Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Pharmacol Res Perspect ; 4(1): e00209, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26977299

ABSTRACT

The chemical modification 2'-O-methyl of nucleosides is often used to increase siRNA stability towards nuclease activities. However, the metabolic fate of modified nucleosides remains unclear. Therefore, the aim of this study was to determine the mass balance, pharmacokinetic, and absorption, distribution, metabolism, and excretion (ADME)-properties of tritium-labeled 2'-O-methyluridine, following a single intravenous dose to male CD-1 mice. The single intravenous administration of [5-(3)H]-2'-O-methyluridine was well tolerated in mice. Radioactivity was rapidly and widely distributed throughout the body and remained detectable in all tissues investigated throughout the observation period of 48 h. After an initial rapid decline, blood concentrations of total radiolabeled components declined at a much slower rate. [(3)H]-2'-O-Methyluridine represented a minor component of the radioactivity in plasma (5.89% of [(3)H]-AUC 0-48 h). Three [(3)H]-2'-O-methyluridine metabolites namely uridine (M1), cytidine (M2), and uracil (M3) were the major circulating components representing 32.8%, 8.11%, and 23.6% of radioactivity area under the curve, respectively. The highest concentrations of total radiolabeled components and exposures were observed in kidney, spleen, pineal body, and lymph nodes. The mass balance, which is the sum of external recovery of radioactivity in excreta and remaining radioactivity in carcass and cage wash, was complete. Renal excretion accounted for about 52.7% of the dose with direct renal excretion of the parent in combination with metabolism to the endogenous compounds cytidine, uracil, cytosine, and cytidine.

2.
Drug Metab Dispos ; 42(3): 431-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24389421

ABSTRACT

Absorption, distribution, metabolism, and excretion properties of a small interfering RNA (siRNA) formulated in a lipid nanoparticle (LNP) vehicle were determined in male CD-1 mice following a single intravenous administration of LNP-formulated [(3)H]-SSB siRNA, at a target dose of 2.5 mg/kg. Tissue distribution of the [(3)H]-SSB siRNA was determined using quantitative whole-body autoradiography, and the biostability was determined by both liquid chromatography mass spectrometry (LC-MS) with radiodetection and reverse-transcriptase polymerase chain reaction techniques. Furthermore, the pharmacokinetics and distribution of the cationic lipid (one of the main excipients of the LNP vehicle) were investigated by LC-MS and matrix-assisted laser desorption ionization mass spectrometry imaging techniques, respectively. Following i.v. administration of [(3)H]-SSB siRNA in the LNP vehicle, the concentration of parent guide strand could be determined up to 168 hours p.d. (post dose), which was ascribed to the use of the vehicle. This was significantly longer than what was observed after i.v. administration of the unformulated [(3)H]-SSB siRNA, where no intact parent guide strand could be observed 5 minutes post dosing. The disposition of the siRNA was determined by the pharmacokinetics of the formulated LNP vehicle itself. In this study, the radioactivity was widely distributed throughout the body, and the total radioactivity concentration was determined in selected tissues. The highest concentrations of radioactivity were found in the spleen, liver, esophagus, stomach, adrenal, and seminal vesicle wall. In conclusion, the LNP vehicle was found to drive the kinetics and biodistribution of the SSB siRNA. The renal clearance was significantly reduced and its exposure in plasma significantly increased compared with the unformulated [(3)H]-SSB siRNA.


Subject(s)
Drug Carriers/metabolism , Lipids/pharmacokinetics , Nanoparticles/metabolism , RNA, Small Interfering/metabolism , Animals , Autoradiography , Chromatography, High Pressure Liquid , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Stability , Injections, Intravenous , Lipids/blood , Lipids/chemistry , Male , Mice , Mice, Inbred Strains , Nanoparticles/chemistry , RNA, Small Interfering/blood , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Distribution , Tritium , Whole-Body Counting
3.
Drug Metab Dispos ; 41(6): 1211-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23524663

