Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Technol Cancer Res Treat ; 23: 15330338241245939, 2024.
Article in English | MEDLINE | ID: mdl-38752263

ABSTRACT

OBJECTIVES: Small nucleolar RNAs (snoRNAs) form clusters within the genome, representing a mysterious category of small non-coding RNAs. Research has demonstrated that aberrant snoRNAs can contribute to the development of various types of cancers. Recent studies have identified snoRNAs as potentially valuable biomarkers for the diagnosis or/and prognosis of cancers. However, there has been a lack of comprehensive reviews on prognostic and diagnostic snoRNAs across different types of cancers. METHODS: We conducted a systematic search of various databases including Google Scholar, Medline, Cochrane, Scopus, PubMed, Embase, ScienceDirect, Ovid-Medline, Chinese National Knowledge Infrastructure, WanFang, and SinoMed with a time frame reception to December 30, 2022. A total of 49 relevant articles were included in our analysis, consisting of 21 articles focusing on diagnostic aspects and 41 articles focusing on prognostic aspects. Pooled odds ratio, 95% confidence intervals (CIs), and hazard ratio (HR) were utilized to evaluate clinical parameters and overall survival (OS), respectively. RESULT: The findings indicated that area under the curve, sensitivity, and specificity were 0.85, 75%, and 80% in cancer, respectively. There was a possibility that snoRNAs had a positive impact on the diagnosis (risk ratio, RR = 2.95, 95% CI: 2.75-3.16, P = 0.000) and OS (HR = 1) in cancer. Additionally, abnormally expressed snoRNAs were associated with a positive impact on OS time for chronic lymphocytic leukemia (HR: 0.88, 95%Cl: 0.69-1.11, P < 0.00001), colon adenocarcinoma (HR: 0.97, 95%Cl: 0.91-1.03, P < 0.0001), and ovarian cancer (HR: 0.98, 95%Cl: 0.98-0.99, P < 0.00001). However, dysregulated snoRNAs of colon cancer and colorectal cancer had a negative impact on OS time (HR = 3.01 and 1.01 respectively, P < 0.0001). CONCLUSION: The results strongly suggested that snoRNAs could serve as potential novel indicators for prognosis and diagnosis in cancers. This systematic review followed the guidelines of the Transparent Reporting of Systematic Review and Meta-Analyses (PROSPERO register: CRD42020209096).


Subject(s)
Biomarkers, Tumor , Neoplasms , RNA, Small Nucleolar , Humans , RNA, Small Nucleolar/genetics , Biomarkers, Tumor/genetics , Prognosis , Neoplasms/genetics , Neoplasms/diagnosis , Neoplasms/mortality , ROC Curve
2.
Hum Exp Toxicol ; 42: 9603271231183359, 2023.
Article in English | MEDLINE | ID: mdl-37303070

ABSTRACT

Various circular RNAs (circRNAs) are novel class of non-coding RNAs, which are pervasively transcribed in the genome. CircRNAs play important roles in human, animals and plants. Up to now, there was no report regarding circRNAs of cleft palate by 2,3,7,8-tetrachlorodibenzo-pdioxin (TCDD) induce. The present study screened identification and characterization of differential expressed-circRNAs in TCDD-induced cleft palate. 6903 circRNAs candidates came from cleft palates. Among them, 3525 circRNAs are up-regulation, and 3378 circRNAs are down-regulation by TCDD induce. The cluster and GO analysis found that circRNAs involved in biological process, cellular component, and molecular function. Through the analysis of KEGG Pathway, circRNAs made functions via classical signaling pathway in cleft palate, such as TGF-beta signaling pathway, BMP signal pathway, MAPK signaling pathway. In addition, we found down-regulated circRNA224, circRNA3302 and up-regulated circRNA5021 targeted tgfbr3, but up-regulated circRNA4451 targeted tgfbr2. circRNA4451 may make functions through TGF-beta signaling pathway. These results suggested that many different circRNAs may make important role in TCDD-induced cleft palate, which provided a theoretical basis for further research.


