Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS One ; 10(8): e0135365, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26270474

RESUMO

Disease modifying treatments for Alzheimer's disease (AD) constitute a major goal in medicine. Current trends suggest that biomarkers reflective of AD neuropathology and modifiable by treatment would provide supportive evidence for disease modification. Nevertheless, a lack of quantitative tools to assess disease modifying treatment effects remains a major hurdle. Cerebrospinal fluid (CSF) biochemical markers such as total tau, p-tau and Ab42 are well established markers of AD; however, global quantitative biochemical changes in CSF in AD disease progression remain largely uncharacterized. Here we applied a high resolution open discovery platform, dMS, to profile a cross-sectional cohort of lumbar CSF from post-mortem diagnosed AD patients versus those from non-AD/non-demented (control) patients. Multiple markers were identified to be statistically significant in the cohort tested. We selected two markers SME-1 (p<0.0001) and SME-2 (p = 0.0004) for evaluation in a second independent longitudinal cohort of human CSF from post-mortem diagnosed AD patients and age-matched and case-matched control patients. In cohort-2, SME-1, identified as neuronal secretory protein VGF, and SME-2, identified as neuronal pentraxin receptor-1 (NPTXR), in AD were 21% (p = 0.039) and 17% (p = 0.026) lower, at baseline, respectively, than in controls. Linear mixed model analysis in the longitudinal cohort estimate a decrease in the levels of VGF and NPTXR at the rate of 10.9% and 6.9% per year in the AD patients, whereas both markers increased in controls. Because these markers are detected by mass spectrometry without the need for antibody reagents, targeted MS based assays provide a clear translation path for evaluating selected AD disease-progression markers with high analytical precision in the clinic.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Proteína C-Reativa/líquido cefalorraquidiano , Espectrometria de Massas , Fatores de Crescimento Neural/líquido cefalorraquidiano , Proteínas do Tecido Nervoso/líquido cefalorraquidiano , Proteômica , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/líquido cefalorraquidiano , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
2.
Lipids ; 46(11): 991-1003, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21826528

RESUMO

The association between hypercholesterolemia and elevated serum apolipoprotein B (APOB) has generated interest in APOB as a therapeutic target for patients at risk of developing cardiovascular disease. In the clinic, mipomersen, an antisense oligonucleotide (ASO) APOB inhibitor, was associated with a trend toward increased hepatic triglycerides, and liver steatosis remains a concern. We found that siRNA-mediated knockdown of ApoB led to elevated hepatic triglycerides and liver steatosis in mice engineered to exhibit a human-like lipid profile. Many genes required for fatty acid synthesis were reduced, suggesting that the observed elevation in hepatic triglycerides is maintained by the cell through fatty acid uptake as opposed to fatty acid synthesis. Fatty acid transport protein 5 (Fatp5/Slc27a5) is required for long chain fatty acid (LCFA) uptake and bile acid reconjugation by the liver. Fatp5 knockout mice exhibited lower levels of hepatic triglycerides due to decreased fatty acid uptake, and shRNA-mediated knockdown of Fatp5 protected mice from diet-induced liver steatosis. Here, we evaluated if siRNA-mediated knockdown of Fatp5 was sufficient to alleviate ApoB knockdown-induced steatosis. We determined that, although Fatp5 siRNA treatment was sufficient to increase the proportion of unconjugated bile acids 100-fold, consistent with FATP5's role in bile acid reconjugation, Fatp5 knockdown failed to influence the degree, zonal distribution, or composition of the hepatic triglycerides that accumulated following ApoB siRNA treatment.


Assuntos
Apolipoproteínas B/genética , Proteínas de Transporte de Ácido Graxo/genética , Fígado Gorduroso/genética , Animais , Apolipoproteínas B/sangue , Ácidos e Sais Biliares/metabolismo , Colesterol/sangue , HDL-Colesterol/sangue , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno , Triglicerídeos/sangue
3.
J Histochem Cytochem ; 59(8): 727-40, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21804077