ABSTRACT

Absorption, distribution, metabolism, and excretion properties of two unformulated model short interfering RNA (siRNAs) were determined using a single internal [(3)H]-radiolabeling procedure, in which the full-length oligonucleotides were radiolabeled by Br/(3)H -exchange. Tissue distribution, excretion, and mass balance of radioactivity were investigated in male CD-1 mice after a single intravenous administration of the [(3)H]siRNAs, at a target dose level of 5 mg/kg. Quantitative whole-body autoradiography and liquid scintillation counting techniques were used to determine tissue distribution. Radiochromatogram profiles were determined in plasma, tissue extracts, and urine. Metabolites were separated by liquid chromatography and identified by radiodetection and high-resolution accurate mass spectrometry. In general, there was little difference in the distribution of total radiolabeled components after administration of the two unformulated [(3)H]siRNAs. The radioactivity was rapidly and widely distributed throughout the body and remained detectable in all tissues investigated at later time points (24 and 48 hours for [(3)H]MRP4 (multidrug resistance protein isoform 4) and [(3)H]SSB (Sjögren Syndrome antigen B) siRNA, respectively). After an initial rapid decrease, concentrations of total radiolabeled components in dried blood decreased at a much slower rate. A nearly complete mass balance was obtained for the [(3)H]SSB siRNA, and renal excretion was the main route of elimination (38%). The metabolism of the two model siRNAs was rapid and extensive. Five minutes after administration, no parent compound could be detected in plasma. Instead, radiolabeled nucleosides resulting from nuclease hydrolysis were observed. In the metabolism profiles obtained from various tissues, only radiolabeled nucleosides were found, suggesting that siRNAs are rapidly metabolized and that the distribution pattern of total radiolabeled components can be ascribed to small molecular weight metabolites.


Subject(s)
RNA, Small Interfering/metabolism , Tritium/metabolism , Animals , Female , Injections, Intravenous , Male , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Tissue Distribution/drug effects , Tissue Distribution/genetics , Tritium/administration & dosage
4.
Nat Cell Biol ; 11(4): 501-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287375

ABSTRACT

Impaired ribosome biogenesis is attributed to nucleolar disruption and diffusion of a subset of 60S ribosomal proteins, particularly ribosomal protein (rp)L11, into the nucleoplasm, where they inhibit MDM2, leading to p53 induction and cell-cycle arrest. Previously, we demonstrated that deletion of the 40S rpS6 gene in mouse liver prevents hepatocytes from re-entering the cell cycle after partial hepatectomy. Here, we show that this response leads to an increase in p53, which is recapitulated in culture by rpS6-siRNA treatment and rescued by the simultaneous depletion of p53. However, disruption of biogenesis of 40S ribosomes had no effect on nucleolar integrity, although p53 induction was mediated by rpL11, leading to the finding that the cell selectively upregulates the translation of mRNAs with a polypyrimidine tract at their 5'-transcriptional start site (5'-TOP mRNAs), including that encoding rpL11, on impairment of 40S ribosome biogenesis. Increased 5'-TOP mRNA translation takes place despite continued 60S ribosome biogenesis and a decrease in global translation. Thus, in proliferative human disorders involving hypomorphic mutations in 40S ribosomal proteins, specific targeting of rpL11 upregulation would spare other stress pathways that mediate the potential benefits of p53 induction.


Subject(s)
Cell Nucleolus/metabolism , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Humans , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6/deficiency , Ribosomal Protein S6/metabolism , Ribosomal Proteins/genetics , Transcription Initiation Site , Up-Regulation
5.
Brain Res ; 1243: 124-33, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-18823960

ABSTRACT

Genetic abnormalities in amyloid precursor protein (APP) are associated with Down's syndrome and familial Alzheimer's disease where hallmark plaques contain A beta peptides derived from APP. Both APP and its derivatives are implicated in neurodegenerative processes and may play important physiological and pathophysiological roles in synaptic function. Here, we show that young APP23 transgenic mice overexpressing human APP with the Swedish double mutation display altered novelty seeking behavior before the age of plaque onset. Using short interfering RNA (siRNA) targeted against APP, we investigate the direct contribution of APP and its derivatives to this behavioral deficit. After validating siRNAs targeting human APP in vitro, siRNAs were infused directly into the brain of APP23 mice for 2 weeks. Behavioral analysis shows that infusion of siRNA targeted against APP completely reverses increased exploratory activity in APP23 mice. Collectively, these data suggest that excessive APP and/or its derivatives, causes a hyperactive phenotype in APP23 mice when placed in a novel environment, which is fully reversible and not linked to plaque deposits.