Subject(s)
Cleft Palate , Polychlorinated Dibenzodioxins , Animals , Humans , Cleft Palate/chemically induced , Cleft Palate/genetics , RNA, Circular/genetics , Down-Regulation , Polychlorinated Dibenzodioxins/toxicity , Transforming Growth Factor beta
3.
Folia Histochem Cytobiol ; 60(1): 89-100, 2022.
Article in English | MEDLINE | ID: mdl-35212388

ABSTRACT

INTRODUCTION: With cellular lipid storage varying, the balance between lipid intake and lipid degradation was a must to keep healthy and determined the level of lipid droplets. Although lipid droplets accumulation had been well demonstrated in adipocytes, gene expression profiling and gene function during adipogenesis and osteoblastogenesis remain unknown. MATERIAL AND METHODS: Here, this work profiled gene transcriptional landscapes of lipid droplets formation during adipogenesis from human mesenchymal stem cells (hMSCs) using RNA-Seq technique. By using RNA interference (RNAi) we investigated the function of candidate genes during adipogenesis and osteoblastogenesis using Oil Red/Alizarin Red/alkaline phosphatase (ALPL) staining and qRT-PCR (quantitative real-time PCR). RESULTS: Eleven differentially up-regulated genes associated with lipid droplets formation were identified at 3, 5, 7, 14, 21, and 28 days during adipogenesis. Unexpectedly, APOB per se inhibiting adipogenesis weakened osteoblastogenesis and METTL7A facilitating adipogenesis negligibly inhibited osteoblastogenesis according to the phenotypic characterization of adipocytes and osteoblasts and transcriptional condition of biomarkers through lentivirus transfection assays. CONCLUSIONS: The establishment of the gene transcriptional profiling of lipid droplets formation would provide the molecular switches of hMSCs cell fate determination and the study targets for fat metabolic diseases.


Subject(s)
Adipogenesis , Mesenchymal Stem Cells , Adipocytes , Adipogenesis/genetics , Cell Differentiation , Cells, Cultured , Humans , Lipid Droplets , Mesenchymal Stem Cells/metabolism
4.
Stem Cell Res Ther ; 11(1): 62, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32059748

ABSTRACT

BACKGROUND: The process of bone repair is heavily dependent on the ability of human bone marrow mesenchymal stem cells (hMSCs) to undergo osteogenic differentiation. MicroRNAs have been shown to regulate this osteogenic process. This study aimed to investigate the role of miR-765 in the osteogenic differentiation of hMSCs. METHODS: We transfected hMSCs with lentiviral constructs to knock down or overexpress this miRNA, allowing us to assess its role in osteogenesis via assessing the expression of the relevant markers alkaline phosphatase (ALP), runt-related gene-2 (RUNX-2), and osteocalcin (OCN), with further functional measurements made via quantifying ALP activity and conducting Alizarin Red S staining. The targeting of the 3'-untranslated region (UTR) of BMP6 by miR-765 was examined via luciferase assay. We used hMSCs with altered miR-765 expression to assess p-Smad1/5/9 levels via Western blotting over the course of osteogenic differentiation. We also assessed the osteogenic differentiation of hMSCs in which miR-765 was knocked down and at the same time as a BMP/Smad signaling inhibitor was added to disrupt Smad1/5/9 phosphorylation. RESULTS: We found miR-765 overexpression to inhibit osteogenesis-associated gene upregulation during osteogenic differentiation of hMSCs, whereas knockdown of this miRNA was associated with increased expression of these genes. Using luciferase reporter assays, we confirmed direct miR-765 binding to the 3'-untranslated region (UTR) of BMP6. We also found that miR-765 overexpression reduced Smad1/5/9 phosphorylation, and knockdown of this miRNA enhanced this phosphorylation on BMP6/Smad1/5/9 signaling. The osteogenic differentiation of hMSCs in which miR-765 had been knocked down was further weakened upon the addition of a BMP/Smad signaling inhibitor relative to miR-765 knockdown alone. CONCLUSIONS: Together, these results thus suggest that miR-765 is able to inhibit hMSC osteogenic differentiation by targeting BMP6 via regulation of the BMP6/Smad1/5/9 signaling pathway. Our findings may offer molecular insights of value for the development of novel therapeutic treatments for bone diseases including osteoporosis.