RESUMO

Chemically stabilized small interfering RNA (siRNA) can be delivered systemically by intravenous injection of lipid nanoparticles (LNPs) in rodents and primates. The biodistribution and kinetics of LNP-siRNA delivery in mice at organ and cellular resolution have been studied using immunofluorescence (IF) staining and quantitative polymerase chain reaction (qPCR). At 0.5 and 2 hr post tail vein injection of Cy5-labeled siRNA encapsulated in LNP, the organ rank-order of siRNA levels is liver > spleen > kidney, with only negligible accumulation in duodenum, lung, heart, and brain. Similar conclusions were drawn by using qPCR to measure tissue siRNA levels as a secondary end point. siRNA levels in these tissues decreased by more than 10-fold after 24 hr. Within the liver, LNPs delivered siRNA to hepatocytes, Kupffer cells, and sinusoids in a time-dependent manner, as revealed by IF staining and signal quantitation methods established using OPERA/Columbus software. siRNA first accumulated in liver sinusoids and trafficked to hepatocytes by 2 hr post dose, corresponding to the onset of target mRNA silencing. Fluorescence in situ hybridization methods were used to detect both strands of siRNA in fixed tissues. Collectively, the authors have implemented a platform to evaluate biodistribution of siRNA across cell types and across tissues in vivo, with the objective of elucidating the pharmacokinetic and pharmacodynamic relationship to guide optimization of delivery vehicles.


Assuntos
Autoantígenos/metabolismo , Lipídeos , Nanopartículas , RNA Interferente Pequeno/metabolismo , Ribonucleoproteínas/metabolismo , Animais , Autoantígenos/genética , Portadores de Fármacos , Imunofluorescência , Técnicas de Silenciamento de Genes , Hibridização in Situ Fluorescente , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/administração & dosagem , Ribonucleoproteínas/genética , Distribuição Tecidual , Antígeno SS-B
4.
J Lipid Res ; 52(6): 1084-1097, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21398511

RESUMO

Increased serum apolipoprotein (apo)B and associated LDL levels are well-correlated with an increased risk of coronary disease. ApoE⁻/⁻ and low density lipoprotein receptor (LDLr)⁻/⁻ mice have been extensively used for studies of coronary atherosclerosis. These animals show atherosclerotic lesions similar to those in humans, but their serum lipids are low in apoB-containing LDL particles. We describe the development of a new mouse model with a human-like lipid profile. Ldlr CETP⁺/⁻ hemizygous mice carry a single copy of the human CETP transgene and a single copy of a LDL receptor mutation. To evaluate the apoB pathways in this mouse model, we used novel short-interfering RNAs (siRNA) formulated in lipid nanoparticles (LNP). ApoB siRNAs induced up to 95% reduction of liver ApoB mRNA and serum apoB protein, and a significant lowering of serum LDL in Ldlr CETP⁺/⁻ mice. ApoB targeting is specific and dose-dependent, and it shows lipid-lowering effects for over three weeks. Although specific triglycerides (TG) were affected by ApoB mRNA knockdown (KD) and the total plasma lipid levels were decreased by 70%, the overall lipid distribution did not change. Results presented here demonstrate a new mouse model for investigating additional targets within the ApoB pathways using the siRNA modality.


Assuntos
Apolipoproteínas B/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/genética , LDL-Colesterol/sangue , Modelos Animais de Doenças , Receptores de LDL/genética , Animais , Apolipoproteínas B/sangue , Apolipoproteínas E/sangue , Apolipoproteínas E/genética , Aterosclerose/patologia , Linhagem Celular Tumoral , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Efeito Fundador , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hemizigoto , Humanos , Metabolismo dos Lipídeos/genética , Lipossomos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nanopartículas/administração & dosagem , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores de LDL/metabolismo , Triglicerídeos/sangue
5.
Mol Pharmacol ; 79(6): 953-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21427169

RESUMO

Deeper knowledge of pharmacokinetic and pharmacodynamic (PK/PD) concepts for RNA therapeutics is important to streamline the drug development process and for rigorous selection of best performing drug candidates. Here we characterized the PK/PD relationship for small interfering RNAs (siRNAs) targeting luciferase by examining siRNA concentration in plasma and liver, the temporal RNA-induced silencing complex binding profiles, mRNA reduction, and protein inhibition measured by noninvasive bioluminescent imaging. A dose-dependent and time-related decrease in bioluminescence was detected over 25 days after a single treatment of a lipid nanoparticle-formulated siRNA targeting luciferase messenger RNA. A direct relationship was observed between the degree of in vivo mRNA and protein reduction and the Argonaute2 (Ago2)-bound siRNA fraction but not with the total amount of siRNA found in the liver, suggesting that the Ago2-siRNA complex is the key determinant of target inhibition. These observations were confirmed for an additional siRNA that targets endogenously expressed Sjögren syndrome antigen B (Ssb) mRNA, indicating that our observations are not limited to a transgenic mouse system. Our data provide detailed information of the temporal regulation of siRNA liver delivery, Ago2 loading, mRNA reduction, and protein inhibition that are essential for the rapid and cost-effective clinical development of siRNAs therapeutics.


