Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 93: 37-44, 2018 09.
Article in English | MEDLINE | ID: mdl-29895432

ABSTRACT

Glomerulonephritis (GN) is a typical lesion in autoantibody and immune complex disorders, including SLE. Because the Gas6/Axl pathway has been implicated in the pathogenesis of many types of GN, targeting this pathway might ameliorate GN. Consequently, we have studied the efficacy and mechanism of R428, a potent selective Axl inhibitor, in the prevention of experimental anti-GBM nephritis. Axl upregulation was investigated with Sp1/3 siRNA in the SV40-transformed mesangial cells. For Axl inhibition, a daily dose of R428 (125 mg/kg) or vehicle was administered orally. GN was induced with anti-GBM sera. Renal disease development was followed by serial blood urine nitrogen (BUN) determinations and by evaluation of kidney histology at the time of sacrifice. Axl-associated signaling proteins were analyzed by Western blotting and inflammatory cytokine secretion was analyzed by Proteome array. SiRNA data revealed the transcription factor Sp1 to be an important regulator of mesangial Axl expression. Anti-GBM serum induced severe nephritis with azotemia, protein casts and necrotic cell death. R428 treatment diminished renal Axl expression and improved kidney function, with significantly decreased BUN and glomerular proliferation. R428 treatment inhibited Axl and significantly decreased Akt phosphorylation and renal inflammatory cytokine and chemokine expression; similar effects were observed in anti-GBM antiserum-treated Axl-KO mice. These studies support a role for Axl inhibition in glomerulonephritis.


Subject(s)
Benzocycloheptenes/pharmacology , Immunologic Factors/pharmacology , Lupus Nephritis/drug therapy , Mesangial Cells/drug effects , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Triazoles/pharmacology , Administration, Oral , Animals , Antibodies/administration & dosage , Cell Line, Transformed , Drug Administration Schedule , Gene Expression Regulation , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Lupus Nephritis/chemically induced , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Mesangial Cells/immunology , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/immunology , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/immunology , Axl Receptor Tyrosine Kinase
2.
Gene ; 536(1): 135-44, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24315822

ABSTRACT

Microsomal epoxide hydrolase (mEH, EPHX1) is a critical biotransformation enzyme, catalyzing the metabolism of many xenobiotics. Human mEH is transcribed using alternative promoters. The upstream E1 promoter is active in liver while the far upstream E1b promoter drives the expression of mEH in all tissues, including liver. Although several liver-specific transcription factors have been identified in the regulation of E1 transcription, little is known regarding the mechanisms of E1b transcriptional regulation. Genome-wide mapping of DNase I hypersensitive sites revealed an open chromatin region between nucleotide -300 upstream and +400 downstream of E1b. This area coincides with a previously described promoter region responsible for maintaining high basal promoter activity. In silico analysis of this location revealed several Sp1/Sp3 binding sites. Site-directed mutagenesis of these motifs suppressed the transactivation activity of the E1b proximal promoter, indicating their importance as contributors to E1b promoter regulation. Further, E1b promoter activities were increased significantly following Sp1 and Sp3 overexpression, while Mithramycin A, a selective Sp1 inhibitor, reduced the promoter activities. EMSA studies demonstrated that Sp1 bound to two putative Sp1/Sp3 binding sites. ChIP analysis confirmed that both endogenous Sp1 and Sp3 were bound to the proximal promoter region of E1b. Knockdown of Sp1 expression using siRNA did not alter the endogenous E1b transcriptional level, while knockdown of Sp3 greatly decreased E1b expression in different human cell lines. Taken together, these results support the concept that Sp1 and Sp3 are functionally involved as transcriptional integrators regulating the basal expression of the derived mEH E1b variant transcript.


Subject(s)
Epoxide Hydrolases/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor/physiology , Binding Sites/genetics , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Hep G2 Cells , Humans , Isoenzymes/genetics , Protein Binding/physiology , RNA, Small Interfering/pharmacology , Sp1 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/antagonists & inhibitors
3.
PLoS One ; 8(12): e80638, 2013.
Article in English | MEDLINE | ID: mdl-24324617

ABSTRACT

Mina is an epigenetic gene regulatory protein known to function in multiple physiological and pathological contexts, including pulmonary inflammation, cell proliferation, cancer and immunity. We showed previously that the level of Mina gene expression is subject to natural genetic variation linked to 21 SNPs occurring in the Mina 5' region. In order to explore the mechanisms regulating Mina gene expression, we set out to molecularly characterize the Mina promoter in the region encompassing these SNPs. We used three kinds of assays--reporter, gel shift and chromatin immunoprecipitation--to analyze a 2 kb genomic fragment spanning the upstream and intron 1 regions flanking exon 1. Here we discovered a pair of Mina promoters (P1 and P2) and a P1-specific enhancer element (E1). Pharmacologic inhibition and siRNA knockdown experiments suggested that Sp1/3 transcription factors trigger Mina expression through additive activity targeted to a cluster of four Sp1/3 binding sites forming the P1 promoter. These results set the stage for comprehensive analysis of Mina gene regulation from the context of tissue specificity, the impact of inherited genetic variation and the nature of upstream signaling pathways.