Subject(s)
Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 6/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis , Signal Transduction , Transfection
5.
Molecules ; 23(10)2018 10 11.
Article in English | MEDLINE | ID: mdl-30314388

ABSTRACT

Skin wound healing is a complex and dynamic process that involves angiogenesis and growth factor secretion. Newly formed vessels can provide nutrition and oxygen for skin wound healing. Growth factors in skin wounds are important for keratinocytes and fibroblasts proliferation, epithelialization, extracellular matrix remodeling, and angiogenesis, which accelerate skin wound healing. Therefore, treatment strategies that enhance angiogenesis and growth factors secretion in skin wounds can accelerate skin wound healing. This study investigated the effects of a SIKVAV (Ser-Ile-Lys-Val-Ala-Val) peptide-modified chitosan hydrogel on skin wound healing. Hematoxylin and eosin (H&E) staining demonstrated that the SIKVAV-modified chitosan hydrogel accelerated the re-epithelialization of wounds compared with that seen in the negative and positive controls. Masson's trichrome staining showed that more collagen fibers were deposited in the skin wounds treated with the SIKVAV-modified chitosan hydrogel than in the negative and positive controls. Immunohistochemistry assays demonstrated that more myofibroblasts were deposited and more angiogenesis occurred in skin wounds treated with the SIKVAV-modified chitosan hydrogel than in the negative and positive controls. In addition, ELISA assays showed that the SIKVAV-modified chitosan hydrogels promoted the secretion of growth factors in skin wounds. Taken together, these results suggest that the SIKVAV-modified chitosan hydrogel has the potential to be developed as synthesized biomaterials for the treatment of skin wounds.


Subject(s)
Biocompatible Materials/chemistry , Chitosan/chemistry , Hydrogels/chemistry , Oligopeptides/chemistry , Skin, Artificial , Wound Healing , Animals , Biocompatible Materials/chemical synthesis , Collagen/biosynthesis , Fibroblasts/metabolism , Hydrogels/chemical synthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Mice , Neovascularization, Physiologic , Re-Epithelialization
6.
Am J Transl Res ; 10(12): 4258-4268, 2018.
Article in English | MEDLINE | ID: mdl-30662668

ABSTRACT

Skin wound healing is complex and involves the processes of many factors, among which angiogenesis and inflammatory responses play important roles. New blood vessels provide nutrition and oxygen for skin wound repair. Cytokines in skin wounds, which include pro-inflammatory and anti-inflammatory factors, can modulate the inflammatory response. Therefore, treatment strategies that promote angiogenesis and modulate the inflammatory response in skin wounds can accelerate skin wound healing. This study explored the effects of peptide Ser-Ile-Lys-Val-Ala-Val (SIKVAV)-modified chitosan hydrogels in skin wound healing. General observation demonstrated that SIKVAV-modified chitosan hydrogels promoted the contraction of skin wounds compared with the negative and positive controls. Masson's trichrome staining indicated that peptide-modified chitosan hydrogels accelerated the deposition of more collagen fibers in the skin wounds compared with the negative and positive controls. Immunohistochemistry assays showed that more myofibroblasts were deposited and more angiogenesis was found in skin wounds treated with peptide-modified chitosan hydrogels compared with the negative and positive controls. In addition, qRT-PCR assays showed that peptide-modified chitosan hydrogels promoted the expression of TGF-ß1 (transforming growth factor-ß1) mRNA and inhibited the expression of TNF-α (tumor necrosis factor-α) mRNA and IL-1ß (Interleukin-1ß) mRNA and IL-6 (Interleukin-6) mRNA in skin wounds. Taken together, these results indicate the potential of SIKVAV-modified chitosan hydrogels in skin wound healing as complex biomaterials.

7.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 48(6): 824-827, 2017 Nov.
Article in Chinese | MEDLINE | ID: mdl-29260514

ABSTRACT

OBJECTIVE: To construct the expression vectors for red fluorescent protein fused with inhibiting peptides for nuclear import (Bimax),and explore the location of Bimax and its potential effects on cell proliferation and migration in HeLa cells. METHODS: Two kinds of polynucleotide encoding inhibiting peptides for nuclear import were synthesis respectively and subsequently annealed for inserting into vector pDs-Red-C1. The recombinant plasmids were transfected into competent bacterial DH-5α. After transfection,the positive bacteria were picked up for DNA sequencing. The recombinant plasmids pDs-Red-Bimax2,pDs-Red-Bimax1 and negative plasmid pDs-Red-C1 were transfected into HeLa cells respectively according to Lipofectamine2000 protocol. After transfection,the expression and location of red fluorescent protein were observed with fluorescence microscope. Furthermore,MTT assay and cell-migration assay were used to detect the proliferation and migration of Bimax transducted cells. RESULTS: DNA sequencing showed that the polynucleotides encoding Bimax1 or Bimax2 were inserted into pDs-Red-C1 vector successfully. After transfected into HeLa cells,the inhibiting peptide induced red fluorescent protein locating in nuclear. Furthermore,either the fusion protein RFP-Bimax1 or RFP-Bimax2 can suppress the proliferation and migration of HeLa cells. CONCLUSION: The expression vectors for red fluorescent protein fused with inhibiting peptides for nuclear import were successfully constructed. In addition,the fusion proteins were expressed and located in nuclear and suppressed the proliferation and migration of tumor cells.