Assuntos
Inativação Gênica , RNA Interferente Pequeno/genética , Animais , Sequência de Bases , Primers do DNA , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase
6.
J Lipid Res ; 52(4): 679-87, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21262787

RESUMO

Reducing circulating LDL-cholesterol (LDL-c) reduces the risk of cardiovascular disease in people with hypercholesterolemia. Current approaches to reduce circulating LDL-c include statins, which inhibit cholesterol synthesis, and ezetimibe, which blocks cholesterol absorption. Both elevate serum PCSK9 protein levels in patients, which could attenuate their efficacy by reducing the amount of cholesterol cleared from circulation. To determine whether PCSK9 inhibition could enhance LDL-c lowering of both statins and ezetimibe, we utilized small interfering RNAs (siRNAs) to knock down Pcsk9, together with ezetimibe, rosuvastatin, and an ezetimibe/rosuvastatin combination in a mouse model with a human-like lipid profile. We found that ezetimibe, rosuvastatin, and ezetimibe/rosuvastatin combined lower serum cholesterol but induce the expression of Pcsk9 as well as the Srebp-2 hepatic cholesterol biosynthesis pathway. Pcsk9 knockdown in combination with either treatment led to greater reductions in serum non-HDL with a near-uniform reduction of all LDL-c subfractions. In addition to reducing serum cholesterol, the combined rosuvastatin/ezetimibe/Pcsk9 siRNA treatment exhibited a significant reduction in serum APOB protein and triglyceride levels. Taken together, these data provide evidence that PCSK9 inhibitors, in combination with current therapies, have the potential to achieve greater reductions in both serum cholesterol and triglycerides.


Assuntos
Anticolesterolemiantes/uso terapêutico , Azetidinas/uso terapêutico , Fluorbenzenos/uso terapêutico , Pirimidinas/uso terapêutico , Serina Endopeptidases/metabolismo , Sulfonamidas/uso terapêutico , Animais , Apolipoproteínas B/sangue , Colesterol/sangue , LDL-Colesterol/sangue , Ensaio de Imunoadsorção Enzimática , Ezetimiba , Hipercolesterolemia/sangue , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/terapia , Camundongos , Camundongos Endogâmicos C57BL , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosuvastatina Cálcica , Serina Endopeptidases/genética , Triglicerídeos/sangue
7.
Mol Ther ; 19(3): 567-75, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21179008

RESUMO

A major hurdle for harnessing small interfering RNA (siRNA) for therapeutic application is an effective and safe delivery of siRNA to target tissues and cells via systemic administration. While lipid nanoparticles (LNPs) composed of a cationic lipid, poly-(ethylene glycol) lipid and cholesterol, are effective in delivering siRNA to hepatocytes via systemic administration, they may induce multi-faceted toxicities in a dose-dependent manner, independently of target silencing. To understand the underlying mechanism of toxicities, pharmacological probes including anti-inflammation drugs and specific inhibitors blocking different pathways of innate immunity were evaluated for their abilities to mitigate LNP-siRNA-induced toxicities in rodents. Three categories of rescue effects were observed: (i) pretreatment with a Janus kinase (Jak) inhibitor or dexamethasone abrogated LNP-siRNA-mediated lethality and toxicities including cytokine induction, organ impairments, thrombocytopenia and coagulopathy without affecting siRNA-mediated gene silencing; (ii) inhibitors of PI3K, mammalian target of rapamycin (mTOR), p38 and IκB kinase (IKK)1/2 exhibited a partial alleviative effect; (iii) FK506 and etoricoxib displayed no protection. Furthermore, knockout of Jak3, tumor necrosis factor receptors (Tnfr)p55/p75, interleukin 6 (IL-6) or interferon (IFN)-γ alone was insufficient to alleviate LNP-siRNA-associated toxicities in mice. These indicate that activation of innate immune response is a primary trigger of systemic toxicities and that multiple innate immune pathways and cytokines can mediate toxic responses. Jak inhibitors are effective in mitigating LNP-siRNA-induced toxicities.