Subject(s)
Enhancer Elements, Genetic , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Transcriptional Activation , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Epigenesis, Genetic , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Polymorphism, Single Nucleotide , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/metabolism
4.
PLoS One ; 8(12): e83426, 2013.
Article in English | MEDLINE | ID: mdl-24391766

ABSTRACT

Uncoupling protein (UCP) 3 is a mitochondrial inner membrane protein implicated in lipid handling and metabolism of reactive oxygen species. Its transcription is mainly regulated by peroxisome proliferator-activated receptors (PPAR), a family of nuclear hormone receptors. Employing bandshift assays, RNA interference and reporter gene assays we examine an intronic region in the UCP3 gene harboring a cis-element essential for expression in brown adipocytes. We demonstrate binding of SP1 and SP3 to this element which is adjacent to a direct repeat 1 element mediating activation of UCP3 expression by PPARγ agonists. Transactivation mediated by these elements is interdependent and indispensable for UCP3 expression. Systematic deletion uncovered a third binding element, a putative NF1 site, in close proximity to the SP1/3 and PPARγ binding elements. Data mining demonstrated binding of MyoD and Myogenin to this third element in C2C12 cells, and, furthermore, revealed recruitment of p300. Taken together, this intronic region is the main enhancer driving UCP3 expression with SP1/3 and PPARγ as the core factors required for expression.


Subject(s)
Adipocytes, Brown/metabolism , Ion Channels/genetics , Mitochondrial Proteins/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cricetinae , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Gene Knockdown Techniques , Introns , Mice , Models, Biological , Molecular Sequence Data , MyoD Protein/metabolism , Myogenin/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Phodopus , Protein Binding , Rats , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Uncoupling Protein 3 , p300-CBP Transcription Factors/metabolism
5.
FEBS J ; 279(15): 2714-26, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22682030

ABSTRACT

The overexpression of centromere protein H (CENPH), one of the fundamental components of the human active kinetochore, has been shown to be closely associated with human cancers. However, the mechanism of its transcriptional regulation has not been reported. The aim of the present study was to investigate the regulatory elements for the transcriptional regulation of CENPH in nasopharyngeal carcinoma cells. To characterize the CENPH promoter and identify regulatory elements, we cloned 1015 bp (-975/+40 bp) of the 5'-flanking region of the CENPH gene from immortalized normal nasopharyngeal epithelial cells (Bmi-1/NPEC). Functional analysis established a minimal region (-140/-87 bp) involved in the regulation of human CENPH promoter activity. Through site-directed mutagenesis, a transactivation assay, chromatin immunoprecipitation, and electrophoretic mobility shift assay, we found that the Sp1/Sp3 transcription factors could bind to the CENPH promoter in vitro and in vivo, and that they regulated CENPH promoter activation in human nasopharyngeal carcinoma cells. Furthermore, Sp1 and Sp3 were highly expressed in nasopharyngeal carcinoma cells. Knockdown of Sp1 and Sp3 by small interfering RNA or inhibition of Sp1 and Sp3 activity by mithramycin A decreased CENPH mRNA expression, whereas the exogenous expression of Sp1 and Sp3 upregulated CENPH mRNA expression. Taken together, our results indicate that Sp1 and Sp3 bind to the CENPH minimal promoter and function as a regulator of the transcription of CENPH in human nasopharyngeal carcinomas.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Base Sequence , Binding Sites/genetics , Carcinoma , Cell Line, Tumor , Cloning, Molecular , Gene Knockdown Techniques , Humans , Mutagenesis, Site-Directed , Nasopharyngeal Carcinoma , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulatory Elements, Transcriptional , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics
6.
Am J Physiol Gastrointest Liver Physiol ; 299(6): G1344-53, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20884886

ABSTRACT

Glutamine plays a key role in intestinal growth and maintenance of gut function, and as we have shown protects the postischemic gut (Kozar RA, Scultz SG, Bick RJ, Poindexter BJ, Desoigne R, Weisbrodt NW, Haber MM, Moore FA. Shock 21: 433-437, 2004). However, the precise mechanisms of the gut protective effects of glutamine have not been well elucidated. In the present study, RNA microarray was performed to obtain differentially expressed genes in intestinal epithelial IEC-6 cells following either 2 mM or 10 mM glutamine. The result demonstrated that specificity protein 3 (Sp3) mRNA expression was downregulated 3.1-fold. PCR and Western blot confirmed that Sp3 expression was decreased by glutamine in a time- and dose-dependent fashion. To investigate the role of Sp3, Sp3 gene siRNA silencing was performed and apoptosis was assessed. Silencing of Sp3 demonstrated a significant increase in Bcl-2 and decrease in Bax protein expression, as well as a decrease in caspase-3, -8, and -9 protein expression and activity. The protein expression of apoptosis-related proteins after hypoxia/reoxygenation was similar to that of normoxia and correlated with a decrease in DNA fragmentation. Importantly, the addition of glutamine to Sp3-silenced cells did not further lessen apoptosis, suggesting that Sp3 plays a major role in the inhibitory effect of glutamine on apoptosis. This novel finding may explain in part the gut-protective effects of glutamine.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Glutamine/pharmacology , Intestinal Mucosa/cytology , Sp3 Transcription Factor/metabolism , Animals , Apoptosis/physiology , Caspases/genetics , Caspases/metabolism , Cell Line , Dose-Response Relationship, Drug , Down-Regulation , Epithelial Cells/physiology , Gene Expression Profiling , Gene Silencing , Genes, bcl-2/physiology , RNA, Small Interfering , Rats , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics
7.
Biosci Rep ; 28(3): 161-76, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18522535

ABSTRACT

Axl is a receptor tyrosine kinase which promotes anti-apoptosis, mitogenesis, invasion, angiogenesis and metastasis, and is highly expressed in cancers. However, the transcriptional regulation of this important gene has never been characterized. The present study was initiated to characterize the promoter, cis-acting elements and promoter methylation driving expression of Axl. The 2.4 kb sequence upstream of the translational start site, and sequential 5'-deletions were cloned and revealed a minimal GC-rich region (-556 to +7) to be sufficient for basal Axl promoter activity in Rko, HCT116 and HeLa cells. Within this minimal region, five Sp (specificity protein)-binding sites were identified. Two sites (Sp a and Sp b) proximal to the translation start site were indispensable for Axl promoter activity, whereas mutation of three additional upstream motifs (Sp c, Sp d and Sp e) was of additional relevance. Gel-shift assays and chromatin immunoprecipitation identified that Sp1 and Sp3 bound to all five motifs, and mutation of all motifs abolished binding. Mithramycin, which inhibits binding of Sp factors to GC-rich sites, dramatically reduced Axl promoter activity and Axl, Sp1 and Sp3 expression. In Drosophila Schneider SL2-cells, exogenous expression of Sp1/Sp3 increased Axl promoter activity. Use of Sp1/Sp3 siRNAs (small interfering RNAs) significantly reduced Axl promoter activity and protein levels in Rko and HeLa cells. Methylation-bisulfite sequencing detected methylated CpG sites within three Sp motifs (Sp a, Sp b and Sp c) and GC-rich flanking sequences, and demethylation by 5-aza-2'-deoxycytidine up-regulated Axl and Sp3 expression in low-Axl-expressing Colo206f/WiDr cells, but not in high-Axl-expressing Rko cells. The results of the present study suggest that Axl gene expression in cancer cells is (1) constitutively driven by Sp1/Sp3 bound to five core promoter motifs, and (2) restricted by methylation within/around Sp-binding sites. This might enhance the understanding and treatment of essential mechanisms associated with cancer and other diseases.


Subject(s)
CpG Islands/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Promoter Regions, Genetic/genetics , Receptor Protein-Tyrosine Kinases/genetics , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor/physiology , Amino Acid Motifs , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor/metabolism , Colorectal Neoplasms/pathology , DNA Methylation/drug effects , Decitabine , Enzyme Induction/drug effects , Enzyme Induction/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Mutagenesis, Site-Directed , Neoplasm Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins , RNA Interference , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Axl Receptor Tyrosine Kinase
8.
Gene ; 413(1-2): 58-66, 2008 Apr 30.
Article in English | MEDLINE | ID: mdl-18343609

ABSTRACT

The LAT gene encodes an adaptor molecule that links receptor engagement to critical downstream signaling events. Previously, we identified the proximal promoter for the human LAT gene and found that it contains binding sites for members of the Ets and Runx transcription factor families. In the present study, we show that the promoter also contains 5 GC-rich elements that contribute to promoter activity and that are capable of binding the transcription factors Sp1 and Sp3. Overexpression of either Sp1 or full-length Sp3 was shown to augment LAT promoter activity, while siRNA-mediated knockdown of each transcription factor was demonstrated to have an inhibitory effect. We also discovered a cell-type specific DNase hypersensitive site that maps to the Sp1/Sp3 and adjacent Ets and Runx binding sites. Collectively, these results provide compelling data that implicates Sp1 and Sp3 in the transcriptional regulation of the human LAT gene.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Membrane Proteins/genetics , Regulatory Elements, Transcriptional , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , Drosophila , GC Rich Sequence , HeLa Cells , Humans , Jurkat Cells , Molecular Sequence Data , Mutagenesis, Site-Directed , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Transcription, Genetic
9.
Nucleic Acids Res ; 36(1): 330-41, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18033804