Subject(s)
Amyloid beta-Protein Precursor/antagonists & inhibitors , Behavior, Animal/physiology , Brain/metabolism , Exploratory Behavior/physiology , Psychomotor Agitation/genetics , RNA, Small Interfering/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/physiopathology , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Environment , Exploratory Behavior/drug effects , Genetic Therapy/methods , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Mutation/genetics , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Psychomotor Agitation/metabolism , Psychomotor Agitation/physiopathology , RNA Interference/physiology , RNA, Small Interfering/therapeutic use
6.
Cell Metab ; 7(5): 456-65, 2008 May.
Article in English | MEDLINE | ID: mdl-18460336

ABSTRACT

Excess levels of circulating amino acids (AAs) play a causal role in specific human pathologies, including obesity and type 2 diabetes. Moreover, obesity and diabetes are contributing factors in the development of cancer, with recent studies suggesting that this link is mediated in part by AA activation of mammalian target of rapamycin (mTOR) Complex 1. AAs appear to mediate this response through class III phosphatidylinositol 3-kinase (PI3K), or human vacuolar protein sorting 34 (hVps34), rather than through the canonical class I PI3K pathway used by growth factors and hormones. Here we show that AAs induce a rise in intracellular Ca(2+) ([Ca(2+)](i)), which triggers mTOR Complex 1 and hVps34 activation. We demonstrate that the rise in [Ca(2+)](i) increases the direct binding of Ca(2+)/calmodulin (CaM) to an evolutionarily conserved motif in hVps34 that is required for lipid kinase activity and increased mTOR Complex 1 signaling. These findings have important implications regarding the basic signaling mechanisms linking metabolic disorders with cancer progression.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Leucine/pharmacology , Protein Kinases/metabolism , Signal Transduction , Vesicular Transport Proteins/metabolism , Binding Sites , Blotting, Western , Cells, Cultured , HeLa Cells , Humans , Immunoprecipitation , Kidney/metabolism , Mutagenesis, Site-Directed , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases , Transfection
7.
Genome Biol ; 9(2): R44, 2008.
Article in English | MEDLINE | ID: mdl-18302737

ABSTRACT

BACKGROUND: The mitotic spindle is a complex mechanical apparatus required for accurate segregation of sister chromosomes during mitosis. We designed a genetic screen using automated microscopy to discover factors essential for mitotic progression. Using a RNA interference library of 49,164 double-stranded RNAs targeting 23,835 human genes, we performed a loss of function screen to look for small interfering RNAs that arrest cells in metaphase. RESULTS: Here we report the identification of genes that, when suppressed, result in structural defects in the mitotic spindle leading to bent, twisted, monopolar, or multipolar spindles, and cause cell cycle arrest. We further describe a novel analysis methodology for large-scale RNA interference datasets that relies on supervised clustering of these genes based on Gene Ontology, protein families, tissue expression, and protein-protein interactions. CONCLUSION: This approach was utilized to classify functionally the identified genes in discrete mitotic processes. We confirmed the identity for a subset of these genes and examined more closely their mechanical role in spindle architecture.


Subject(s)
Genome, Human , Mitosis/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Humans , RNA Interference
8.
J Bone Miner Res ; 22(12): 1957-67, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17696759