Assuntos
Inibidores Enzimáticos/metabolismo , Janus Quinases/antagonistas & inibidores , Lipídeos , Nanopartículas , RNA Interferente Pequeno/toxicidade , Animais , Citocinas/sangue , Dexametasona/metabolismo , Etoricoxib , Feminino , Técnicas de Inativação de Genes , Quinase I-kappa B/antagonistas & inibidores , Interferon gama/genética , Interleucina-6/genética , Janus Quinases/genética , Lipídeos/química , Lipídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores de Fosfoinositídeo-3 Quinase , Piridinas/metabolismo , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Tipo II do Fator de Necrose Tumoral/genética , Sulfonas/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Tacrolimo/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
8.
RNA ; 16(12): 2553-63, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20940339

RESUMO

Effective small interfering RNA (siRNA)-mediated therapeutics require the siRNA to be delivered into the cellular RNA-induced silencing complex (RISC). Quantitative information of this essential delivery step is currently inferred from the efficacy of gene silencing and siRNA uptake in the tissue. Here we report an approach to directly quantify siRNA in the RISC in rodents and monkey. This is achieved by specific immunoprecipitation of the RISC from tissue lysates and quantification of small RNAs in the immunoprecipitates by stem-loop PCR. The method, expected to be independent of delivery vehicle and target, is label-free, and the throughput is acceptable for preclinical animal studies. We characterized a lipid-formulated siRNA by integrating these approaches and obtained a quantitative perspective on siRNA tissue accumulation, RISC loading, and gene silencing. The described methodologies have utility for the study of silencing mechanism, the development of siRNA therapeutics, and clinical trial design.


Assuntos
Técnicas de Transferência de Genes , RNA Interferente Pequeno/genética , Animais , Animais Geneticamente Modificados , Anticorpos/isolamento & purificação , Anticorpos/metabolismo , Anticorpos/farmacologia , Especificidade de Anticorpos , Proteínas Argonautas , Células Cultivadas , Fator de Iniciação 2 em Eucariotos/imunologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Estudos de Avaliação como Assunto , Feminino , Inativação Gênica/fisiologia , Marcação de Genes/métodos , Técnicas de Transferência de Genes/normas , Humanos , Imunoprecipitação/métodos , Imunoprecipitação/normas , Macaca mulatta , Camundongos , Camundongos Endogâmicos ICR , Ligação Proteica , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Roedores
9.
RNA ; 16(12): 2336-40, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20971811

RESUMO

microRNAs are ∼ 22 nucleotide regulatory RNAs that are processed into duplexes from hairpin structures and incorporated into Argonaute proteins. Here, we show that a nick in the middle of the guide strand of an miRNA sequence allows for seed-based targeting characteristic of miRNA activity. Insertion of an inverted abasic, a dye, or a small gap between the two segments still permits target knockdown. While activity from the seed region of the segmented miRNA is apparent, activity from the 3' half of the guide strand is impaired, suggesting that an intact guide backbone is required for contribution from the 3' half. miRNA activity was also observed following nicking of a miRNA precursor. These results illustrate a structural flexibility in miRNA duplexes and may have applications in the design of miRNA mimetics.


Assuntos
Marcação de Genes/métodos , MicroRNAs/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , Deleção de Sequência/fisiologia , Sequência de Bases/genética , Sequência de Bases/fisiologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , MicroRNAs/química , MicroRNAs/genética , MicroRNAs/fisiologia , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Ribonuclease III/metabolismo , Ribonuclease III/fisiologia , Deleção de Sequência/genética , Moldes Genéticos , Transfecção , Pequeno RNA não Traduzido
10.
J Proteome Res ; 9(3): 1392-401, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20095649

