Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
SLAS Discov ; 27(6): 349-357, 2022 09.
Article in English | MEDLINE | ID: mdl-35580766

ABSTRACT

Small-molecule high-throughput screening (HTS) campaigns have frequently been used to identify lead molecules that can alter expression of disease-relevant proteins in cell-based assays. However, most cell-based HTS assays require short compound exposure periods to avoid toxicity and ensure that compounds are stable in media for the duration of the exposure. This limits the ability of HTS assays to detect inhibitors of the synthesis of target proteins with long half-lives, which can often exceed the exposure times utilized in most HTS campaigns. One such target is alpha-synuclein (α-syn)-a protein well-known for its pathological aggregation in Parkinson's Disease (PD) and other forms of neurodegeneration known collectively as synucleinopathies. Here, we report the development of an HTS assay using a CRISPR-engineered neuroblastoma cell line expressing a destabilized luciferase reporter inserted at the end of the coding region of the SNCA locus. The resultant destabilized fusion protein exhibited a significant reduction in half-life compared to the endogenous, unmodified α-syn protein, and accurately reported reductions in α-syn levels due to known protein translation inhibitors and specific α-syn siRNAs. The robustness and utility of this approach was shown by using the resulting cell line (dsLuc-Syn) to screen a focused library of 3,192 compounds for reduction of α-syn. These data demonstrate the general utility of converting endogenous loci into destabilized reporter genes capable of identifying inhibitors of gene expression of highly stable proteins even in short-term assays.


Subject(s)
Parkinson Disease , alpha-Synuclein , Cell Line , Gene Expression , High-Throughput Screening Assays/methods , Humans , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
2.
ACS Chem Biol ; 15(12): 3149-3158, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33206504

ABSTRACT

There is a growing interest in using targeted protein degradation as a therapeutic modality in view of its potential to expand the druggable proteome. One avenue to using this modality is via molecular glue based Cereblon E3 Ligase Modulating Drug compounds. Here, we report the identification of the transcription factor ZBTB16 as a Cereblon neosubstrate. We also report two new Cereblon modulators, CC-3060 and CC-647, that promote ZBTB16 degradation. Unexpectedly, CC-3060 and CC-647 target ZBTB16 for degradation by primarily engaging distinct structural degrons on different zinc finger domains. The reciprocal fusion proteins, ZBTB16-RARα and RARα-ZBTB16, which cause a rare acute promyelocytic leukemia, contain these same structural degrons and can be targeted for proteasomal degradation with Cereblon modulator treatment. Thus, a targeted protein degradation approach via Cereblon modulators may represent a novel therapeutic strategy in acute promyelocytic leukemia where ZBTB16/RARA rearrangements are critical disease drivers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Oncogene Proteins, Fusion/metabolism , Promyelocytic Leukemia Zinc Finger Protein/drug effects , Ubiquitin-Protein Ligases/metabolism , Humans , Leukemia, Promyelocytic, Acute/metabolism , Proteolysis , Retinoic Acid Receptor alpha/metabolism , Substrate Specificity
3.
Chem Biol ; 22(9): 1228-37, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26364931

ABSTRACT

In an attempt to identify novel therapeutics and mechanisms to differentially kill tumor cells using phenotypic screening, we identified N-benzyl indole carbinols (N-BICs), synthetic analogs of the natural product indole-3-carbinol (I3C). To understand the mode of action for the molecules we employed Cancer Cell Line Encyclopedia viability profiling and correlative informatics analysis to identify and ultimately confirm the phase II metabolic enzyme sulfotransferase 1A1 (SULT1A1) as the essential factor for compound selectivity. Further studies demonstrate that SULT1A1 activates the N-BICs by rendering the compounds strong electrophiles which can alkylate cellular proteins and thereby induce cell death. This study demonstrates that the selectivity profile for N-BICs is through conversion by SULT1A1 from an inactive prodrug to an active species that induces cell death and tumor suppression.