ABSTRACT

UNLABELLED: Expression of the osteocyte-derived bone formation inhibitor sclerostin in adult bone requires a distant enhancer. We show that MEF2 transcription factors control this enhancer and mediate inhibition of sclerostin expression by PTH. INTRODUCTION: Sclerostin encoded by the SOST gene is a key regulator of bone formation. Lack of SOST expression is the cause for the progressive bone overgrowth disorders sclerosteosis and Van Buchem disease. We have previously identified a distant enhancer within the 52-kb Van Buchem disease deletion downstream of the SOST gene that is essential for its expression in adult bone. Furthermore, we and others have reported that SOST expression is suppressed by PTH. The aim of this study was to identify transcription factors involved in SOST bone enhancer activity and mediating PTH responsiveness. MATERIALS AND METHODS: Regulation of the SOST enhancer and promoter was studied by luciferase reporter gene assays. Transcription factor binding sites were mapped by footprint analysis and functional mutation analyses using transient transfections of osteoblast-like UMR-106 cells that exhibit endogenous SOST expression. Specific transcription factor binding was predicted by sequence analysis and shown by gel retardation assays and antibody-induced supershifts. Expression of myocyte enhancer factors 2 (MEF2) was detected by in situ hybridization, quantitative RT-PCR (qPCR), and immunohistochemistry. The role of MEF2s in SOST expression was assessed by reporter gene assays and siRNA-mediated RNA knockdown. RESULTS: PTH completely suppressed the transcriptional activity of the SOST bone enhancer but did not affect the SOST promoter. A MEF2 response element was identified in the bone enhancer. It was essential for transcriptional activation, bound MEF2 transcription factors, and mediated PTH responsiveness. Expression of MEF2s in bone was shown by qPCR, in situ hybridization, and immunohistochemistry. MEF2s and sclerostin co-localized in osteocytes. Enhancer activity was stimulated by MEF2C overexpression and inhibited by co-expression of a dominant negative MEF2C mutant. Finally, siRNA-mediated knockdown of MEF2A, C, and D suppressed endogenous SOST expression in UMR-106 osteoblast-like cells. CONCLUSIONS: These data strongly suggest that SOST expression in osteocytes of adult bone and its inhibition by PTH is mediated by MEF2A, C, and D transcription factors controlling the SOST bone enhancer. Hence, MEF2s are implicated in the regulation of adult bone mass.


Subject(s)
Bone Development/drug effects , Bone Morphogenetic Proteins/biosynthesis , Enhancer Elements, Genetic , Gene Expression Regulation/drug effects , Myogenic Regulatory Factors/metabolism , Parathyroid Hormone/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Base Sequence/genetics , Bone Development/genetics , Bone Diseases/genetics , Bone Diseases/metabolism , Bone Diseases/pathology , Bone Morphogenetic Proteins/genetics , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Genetic Markers/genetics , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors/genetics , Rats , Sequence Deletion , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
9.
Curr Opin Mol Ther ; 9(3): 242-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17608022

ABSTRACT

'Omics' technologies, combined with knowledge management, have changed the way pharmaceutical companies approach drug discovery activities. The rapid generation of more reliable putative drug targets from gain- or loss-of-function genome scale screens means that there is a strong need to filter and prioritize targets that will be considered in the drug discovery process. Short interfering RNAs (siRNAs) can be used to validate genomic drug targets. The outlook for this approach is promising, provided that special attention is given to the use of optimal reagents and to careful development of animal models.


Subject(s)
Drug Design , RNA Interference , RNA, Small Interfering/genetics , Animals , Drug Evaluation, Preclinical , Genomics/methods , Genomics/trends , Humans , Reproducibility of Results , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends
10.
J Neurochem ; 102(6): 1928-1940, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17540010

ABSTRACT

The cleavage-product of amyloid precursor protein (APP) constitutes the core component of plaques found in the brains of Alzheimer's disease (AD) patients. APP is ubiquitously expressed and its precise physiological functions remain unclear. This protein has been proposed to regulate synaptic function and processes underlying learning and memory. While APP knockout mice show behavioral impairments, these may occur due to early changes during development and/or due to abolition of APP function in adult. To investigate the acute effects of APP knockdown without involving developmental processes, APP expression was reduced using RNA interference in adult mouse brain. Small interfering RNAs (siRNAs) that down-regulated mouse APP protein levels (APP-siRNA) were identified using an APP plasmid-siRNA co-transfection assay in mouse NIH/3T3 fibroblast cells. Infusion of APP-siRNAs into the ventricular system for 2 weeks also down-regulated APP mRNA in mouse brain. Highest knockdown of APP mRNA levels was found in the CA2-CA3 regions of the hippocampus. Mice treated with the most active APP-siRNA showed a significant reduction in spontaneous alternation rate in the Y-maze, without effects on forelimb grip strength or locomotor activity. These data suggest that acute knockdown of APP in adult mouse brain impairs hippocampus-dependent spatial working memory.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Down-Regulation/genetics , Hippocampus/metabolism , Memory Disorders/metabolism , RNA, Small Interfering/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , RNA Interference/physiology , RNA, Small Interfering/pharmacology
11.
Nucleic Acids Res ; 35(7): e52, 2007.
Article in English | MEDLINE | ID: mdl-17355992

ABSTRACT

Microarrays to examine the global expression levels of microRNAs (miRNAs) in a systematic in-parallel manner have become important tools to help unravel the functions of miRNAs and to understand their roles in RNA-based regulation and their implications in human diseases. We have established a novel miRNA-specific microarray platform that enables the simultaneous expression analysis of both known and predicted miRNAs obtained from human or mouse origin. Chemically modified 2'-O-(2-methoxyethyl)-(MOE) oligoribonucleotide probes were arrayed onto Evanescent Resonance (ER) microchips by robotic spotting. Supplementing the complementary probes against miRNAs with carefully designed mismatch controls allowed for accurate sequence-specific determination of miRNA expression profiles obtained from a panel of mouse tissues. This revealed new expression signatures of known miRNAs as well as of novel miRNAs previously predicted using bioinformatic methods. Systematic confirmation of the array data with northern blotting and, in particular, real-time PCR suggests that the described microarray platform is a powerful tool to analyze miRNA expression patterns with rapid throughput and high fidelity.


Subject(s)
Gene Expression Profiling/methods , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis/methods , Animals , HeLa Cells , Humans , Mice , Oligonucleotide Probes/chemistry , RNA/chemistry , Tissue Distribution
12.
J Recept Signal Transduct Res ; 26(5-6): 527-47, 2006.
Article in English | MEDLINE | ID: mdl-17118797

ABSTRACT

RNA interference (RNAi) is a natural mechanism for regulating gene expression, which exists in plants, invertebrates, and mammals. We investigated whether non-viral infusion of short interfering RNA (siRNA) by the intracerebroventricular route would enable a sequence-specific gene knockdown in the mouse brain and whether the knockdown translates into disease-relevant behavioral changes. Initially, we targeted enhanced green fluorescent protein (EGFP) in mice overexpressing EGFP. A selective knockdown of both EGFP protein and mRNA was observed throughout the brain, with lesser down-regulation in regions distal to the infusion site. We then targeted endogenous genes, encoding the dopamine (DAT) and serotonin transporters (SERT). DAT-siRNA infusion in adult mice produced a significant down-regulation of DAT mRNA and protein and elicited hyperlocomotion similar, but delayed, to that produced on infusion of GBR-12909, a potent and selective DAT inhibitor. Similarly, SERT-siRNA infusion resulted in significant knockdown of SERT mRNA and protein and elicited reduced immobility in the forced swim test similar to that obtained on infusion of citalopram, a very selective and potent SSRI. Application of this non-viral RNAi approach may accelerate target validation for neuropsychiatric disorders that involve a complex interplay of gene(s) from various brain regions.


Subject(s)
Brain/drug effects , Gene Expression Regulation/drug effects , Plasma Membrane Neurotransmitter Transport Proteins/genetics , RNA Interference/drug effects , Receptors, G-Protein-Coupled/genetics , Animals , Brain/metabolism , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/genetics , Down-Regulation/genetics , Green Fluorescent Proteins/genetics , Locomotion/drug effects , Mental Disorders/drug therapy , Mice , Nervous System Diseases/drug therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics
13.
Proc Natl Acad Sci U S A ; 103(40): 14819-24, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17001007