RESUMO

The rapid identification of protein biomarkers in biofluids is important to drug discovery and development. Here, we describe a general proteomic approach for the discovery and identification of proteins that exhibit a statistically significant difference in abundance in cerebrospinal fluid (CSF) before and after pharmacological intervention. This approach, differential mass spectrometry (dMS), is based on the analysis of full scan mass spectrometry data. The dMS workflow does not require complex mixing and pooling strategies, or isotope labeling techniques. Accordingly, clinical samples can be analyzed individually, allowing the use of longitudinal designs and within-subject data analysis in which each subject acts as its own control. As a proof of concept, we performed multifactorial dMS analyses on CSF samples drawn at 6 time points from n = 6 cisterna magna ported (CMP) rhesus monkeys treated with 2 potent gamma secretase inhibitors (GSI) or comparable vehicle in a 3-way crossover study that included a total of 108 individual CSF samples. Using analysis of variance and statistical filtering on the aligned and normalized LC-MS data sets, we detected 26 features that were significantly altered in CSF by drug treatment. Of those 26 features, which belong to 10 distinct isotopic distributions, 20 were identified by MS/MS as 7 peptides from CD99, a cell surface protein. Six features from the remaining 3 isotopic distributions were not identified. A subsequent analysis showed that the relative abundance of these 26 features showed the same temporal profile as the ELISA measured levels of CSF A beta 42 peptide, a known pharmacodynamic marker for gamma-secretase inhibition. These data demonstrate that dMS is a promising approach for the discovery, quantification, and identification of candidate target engagement biomarkers in CSF.


Assuntos
Proteínas do Líquido Cefalorraquidiano/análise , Espectrometria de Massas/métodos , Proteômica/métodos , Algoritmos , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/metabolismo , Análise de Variância , Animais , Área Sob a Curva , Biomarcadores/líquido cefalorraquidiano , Proteínas do Líquido Cefalorraquidiano/metabolismo , Macaca mulatta , Dados de Sequência Molecular , Oligopeptídeos/farmacocinética , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/metabolismo
11.
Nucleic Acids Res ; 38(2): 660-71, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19917641

RESUMO

Small interfering RNAs (siRNAs) are short, double-stranded RNAs that use the endogenous RNAi pathway to mediate gene silencing. Phosphorylation facilitates loading of a siRNA into the Ago2 complex and subsequent cleavage of the target mRNA. In this study, 2', 3' seco nucleoside modifications, which contain an acylic ribose ring and are commonly called unlocked nucleic acids (UNAs), were evaluated at all positions along the guide strand of a siRNA targeting apolipoprotein B (ApoB). UNA modifications at positions 1, 2 and 3 were detrimental to siRNA activity. UNAs at positions 1 and 2 prevented phosphorylation by Clp1 kinase, abrogated binding to Ago2, and impaired Ago2-mediated cleavage of the mRNA target. The addition of a 5'-terminal phosphate to siRNA containing a position 1 UNA restored ApoB mRNA silencing, Ago2 binding, and Ago2 mediated cleavage activity. Position 1 UNA modified siRNA containing a 5'-terminal phosphate exhibited a partial restoration of siRNA silencing activity in vivo. These data reveal the complexity of interpreting the effects of chemical modification on siRNA activity, and exemplify the importance of using multiple biochemical, cell-based and in vivo assays to rationally design chemically modified siRNA destined for therapeutic use.


Assuntos
Nucleosídeos/química , Interferência de RNA , RNA Interferente Pequeno/química , Animais , Apolipoproteínas B/genética , Linhagem Celular , Fator de Iniciação 2 em Eucariotos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Fosforilação , Fosfotransferases/metabolismo , RNA Interferente Pequeno/metabolismo , Termodinâmica , Fatores de Transcrição/metabolismo , Pequeno RNA não Traduzido
12.
Mol Ther ; 18(1): 171-80, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19738601

RESUMO

Despite recent progress, systemic delivery remains the major hurdle for development of safe and effective small inhibitory RNA (siRNA)-based therapeutics. Encapsulation of siRNA into liposomes is a promising option to overcome obstacles such as low stability in serum and inefficient internalization by target cells. However, a major liability of liposomes is the potential to induce an acute inflammatory response, thereby increasing the risk of numerous adverse effects. In this study, we characterized a liposomal siRNA delivery vehicle, LNP201, which is capable of silencing an mRNA target in mouse liver by over 80%. The biodistribution profile, efficacy after single and multiple doses, mechanism of action, and inflammatory toxicity are characterized for LNP201. Furthermore, we demonstrate that the glucocorticoid receptor (GR) agonist dexamethasone (Dex) inhibits LNP201-induced cytokine release, inflammatory gene induction, and mitogen-activated protein kinase (MAPK) phosphorylation in multiple tissues. These data present a possible clinical strategy for increasing the safety profile of siRNA-based drugs while maintaining the potency of gene silencing.


