Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 8(3): e59045, 2013.
Article in English | MEDLINE | ID: mdl-23527084

ABSTRACT

Bone Morphogenetic Proteins (BMPs) are morphogens that play a major role in regulating development and homeostasis. Although BMPs are used for the treatment of bone and kidney disorders, their clinical use is limited due to the supra-physiological doses required for therapeutic efficacy causing severe side effects. Because recombinant BMPs are expensive to produce, small molecule activators of BMP signaling would be a cost-effective alternative with the added benefit of being potentially more easily deliverable. Here, we report our efforts to identify small molecule activators of BMP signaling. We have developed a cell-based assay to monitor BMP signaling by stably transfecting a BMP-responsive human cervical carcinoma cell line (C33A) with a reporter construct in which the expression of luciferase is driven by a multimerized BMP-responsive element from the Id1 promoter. A BMP-responsive clone C33A-2D2 was used to screen a bioactive library containing ∼5,600 small molecules. We identified four small molecules of the family of flavonoids all of which induced luciferase activity in a dose-dependent manner and ventralized zebrafish embryos. Two of the identified compounds induced Smad1, 5 phosphorylation (P-Smad), Id1 and Id2 expression in a dose-dependent manner demonstrating that our assays identified small molecule activators of BMP signaling.


Subject(s)
Bone Morphogenetic Proteins/agonists , Bone Morphogenetic Proteins/metabolism , Drug Discovery , Signal Transduction/drug effects , Small Molecule Libraries , Animals , Cell Line, Tumor , Chalcone/pharmacology , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Flavones/pharmacology , Genes, Reporter , High-Throughput Screening Assays , Humans , Mice , Mice, Knockout , Myoblasts/cytology , Myoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Zebrafish
2.
BMC Cancer ; 12: 154, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22533991

ABSTRACT

BACKGROUND: Alveolar rhabdomyosarcoma (ARMS) has a high propensity to metastasize, leading to its aggressiveness and a poor survival rate among those with the disease. More than 80% of aggressive ARMSs harbor a PAX3-FKHR fusion transcription factor, which regulates cell migration and promotes metastasis, most likely by regulating the fusion protein's transcriptional targets. Therefore, identifying druggable transcription targets of PAX3-FKHR that are also downstream effectors of PAX3-FKHR-mediated cell migration and metastasis may lead to novel therapeutic approaches for treating ARMS. METHODS: To identify genes whose expression is directly affected by the level of PAX3-FKHR in an ARMS cellular-context, we first developed an ARMS cell line in which PAX3-FKHR is stably down-regulated, and showed that stably downregulating PAX3-FKHR in ARMS cells significantly decreased the cells' motility. We used microarray analysis to identify genes whose expression level decreased when PAX3-FKHR was downregulated. We used mutational analysis, promoter reporter assays, and electrophoretic mobility shift assays to determine whether PAX3-FKHR binds to the promoter region of the target gene. We used siRNA and pharmacologic inhibitor to downregulate the target gene of PAX3-FKHR and investigated the effect of such downregulation on cell motility. RESULTS: We found that when PAX3-FKHR was downregulated, the expression of carnitine palmitoyltransferase 1A (CPT1A) decreased. We showed that PAX3-FKHR binds to a paired-domain binding-site in the CPT1A promoter region, indicating that CPT1A is a novel transcriptional target of PAX3-FKHR. Furthermore, downregulating CPT1A decreased cell motility in ARMS cells, indicating that CPT1A is a downstream effector of PAX3-FKHR-mediated cell migration and metastasis. CONCLUSIONS: Taken together, we have identified CPT1A as a novel transcriptional target of PAX3-FKHR and revealed the novel function of CPT1A in promoting cell motility. CPT1A may represent a novel therapeutic target for the treatment of ARMS.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/metabolism , Transcription, Genetic , Binding Sites , Cell Line, Tumor , Down-Regulation/genetics , Humans , Oncogene Proteins, Fusion/genetics , Paired Box Transcription Factors/metabolism , Response Elements , Transcriptional Activation
3.
Curr Chem Genomics ; 5: 42-7, 2011.
Article in English | MEDLINE | ID: mdl-21804905

ABSTRACT

Large-scale screening of small organic compounds has become a standard and essential practice in the early discovery of chemical entities with potential therapeutic use. To effectively support high-throughput screening campaigns, compound collections have to be in suitable formats, which requires a process known as compound reformatting. Here we report our approach to reformat the newly-established chemical repository of a large-scale screening facility at St. Jude Children's Research Hospital, which comprises more than half a million compounds, mostly from commercial sources. We highlight the timeline for a reformatting process, the importance of standardizing the operational procedures, and the advantages and disadvantages of using automation. The end result of our reformatting process is the concurrent generation of copies for long-term storage, screening, and "cherry-picking"; all of which facilitate compound management and high-throughput screening.

4.
J Biomol Screen ; 16(4): 394-404, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21471460

ABSTRACT

Class O forkhead box (FOXO) transcription factors are downstream targets of the PI3K/AKT signaling pathway, which is upregulated in many tumors. AKT phosphorylates and inactivates FOXO1 by relocating it to the cytoplasm. Because FOXO1 functions as a tumor suppressor by negatively regulating cell cycle progression and cell survival, compounds that promote FOXO1 localization to the nucleus might have therapeutic value in oncology. Here the authors describe the identification of such compounds by using an image-based, high-content screen. Compounds that were active in retaining FOXO1 in the nucleus were tested to determine their pathway specificity and isoform specificity by using high-content assays for Rev and FOXO3, respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Forkhead Transcription Factors/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Line , Cell Nucleus/metabolism , Humans , Protein Transport/drug effects , Small Molecule Libraries
5.
Chem Biol ; 17(8): 892-902, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797618

ABSTRACT

Pantothenate kinase (PanK) catalyzes the rate-controlling step in coenzyme A (CoA) biosynthesis. PanK3 is stringently regulated by acetyl-CoA and uses an ordered kinetic mechanism with ATP as the leading substrate. Biochemical analysis of site-directed mutants indicates that pantothenate binds in a tunnel adjacent to the active site that is occupied by the pantothenate moiety of the acetyl-CoA regulator in the PanK3acetyl-CoA binary complex. A high-throughput screen for PanK3 inhibitors and activators was applied to a bioactive compound library. Thiazolidinediones, sulfonylureas and steroids were inhibitors, and fatty acyl-amides and tamoxifen were activators. The PanK3 activators and inhibitors either stimulated or repressed CoA biosynthesis in HepG2/C3A cells. The flexible allosteric acetyl-CoA regulatory domain of PanK3 also binds the substrates, pantothenate and pantetheine, and small molecule inhibitors and activators to modulate PanK3 activity.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Small Molecule Libraries/metabolism , Acetyl Coenzyme A/metabolism , Allosteric Regulation/drug effects , Binding Sites , Coenzyme A/biosynthesis , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hep G2 Cells , High-Throughput Screening Assays , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Substrate Specificity , Sulfonylurea Compounds/metabolism , Thiazolidinediones/metabolism
6.
Drug Metab Dispos ; 38(9): 1411-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20538721

ABSTRACT

The human pregnane X receptor (hPXR) regulates the expression of CYP3A4, which plays a vital role in hepatic drug metabolism and has considerably reduced expression levels in proliferating hepatocytes. We have recently shown that cyclin-dependent kinase 2 (CDK2) negatively regulates hPXR-mediated CYP3A4 gene expression. CDK2 can be dephosphorylated and inactivated by protein phosphatase type 2C beta isoform long (PP2Cbetal), a unique phosphatase that was originally cloned from human liver. In this study, we sought to determine whether PP2Cbetal is involved in regulating hPXR's transactivation activity and whether PP2Cbetal affects CDK2 regulation of this activity in HepG2 liver carcinoma cells. In transactivation assays, transiently coexpressed PP2Cbetal significantly enhanced the hPXR-mediated CYP3A4 promoter activity and decreased the inhibitory effect of CDK2 on hPXR transactivation activity. In addition, shRNA-mediated down-regulation of endogenous PP2Cbetal promoted cell proliferation, inhibited the interaction of hPXR with steroid receptor coactivator-1, and attenuated the hPXR transcriptional activity. The levels of PP2Cbetal did not affect hPXR expression. Our results show for the first time that PP2Cbetal is essential for hPXR activity and can positively regulate this activity by counteracting the inhibitory effect of CDK2. Our results implicate a novel and important role for PP2Cbetal in regulating hPXR activity and CYP3A4 expression by inhibiting or desensitizing signaling pathways that negatively regulate the function of pregnane X receptor in liver cells and are consistent with the notion that both the activity of hPXR and the expression of CYP3A4 are regulated in a cell cycle-dependent and cell proliferation-dependent manner.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Gene Expression Regulation, Enzymologic , Liver Neoplasms/enzymology , Phosphoprotein Phosphatases/metabolism , Receptors, Steroid/physiology , Base Sequence , Cell Cycle , Cell Line, Tumor , DNA Primers , Down-Regulation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Pregnane X Receptor , Promoter Regions, Genetic , Protein Phosphatase 2C , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Two-Hybrid System Techniques
7.
Biochem Biophys Res Commun ; 391(1): 1049-55, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19995556

ABSTRACT

Patients with alveolar rhabdomyosarcoma (ARMS) have poorer response to conventional chemotherapy and lower survival rates than those with embryonal RMS (ERMS). To identify compounds that preferentially block the growth of ARMS, we conducted a small-scale screen of 160 kinase inhibitors against the ARMS cell line Rh30 and ERMS cell line RD and identified inhibitors of glycogen synthase kinase 3 (GSK3), including TWS119 as ARMS-selective inhibitors. GSK3 inhibitors inhibited cell proliferation and induced apoptosis more effectively in Rh30 than RD cells. Ectopic expression of fusion protein PAX3-FKHR in RD cells significantly increased their sensitivity to TWS119. Down-regulation of GSK3 by GSK3 inhibitors or siRNA significantly reduced the transcriptional activity of PAX3-FKHR. These results suggest that GSK3 is directly involved in regulating the transcriptional activity of PAX3-FKHR. Also, GSK3 phosphorylated PAX3-FKHR in vitro, suggesting that GSK3 might regulate PAX3-FKHR activity via phosphorylation. These findings support a novel mechanism of PAX3-FKHR regulation by GSK3 and provide a novel strategy to develop GSK inhibitors as anti-ARMS therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Paired Box Transcription Factors/metabolism , Protein Kinase Inhibitors/pharmacology , Rhabdomyosarcoma, Alveolar/enzymology , Antineoplastic Agents/isolation & purification , Drug Screening Assays, Antitumor , Forkhead Box Protein O1 , Glycogen Synthase Kinase 3/metabolism , Humans , PAX3 Transcription Factor , Phosphorylation , Protein Kinase Inhibitors/isolation & purification , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rhabdomyosarcoma, Alveolar/pathology
8.
Cancer Lett ; 284(2): 157-64, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19442434

ABSTRACT

Patients with alveolar rhabdomyosarcoma (ARMS) have poorer response to conventional chemotherapy and lower survival rates than those with embryonal RMS (ERMS). By high-throughput screening, we identified camptothecin as an ARMS-selective inhibitor. Camptothecin more efficiently inhibited proliferation and induced apoptosis in Rh30 (ARMS) than RD (ERMS) cells. Ectopic expression of the PAX3-FKHR (PF) fusion protein in RD cells significantly increased sensitivity, whereas siRNA knockdown of PF decreased sensitivity of Rh30 cells to camptothecin. The sensitization required a transcriptionally active PF, and camptothecin downregulated levels of PF protein. These findings suggest that it is feasible to develop agents that preferentially block the growth of ARMS.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Forkhead Transcription Factors/physiology , Rhabdomyosarcoma, Alveolar/metabolism , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , RNA Interference , RNA, Small Interfering/pharmacology , Rhabdomyosarcoma, Alveolar/drug therapy , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Transcription, Genetic
9.
Phys Med Rehabil Clin N Am ; 16(2): 351-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15893676

ABSTRACT

The precise cause of MS remains unknown, although immunopathologic studies provide solid evidence of participation of activated immune cells in the formation of MS lesions. Cell-mediated and antibody-mediated(humoral) immune responses are involved in the immunopathogenesis of MS. The Thl and Th2 cells, by releasing a variety of pro- or anti-inflammatory cytokines, work in opposition to balance the immune response and to determine the net effect of the inflammation. The current understanding of immunopathogenesis shapes the contemporary approach to the treatment of MS. The article also reviewed the mechanisms of action of commonly used immunotherapy agents and their impact on the immune response of patients who have MS.


Subject(s)
Multiple Sclerosis/immunology , Antibody Formation/immunology , Cytokines/immunology , Humans , Immunity, Cellular/immunology , Immunotherapy , Multiple Sclerosis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...