ABSTRACT

Human cells have evolved complex signaling networks to coordinate the cell cycle. A detailed understanding of the global regulation of this fundamental process requires comprehensive identification of the genes and pathways involved in the various stages of cell-cycle progression. To this end, we report a genome-wide analysis of the human cell cycle, cell size, and proliferation by targeting >95% of the protein-coding genes in the human genome using small interfering RNAs (siRNAs). Analysis of >2 million images, acquired by quantitative fluorescence microscopy, showed that depletion of 1,152 genes strongly affected cell-cycle progression. These genes clustered into eight distinct phenotypic categories based on phase of arrest, nuclear area, and nuclear morphology. Phase-specific networks were built by interrogating knowledge-based and physical interaction databases with identified genes. Genome-wide analysis of cell-cycle regulators revealed a number of kinase, phosphatase, and proteolytic proteins and also suggests that processes thought to regulate G(1)-S phase progression like receptor-mediated signaling, nutrient status, and translation also play important roles in the regulation of G(2)/M phase transition. Moreover, 15 genes that are integral to TNF/NF-kappaB signaling were found to regulate G(2)/M, a previously unanticipated role for this pathway. These analyses provide systems-level insight into both known and novel genes as well as pathways that regulate cell-cycle progression, a number of which may provide new therapeutic approaches for the treatment of cancer.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Genome, Human/genetics , Cluster Analysis , Cytokinesis/genetics , Gene Expression , Genes, cdc , Genomic Library , Humans , Mitosis/genetics , Neoplasms/genetics , Phenotype , Protein Interaction Mapping , RNA Interference , RNA, Small Interfering/metabolism
14.
Proc Natl Acad Sci U S A ; 102(40): 14238-43, 2005 Oct 04.
Article in English | MEDLINE | ID: mdl-16176982

ABSTRACT

During the evolution of metazoans and the rise of systemic hormonal regulation, the insulin-controlled class 1 phosphatidylinositol 3OH-kinase (PI3K) pathway was merged with the primordial amino acid-driven mammalian target of rapamycin (mTOR) pathway to control the growth and development of the organism. Insulin regulates mTOR function through a recently described canonical signaling pathway, which is initiated by the activation of class 1 PI3K. However, how the amino acid input is integrated with that of the insulin signaling pathway is unclear. Here we used a number of molecular, biochemical, and pharmacological approaches to address this issue. Unexpectedly, we found that a major pathway by which amino acids control mTOR signaling is distinct from that of insulin and that, instead of signaling through components of the insulin/class 1 PI3K pathway, amino acids mediate mTOR activation by signaling through class 3 PI3K, hVps34.


Subject(s)
Amino Acids/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Blotting, Western , Cell Line, Tumor , Humans , Microscopy, Fluorescence , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , RNA, Small Interfering/genetics , Ras Homolog Enriched in Brain Protein , Regulatory-Associated Protein of mTOR , TOR Serine-Threonine Kinases
15.
Cell Signal ; 17(10): 1203-17, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16038795

ABSTRACT

Sphingosine-1-phosphate, a lipid mediator produced by sphingosine kinases, regulates diverse cellular processes, ranging from cell growth and survival to effector functions, such as proinflammatory mediator synthesis. Using human A549 epithelial lung carcinoma cells as a model system, we observed transient upregulation of sphingosine kinase type 1 (SPHK1) enzyme activity upon stimulation with both TNF-alpha or IL-1beta. This transient activation of SPHK1 was found to be required for cytokine-induced COX-2 transcription and PGE2 production, since not only specific siRNA (abolishing both basal and induced SPHK1 enzyme activity), but also a dominant-negative SPHK1 mutant (suppressing induced SPHK1 activity only) both reduced COX-2 and PGE2. Furthermore, TNF-alpha- or IL-1beta-induced transcription of selected cytokines, chemokines, and adhesion molecules (IL-6, RANTES, MCP-1, and VCAM-1) was found to require SPHK1 activation. Suppression of SPHK1 activation led to reduction of cytokine-induced IkappaBalpha phosphorylation and consequently diminished NFkappaB activity due to reduced nuclear translocation of RelA (p65), explaining the dependence of inflammatory mediator production on SPHK1 activation. Inhibition of basal SPHK1 activity by N,N-dimethylsphingosine or by downregulation of its expression using siRNA induced spontaneous apoptosis in A549 cells, an effect that can be explained through interference with constitutive NFkappaB activity in this cell type. In contrast, expression of the dominant-negative mutant did not induce apoptosis. Taken together, these findings demonstrate a role of SPHK1 activation in proinflammatory signalling and of SPHK1 basal activity in survival of A549 lung carcinoma cells.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-1/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Arachidonic Acid/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/physiology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Cyclooxygenase 2 , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Humans , I-kappa B Proteins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Ionomycin/pharmacology , Membrane Proteins , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B p50 Subunit , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Prostaglandin-Endoperoxide Synthases/genetics , Protein Precursors/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA , Transfection , Vascular Cell Adhesion Molecule-1/genetics
16.
Nat Biotechnol ; 23(8): 995-1001, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025102

ABSTRACT

The largest gene knock-down experiments performed to date have used multiple short interfering/short hairpin (si/sh)RNAs per gene. To overcome this burden for design of a genome-wide siRNA library, we used the Stuttgart Neural Net Simulator to train algorithms on a data set of 2,182 randomly selected siRNAs targeted to 34 mRNA species, assayed through a high-throughput fluorescent reporter gene system. The algorithm, (BIOPREDsi), reliably predicted activity of 249 siRNAs of an independent test set (Pearson coefficient r = 0.66) and siRNAs targeting endogenous genes at mRNA and protein levels. Neural networks trained on a complementary 21-nucleotide (nt) guide sequence were superior to those trained on a 19-nt sequence. BIOPREDsi was used in the design of a genome-wide siRNA collection with two potent siRNAs per gene. When this collection of 50,000 siRNAs was used to identify genes involved in the cellular response to hypoxia, two of the most potent hits were the key hypoxia transcription factors HIF1A and ARNT.


Subject(s)
Algorithms , Gene Silencing , Models, Genetic , Nerve Net , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Sequence Alignment/methods , Sequence Analysis, RNA/methods , Base Sequence , Computer Simulation , Computer-Aided Design , Gene Library , Models, Statistical , Molecular Sequence Data
17.
Cell ; 120(6): 747-59, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15797377

ABSTRACT

Although DNA damaging agents have revolutionized chemotherapy against solid tumors, a narrow therapeutic window combined with severe side effects has limited their broader use. Here we show that RAD001 (everolimus), a rapamycin derivative, dramatically enhances cisplatin-induced apoptosis in wild-type p53, but not mutant p53 tumor cells. The use of isogenic tumor cell lines expressing either wild-type mTOR cDNA or a mutant that does not bind RAD001 demonstrates that the effects of RAD001 are through inhibition of mTOR function. We further show that RAD001 sensitizes cells to cisplatin by inhibiting p53-induced p21 expression. Unexpectedly, this effect is attributed to a small but significant inhibition of p21 translation combined with its short half-life. These findings provide the molecular rationale for combining DNA damaging agents with RAD001, showing that a general effect on a major anabolic process may dramatically enhance the efficacy of an established drug protocol in the treatment of cancer patients with solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle Proteins/antagonists & inhibitors , Cisplatin/pharmacology , Protein Biosynthesis/drug effects , Protein Kinases/metabolism , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p21 , DNA Damage/drug effects , DNA Damage/genetics , DNA Damage/physiology , Drug Synergism , Everolimus , Humans , Mutation , Polyribosomes/drug effects , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Protein Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , TOR Serine-Threonine Kinases , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Exp Cell Res ; 303(2): 482-93, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15652359

ABSTRACT

Ubiquitin-mediated degradation of the cyclin-dependent kinase inhibitor p27Kip1 was shown to be required for the activation of key cyclin-dependent kinases, thereby triggering the onset of DNA replication and cell cycle progression. Although the SCFSkp2 ubiquitin ligase has been reported to mediate p27Kip1 degradation, the nature of the human ubiquitin-conjugating enzyme involved in this process has not yet been determined at the cellular level. Here, we show that antisense oligonucleotides targeting the human ubiquitin-conjugating enzyme Cdc34 downregulate its expression, inhibit the degradation of p27Kip1, and prevent cellular proliferation. Elevation of p27Kip1 protein level is found to be the sole requirement for the inhibition of cellular proliferation induced upon downregulation of Cdc34. Indeed, reducing the expression of p27Kip1 with a specific antisense oligonucleotide is sufficient to reverse the anti-proliferative phenotype elicited by the Cdc34 antisense. Furthermore, downregulation of Cdc34 is found to specifically increase the abundance of the SCFSkp2) ubiquitin ligase substrate p27Kip1, but has no concomitant effect on the level of IkBalpha and beta-catenin, which are known substrates of a closely related SCF ligase.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Base Sequence , Carrier Proteins/genetics , Cell Cycle , Cell Line , Cyclin-Dependent Kinase Inhibitor p27 , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligase Complexes/genetics
19.
Proc Natl Acad Sci U S A ; 101(49): 17270-5, 2004 Dec 07.
Article in English | MEDLINE | ID: mdl-15569935