Assuntos
Dexametasona/uso terapêutico , Nanopartículas/efeitos adversos , RNA Interferente Pequeno/imunologia , RNA Interferente Pequeno/metabolismo , Animais , Feminino , Inativação Gênica , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Camundongos , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Receptores de Glucocorticoides/agonistas
13.
RNA ; 15(2): 308-15, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19144911

RESUMO

siRNAs mediate sequence-specific gene silencing in cultured mammalian cells but also silence unintended transcripts. Many siRNA off-target transcripts match the guide-strand "seed region," similar to the way microRNAs match their target sites. The extent to which this seed-matched, microRNA-like, off-target silencing affects the specificity of therapeutic siRNAs in vivo is currently unknown. Here, we compare microRNA-like off-target regulations in mouse liver in vivo with those seen in cell culture for a series of therapeutic candidate siRNAs targeting Apolipoprotein B (APOB). Each siRNA triggered regulation of consistent microRNA-like off-target transcripts in mouse livers and in cultured mouse liver tumor cells. In contrast, there was only random overlap between microRNA-like off-target transcripts from cultured human and mouse liver tumor cells. Therefore, siRNA therapeutics may trigger microRNA-like silencing of many unintended targets in vivo, and the potential toxicities caused by these off-target gene regulations cannot be accurately assessed in rodent models.


Assuntos
Regiões 3' não Traduzidas/genética , Apolipoproteínas B/genética , Inativação Gênica , MicroRNAs/metabolismo , RNA Interferente Pequeno/metabolismo , Regiões 3' não Traduzidas/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Camundongos , MicroRNAs/genética , RNA Interferente Pequeno/genética , Seleção Genética , Especificidade da Espécie , Transcrição Gênica
14.
Proc Natl Acad Sci U S A ; 103(47): 17967-72, 2006 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-17098871

RESUMO

Rare familial forms of Alzheimer's disease (AD) are thought to be caused by elevated proteolytic production of the Abeta42 peptide from the beta-amyloid-precursor protein (APP). Although the pathogenesis of the more common late-onset AD (LOAD) is not understood, BACE1, the protease that cleaves APP to generate the N terminus of Abeta42, is more active in patients with LOAD, suggesting that increased amyloid production processing might also contribute to the sporadic disease. Using high-throughput siRNA screening technology, we assessed 15,200 genes for their role in Abeta42 secretion and identified leucine-rich repeat transmembrane 3 (LRRTM3) as a neuronal gene that promotes APP processing by BACE1. siRNAs targeting LRRTM3 inhibit the secretion of Abeta40, Abeta42, and sAPPbeta, the N-terminal APP fragment produced by BACE1 cleavage, from cultured cells and primary neurons by up to 60%, whereas overexpression increases Abeta secretion. LRRTM3 is expressed nearly exclusively in the nervous system, including regions affected during AD, such as the dentate gyrus. Furthermore, LRRTM3 maps to a region of chromosome 10 linked to both LOAD and elevated plasma Abeta42, and is structurally similar to a family of neuronal receptors that includes the NOGO receptor, an inhibitor of neuronal regeneration and APP processing. Thus, LRRTM3 is a functional and positional candidate gene for AD, and, given its receptor-like structure and restricted expression, a potential therapeutic target.


Assuntos
Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Proteínas , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Cromossomos Humanos Par 10 , Ativação Enzimática , Humanos , Proteínas de Repetições Ricas em Leucina , Proteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares , Fragmentos de Peptídeos/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
15.
Genomics ; 86(6): 648-56, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16297596