Subject(s)
Arylsulfotransferase/metabolism , Benzyl Compounds/pharmacology , Indoles/pharmacology , Animals , Benzyl Compounds/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , HCT116 Cells , Humans , Indoles/pharmacokinetics , Mice , Mice, Nude , Random Allocation , Xenograft Model Antitumor Assays
4.
PLoS One ; 10(5): e0127480, 2015.
Article in English | MEDLINE | ID: mdl-25996873

ABSTRACT

Intracellular calcium signaling is critical for initiating and sustaining diverse cellular functions including transcription, synaptic signaling, muscle contraction, apoptosis and fertilization. Trans-membrane 203 (TMEM203) was identified here in cDNA overexpression screens for proteins capable of modulating intracellular calcium levels using activation of a calcium/calcineurin regulated transcription factor as an indicator. Overexpression of TMEM203 resulted in a reduction of Endoplasmic Reticulum (ER) calcium stores and elevation in basal cytoplasmic calcium levels. TMEM203 protein was localized to the ER and found associated with a number of ER proteins which regulate ER calcium entry and efflux. Mouse Embryonic Fibroblasts (MEFs) derived from Tmem203 deficient mice had reduced ER calcium stores and altered calcium homeostasis. Tmem203 deficient mice were viable though male knockout mice were infertile and exhibited a severe block in spermiogenesis and spermiation. Expression profiling studies showed significant alternations in expression of calcium channels and pumps in testes and concurrently Tmem203 deficient spermatocytes demonstrated significantly altered calcium handling. Thus Tmem203 is an evolutionarily conserved regulator of cellular calcium homeostasis, is required for spermatogenesis and provides a causal link between intracellular calcium regulation and spermiogenesis.


Subject(s)
Calcium/metabolism , Homeostasis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Spermatogenesis , Animals , Calcineurin/metabolism , Calcium Signaling , Cell Line , Endoplasmic Reticulum/metabolism , Epididymis/metabolism , Epididymis/pathology , Female , Gene Expression , Gene Expression Regulation , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Intracellular Space/metabolism , Male , Mice , Mice, Knockout , Protein Binding , Transcription Factors/metabolism
5.
Nat Cell Biol ; 16(11): 1069-79, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25327288

ABSTRACT

Cells rely on autophagy to clear misfolded proteins and damaged organelles to maintain cellular homeostasis. In this study we use the new autophagy inhibitor PIK-III to screen for autophagy substrates. PIK-III is a selective inhibitor of VPS34 that binds a unique hydrophobic pocket not present in related kinases such as PI(3)Kα. PIK-III acutely inhibits autophagy and de novo lipidation of LC3, and leads to the stabilization of autophagy substrates. By performing ubiquitin-affinity proteomics on PIK-III-treated cells we identified substrates including NCOA4, which accumulates in ATG7-deficient cells and co-localizes with autolysosomes. NCOA4 directly binds ferritin heavy chain-1 (FTH1) to target the iron-binding ferritin complex with a relative molecular mass of 450,000 to autolysosomes following starvation or iron depletion. Interestingly, Ncoa4(-/-) mice exhibit a profound accumulation of iron in splenic macrophages, which are critical for the reutilization of iron from engulfed red blood cells. Taken together, the results of this study provide a new mechanism for selective autophagy of ferritin and reveal a previously unappreciated role for autophagy and NCOA4 in the control of iron homeostasis in vivo.


Subject(s)
Autophagy/physiology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Ferritins/metabolism , Homeostasis/physiology , Iron/metabolism , Nuclear Receptor Coactivators/metabolism , Animals , Autophagy/drug effects , Cells, Cultured , Humans , Lysosomes/metabolism , Mice , Phagosomes/metabolism , Protein Binding
6.
Chem Biol ; 20(7): 912-21, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23890009

ABSTRACT

Toll-like receptor (TLR) signaling is a key component of innate immunity. Aberrant TLR activation leads to immune disorders via dysregulation of cytokine production, such as IL-12/IL-23. Herein, we identify and characterize PIKfyve, a lipid kinase, as a critical player in TLR signaling using apilimod as an affinity tool. Apilimod is a potent small molecular inhibitor of IL-12/IL-23 with an unknown target and has been evaluated in clinical trials for patients with Crohn's disease or rheumatoid arthritis. Using a chemical genetic approach, we show that it binds to PIKfyve and blocks its phosphotransferase activity, leading to selective inhibition of IL-12/IL-23p40. Pharmacological or genetic inactivation of PIKfyve is necessary and sufficient for suppression of IL-12/IL-23p40 expression. Thus, we have uncovered a phosphoinositide-mediated regulatory mechanism that controls TLR signaling.


Subject(s)
Interleukin-12/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Triazines/pharmacology , Animals , Cell Line , Cytokines/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Hydrazones , Mice , Morpholines/metabolism , Protein Binding , Pyrimidines , Substrate Specificity , Triazines/metabolism
7.
PLoS One ; 8(4): e60913, 2013.
Article in English | MEDLINE | ID: mdl-23637775

ABSTRACT

Lysine-specific demethylase 1 (Lsd1/Aof2/Kdm1a), the first enzyme with specific lysine demethylase activity to be described, demethylates histone and non-histone proteins and is essential for mouse embryogenesis. Lsd1 interacts with numerous proteins through several different domains, most notably the tower domain, an extended helical structure that protrudes from the core of the protein. While there is evidence that Lsd1-interacting proteins regulate the activity and specificity of Lsd1, the significance and roles of such interactions in developmental processes remain largely unknown. Here we describe a hypomorphic Lsd1 allele that contains two point mutations in the tower domain, resulting in a protein with reduced interaction with known binding partners and decreased enzymatic activity. Mice homozygous for this allele die perinatally due to heart defects, with the majority of animals suffering from ventricular septal defects. Molecular analyses revealed hyperphosphorylation of E-cadherin in the hearts of mutant animals. These results identify a previously unknown role for Lsd1 in heart development, perhaps partly through the control of E-cadherin phosphorylation.


Subject(s)
Alleles , Heart Defects, Congenital/genetics , Oxidoreductases, N-Demethylating/genetics , Animals , Cadherins/metabolism , Disease Models, Animal , Enzyme Activation , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Septal Defects, Ventricular/genetics , Heart Septal Defects, Ventricular/metabolism , Heart Septal Defects, Ventricular/pathology , Histone Demethylases , Homozygote , Mice , Mice, Knockout , Oxidoreductases, N-Demethylating/metabolism , Phosphorylation , Point Mutation , Pregnancy , Protein Binding
8.
Proc Natl Acad Sci U S A ; 110(11): E1026-34, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23431153

ABSTRACT

The calcium-activated chloride channel anoctamin 1 (ANO1) is located within the 11q13 amplicon, one of the most frequently amplified chromosomal regions in human cancer, but its functional role in tumorigenesis has remained unclear. The 11q13 region is amplified in ∼15% of breast cancers. Whether ANO1 is amplified in breast tumors, the extent to which gene amplification contributes to ANO1 overexpression, and whether overexpression of ANO1 is important for tumor maintenance have remained unknown. We have found that ANO1 is amplified and highly expressed in breast cancer cell lines and primary tumors. Amplification of ANO1 correlated with disease grade and poor prognosis. Knockdown of ANO1 in ANO1-amplified breast cancer cell lines and other cancers bearing 11q13 amplification inhibited proliferation, induced apoptosis, and reduced tumor growth in established cancer xenografts. Moreover, ANO1 chloride channel activity was important for cell viability. Mechanistically, ANO1 knockdown or pharmacological inhibition of its chloride-channel activity reduced EGF receptor (EGFR) and calmodulin-dependent protein kinase II (CAMKII) signaling, which subsequently attenuated AKT, v-src sarcoma viral oncogene homolog (SRC), and extracellular signal-regulated kinase (ERK) activation in vitro and in vivo. Our results highlight the involvement of the ANO1 chloride channel in tumor progression and provide insights into oncogenic signaling in human cancers with 11q13 amplification, thereby establishing ANO1 as a promising target for therapy in these highly prevalent tumor types.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Chloride Channels/metabolism , Chromosomes, Human, Pair 11/metabolism , Gene Amplification , Neoplasm Proteins/metabolism , Animals , Anoctamin-1 , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line, Tumor , Cell Survival/genetics , Chloride Channels/genetics , Chromosomes, Human, Pair 11/genetics , Enzyme Activation/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Transplantation , Signal Transduction/genetics , Transplantation, Heterologous
10.
Cell Res ; 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22143567

ABSTRACT

Lysine-specific demethylase 1 (LSD1/AOF2/KDM1A), the first enzyme with specific lysine demethylase activity to be described, demethylates histone and non-histone proteins and is essential for mouse embryogenesis. LSD1 interacts with numerous proteins through several different domains, most notably the tower domain, an extended helical structure that protrudes from the core of the protein. While there is evidence that LSD1-interacting proteins regulate the activity and specificity of LSD1, the significance and roles of such interactions in developmental processes remain largely unknown. Here we describe a hypomorphic LSD1 allele that contains two point mutations in the tower domain, resulting in a protein with reduced interaction with known binding partners and decreased enzymatic activity. Mice homozygous for this allele die perinatally due to heart defects, with the majority of animals suffering from ventricular septal defects. Transcriptional profiling revealed altered expression of a limited subset of genes in the hearts. This includes an increase in calmodulin kinase (CK) 2ß, the regulatory subunit of the CK2 kinase, which correlates with E-cadherin hyperphosphorylation. These results identify a previously unknown role for LSD1 in heart development, perhaps partly through the control of E-cadherin phosphorylation.Cell Research advance online publication 6 December 2011; doi:10.1038/cr.2011.194.

11.
Cancer Res ; 71(12): 4280-91, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21527556

ABSTRACT

RAS mutations occur in more than 30% of all human cancers but efforts to directly target mutant RAS signaling as a cancer therapy have yet to succeed. As alternative strategies, RAF and MEK inhibitors have been developed to block oncogenic signaling downstream of RAS. As might be expected, studies of these inhibitors have indicated that tumors with RAS or BRAF mutations display resistance RAF or MEK inhibitors. In order to better understand the mechanistic basis for this resistance, we conducted a RNAi-based screen to identify genes that mediated chemoresistance to the RAF kinase inhibitor RAF265 in a BRAF (V600E) mutant melanoma cell line that is resistant to this drug. In this way, we found that knockdown of protein kinase D3 (PRKD3) could enhance cell killing of RAF and MEK inhibitors across multiple melanoma cell lines of various genotypes and sensitivities to RAF265. PRKD3 blockade cooperated with RAF265 to prevent reactivation of the MAPK signaling pathway, interrupt cell cycle progression, trigger apoptosis, and inhibit colony formation growth. Our findings offer initial proof-of-concept that PRKD3 is a valid target to overcome drug resistance being encountered widely in the clinic with RAF or MEK inhibitors.


Subject(s)
Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Protein Kinase C/physiology , Pyridines/pharmacology , raf Kinases/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin D1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/physiology , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase C/antagonists & inhibitors , RNA, Small Interfering/genetics
12.
J Immunol ; 186(10): 6014-23, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21471444

ABSTRACT

Inflammatory cytokines like TNF play a central role in autoimmune disorders such as rheumatoid arthritis. We identified the tyrosine kinase bone marrow kinase on chromosome X (BMX) as an essential component of a shared inflammatory signaling pathway. Transient depletion of BMX strongly reduced secretion of IL-8 in cell lines and primary human cells stimulated by TNF, IL-1ß, or TLR agonists. BMX was required for phosphorylation of p38 MAPK and JNK, as well as activation of NF-κB. The following epistasis analysis indicated that BMX acts downstream of or at the same level as the complex TGF-ß activated kinase 1 (TAK1)-TAK1 binding protein. At the cellular level, regulation of the IL-8 promoter required the pleckstrin homology domain of BMX, which could be replaced by an ectopic myristylation signal, indicating a requirement for BMX membrane association. In addition, activation of the IL-8 promoter by in vitro BMX overexpression required its catalytic activity. Genetic ablation of BMX conferred protection in the mouse arthritis model of passive K/BxN serum transfer, confirming that BMX is an essential mediator of inflammation in vivo. However, genetic replacement with a catalytically inactive BMX allele was not protective in the same arthritis animal model. We conclude that BMX is an essential component of inflammatory cytokine signaling and that catalytic, as well as noncatalytic functions of BMX are involved.


Subject(s)
Arthritis/immunology , Protein-Tyrosine Kinases/metabolism , Animals , Arthritis/metabolism , Blood Proteins , Cell Line , Disease Models, Animal , HeLa Cells , Humans , Immunoblotting , Interleukin-1beta/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/metabolism , Phosphoproteins , Phosphorylation , Protein-Tyrosine Kinases/genetics , Signal Transduction , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Stem Cells ; 28(2): 201-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20014010

ABSTRACT

Dynamic regulation of histone methylation by methyltransferases and demethylases plays a central role in regulating the fate of embryonic stem (ES) cells. The histone H3K9 methyltransferase KMT1E, formerly known as ESET or Setdb1, is essential to embryonic development as the ablation of the Setdb1 gene results in peri-implantation lethality and prevents the propagation of ES cells. However, Setdb1-null blastocysts do not display global changes in H3K9 methylation or DNA methylation, arguing against a genome-wide defect. Here we show that conditional deletion of the Setdb1 gene in ES cells results in the upregulation of lineage differentiation markers, especially trophectoderm-specific factors, similar to effects observed upon loss of Oct3/4 expression in ES cells. We demonstrate that KMT1E deficiency in ES cells leads to a decrease in histone H3K9 methylation at and derepression of trophoblast-associated genes such as Cdx2. Furthermore, we find genes that are derepressed upon Setdb1 deletion to overlap with known targets of polycomb mediated repression, suggesting that KMT1E mediated H3K9 methylation acts in concert with polycomb controlled H3K27 methylation. Our studies thus demonstrate an essential role for KMT1E in the control of developmentally regulated gene expression programs in ES cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Protein Methyltransferases/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Histone-Lysine N-Methyltransferase , Histones/metabolism , Immunoblotting , Methylation , Mice , Oligonucleotide Array Sequence Analysis , Protein Methyltransferases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology
14.
J Virol ; 83(19): 10058-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19605471

ABSTRACT

Host factor pathways are known to be essential for hepatitis C virus (HCV) infection and replication in human liver cells. To search for novel host factor proteins required for HCV replication, we screened a subgenomic genotype 1b replicon cell line (Luc-1b) with a kinome and druggable collection of 20,779 siRNAs. We identified and validated several enzymes required for HCV replication, including class III phosphatidylinositol 4-kinases (PI4KA and PI4KB), carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD), and mevalonate (diphospho) decarboxylase. Knockdown of PI4KA could inhibit the replication and/or HCV RNA levels of the two subgenomic genotype 1b clones (SG-1b and Luc-1b), two subgenomic genotype 1a clones (SG-1a and Luc-1a), JFH-1 genotype 2a infectious virus (JFH1-2a), and the genomic genotype 1a (FL-1a) replicon. In contrast, PI4KB knockdown inhibited replication and/or HCV RNA levels of Luc-1b, SG-1b, and Luc-1a replicons. The small molecule inhibitor, PIK93, was found to block subgenomic genotype 1b (Luc-1b), subgenomic genotype 1a (Luc-1a), and genomic genotype 2a (JFH1-2a) infectious virus replication in the nanomolar range. PIK93 was characterized by using quantitative chemical proteomics and in vitro biochemical assays to demonstrate PIK93 is a bone fide PI4KA and PI4KB inhibitor. Our data demonstrate that genetic or pharmacological modulation of PI4KA and PI4KB inhibits multiple genotypes of HCV and represents a novel druggable class of therapeutic targets for HCV infection.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Hepacivirus/genetics , Hepacivirus/metabolism , Liver/virology , Virus Replication , 1-Phosphatidylinositol 4-Kinase/chemistry , Antiviral Agents/pharmacology , Binding, Competitive , Cell Line , Gene Silencing , Genotype , Humans , Inhibitory Concentration 50 , Mass Spectrometry/methods , Proteomics/methods , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/pharmacology
15.
Virology ; 389(1-2): 8-19, 2009 Jun 20.
Article in English | MEDLINE | ID: mdl-19419745

ABSTRACT

We performed a focused siRNA screen in an A549 dengue type 2 New Guinea C subgenomic replicon cell line (Rluc-replicon) that contains a Renilla luciferase cassette. We found that siRNA mediated knock down of mevalonate diphospho decarboxylase (MVD) inhibited viral replication of the Rluc-replicon and DEN-2 NGC live virus replication in A549 cells. When the Rluc-replicon A459 cells were grown in delipidated media the replicon expression was suppressed and MVD knock down could further sensitize Renilla expression. Hymeglusin and zaragozic acid A could inhibit DEN-2 NGC live virus replication in K562 cells, while lovastatin could inhibit DEN-2 NGC live virus replication in human peripheral blood mononuclear cells. Renilla expression could be rescued in fluvastatin treated A549 Rluc-replicon cells after the addition of mevalonate, and partially restored with geranylgeranyl pyrophosphate, or farnesyl pyrophosphate. Our data suggest genetic and pharmacological modulation of cholesterol biosynthesis can regulate dengue virus replication.


Subject(s)
Carboxy-Lyases/metabolism , Cholesterol/biosynthesis , Dengue Virus/physiology , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carboxy-Lyases/genetics , Dengue Virus/drug effects , Dengue Virus/genetics , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Gene Knockdown Techniques , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Indoles/pharmacology , K562 Cells , Mevalonic Acid/pharmacology , Polyisoprenyl Phosphates/pharmacology , Replicon/drug effects , Sesquiterpenes/pharmacology , Tricarboxylic Acids/pharmacology
16.
PLoS One ; 4(4): e5197, 2009.
Article in English | MEDLINE | ID: mdl-19381295

ABSTRACT

BACKGROUND: Lipid metabolism in mammals is orchestrated by a family of transcription factors called sterol regulatory element-binding proteins (SREBPs) that control the expression of genes required for the uptake and synthesis of cholesterol, fatty acids, and triglycerides. SREBPs are thus essential for insulin-induced lipogenesis and for cellular membrane homeostasis and biogenesis. Although multiple players have been identified that control the expression and activation of SREBPs, gaps remain in our understanding of how SREBPs are coordinated with other physiological pathways. METHODOLOGY: To identify novel regulators of SREBPs, we performed a genome-wide cDNA over-expression screen to identify proteins that might modulate the transcription of a luciferase gene driven from an SREBP-specific promoter. The results were verified through secondary biological assays and expression data were analyzed by a novel application of the Gene Set Enrichment Analysis (GSEA) method. CONCLUSIONS/SIGNIFICANCE: We screened 10,000 different cDNAs and identified a number of genes and pathways that have previously not been implicated in SREBP control and cellular cholesterol homeostasis. These findings further our understanding of lipid biology and should lead to new insights into lipid associated disorders.


Subject(s)
Sterol Regulatory Element Binding Proteins/physiology , Transcription, Genetic , Base Sequence , Cell Line , Cholesterol/metabolism , DNA, Complementary , Homeostasis , Humans , Promoter Regions, Genetic , Signal Transduction , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
17.
J Biol Chem ; 284(6): 3709-18, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19036726

ABSTRACT

Nanog, Oct4, and Sox2 form the core of a transcription factor network that maintains embryonic stem cells in the pluripotent state in both humans and mice. These critical factors have been implicated as both positive and negative regulators of transcription, varying by promoter and differentiation state of the cell. The Mediator complex, a ubiquitous conserved complex of approximately 30 subunits, facilitates transcription by coordinating RNA polymerase II binding to target promoters via gene-specific activators and can be divided into several functional subcomplexes. Med12 is part of a subcomplex of four proteins associated with the core Mediator complex and has been found to function both in repressing and activating transcription when recruited to target promoters. We identified an interaction between Med12 and Nanog and present evidence of involvement of Med12 in regulation of Nanog function. Gene expression analysis of embryonic stem cells knocked down for Med12 showed a similarity to Nanog knockdown, with increased expression of Nanog-repressed targets and decreased expression of Nanog-activated targets. Using chromatin immunoprecipitation, we found that Med12 and Nanog co-occupied Nanog target promoters in embryonic stem cells and that Med12 dissociated from target promoters upon differentiation with kinetics similar to Nanog. Our results indicate that Nanog and Med12 function in concert to regulate Nanog target genes and identify a novel role for Med12 in embryonic stem cell regulation.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Multiprotein Complexes/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Embryonic Stem Cells/cytology , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Mediator Complex , Mice , Multiprotein Complexes/genetics , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics
18.
Nat Chem Biol ; 4(1): 59-68, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18066055

ABSTRACT

High-content screening is transforming drug discovery by enabling simultaneous measurement of multiple features of cellular phenotype that are relevant to therapeutic and toxic activities of compounds. High-content screening studies typically generate immense datasets of image-based phenotypic information, and how best to mine relevant phenotypic data is an unsolved challenge. Here, we introduce factor analysis as a data-driven tool for defining cell phenotypes and profiling compound activities. This method allows a large data reduction while retaining relevant information, and the data-derived factors used to quantify phenotype have discernable biological meaning. We used factor analysis of cells stained with fluorescent markers of cell cycle state to profile a compound library and cluster the hits into seven phenotypic categories. We then compared phenotypic profiles, chemical similarity and predicted protein binding activities of active compounds. By integrating these different descriptors of measured and potential biological activity, we can effectively draw mechanism-of-action inferences.


Subject(s)
Antineoplastic Agents , Computational Biology/methods , Drug Design , Small Molecule Libraries , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Proliferation/drug effects , Cluster Analysis , Computational Biology/statistics & numerical data , DNA Replication/drug effects , Dose-Response Relationship, Drug , HeLa Cells , Humans , Ligands , Models, Statistical , Molecular Structure , Predictive Value of Tests , Protein Binding , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
19.
Cancer Res ; 67(24): 11493-8, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18089776

ABSTRACT

Smac mimetic compounds targeting the inhibitor of apoptosis proteins (IAP) baculoviral IAP repeat-3 domain are presumed to reduce the threshold for apoptotic cell death by alleviating caspase-9 repression. We explored this tenet in an unbiased manner by searching for small interfering RNAs that are able to confer resistance to the Smac mimetic compound LBW242. Among the screening hits were multiple components of the tumor necrosis factor alpha (TNFalpha) signaling pathway as well as X-linked inhibitor of apoptosis (XIAP) itself. Here, we show that in a subset of highly sensitive tumor cell lines, activity of LBW242 is dependent on TNFalpha signaling. Mechanistic studies indicate that in this context, XIAP is a positive modulator of TNFalpha induction whereas cellular inhibitor of apoptosis protein 1 negatively regulates TNFalpha-mediated apoptosis.


Subject(s)
Inhibitor of Apoptosis Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Mitochondrial Proteins/physiology , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/physiology , X-Linked Inhibitor of Apoptosis Protein/physiology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins , Cell Death , Cell Line, Tumor , Conserved Sequence , Female , Humans , Oligopeptides/pharmacology , Ovarian Neoplasms , RNA Interference/physiology , RNA, Neoplasm/genetics , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein/genetics
20.
J Biomol Screen ; 12(4): 546-59, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17435168

ABSTRACT

High-throughput screening of RNAi libraries has become an essential part of functional analysis in academic and industrial settings. The transition of a cell-based RNAi assay into a 384-well format requires several optimization steps to ensure the phenotype being screened is appropriately measured and that the signal-to-background ratio is above a certain quantifiable threshold. Methods currently used to assess small interfering RNA (siRNA) efficacy after transfection, including quantitative PCR or branch DNA analysis, face several technical limitations preventing the accurate measurement of mRNA levels in a 384-well format. To overcome these difficulties, the authors developed an approach using a viral-based transfection system that measures siRNA efficacy in a standardized 384-well assay. This method allows measurement of siRNA activity in a phenotypically neutral manner by quantifying the knockdown of an exogenous luciferase gene delivered by a lentiviral vector. In this assay, the efficacy of a luciferase siRNA is compared to a negative control siRNA across many distinct assay parameters including cell type, cell number, lipid type, lipid volume, time of the assay, and concentration of siRNA. Once the siRNA transfection is optimized as a 384-well luciferase knockdown, the biologically relevant phenotypic analysis can proceed using the best siRNA transfection conditions. This approach provides a key technology for 384-well assay development when direct measurement of mRNA knockdown is not possible. It also allows for direct comparison of siRNA activity across cell lines from almost any mammalian species. Defining optimal conditions for siRNA delivery into mammalian cells will greatly increase the speed and quality of large-scale siRNA screening campaigns.


Subject(s)
RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Cell Line , Cell Line, Tumor , HeLa Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...