Subject(s)
Active Transport, Cell Nucleus , Genetic Vectors , Peptides , Plasmids , Cell Movement , HeLa Cells , Humans , Luminescent Proteins , Transfection , Red Fluorescent Protein
8.
Mol Med Rep ; 14(2): 1418-24, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27277970

ABSTRACT

Runt-related transcription factor 2 (RUNX2) is a vital regulatory factor that controls osteoblast-specific gene expression; however, RUNX2­regulated genes in human mesenchymal stem cells (hMSCs) remain to be fully elucidated. In the present study, chromatin immunoprecipitation (ChIP)-on-chip analysis of RUNX2 in hMSCs demonstrated that importin 8 (IPO8) may be a novel target gene. The 5' flanking region of the IPO8 gene, which is ~3,300 bp in length, was cloned and inserted into the pGL3­basic luciferase reporter vector. The results of dual luciferase reporter assays indicated that this segment possessed strong basal promoter activity. Furthermore, the RUNX2 binding site, which encompasses positions ­496 to ­501 bp, was required to achieve maximal IPO8 promoter activity in Saos­2 human osteosarcoma cells. In addition, ChIP analysis indicated that RUNX2 uniquely binds to this specific IPO8 sequence motif. Cells with a knockdown in RUNX2 expression exhibited downregulated IPO8 transcription. Finally, synchronization of IPO8 and RUNX2 expression was observed in Saos­2 cells cultured in osteoblast­induction medium. Taken together, these results indicated that RUNX2 regulates IPO8 gene transcription, and may have a contributory role in osteoblast differentiation.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Neoplastic , Transcription, Genetic , beta Karyopherins/genetics , Binding Sites , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Mesenchymal Stem Cells/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , Promoter Regions, Genetic , Protein Binding
9.
J Mater Chem B ; 2(15): 2114-2126, 2014 Apr 21.
Article in English | MEDLINE | ID: mdl-32261495

ABSTRACT

Chemotherapy is a major therapeutic approach for cancer patients. The action sites of cancer drugs are intracellular compartments including cytoplasm or nucleus. However, targeting drug delivery into the nucleus of specific tumor cells remains a challenging task. Herein, we developed dual-decorated polymeric micelles with folic acid (FA) and a nuclear localization signal (NLS) for specific tumor-targeted drug delivery. Cholesterol-modified glycol chitosan (CHGC) was synthesized. NLS and FA conjugated CHGC (NFCHGC) micelles were constructed. Doxorubicin (DOX) was chosen as a model anticancer drug and coumarin 6 (C6) was used as a hydrophobic fluorescence probe. The drug-loaded polymeric micelles were prepared and characterized. C6-loaded NFCHGC (C6/NFCHGC) showed efficient intracellular trafficking including endosomal/lysosomal escape and nucleus transportation in folate receptor (FR)-positive KB cells investigated by confocal laser scanning microscopy (CLSM). DOX-loaded NFCHGC (DOX/NFCHGC) exhibited stronger cytotoxicity against KB cells than other DOX formulations. Furthermore, blank polymeric micelles displayed low toxicity and good biocompatibility in vivo. DOX/NFCHGC micelles had the strongest anti-tumor efficacy against KB tumor xenograft models in vivo. These findings demonstrated that NFCHGC micelles were deemed as a potential drug nanocarrier for cancer therapy, especially used in FR-positive tumor cells and nucleus-targeting delivery.

10.
PLoS One ; 8(12): e80644, 2013.
Article in English | MEDLINE | ID: mdl-24349009

ABSTRACT

OBJECTIVE: Experimental and clinical studies have shown that administration of insulin during reperfusion is cardioprotective, but the mechanisms underlying this effect are still unknown. In this study, the ability of insulin to protect apoptotic cardiomyocytes from hypoxia/reoxygenation injury using the sphingosine kinase/sphingosine 1-phosphate axis was investigated. METHODS AND RESULTS: Rat cardiomyocytes were isolated and subjected to hypoxia and reoxygenation. [γ-32P] ATP was used to assess sphingosine kinase activity. Insulin was found to increase sphingosine kinase activity. Immunocytochemistry and Western blot analysis showed changes in the subcellular location of sphingosine kinase 1 from cytosol to the membrane in cardiomyocytes. Insulin caused cardiomyocytes to accumulate of S1P in a dose-dependent manner. FRET efficiency showed that insulin also transactivates the S1P1 receptor. TUNEL staining showed that administration of insulin during reoxygenation could to reduce the rate of reoxygenation-induced apoptosis, which is a requirement for SphK 1 activity. It also reduced the rate of activation of the S1P receptor and inhibited hypoxia/reoxygenation-induced cell death in cardiomyocytes. CONCLUSION: The sphingosine kinase 1/sphingosine 1-phosphate/S1P receptor axis is one pathway through which insulin protects rat cardiomyocytes from apoptosis induced by hypoxia/reoxygenation injury.


Subject(s)
Insulin/pharmacology , Lysophospholipids/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Hypoxia/physiology , Cells, Cultured , In Situ Nick-End Labeling , Rats , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(4): 516-9, 2013 Apr.
Article in Chinese | MEDLINE | ID: mdl-23644110

ABSTRACT

OBJECTIVE: To observe the variations of intracellular localization and expression of importin 8 (IPO8) during osteoblast differentiation. METHODS: Alizarin red staining, immunocytochemistry and real-time PCR were employed to examine the changes in the intracellular localization and expression of IPO8 mRNA during induced osteogenic differentiation of human osteoblast-like SaOS-2 cells. RESULTS: Numerous red mineralized nodules were observed on day 10 in the induced cells with alizarin red staining. Immunocytochemical staining showed that IPO8 immunoreactivity was the strongest in the perinuclear cytoplasm of the cells. On day 3 of osteoblast differentiation, IPO8 immunoreactivity in the cell nuclei became stronger. On day 7, IPO8 was located mainly in the nuclei, and on day 10 the cells were osteocyte-like and IPO8 was distributed in the cytoplasm. Real-time PCR showed a significantly increased expression of OPN mRNA during osteoblast differentiation, and the expression level of IPO8 mRNA was the highest on day 3 and declined on days 7 and 10. CONCLUSION: The intracellular localization and expression level of IPO8 undergo significant changes during osteogenesis, indicating its role in regulating osteoblast differentiation.


Subject(s)
Cell Differentiation , Osteoblasts/cytology , Osteoblasts/metabolism , beta Karyopherins/metabolism , Cell Line , Humans , Osteogenesis
12.
J Biomater Sci Polym Ed ; 24(5): 606-20, 2013.
Article in English | MEDLINE | ID: mdl-23565871

ABSTRACT

For folate receptor (FR) targeted anticancer therapy, novel folic acid (FA) conjugated cholesterol-modified glycol chitosan (FCHGC) micelles were synthesized and characterized by (1)H NMR, dynamic light scattering, transmission electron microscopy, and fluorescence spectroscopy. The degree of substitution was 1.4 FA groups and 7.7 cholesterol groups per 100 sugar residues of glycol chitosan. The critical aggregation concentration of FCHGC micelles in aqueous solution was 0.0169 mg/ml. The doxorubicin (DOX)-loaded FCHGC (DFCHGC) micelles were prepared by an emulsion/solvent evaporation method. The DFCHGC micelles were almost spherical in shape and their size increased from 282 to 320 nm with the DOX-loading content increasing from 4.53 to 11.4%. DOX released from DOX-loaded micelles displayed sustained release behavior. The targeted micelles encapsulated DOX showed significantly greater cytotoxicity against FR-positive HeLa cells than the nontargeted DOX-loaded micelles and free DOX. These results suggested that FCHGC micelles could be a potential carrier for targeted drug delivery.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Chitosan/chemistry , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Folic Acid/chemistry , Micelles , Antibiotics, Antineoplastic/pharmacology , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Delivery Systems , HeLa Cells , Humans , Neoplasms/drug therapy
13.
Int J Nanomedicine ; 7: 5079-90, 2012.
Article in English | MEDLINE | ID: mdl-23049255

ABSTRACT

BACKGROUND: Supramolecular micelles as drug-delivery vehicles are generally unable to enter the nucleus of nondividing cells. In the work reported here, nuclear localization signal (NLS)-modified polymeric micelles were studied with the aim of improving nuclear drug delivery. METHODS: In this research, cholesterol-modified glycol chitosan (CHGC) was synthesized. NLS-conjugated CHGC (NCHGC) was synthesized and characterized using proton nuclear magnetic resonance spectroscopy, dynamic light scattering, and fluorescence spectroscopy. Doxorubicin (DOX), an anticancer drug with an intracellular site of action in the nucleus, was chosen as a model drug. DOX-loaded micelles were prepared by an emulsion/solvent evaporation method. The cellular uptake of different DOX formulations was analyzed by flow cytometry and confocal laser scanning microscopy. The cytotoxicity of blank micelles, free DOX, and DOX-loaded micelles in vitro was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in HeLa and HepG2 cells. RESULTS: The degree of substitution was 5.9 cholesterol and 3.8 NLS groups per 100 sugar residues of the NCHGC conjugate. The critical aggregation concentration of the NCHGC micelles in aqueous solution was 0.0209 mg/mL. The DOX-loaded NCHGC (DNCHGC) micelles were observed as being almost spherical in shape under transmission electron microscopy, and the size was determined as 248 nm by dynamic light scattering. The DOX-loading content of the DNCHGC micelles was 10.1%. The DOX-loaded micelles showed slow drug-release behavior within 72 hours in vitro. The DNCHGC micelles exhibited greater cellular uptake and higher amounts of DOX in the nuclei of HeLa cells than free DOX and DOX-loaded CHGC (DCHGC) micelles. The half maximal inhibitory concentration (IC(50)) values of free DOX, DCHGC, and DNCHGC micelles against HepG2 cells were 4.063, 0.591, and 0.171 µg/mL, respectively. Moreover, the IC(50) values of free DOX (3.210 µg/mL) and the DCHGC micelles (1.413 µg/mL) against HeLa cells were nearly 6.96- and 3.07-fold (P < 0.01), respectively, higher than the IC(50) value of the DNCHGC micelles (0.461 µg/mL). CONCLUSION: The results of this study suggest that novel NCHGC micelles could be a potential carrier for nucleus-targeting delivery.


Subject(s)
Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chitosan/chemistry , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Nanocapsules/chemistry , Nuclear Localization Signals/pharmacokinetics , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/chemistry , Diffusion , HeLa Cells , Hep G2 Cells , Humans , Micelles , Nanocapsules/administration & dosage , Nuclear Localization Signals/chemistry , Polyethylene Glycols/chemistry
14.
PLoS One ; 7(12): e52716, 2012.
Article in English | MEDLINE | ID: mdl-23300749

ABSTRACT

Ubiquitin-specific processing enzyme 22 (USP22) plays a direct role in regulating cell cycle, and its overexpression has been reported to be involved in tumor progression. However, little is known about the regulation of USP22 transcription. In this study, we cloned and characterized the human USP22 promoter. Using 5' RACE (rapid amplification of cDNA ends) analysis, the transcriptional initiation site was identified. Promoter deletion analysis showed that the sequence between -210 and -7 contains the basal promoter for USP22 in human fibroblast and tumor cells. Surprisingly, mutations in a putative Sp1 binding site immediately upstream of the USP22 transcriptional start site (-13 to -7) resulted in a significant induction of promoter activity. Further study revealed that Sp1 binds to this site in human normal fibroblast cells, and treatment with the Sp1 inhibitor mithramycin A led to a marked increase in USP22 transcript levels. Forced expression of exogenous Sp1 repressed the USP22 promoter activity in HeLa cells. In contrast, knockdown of Sp1 enhanced USP22 promoter activity and mRNA levels. These data suggest that Sp1 is a crucial regulator of USP22 transcription.


Subject(s)
Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Thiolester Hydrolases/genetics , 5' Flanking Region , Base Sequence , Binding Sites , Cloning, Molecular , Codon, Initiator , Enzyme Induction , Genes, Reporter , HeLa Cells , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Sequence Analysis, DNA , Sp1 Transcription Factor/physiology , Thiolester Hydrolases/metabolism , Transcriptional Activation , Ubiquitin Thiolesterase
SELECTION OF CITATIONS
SEARCH DETAIL