RESUMO

Niemann-Pick C1-like 1 (NPC1L1) is an intestinal cholesterol transporter and the molecular target of ezetimibe, a cholesterol absorption inhibitor demonstrated to reduce LDL-cholesterol (LDL-C) both as monotherapy and when co-administered with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins). Interestingly, significant interindividual variability has been observed for rates of intestinal cholesterol absorption and LDL-C reductions at both baseline and post ezetimibe treatment. To test the hypothesis that genetic variation in NPC1L1 could influence the LDL-C response to ezetimibe, we performed extensive resequencing of the gene in 375 apparently healthy individuals and genotyped hypercholesterolemic patients from clinical trial cohorts. No association was observed between NPC1L1 single-nucleotide polymorphism and baseline cholesterol. However, significant associations to LDL-C response to treatment with ezetimibe were observed in patients treated with ezetimibe in two large clinical trials. Our data demonstrate that DNA sequence variants in NPC1L1 are associated with an improvement in response to ezetimibe pharmacotherapy and suggest that detailed analysis of genetic variability in clinical trial cohorts can lead to improved understanding of factors contributing to variable drug response.


Assuntos
Anticolesterolemiantes/farmacologia , Azetidinas/farmacologia , LDL-Colesterol/metabolismo , Variação Genética , Proteínas de Membrana/genética , Negro ou Afro-Americano/genética , Ezetimiba , Frequência do Gene , Hispânico ou Latino/genética , Humanos , Absorção Intestinal/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Fitosteróis/sangue , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Estados Unidos , População Branca/genética
16.
Curr Opin Biotechnol ; 16(6): 687-90, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16257524

RESUMO

The integration of biomarkers reporting on drug pharmacodynamics or on a disease state is becoming necessary in the cost-effective design of clinical trials. Transcriptional changes in skin measured using whole-genome arrays have been useful in assessing the disease state in dermatology. Not only are skin biopsies well-tolerated and easy to obtain, but they sample a system in which many complex signaling and developmental networks are active and in which a wide variety of drug targets may be expected to play a role, either directly or indirectly. Recent advances have led to the use of transcriptional profiling of skin for making pharmacodynamic measurements in clinical trials, even for non-dermatological conditions.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Preparações Farmacêuticas/metabolismo , Farmacocinética , Pele/metabolismo , Animais , Biópsia/métodos , Biópsia/tendências , Ensaios Clínicos como Assunto/métodos , Ensaios Clínicos como Assunto/tendências , Desenho de Fármacos , Perfilação da Expressão Gênica/métodos , Perfilação da Expressão Gênica/tendências , Humanos , Preparações Farmacêuticas/administração & dosagem , Pele/patologia
17.
Genomics ; 86(5): 505-17, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16126366

RESUMO

The use of inbred strains of mice to dissect the genetic complexity of common diseases offers a viable alternative to human studies, given the control over experimental parameters that can be exercised. Central to efforts to map susceptibility loci for common diseases in mice is a comprehensive map of DNA variation among the common inbred strains of mice. Here we present one of the most comprehensive high-density, single nucleotide polymorphism (SNP) maps of mice constructed to date. This map consists of 10,350 SNPs genotyped in 62 strains of inbred mice. We demonstrate the utility of these data via a novel integrative genomics approach to mapping susceptibility loci for complex traits. By integrating in silico quantitative trait locus (QTL) mapping with progressive QTL mapping strategies in segregating mouse populations that leverage large-scale mapping of the genetic determinants of gene expression traits, we not only facilitate identification of candidate quantitative trait genes, but also protect against spurious associations that can arise in genetic association studies due to allelic association among unlinked markers. Application of this approach to our high-density SNP map and two previously described F2 crosses between strains C57BL/6J (B6) and DBA/2J and between B6 ApoE(-/-) and C3H/HeJ ApoE(-/-) results in the identification of Insig2 as a strong candidate susceptibility gene for total plasma cholesterol levels.


Assuntos
Colesterol/sangue , Mapeamento Cromossômico/métodos , Predisposição Genética para Doença , Hipercolesterolemia/genética , Proteínas de Membrana/genética , Animais , Marcadores Genéticos , Hipercolesterolemia/veterinária , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas
18.
Cell ; 115(6): 739-50, 2003 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-14675538

