Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 50(20): 11401-11414, 2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-35944903

RESUMO

Current therapies for Duchenne muscular dystrophy (DMD) use phosphorodiamidate morpholino oligomers (PMO) to induce exon skipping in the dystrophin pre-mRNA, enabling the translation of a shortened but functional dystrophin protein. This strategy has been hampered by insufficient delivery of PMO to cardiac and skeletal muscle. To overcome these limitations, we developed the FORCETM platform consisting of an antigen-binding fragment, which binds the transferrin receptor 1, conjugated to an oligonucleotide. We demonstrate that a single dose of the mouse-specific FORCE-M23D conjugate enhances muscle delivery of exon skipping PMO (M23D) in mdx mice, achieving dose-dependent and robust exon skipping and durable dystrophin restoration. FORCE-M23D-induced dystrophin expression reached peaks of 51%, 72%, 62%, 90% and 77%, of wild-type levels in quadriceps, tibialis anterior, gastrocnemius, diaphragm, and heart, respectively, with a single 30 mg/kg PMO-equivalent dose. The shortened dystrophin localized to the sarcolemma, indicating expression of a functional protein. Conversely, a single 30 mg/kg dose of unconjugated M23D displayed poor muscle delivery resulting in marginal levels of exon skipping and dystrophin expression. Importantly, FORCE-M23D treatment resulted in improved functional outcomes compared with administration of unconjugated M23D. Our results suggest that FORCE conjugates are a potentially effective approach for the treatment of DMD.


The biggest problem confronting oligonucleotide therapeutics is a lack of compounds capable of targeting compounds to diseased tissues. This paper reports a major advance targeting the transferrin receptor to increase the delivery of morpholine oligomers to muscle cells in vivo. This work suggests the possibility for improved treatments of muscular dystrophy and other diseases.


Assuntos
Distrofina , Éxons , Morfolinos , Distrofia Muscular de Duchenne , Oligonucleotídeos Antissenso , Animais , Camundongos , Distrofina/genética , Éxons/genética , Camundongos Endogâmicos mdx , Morfolinos/farmacologia , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/farmacologia , Receptores da Transferrina/genética
2.
Nucleic Acid Ther ; 29(2): 104-113, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30676254

RESUMO

Primary Hyperoxaluria Type 1 (PH1) is an autosomal recessive disorder of glyoxylate metabolism. Loss of alanine glyoxylate aminotransferase (AGT) function to convert intermediate metabolite glyoxylate to glycine causes the accumulation and reduction of glyoxylate to glycolate, which eventually is oxidized to oxalate. Excess oxalate in PH1 patients leads to the formation and deposition of calcium oxalate crystals in the kidney and urinary tract. Oxalate crystal deposition causes a decline in renal function, systemic oxalosis, and eventually end-stage renal disease and premature death. mRNA-based therapies are a new class of drugs that work by replacing the missing enzyme. mRNA encoding AGT has the potential to restore normal glyoxylate to glycine metabolism, thus preventing the buildup of calcium oxalate in various organs. Panels of codon-optimized AGT mRNA constructs were screened in vitro and in wild-type mice for the production of a functional AGT enzyme. Two human constructs, wild-type and engineered AGT (RHEAM), were tested in Agxt-/- mice. Repeat dosing in Agxt-/- mice resulted in a 40% reduction in urinary oxalate, suggesting therapeutic benefit. These studies suggest that mRNA encoding AGT led to increased expression and activity of the AGT enzyme in liver that translated into decrease in urinary oxalate levels. Taken together, our data indicate that AGT mRNA may have the potential to be developed into a therapeutic for PH1.


Assuntos
Hiperoxalúria Primária/genética , Fígado/efeitos dos fármacos , RNA Mensageiro/farmacologia , Transaminases/farmacologia , Animais , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Glioxilatos/metabolismo , Humanos , Hiperoxalúria Primária/terapia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Oxalatos/metabolismo , RNA Mensageiro/genética , Transaminases/genética
3.
J Nucleic Acids ; 2018: 8247935, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30009048

RESUMO

Alpha-1-antitrypsin (AAT) deficiency is a genetic disorder that produces inactive/defective AAT due to mutations in the SERPINA1 gene encoding AAT. This disease is associated with decreased activity of AAT in the lungs and deposition of excessive defective AAT protein in the liver. Currently there is no specific treatment for liver disease associated with AAT deficiency. AAT lung disease is often treated with one of several serum protein replacement products; however, long-term studies of the effectiveness of SerpinA1 replacement therapy are not available, and it does not reduce liver damage in AAT deficiency. mRNA therapy could potentially target both the liver and lungs of AAT deficient patients. AAT patient fibroblasts and AAT patient fibroblast-derived hepatocytes were transfected with SERPINA1-encoding mRNA and cell culture media were tested for SerpinA1 expression. Our data demonstrates increased SerpinA1 protein in culture media from treated AAT patient fibroblasts and AAT patient fibroblast-derived hepatocytes. In vivo studies in wild type mice demonstrate SERPINA1 mRNA biodistribution in liver and lungs, as well as SerpinA1 protein expression in these two target organs which are critically affected in AAT deficiency. Taken together, our data suggests that SerpinA1 mRNA therapy has the potential to benefit patients suffering from AAT deficiency.

4.
RNA Biol ; 15(7): 914-922, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29923457

RESUMO

Arginase I (ARG1) deficiency is an autosomal recessive urea cycle disorder, caused by deficiency of the enzyme Arginase I, resulting in accumulation of arginine in blood. Current Standard of Care (SOC) for ARG1 deficiency in patients or those having detrimental mutations of ARG1 gene is diet control. Despite diet and drug therapy with nitrogen scavengers, ~25% of patients suffer from severe mental deficits and loss of ambulation. 75% of patients whose symptoms can be managed through diet therapy continue to suffer neuro-cognitive deficits. In our research, we demonstrate in vitro and in vivo that administration of ARG1 mRNA increased ARG1 protein expression and specific activity in relevant cell types, including ARG1-deficient patient cell lines, as well as in wild type mice for up to 4 days. These studies demonstrate that ARG1 mRNA treatment led to increased functional protein expression of ARG1 and subsequently an increase in urea. Hence, ARG1 mRNA therapy could be a potential treatment option to develop for patients.


Assuntos
Arginase/metabolismo , Arginina/metabolismo , Terapia Biológica/métodos , Hiperargininemia/terapia , RNA Mensageiro/administração & dosagem , Animais , Arginase/genética , Células HeLa , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Ureia/metabolismo
5.
Sci Rep ; 8(1): 7859, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29777164

RESUMO

Thrombotic thrombocytopenic purpura (TTP) is primarily caused by deficiency of ADAMTS13 within the blood stream due to either genetic defects or presence of inhibitory autoantibodies. Preclinical and clinical studies suggest that enzyme replacement therapy with recombinant human ADAMTS13 protein (rhADAMTS13) is effective and safe in treatment of TTP. However, frequent dosing would be required due to the relatively short half-life of rhADAMTS13 in circulation as well as the presence of inhibitory autoantibodies that collectively result in the poor pharmacological profile of rhADAMTS13. With technical breakthroughs in exploring mRNA as therapeutics, we hypothesized that restoration of ADAMTS13 activity for a prolonged duration of time can be achieved through systemic dosing of mRNA, wherein the dosed mRNA would utilize hepatic cells as bioreactors for continuous production of ADAMTS13. To test this hypothesis, mRNA encoding human ADAMTS13 WT or an ADAMTS13 variant, that had demonstrated resistance to predominant clinical TTP autoantibodies, was formulated in lipid nano-particles for liver-targeted delivery. In both ADAMTS13-sufficient and -deficient mice, a single dose of the formulated mRNAs at 1 mg/kg resulted in expression of hADAMTS13 at or above therapeutically relevant levels in mice for up to five days. This proof-of-concept study suggests that mRNA therapy could provide a novel approach for TTP treatment.


Assuntos
Proteína ADAMTS13/genética , Terapia Genética/métodos , RNA Mensageiro/genética , Proteína ADAMTS13/sangue , Proteína ADAMTS13/metabolismo , Animais , Autoanticorpos/sangue , Portadores de Fármacos/química , Células HEK293 , Humanos , Lipídeos/química , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Mutagênese , Nanopartículas/química , Púrpura Trombocitopênica Trombótica/terapia , Púrpura Trombocitopênica Trombótica/veterinária , RNA Mensageiro/sangue , RNA Mensageiro/química , RNA Mensageiro/uso terapêutico
6.
RNA Biol ; 15(6): 756-762, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29578372

RESUMO

mRNA based therapies hold great promise for the treatment of genetic diseases. However, this therapeutic approach suffers from multiple challenges including the short half-life of exogenously administered mRNA and subsequent protein production. Modulation of untranslated regions (UTR) represents one approach to enhance both mRNA stability and translation efficiency. The current studies describe and validate screening methods using a diverse set of 5'UTR and 3'UTR combinations for improved expression of the Arginase 1 (ARG1) protein, a potential therapeutic mRNA target. Data revealed a number of critical aspects which need to be considered when developing a screening approach for engineering mRNA improvements. First, plasmid-based screening methods do not correlate with protein expression driven by exogenously expressed mRNA. Second, improved ARG1 protein production was driven by increased translation and not improved mRNA stability. Finally, the 5' UTR appears to be the key driver in protein expression for exogenously delivered mRNA. From the testing of the combinatorial library, the 5'UTR for complement factor 3 (C3) and cytochrome p4502E1 (CYP2E1) showed the largest and most consistent increase in protein expression relative to a reference UTR. Collectively, these data provide important information for the development and optimization of therapeutic mRNAs.


Assuntos
Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Arginase , Complemento C3/genética , Citocromo P-450 CYP2E1/genética , Biossíntese de Proteínas/genética , Arginase/biossíntese , Arginase/genética
7.
Mol Ther ; 26(3): 814-821, 2018 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-29428299

RESUMO

Glycogen storage disease type Ia (GSD1a) is an inherited metabolic disorder caused by the deficiency of glucose-6-phosphatase (G6Pase). GSD1a is associated with life-threatening hypoglycemia and long-term liver and renal complications. We examined the efficacy of mRNA-encoding human G6Pase in a liver-specific G6Pase-/- mouse model (L-G6PC-/-) that exhibits the same hepatic biomarkers associated with GSD1a patients, such as fasting hypoglycemia, and elevated levels of hepatic glucose-6-phosphate (G6P), glycogen, and triglycerides. We show that a single systemic injection of wild-type or native human G6PC mRNA results in significant improvements in fasting blood glucose levels for up to 7 days post-dose. These changes were associated with significant reductions in liver mass, hepatic G6P, glycogen, and triglycerides. In addition, an engineered protein variant of human G6Pase, designed for increased duration of expression, showed superior efficacy to the wild-type sequence by maintaining improved fasting blood glucose levels and reductions in liver mass for up to 12 days post-dose. Our results demonstrate for the first time the effectiveness of mRNA therapy as a potential treatment in reversing the hepatic abnormalities associated with GSD1a.


Assuntos
Glicemia , Terapia Genética , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Fígado/metabolismo , RNA Mensageiro/genética , Animais , Biomarcadores , Modelos Animais de Doenças , Jejum , Expressão Gênica , Glucose-6-Fosfatase/metabolismo , Doença de Depósito de Glicogênio/patologia , Doença de Depósito de Glicogênio/terapia , Imuno-Histoquímica , Fígado/patologia , Masculino , Redes e Vias Metabólicas , Camundongos , Camundongos Knockout , Engenharia de Proteínas
8.
Nucleic Acid Ther ; 28(2): 74-85, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29437538

RESUMO

Messenger RNA (mRNA) is a promising new class of therapeutics that has potential for treatment of diseases in fields such as immunology, oncology, vaccines, and inborn errors of metabolism. mRNA therapy has several advantages over DNA-based gene therapy, including the lack of the need for nuclear import and transcription, as well as limited possibility of genomic integration. One drawback of mRNA therapy, especially in cases such as metabolic disorders where repeated dosing will be necessary, is the relatively short in vivo half-life of mRNA (∼6-12 h). We hypothesize that protein engineering designed to improve translation, yielding longer-lasting protein, or modifications that would increase enzymatic activity would be helpful in alleviating this issue. In this study, we present two examples where sequence engineering improved the expression and duration, as well as enzymatic activity of target proteins in vitro. We then confirmed these findings in wild-type mice. This work shows that rational engineering of proteins can lead to improved therapies in vivo.


Assuntos
Arginase/genética , Hiperargininemia/terapia , Hipoxantina Fosforribosiltransferase/genética , Síndrome de Lesch-Nyhan/terapia , RNA Mensageiro/uso terapêutico , Sequência de Aminoácidos , Animais , Arginase/isolamento & purificação , Arginase/metabolismo , Células HeLa , Humanos , Hiperargininemia/sangue , Hipoxantina Fosforribosiltransferase/isolamento & purificação , Hipoxantina Fosforribosiltransferase/metabolismo , Síndrome de Lesch-Nyhan/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Nanopartículas/uso terapêutico , Engenharia de Proteínas , RNA Mensageiro/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
9.
J Cardiovasc Pharmacol ; 67(5): 433-41, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26841069

RESUMO

Leukocyte infiltration of adventitial and perivascular tissues is an early event in the development of vascular remodeling after injury. We investigated whether Slit/Robo-an axonal chemorepellent system in vertebrate and invertebrate development-is activated during the inflammatory phase that follows endothelial denudation. Using the rat carotid artery model of angioplasty, we conducted a time course analysis of mRNAs encoding Slit ligands (Slit2 and Slit3) and Robo receptors (Robo1, Robo2, and Robo4), as well as proinflammatory cell adhesion molecule (CAM) genes. Adventitial inflammatory cells were counted in immunostained arterial sections. E-selectin, vascular CAM-1, and intercellular CAM-1 were upregulated 2-3 hours after injury, followed by infiltration of neutrophils and monocytes as evidenced by real-time polymerase chain reaction, in situ hybridization, and immunohistochemistry. Slit2, Slit3, and Robo genes exhibited no expression changes at 3 hours; however, they were markedly upregulated 1 day after angioplasty. Intercellular CAM-1 expression was reduced by 50%, and the number of adventitial neutrophils decreased by >75% 1 day after angioplasty. Slit2 has been shown to be a potent chemorepelent of leukocytes, endothelial cells, and smooth muscle cells. Thus, we decided to further investigate the localization of Slit2 in injured vessels. Immunohistochemical stainings revealed the presence of Slit2 within the vessel wall and in the perivascular vasa vasorum of naive and injured arteries. Double immunohistochemical analyses showed that infiltrating monocytes expressed Slit2 in the perivascular and adventitial tissues of injured arteries 1 and 3 days postangioplasty. In addition, recombinant full-length Slit2 and Slit2-N/1118, an N-terminal fragment of Slit2, inhibited stromal cell-derived factor 1-mediated migration of circulating rat peripheral blood mononuclear cells. In summary, adventitial activation of CAM genes and neutrophil infiltration preceded upregulation of Slit/Robo genes. Sli2 expression colocalized with infiltrating inflammatory cells in the adventitial layer. This temporospatial association suggests that leukocyte chemorepellent Slit2 may be involved in halting the adventitial accumulation of inflammatory cells in injured vessels.


Assuntos
Artérias Carótidas/fisiopatologia , Células Endoteliais/metabolismo , Mediadores da Inflamação/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Animais , Moléculas de Adesão Celular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Proteínas de Membrana/biossíntese , RNA Mensageiro , Ratos , Receptores Imunológicos/biossíntese , Regulação para Cima , Proteínas Roundabout
10.
Nucleic Acids Res ; 43(19): 9123-32, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26446989

RESUMO

The in vivo potency of antisense oligonucleotides (ASO) has been significantly increased by reducing their length to 8-15 nucleotides and by the incorporation of high affinity RNA binders such as 2', 4'-bridged nucleic acids (also known as locked nucleic acid or LNA, and 2',4'-constrained ethyl [cET]). We now report the development of a novel ASO design in which such short ASO monomers to one or more targets are co-synthesized as homo- or heterodimers or multimers via phosphodiester linkers that are stable in plasma, but cleaved inside cells, releasing the active ASO monomers. Compared to current ASOs, these multimers and multi-targeting oligonucleotides (MTOs) provide increased plasma protein binding and biodistribution to liver, and increased in vivo efficacy against single or multiple targets with a single construct. In vivo, MTOs synthesized in both RNase H-activating and steric-blocking oligonucleotide designs provide ≈4-5-fold increased potency and ≈2-fold increased efficacy, suggesting broad therapeutic applications.


Assuntos
Oligonucleotídeos Antissenso/química , Animais , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Dimerização , Feminino , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos Antissenso/farmacologia , Distribuição Tecidual
11.
Int J Oncol ; 41(5): 1855-62, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22922669

RESUMO

Members of the Ras protooncogene family are mutated in approximately 75% of colon cancers. The Raf kinases (Raf-1, b-Raf and a-Raf) directly interact with Ras and serve as mediators of mitogenic signals. Expression of the constitutively active alleles of Raf or Ras gene families results in oncogenesis in a number of model systems. Previous studies emphasized the importance of Raf-1 and b-Raf in preventing apoptosis in addition to their roles in cell growth. In the present study, we examined whether inhibition of the Raf-1 or b-Raf kinase decreases cell growth and increases apoptosis in colon cancer cells. c-Raf and b-Raf were depleted in colon cancer cell lines, such as HCT116, HT29 and Colo205, containing Ras or b-Raf mutations by RNA interference (RNAi). The results showed that colon cancer cells with activating Ras mutations undergo apoptosis following Raf-1 inhibition, as determined by cell cycle analysis and the release of cytochrome c. Moreover, in b-Raf mutant colon cancers, the inhibition of b-Raf as compared to Raf-1 is crucial for cancer cell death. There is increasing evidence for both MEK-independent Raf signaling and Raf-independent MEK signaling. Thus, we investigated whether targeting multiple points of the mitogen-activated protein kinase (MAPK) pathway with a MEK inhibitor and Raf RNAi increases cancer cell death. The results showed that combination therapy, inhibiting Raf and MEK kinases simultaneously, increased apoptosis in colon cancer cells. Taken together, our data demonstrate that combination therapy targeting the MAPK pathway at two distinct points, Raf kinase and MEK, has greater efficacy in increasing cancer cell death and is likely to improve therapeutic outcomes for patients.


Assuntos
Apoptose , Neoplasias do Colo/terapia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Interferência de RNA , Animais , Apoptose/genética , Proliferação de Células , Chlorocebus aethiops , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo
12.
J Cancer ; 2: 490-502, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21980324

RESUMO

Investigation of therapy naïve human tumor and adjacent normal tissue biopsies demonstrated that expression levels of miRNAs are altered at and between stages of CRC. Targets of these altered miRNAs are members of the Insulin signaling pathways. Phosphorylation states of several molecules in the Insulin signaling pathways were altered between stages of CRC, and significantly the change in molecular phosphorylation state correlated with decreases in specific miRNAs that target them. This data establishes a direct relationship between decreased expression of specific miRNAs and increased phosphorylation events in the IGF-1 pathway and identifies the IGF-1 pathway as a critical driver of colorectal cancer.The expression levels of 319 miRNAs and phosphorylation levels of major signaling proteins were determined. Interestingly, we observed that miRNAs were altered in expression and several signaling molecules were altered in phosphorylation levels at and between each stage of CRC. Furthermore, many of the miRNAs that are differentially expressed at each CRC stage were targeting these same signaling proteins identified to be altered in phosphorylation level. Thus, our studies define a subset of important miRNAs to classify CRC stage and a relationship between miRNA depression and elevated phosphorylation of IGF-1R pathway signaling molecules.

13.
Mol Ther ; 19(12): 2163-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21971426

RESUMO

Antisense oligonucleotides (ASOs) and small interfering RNA (siRNA) promise specific correction of disease-causing gene expression. Therapeutic implementation, however, has been forestalled by poor delivery to the appropriate tissue, cell type, and subcellular compartment. Topical administration is considered to circumvent these issues. The availability of inhalation devices and unmet medical need in lung disease has focused efforts in this tissue. We report the development of a novel cell sorting method for quantitative, cell type-specific analysis of siRNA, and locked nucleic acid (LNA) ASO uptake and efficacy after intratracheal (i.t.) administration in mice. Through fluorescent dye labeling, we compare the utility of this approach to whole animal and whole tissue analysis, and examine the extent of tissue distribution. We detail rapid systemic access and renal clearance for both therapeutic classes and lack of efficacy at the protein level in lung macrophages, epithelia, or other cell types. We nevertheless observe efficient redirection of i.t. administered phosphorothioate (PS) LNA ASO to the liver and kidney leading to targeted gene knockdown. These data suggest delivery remains a key obstacle to topically administered, naked oligonucleotide efficacy in the lung and introduce inhalation as a potentially viable alternative to injection for antisense administration to the liver and kidneys.


Assuntos
Apolipoproteínas B/antagonistas & inibidores , Inativação Gênica , Luciferases/antagonistas & inibidores , Oligonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos/farmacocinética , RNA Interferente Pequeno/genética , Administração por Inalação , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Células Cultivadas , Marcação de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Rim/citologia , Rim/metabolismo , Fígado/citologia , Fígado/metabolismo , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos Antissenso/administração & dosagem , Distribuição Tecidual
14.
PLoS One ; 6(7): e21503, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21750714

RESUMO

Small interfering RNAs (siRNAs) are routinely used to reduce mRNA levels for a specific gene with the goal of studying its function. Several studies have demonstrated that siRNAs are not always specific and can have many off-target effects. The 3' UTRs of off-target mRNAs are often enriched in sequences that are complementary to the seed-region of the siRNA. We demonstrate that siRNA off-targets can be significantly reduced when cells are treated with a dose of siRNA that is relatively low (e.g. 1 nM), but sufficient to effectively silence the intended target. The reduction in off-targets was demonstrated for both modified and unmodified siRNAs that targeted either STAT3 or hexokinase II. Low concentrations reduced silencing of transcripts with complementarity to the seed region of the siRNA. Similarly, off-targets that were not complementary to the siRNA were reduced at lower doses, including up-regulated genes that are involved in immune response. Importantly, the unintended induction of caspase activity following treatment with a siRNA that targeted hexokinase II was also shown to be a concentration-dependent off-target effect. We conclude that off-targets and their related phenotypic effects can be reduced for certain siRNA that potently silence their intended target at low concentrations.


Assuntos
Hexoquinase/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Fator de Transcrição STAT3/genética , Regiões 3' não Traduzidas/genética , Sequência de Bases , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Lipid Res ; 52(6): 1265-1271, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21371998

RESUMO

Apolipoprotein CIII (apoCIII), a major constituent of triglyceride-rich lipoprotein, has been proposed as a key contributor to hypertriglyceridemia on the basis of its inhibitory effects on lipoprotein lipase. Many immunochemical methods have been developed for human apoCIII quantification, including ELISA. However, a sensitive and quantitative assay for nonhuman primates is not commercially available. We developed a sensitive, quantitative, and highly specific sandwich ELISA to measure apoCIII in both nonhuman primate and human serum. Our assay generates a linear calibration curve from 0.01 µg/ml to 10 µg/ml using an apoCIII standard that was purified from cynomolgus monkey serum. It is highly reproducible (intra- and interplate CV < 5% and < 8%, respectively), sensitive enough to distinguish 10% difference of apoCIII present in serum, and has no interference from purified human apolipoprotein AI, AII, B, CI, CII, or E. The same assay can also be used to measure human apoCIII with a linear calibration curve from 0.005 µg/ml to 1 µg/ml using purified human apoCIII as the standard. This fast and highly sensitive ELISA could be a useful tool to investigate the role of apoCIII in lipoprotein transport and cardiovascular disease.


Assuntos
Anticorpos/metabolismo , Apolipoproteína C-III/sangue , Biomarcadores/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Hipertrigliceridemia/sangue , Animais , Anticorpos/imunologia , Western Blotting , Humanos , Hipertrigliceridemia/fisiopatologia , Limite de Detecção , Macaca fascicularis , Masculino , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
16.
Clin Cancer Res ; 15(24): 7571-7581, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19996220

RESUMO

PURPOSE: Androgen receptor abundance and androgen receptor-regulated gene expression in castration-recurrent prostate cancer are indicative of androgen receptor activation in the absence of testicular androgen. Androgen receptor transactivation of target genes in castration-recurrent prostate cancer occurs in part through mitogen signaling that amplifies the actions of androgen receptor and its coregulators. Herein we report on the role of 14-3-3eta in androgen receptor action. Experimental Design and RESULTS: Androgen receptor and 14-3-3eta colocalized in COS cell nuclei with and without androgen, and 14-3-3eta promoted androgen receptor nuclear localization in the absence of androgen. 14-3-3eta interacted with androgen receptor in cell-free binding and coimmunoprecipitation assays. In the recurrent human prostate cancer cell line, CWR-R1, native endogenous androgen receptor transcriptional activation was stimulated by 14-3-3eta at low dihydrotestosterone concentrations and was increased by epidermal growth factor. Moreover, the dihydrotestosterone- and epidermal growth factor-dependent increase in androgen receptor transactivation was inhibited by a dominant negative 14-3-3eta. In the CWR22 prostate cancer xenograft model, 14-3-3eta expression was increased by androgen, suggesting a feed-forward mechanism that potentiates both 14-3-3eta and androgen receptor actions. 14-3-3eta mRNA and protein decreased following castration of tumor-bearing mice and increased in tumors of castrate mice after treatment with testosterone. CWR22 tumors that recurred 5 months after castration contained 14-3-3eta levels similar to the androgen-stimulated tumors removed before castration. In a human prostate tissue microarray of clinical specimens, 14-3-3eta localized with androgen receptor in nuclei, and the similar amounts expressed in castration-recurrent prostate cancer, androgen-stimulated prostate cancer, and benign prostatic hyperplasia were consistent with androgen receptor activation in recurrent prostate cancer. CONCLUSION: 14-3-3eta enhances androgen- and mitogen-induced androgen receptor transcriptional activity in castration-recurrent prostate cancer. (Clin Cancer Res 2009;15(24):7571-81).

17.
Exp Cell Res ; 294(2): 581-91, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15023544

RESUMO

Apoptosis signal-regulating kinase 1 (ASK1) is a critical mediator of apoptotic signaling pathways initiated by a variety of death stimuli. Its activity is tightly controlled by various mechanisms such as covalent modification and protein-protein interaction. One of the proteins that control ASK1 function is 14-3-3zeta, a member of the 14-3-3 protein family. Here, we report that ASK1 is capable of binding to other isoforms of 14-3-3, suggesting that binding ASK1 is a general property of the 14-3-3 family. In support of this notion, mutational analysis revealed that the ASK1/14-3-3 interaction was mediated by the conserved amphipathic groove of 14-3-3 with some residue selectivity. Functionally, expression of various isoforms of 14-3-3 suppressed ASK1-induced apoptosis. To understand how 14-3-3 controls the ASK1 activity, we examined intracellular localization of ASK1 upon 14-3-3 co-expression. We found that 14-3-3 co-expression is correlated with the translocation of ASK1 from the cytoplasm to a perinuclear localization, likely the ER compartment. Consistent with this notion, ASK1(S967A), a 14-3-3 binding defective mutant of ASK, showed no change in intracellular distribution upon 14-3-3 co-expression. These data support a model that 14-3-3 proteins regulate the proapoptotic function of ASK1 in part by controlling its subcellular distribution.


Assuntos
Apoptose/genética , MAP Quinase Quinase Quinases/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas 14-3-3 , Animais , Sítios de Ligação/genética , Células COS , Compartimento Celular/genética , Análise Mutacional de DNA , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , MAP Quinase Quinase Quinase 5 , MAP Quinase Quinase Quinases/genética , Mutação/genética , Ligação Proteica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico/genética , Tirosina 3-Mono-Oxigenase/genética
18.
Cancer Cell ; 3(5): 483-95, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12781366

RESUMO

Recent studies have demonstrated that introduction of hTERT in combination with SV40 large T antigen (LT), small t antigen (st), and H-rasV12 suffices to transform many primary human cells. In human mammary epithelial cells (HMECs) expressing elevated c-Myc, activated H-Ras is dispensable for anchorage-independent growth. Using this system, we show that st activates the PI3K pathway and that constitutive PI3K signaling substitutes for st in transformation. Moreover, using constitutively active versions of Akt1 and Rac1, we show that these downstream pathways of PI3K synergize to achieve anchorage-independent growth. At lower levels of c-myc expression, activated PI3K also replaces st to complement H-rasV12 and LT and confers both soft agar growth and tumorigenicity. However, elevated c-myc expression cannot replace H-rasV12 for tumorigenesis. These observations begin to define the pathways perturbed during the transformation of HMECs.


Assuntos
Transformação Celular Neoplásica , Glândulas Mamárias Humanas/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas , Ágar/metabolismo , Processamento Alternativo , Divisão Celular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Fibroblastos/metabolismo , Vetores Genéticos , Humanos , Immunoblotting , Fosforilação , Testes de Precipitina , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-myc/metabolismo , Retroviridae/genética , Fatores de Tempo , Transfecção , Proteínas rac1 de Ligação ao GTP/metabolismo
19.
J Biol Chem ; 277(10): 7962-9, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11773070

RESUMO

DEF-1/ASAP1 is an ADP-ribosylation factor GTPase-activating protein (ARF GAP) that localizes to focal adhesions and is involved in cytoskeletal regulation. In this paper, we use a cell-based ARF GAP assay to demonstrate that DEF-1 functions as a GAP for ARF1 and not ARF6 in vivo. This degree of substrate preference was unique to DEF-1, as other ARF GAP proteins, ACAP1, ACAP2, and ARFGAP1, were able to function on both ARF1 and ARF6. Since transient overexpression of DEF-1 has been shown to interfere with focal adhesion formation and platelet-derived growth factor-induced membrane ruffling, we investigated whether NIH 3T3 cells stably expressing DEF-1 have altered cell motility. Here we report that ectopic DEF-1 enhances cell migration toward PDGF as well as IGF-1. This chemotactic effect appears to result from a general increase in cell motility, as DEF-1-expressing cells also exhibit enhanced levels of basal and chemokinetic motility. The increase in cell motility is dependent on DEF-1 GAP activity, since a DEF-1 mutant lacking the GAP domain failed to stimulate motility. This suggests that DEF-1 alters cell motility through the deactivation of ARF1. In contrast, the inhibition of cell spreading by DEF-1 was not dependent on GAP activity, indicating that spreading and motility are altered by DEF-1 through different pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Domínios de Homologia de src , Células 3T3 , Fator 1 de Ribosilação do ADP/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Animais , Western Blotting , Células COS , Linhagem Celular , Movimento Celular , Separação Celular , Proteínas do Citoesqueleto/biossíntese , Citoesqueleto/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Proteínas Ativadoras de GTPase/química , Glutationa Transferase/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Paxilina , Fosfoproteínas/biossíntese , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/metabolismo , Fatores de Tempo , Transfecção , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...