ABSTRACT

Gene expression analysis implicates an increasing number of novel genes in the brain as potential targets for the treatment of neurological and psychiatric disorders. Frequently, these genes are ubiquitously expressed in the brain and, thus, may contribute to a pathophysiological state through actions in several brain nuclei. Current strategies employing genetically modified animals for in vivo validation of such targets are time-consuming and often limited by developmental adaptations. Somatic gene manipulation using viral-mediated RNA interference (RNAi) has emerged recently, although restricting the target validation to specific brain nuclei. We investigated whether nonviral infusion of short interfering RNA (siRNA) into the ventricular system would enable a sequence-specific gene knockdown. The temporality and extent of siRNA-induced down-regulation were analyzed by targeting a transgene, EGFP, in mice overexpressing EGFP. Extensive knockdown of EGFP was observed, especially in regions adjacent or dorsoventrally and mediolaterally distant to the infusion site (dorsal third ventricle), with lesser knockdown in more distal regions. We challenged our RNAi approach to generate a specific knockdown of an endogenous gene, encoding the dopamine transporter (DAT) in regions (ventral midbrain) far distal to the infusion site. DAT-siRNA infusion in adult mice produced a significant down-regulation of DAT mRNA and protein in the brain and also elicited a temporal hyperlocomotor response similar to that (but delayed) obtained upon infusion of GBR-12909, a pharmacologically selective DAT inhibitor. Application of this nonviral RNAi approach may accelerate target validation for neuropsychiatric disorders that involve a complex interplay of gene(s) from various brain regions.


Subject(s)
Brain/drug effects , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Motor Activity/drug effects , Nerve Tissue Proteins/genetics , RNA Interference , Animals , Brain/metabolism , Cerebral Ventricles , Dopamine Plasma Membrane Transport Proteins , Down-Regulation/drug effects , Green Fluorescent Proteins/genetics , Male , Methods , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology
20.
Nucleic Acids Res ; 32(5): e49, 2004 Mar 16.
Article in English | MEDLINE | ID: mdl-15026538

ABSTRACT

Double stranded, short interfering RNAs (siRNA) of 21-22 nt length initiate a sequence-specific, post-trancriptional gene silencing in animals and plants known as RNA interference (RNAi). Here we show that RNAi can block a pathophysiological pain response and provide relief from neuropathic pain in a rat disease model by down regulating an endogenous, neuronally expressed gene. Rats, intrathecally infused with a 21 nt siRNA perfectly complementary to the pain-related cation-channel P2X3, showed diminished pain responses compared to missense (MS) siRNA-treated and untreated controls in models of both agonist-evoked pain and chronic neuropathic pain. This form of delivery caused no adverse effects in any of the animals receiving P2X3 siRNA, MS siRNA or vehicle. Molecular analysis of tissues revealed that P2X3 mRNA expressed in dorsal root ganglia, and P2X3 protein translocated into the dorsal horn of the spinal cord, were significantly diminished. These observations open a path toward use of siRNA as a genetic tool for drug target validation in the mammalian central nervous system, as well as for proof of concept studies and as therapeutic agents in man.


Subject(s)
Neuralgia/therapy , Purinergic P2 Receptor Antagonists , RNA, Small Interfering/therapeutic use , Animals , Chronic Disease , Hyperalgesia/therapy , Neuralgia/metabolism , RNA Interference , Rats , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3
SELECTION OF CITATIONS
SEARCH DETAIL
...