RESUMO

The eukaryotic initiation factor 4G (eIF4G) is the core of a multicomponent switch controlling gene expression at the level of translation initiation. It interacts with the small ribosomal subunit interacting protein, eIF3, and the eIF4E/cap-mRNA complex in order to load the ribosome onto mRNA during cap-dependent translation. We describe the solution structure of the complex between yeast eIF4E/cap and eIF4G (393-490). Binding triggers a coupled folding transition of eIF4G (393-490) and the eIF4E N terminus resulting in a molecular bracelet whereby eIF4G (393-490) forms a right-handed helical ring that wraps around the N terminus of eIF4E. Cofolding allosterically enhances association of eIF4E with the cap and is required for maintenance of optimal growth and polysome distributions in vivo. Our data explain how mRNA, eIF4E, and eIF4G exists as a stable mRNP that may facilitate multiple rounds of ribosomal loading during translation initiation, a key determinant in the overall rate of protein synthesis.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Biossíntese de Proteínas/genética , Capuzes de RNA/metabolismo , Ribossomos/metabolismo , Sítios de Ligação/fisiologia , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação Eucariótico 4G/genética , Guanosina Difosfato/metabolismo , Substâncias Macromoleculares , Modelos Moleculares , Conformação Molecular , Conformação de Ácido Nucleico , Conformação Proteica , Capuzes de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleoproteínas/metabolismo , Ribossomos/genética , Saccharomyces cerevisiae
19.
RNA ; 8(7): 948-58, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12166649

RESUMO

We describe a one-step affinity method for purifying ribosomes from the budding yeast Saccharomyces cerevisiae. Extracts from yeast strains expressing only C-terminally tagged Rpl25 protein or overexpressing this protein in the presence of endogenous Rpl25p were used as the starling materials. The purification was specific for tagged 60S subunits, and resulted in the copurification of 80S subunits and polysomes, as well as ribosome-associated proteins and mRNAs. Two of these associated proteins, Mpt4p and Asc1p, were nearly stoichiometrically bound to the ribosome. In addition, the degree of mRNA association with the purified ribosomes was found to reflect the mRNA's translational status within the cell. The one-step purification of ribosome and its associated components from a crude extract should provide an important tool for future structural and biochemical studies of the ribosome, as well as for expression profiling of translated mRNAs.


Assuntos
RNA Fúngico/isolamento & purificação , RNA Mensageiro/isolamento & purificação , Proteínas Ribossômicas/isolamento & purificação , Ribossomos/química , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Saccharomyces cerevisiae/química , Sequência de Bases , Fracionamento Celular/métodos , Cromatografia de Afinidade , Plasmídeos/genética , RNA Fúngico/genética , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética
20.
RNA ; 8(3): 382-97, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12008673

RESUMO

To provide a bridge between in vivo and in vitro studies of eukaryotic translation initiation, we have developed a reconstituted translation initiation system using components from the yeast Saccharomyces cerevisiae. We have purified a minimal set of initiation factors (elFs) that, together with yeast 80S ribosomes, GTP, and initiator methionyl-tRNA, are sufficient to assemble active initiation complexes on a minimal mRNA template. The kinetics of various steps in the pathway of initiation complex assembly and the formation of the first peptide bond in vitro have been explored. The formation of active initiation complexes in this system is dependent on ribosomes, mRNA, Met-tRNAi, GTP hydrolysis, elF1, elF1A, elF2, elF5, and elF5B. Our data indicate that elF1 and elF1A both facilitate the binding of the elF2 x GTP x Met-tRNAi complex to the 40S ribosomal subunit to form the 43S complex. elF5 stimulates a step after 43S complex formation, consistent with its proposed role in activating GTP hydrolysis by elF2 upon initiation codon recognition. The presence of elF5B is required for the joining of the 40S and 60S subunits to form the 80S initiation complex. The step at which each of these factors acts in this reconstituted system is in agreement with previous data from in vivo studies and work using reconstituted mammalian systems, indicating that the system recapitulates fundamental events in translation initiation in eukaryotic cells. This system should allow us to couple powerful yeast genetic and molecular biological experiments with in vitro kinetic and biophysical experiments, yielding a better understanding of the molecular mechanics of this central, complex process.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/metabolismo , Fatores de Iniciação de Peptídeos/fisiologia , RNA de Transferência de Metionina/metabolismo , Ribossomos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Células Eucarióticas/fisiologia , Proteínas de Ligação ao GTP/química , Humanos , Hidrólise , Cinética , Iniciação Traducional da Cadeia Peptídica , Fatores de Iniciação de Peptídeos/isolamento & purificação , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Transferência de Metionina/genética , Ribossomos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA