were applied to determine the regulatory effect of SP1 and histone deacetylase 1 (HDAC) complex on miR-326 expression. Human OS tissues showed lowly expressed miR-326 but highly expressed Sp1 and HDAC. Sp1 recruited HDAC1 to miR-326 gene promoter, which caused the histone deacetylation and subsequent transcriptional inhibition of miR-326 gene. miR-326 deficiency induced the stimulation of SMO/Hedgehog pathway and promoted the proliferation and invasion of 143B and MG63 cells as well as the growth and metastasis in nude mice. SP1/HDAC1 caused the transcriptional inhibition of miR-326 gene by promoting histone deacetylation; miR-326 deficiency conversely stimulated SMO/Hedgehog pathway that was responsible for the proliferation and metastasis of OS.


Subject(s)
Bone Neoplasms/pathology , Histone Deacetylase 1/physiology , MicroRNAs/antagonists & inhibitors , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/physiology , Osteosarcoma/pathology , RNA, Neoplasm/antagonists & inhibitors , Smoothened Receptor/biosynthesis , Sp1 Transcription Factor/physiology , Adolescent , Adult , Animals , Bone Neoplasms/genetics , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Gene Knockdown Techniques , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Humans , Male , Matrix Metalloproteinase 9/physiology , Mice , Mice, Nude , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Metastasis/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Osteosarcoma/genetics , Osteosarcoma/secondary , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Smoothened Receptor/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Xenograft Model Antitumor Assays , Young Adult , Zinc Finger Protein GLI1/physiology
12.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751694

ABSTRACT

Gastric cancer (GC) is one of the most common and lethal cancers. Alterations in the ubiquitin (Ub) system play key roles in the carcinogenetic process and in metastasis development. Overexpression of transcription factors YY1, HSF1 and SP1, known to regulate Ub gene expression, is a predictor of poor prognosis and shorter survival in several cancers. In this study, we compared a primary (23132/87) and a metastatic (MKN45) GC cell line. We found a statistically significant higher expression of three out of four Ub coding genes, UBC, UBB and RPS27A, in MKN45 compared to 23132/87. However, while the total Ub protein content and the distribution of Ub between the conjugated and free pools were similar in these two GC cell lines, the proteasome activity was higher in MKN45. Ub gene expression was not affected upon YY1, HSF1 or SP1 small interfering RNA (siRNA) transfection, in both 23132/87 and MKN45 cell lines. Interestingly, the simultaneous knockdown of UBB and UBC mRNAs reduced the Ub content in both cell lines, but was more critical in the primary GC cell line 23132/87, causing a reduction in cell viability due to apoptosis induction and a decrease in the oncoprotein and metastatization marker ß-catenin levels. Our results identify UBB and UBC as pro-survival genes in primary gastric adenocarcinoma 23132/87 cells.


Subject(s)
Antigens, Neoplasm/genetics , Ribosomal Proteins/genetics , Stomach Neoplasms/genetics , Ubiquitin/genetics , Ubiquitins/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heat Shock Transcription Factors/antagonists & inhibitors , Heat Shock Transcription Factors/genetics , Humans , Neoplasm Metastasis , RNA, Messenger , RNA, Small Interfering/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Stomach Neoplasms/pathology
13.
J Biol Chem ; 295(14): 4617-4630, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32115405

ABSTRACT

Specificity protein 1 (Sp1) is an important transcription factor implicated in numerous cellular processes. However, whether Sp1 is involved in the regulation of RNA polymerase III (Pol III)-directed gene transcription in human cells remains unknown. Here, we first show that filamin A (FLNA) represses Sp1 expression as well as expression of TFIIB-related factor 1 (BRF1) and general transcription factor III C subunit 2 (GTF3C2) in HeLa, 293T, and SaOS2 cell lines stably expressing FLNA-silencing shRNAs. Both BRF1 promoter 4 (BRF1P4) and GTF3C2 promoter 2 (GTF3C2P2) contain putative Sp1-binding sites, suggesting that Sp1 affects Pol III gene transcription by regulating BRF1 and GTF3C2 expression. We demonstrate that Sp1 knockdown inhibits Pol III gene transcription, BRF1 and GTF3C2 expression, and the proliferation of 293T and HeLa cells, whereas Sp1 overexpression enhances these activities. We obtained a comparable result in a cell line in which both FLNA and Sp1 were depleted. These results indicate that Sp1 is involved in the regulation of Pol III gene transcription independently of FLNA expression. Reporter gene assays showed that alteration of Sp1 expression affects BRF1P4 and GTF3C2P2 activation, suggesting that Sp1 modulates Pol III-mediated gene transcription by controlling BRF1 and GTF3C2 gene expression. Further analysis revealed that Sp1 interacts with and thereby promotes the occupancies of TATA box-binding protein, TFIIAα, and p300 at both BRF1P4 and GTF3C2P2. These findings indicate that Sp1 controls Pol III-directed transcription and shed light on how Sp1 regulates cancer cell proliferation.


Subject(s)
RNA Polymerase III/metabolism , Sp1 Transcription Factor/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factors, TFIII/metabolism , Binding Sites , Cell Line , Cell Proliferation , E1A-Associated p300 Protein/metabolism , Filamins/antagonists & inhibitors , Filamins/genetics , Filamins/metabolism , Humans , Mutagenesis, Site-Directed , Promoter Regions, Genetic , RNA Interference , RNA Polymerase III/genetics , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , TATA-Binding Protein Associated Factors/antagonists & inhibitors , TATA-Binding Protein Associated Factors/genetics , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factors, TFIII/antagonists & inhibitors , Transcription Factors, TFIII/genetics , Transcription, Genetic , Up-Regulation
14.
Eur Rev Med Pharmacol Sci ; 24(2): 535-548, 2020 01.
Article in English | MEDLINE | ID: mdl-32016955

ABSTRACT

OBJECTIVE: Growing evidence has shown that long non-coding RNAs (lncRNAs) play some roles in the progression of osteoarthritis. In this study, we investigated the functions and mechanisms of lncRNA NKILA (NKILA) of chondrocytes in human osteoarthritis (OA). PATIENTS AND METHODS: RT-PCR was used to detect the expressions of NKILA and miR-145 in OA tissues. After transfection of NKILA overexpression lentivirus (LV-NKILA) and NKILA downregulation lentivirus (LV-shNKILA) into primary chondrocytes, MTT assay was carried out to measure the cell proliferation of chondrocytes. The expressions of SP1, Bcl-2, Bax, cleaved caspase-3 and NF-κB signaling factors were detected by Western blot. Moreover, luciferase assay was performed to explore the binding site of NKILA and miR-145, miR-145 and SP1. Finally, JSH, a NF-κB signaling inhibitor, was added into chondrocytes transfected with LV-shNKILA or miR-145 mimic to detect that NKILA functions via miR-145/SP1/NF-κB signaling pathway. RESULTS: We found that NKILA and SP1 were significantly reduced, miR-145 was increased in cartilage tissues of OA patients. After LV-NKILA transfection, the proliferation ability of chondrocytes was improved and cell apoptosis was inhibited; however, the proliferation ability of chondrocytes was repressed, and cell apoptosis was increased in LV-sh NKILA group. MiR-145 was predicted to be a potential target of NKILA and luciferase gene reporter assay confirmed that NKILA could directly bind with miR-145. Furthermore, SP1 was predicted to be a target gene of miR-145 and luciferase gene reporter assay proved that miR-145 could directly bind with SP1. Finally, we added JSH, a NF-κB signaling inhibitor, into chondrocytes with LV-shNKILA or miR-145 mimic. Results showed that the repressed SP1 was reversed after the addition of JSH in both LV-shNKILA and miR-145 mimic group. Further, the repressed proliferation capacities and promoted cell apoptosis were also reversed after the addition of JSH. CONCLUSIONS: According to the results, this study uncovers NKILA is reduced in human osteoarthritic cartilage tissues. Furthermore, we firstly uncover that the reduced NKILA could function as a ceRNA to improve miR-145, which inhibited SP1 expression and regulated NF-κB signaling pathway, thereby promoting tissue inflammation, and inhibiting proliferation and promoting apoptosis of chondrocytes. Thus, it may be used as a promising prognostic marker and a potential target for osteoarthritis.


Subject(s)
Chondrocytes/metabolism , MicroRNAs/biosynthesis , NF-kappa B/biosynthesis , Osteoarthritis/metabolism , RNA, Long Noncoding/metabolism , Sp1 Transcription Factor/biosynthesis , Apoptosis/physiology , Cell Proliferation/physiology , Cells, Cultured , Chondrocytes/pathology , Humans , Osteoarthritis/pathology , Sp1 Transcription Factor/antagonists & inhibitors
15.
Acta Pharmacol Sin ; 41(5): 661-669, 2020 May.
Article in English | MEDLINE | ID: mdl-31932644

ABSTRACT

Chronic tissue injury with fibrosis results in the disruption of tissue architecture, organ dysfunction, and eventual organ failure. Therefore, the development of effective antifibrotic drugs is urgently required. IMB-S7 is novel biphenyl compound derived from bifendate (biphenyldicarboxylate) that is used for the treatment of chronic hepatitis in China. In the current study we investigated the potential of IMB-S7 as an antihepatic fibrosis agent. In bile duct ligation (BDL) rat model, oral administration of IMB-S7 (400 mg· kg-1· d-1, for 14 days) significantly ameliorated BDL-induced liver necrosis, bile duct proliferation, and collagen accumulation. We then showed that IMB-S7 treatment markedly suppressed the TGF-ß/Smad pathway in human hepatic stellate cell line LX2 and mouse primary HSCs, as well as in liver samples of BDL rats, thus inhibiting the transcription of most fibrogenesis-associated genes, including TGF-ß1, COL1A1, and ACTA2. Furthermore, IMB-S7 treatment significantly suppressed the expression of integrin αv at the mRNA and protein levels in TGF-ß-treated LX2 cells and liver samples of BDL rats. Using integrin αv overexpression and silencing, we demonstrated that integrin αv activity correlated positively with the activation of TGF-ß/Smad pathway. Based on dual luciferase assay and DNA affinity precipitation assay, we revealed that IMB-S7 inactivated integrin αv through competitively inhibiting the binding of Sp1, a transcription factor, to the integrin αv (ITGAV) promoter (-173/-163 bp). These results suggest that IMB-S7 inhibits HSCs activation and liver fibrosis through Sp1-integrin αv signaling, and IMB-S7 may be a promising candidate to combat hepatic fibrosis in the future.


Subject(s)
Biphenyl Compounds/pharmacology , Integrin alphaV/genetics , Liver Cirrhosis/drug therapy , Sp1 Transcription Factor/antagonists & inhibitors , Animals , Bile Ducts/surgery , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Integrin alphaV/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/surgery , Molecular Structure , Rats , Sp1 Transcription Factor/metabolism , Structure-Activity Relationship
16.
Clin Exp Pharmacol Physiol ; 47(3): 372-382, 2020 03.
Article in English | MEDLINE | ID: mdl-31732967

ABSTRACT

MicroRNAs (miRNAs) have been shown to have complicated implications in the pathogenesis of Parkinson's disease (PD). However, the role of miR-29c and the underlying mechanism in the development of PD remain not well understood. In this work, the MPTP-treated mice or MPP+ -intoxicated SH-SY5Y cells were established as an in vivo or in vitro PD model. Then the specific agomir of miR-29c was employed to examine its biological function on PD progress. We found that miR-29c was down-expressed but SP1 was high-expressed in substantia nigra pars compacta (SNpc) of MPTP-induced PD mice. Overexpression of miR-29c attenuated dopaminergic neuron loss and α-synuclein accumulation in SNpc of PD mice. Furthermore, the increments of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) and TUNEL-positive apoptotic cells in MPTP-treated mice were ameliorated by miR-29c. Similarly, in SH-SY5Y cell models of PD, we also found that miR-29c inhibited inflammatory cytokine production, reduced apoptotic rate and suppressed pro-apoptotic regulator activity. In addition, the increased expression of SP1 in PD models was found to be inhibited by miR-29c. Luciferase reporter assay confirmed that SP1 was complementary with miR-29c. Knockdown of SP1 with siRNA restored α-synuclein accumulation, inflammation and apoptosis in MPP+ -induced SH-SY5Y cells. Collectively, this current work presents that miR-29c may directly target SP1 to protect against the neuroinflammatory and apoptotic responses in PD, providing a potential biomarker for PD diagnosis and treatment.


Subject(s)
Apoptosis/physiology , MicroRNAs/biosynthesis , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/prevention & control , Sp1 Transcription Factor/biosynthesis , Animals , Cell Line, Tumor , Humans , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/pathology , Random Allocation , Sp1 Transcription Factor/antagonists & inhibitors
17.
Int J Oncol ; 56(1): 273-282, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31746368

ABSTRACT

Oral squamous cell carcinoma (OSCC), the most common malignancy of the oral cavity, accounts for >90% of all diagnosed oral cancer cases. Baicalein, a naturally derived compound, has been shown to alter p65 and the nuclear factor (NF)­κB pathway, thus exerting cytotoxic effects on various tumor cell types. However, the mechanism of action of baicalein in OSCC has not been fully elucidated. In the present study, the proliferation of OSCC cells treated with baicalein was examined using a CCK­8 assay. The effects of baicalein on the cell cycle and apoptosis of OSCC cells were determined by flow cytometric analyses. The expression of specificity protein 1 (Sp1), p65 and p50 at the mRNA and protein levels was determined by reverse transcription­quantitative PCR and western blot analysis, respectively. The results of the present study demonstrated that baicalein suppresses the proliferation of OSCC cell lines in vivo and in vitro. Baicalein also induced apoptosis of OSCC cells and arrested the cell cycle at the G0/G1 phase. Baicalein inhibited the expression of Sp1, p65 and p50 by downregulating the relative mRNA levels. Baicalein reduced the activity of NF­κB in OSCC cells. Knockdown of Sp1 also resulted in reduced expression of p65 and p50. In addition, Sp1 silencing enhanced the effects of baicalein. In conclusion, the present study demonstrated that baicalein suppresses the growth of OSCC cells through an Sp1/NF­κB­dependent mechanism.


Subject(s)
Antioxidants/pharmacology , Carcinoma, Squamous Cell/pathology , Flavanones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Mouth Neoplasms/pathology , Sp1 Transcription Factor/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Eur Urol Oncol ; 2(4): 415-424, 2019 07.
Article in English | MEDLINE | ID: mdl-31277777

ABSTRACT

BACKGROUND: The TMPRSS2-ERG gene fusion is the most frequent genetic rearrangement in prostate cancers and results in broad transcriptional reprogramming and major phenotypic changes. Interaction and cooperation of ERG and SP1 may be instrumental in sustaining the tumorigenic and metastatic phenotype and could represent a potential vulnerability in ERG fusion-positive tumors. OBJECTIVE: To test the activity of EC-8042, a compound able to block SP1, in cellular and mouse models of ERG-positive prostate cancer. DESIGN, SETTING, AND PARTICIPANTS: We evaluated the activity of EC-8042 in cell cultures and ERG/PTEN transgenic/knockout mice that provide reliable models for testing novel therapeutics in this specific disease context. Using a new protocol to generate tumor spheroids from ERG/PTEN mice, we also examined the effects of EC-8042 on tumor-propagating stem-like cancer cells with high self-renewal and tumorigenic capabilities. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The efficacy of EC-8042 was determined by measuring the proliferative capacity and target gene expression in cell cultures, invasive and metastatic capabilities in chick chorioallantoic membrane assays, and tumor development in mice. Significance was determined using statistical test. RESULTS AND LIMITATIONS: EC-8042 blocked transcription of ERG-regulated genes and reverted the invasive and metastatic phenotype of VCaP cells. EC-8042 blocked the expansion of stem-like tumor cells in tumor spheroids from VCaP cells and mouse-derived tumors. In ERG/PTEN mice, systemic treatment with EC-8042 inhibited ERG-regulated gene transcription, tumor progression, and tumor-propagating stem-like tumor cells. CONCLUSIONS: Our data support clinical testing of EC-8042 for the treatment of ERG-positive prostate cancer in precision medicine approaches. PATIENT SUMMARY: In this study, EC-8042, a novel compound with a favorable pharmacological and toxicological profile, exhibited relevant activity in cell cultures and in vivo in a genetically engineered mouse model that closely recapitulates the features of clinically aggressive ERG-positive prostate cancer. Our data indicate that further evaluation of EC-8042 in clinical trials is warranted.


Subject(s)
Plicamycin/analogs & derivatives , Prostatic Neoplasms/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Transcriptional Regulator ERG/genetics , Animals , Cell Line, Tumor , Humans , Male , Mice, Transgenic , Neoplastic Stem Cells , PTEN Phosphohydrolase/genetics , Plicamycin/pharmacology , Plicamycin/therapeutic use , Prostatic Neoplasms/drug therapy
19.
Neuroscience ; 412: 207-215, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31220545

ABSTRACT

High voltage-activated (HVA) Ca2+ (CaV) channels are oligomeric complexes formed by an ion-conducting main subunit (Cavα1) and at least two auxiliary subunits (Cavß and CaVα2δ). It has been reported that the expression of CaVα2δ1 increases in the dorsal root ganglia (DRGs) of animals with mechanical allodynia, and that the transcription factor Sp1 regulates the expression of the auxiliary subunit. Hence, the main aim of this work was to investigate the role of Sp1 as a molecular determinant of the exacerbated expression of CaVα2δ-1 in the nerve ligation-induced model of mechanical allodynia. Our results show that ligation of L5/L6 spinal nerves (SNL) produced allodynia and increased the expression of Sp1 and CaVα2δ-1 in the DRGs. Interestingly, intrathecal administration of the Sp1 inhibitor mithramycin A (Mth) prevented allodynia and decreased the expression of Sp1 and CaVα2δ-1. Likewise, electrophysiological recordings showed that incubation with Mth decreased Ca2+ current density in the DRG neurons, acting mostly on HVA channels. These results suggest that L5/L6 SNL produces mechanical allodynia and increases the expression of the transcription factor Sp1 and the subunit CaVα2δ-1 in the DRGs, while Mth decreases mechanical allodynia and Ca2+ currents through HVA channels in sensory neurons by reducing the functional expression of the CaVα2δ-1 subunit.


Subject(s)
Calcium Channels/metabolism , Ganglia, Spinal/metabolism , Neuralgia/metabolism , Sensory Receptor Cells/metabolism , Sp1 Transcription Factor/metabolism , Animals , Female , Ganglia, Spinal/drug effects , Neuralgia/etiology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/metabolism , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Rats, Wistar , Sensory Receptor Cells/drug effects , Sp1 Transcription Factor/antagonists & inhibitors
20.
Cell Host Microbe ; 25(6): 858-872.e13, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31175045

ABSTRACT

The interferon γ-inducible protein 16 (IFI16) is known as immune sensor of retroviral DNA intermediates. We show that IFI16 restricts HIV-1 independently of immune sensing by binding and inhibiting the host transcription factor Sp1 that drives viral gene expression. This antiretroviral activity and ability to bind Sp1 require the N-terminal pyrin domain and nuclear localization of IFI16, but not the HIN domains involved in DNA binding. Highly prevalent clade C HIV-1 strains are more resistant to IFI16 and less dependent on Sp1 than other HIV-1 subtypes. Furthermore, inhibition of Sp1 by IFI16 or pharmacologically by Mithramycin A suppresses reactivation of latent HIV-1 in CD4+ T cells. Finally, IFI16 also inhibits retrotransposition of LINE-1, known to engage Sp1, and murine IFI16 homologs restrict Friend retrovirus replication in mice. Thus, IFI16 restricts retroviruses and retrotransposons by interfering with Sp1-dependent gene expression, and evasion from this restriction may facilitate spread of HIV-1 subtype C.


Subject(s)
HIV-1/immunology , Immunologic Factors/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Transcription, Genetic , Virus Activation , Virus Latency , Animals , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Genotype , HIV-1/classification , HIV-1/genetics , HIV-1/growth & development , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...