ABSTRACT

Cell-death-inducing DFF45-like effector A (CIDE-A) belongs to a family of proapoptotic proteins, the expression of which is highly restricted in human tissues and cells. Here, the core region of the human CIDE-A promoter was characterized. Surprisingly, two Sp1/Sp3-binding sites, rather than tissue-specific transcription factors, were found to be required for the promoter activity. Although the ubiquitously expressed Sp1 and Sp3 were crucial, they alone could not adequately regulate the specific expression of CIDE-A. We found that the expression of CIDE-A was further regulated by CpG methylation of the promoter region. By performing bisulfite sequencing, we observed dense CpG methylation of the promoter region in tissues and cells with low or no expression of CIDE-A but not in tissues with high level of CIDE-A expression. In vitro methylation of this region showed significantly reduced transcriptional activity. Treatment of CIDE-A-negative cells with 5-aza-2'-deoxycytidine demethylated the CpG sites; this opened the closed chromatin conformation and markedly enhanced the binding affinity of Sp1/Sp3 to the promoter in vivo, thereby restoring CIDE-A expression. These data indicated that CpG methylation plays a crucial role in establishing and maintaining tissue- and cell-specific transcription of the CIDE-A gene through the regulation of Sp1/Sp3 binding.


Subject(s)
Apoptosis Regulatory Proteins/genetics , CpG Islands , Gene Expression Regulation , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Binding Sites , Cell Line , Cell Line, Tumor , Chromatin/drug effects , DNA Methylation , Decitabine , Exons , Gene Expression/drug effects , Humans , Molecular Sequence Data , Promoter Regions, Genetic , RNA Interference , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Tissue Distribution
10.
Nucleic Acids Res ; 35(11): 3784-96, 2007.
Article in English | MEDLINE | ID: mdl-17517765

ABSTRACT

APOBEC3G (A3G), a member of the recently discovered family of human cytidine deaminases, is expressed in peripheral blood lymphocytes and has been shown to be active against HIV-1 and other retroviruses. To gain new insights into the transcriptional regulation of this restriction factor, we cloned and characterized the promoter region of A3G. Transcriptional start sites were identified by 5'-rapid amplification of cDNA ends analysis. Luciferase reporter assays demonstrated that a 1025 bp A3G promoter sequence (from -959 to +66 relative to the major transcriptional start site) displayed constitutive promoter activity. In T cells, the A3G promoter was not inducible by mitogenic stimulation, interferon treatment or expression of HIV-1 proteins. Using a series of 5' deletion promoter constructs in luciferase reporter assays, we identified a 180 bp region that was sufficient for full promoter activity. Transcriptional activity of this A3G core promoter was dependent on a GC-box (located at position -87/-78 relative to the major transcriptional start site) and was abolished after mutation of this DNA element. Electrophoretic mobility shift assays and chromatin immunoprecipitation assays demonstrated that the identified GC-box represented a binding site for the ubiquitous transcription factors specificity protein (Sp) 1 and Sp3.


Subject(s)
Gene Expression Regulation , Nucleoside Deaminases/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , APOBEC-3G Deaminase , Base Sequence , Binding Sites , Cell Line , Cytidine Deaminase , Humans , Molecular Sequence Data , Polymerase Chain Reaction/methods , RNA Interference , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , T-Lymphocytes/metabolism , Transcription Initiation Site , Transcription, Genetic
11.
Cancer Res ; 67(6): 2816-23, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363604

ABSTRACT

Betulinic acid is a pentacyclic triterpene natural product initially identified as a melanoma-specific cytotoxic agent that exhibits low toxicity in animal models. Subsequent studies show that betulinic acid induces apoptosis and antiangiogenic responses in tumors derived from multiple tissues; however, the underlying mechanism of action is unknown. Using LNCaP prostate cancer cells as a model, we now show that betulinic acid decreases expression of vascular endothelial growth (VEGF) and the antiapoptotic protein survivin. The mechanism of these betulinic acid-induced antiangiogenic and proapoptotic responses in both LNCaP cells and in tumors is due to activation of selective proteasome-dependent degradation of the transcription factors specificity protein 1 (Sp1), Sp3, and Sp4, which regulate VEGF and survivin expression. Thus, betulinic acid acts as a novel anticancer agent through targeted degradation of Sp proteins that are highly overexpressed in tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Prostatic Neoplasms/drug therapy , Sp Transcription Factors/antagonists & inhibitors , Triterpenes/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Pentacyclic Triterpenes , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Sp Transcription Factors/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/antagonists & inhibitors , Sp4 Transcription Factor/genetics , Sp4 Transcription Factor/metabolism , Survivin , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Betulinic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...