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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
RNA ; 25(2): 255-263, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30463937

RESUMEN

Hereditary angioedema (HAE) is a genetic disorder mostly caused by mutations in the C1 esterase inhibitor gene (C1INH) that results in poor control of contact pathway activation and excess bradykinin generation. Bradykinin increases vascular permeability and is ultimately responsible for the episodes of swelling characteristic of HAE. We hypothesized that the use of RNA interference (RNAi) to reduce plasma Factor XII (FXII), which initiates the contact pathway signaling cascade, would reduce contact pathway activation and prevent excessive bradykinin generation. A subcutaneously administered GalNAc-conjugated small-interfering RNA (siRNA) targeting F12 mRNA (ALN-F12) was developed, and potency was evaluated in mice, rats, and cynomolgus monkeys. The effect of FXII reduction by ALN-F12 administration was evaluated in two different vascular leakage mouse models. An ex vivo assay was developed to evaluate the correlation between human plasma FXII levels and high-molecular weight kininogen (HK) cleavage. A single subcutaneous dose of ALN-F12 led to potent, dose-dependent reduction of plasma FXII in mice, rats, and NHP. In cynomolgus monkeys, a single subcutaneous dose of ALN-F12 at 3 mg/kg resulted in >85% reduction of plasma FXII. Administration of ALN-F12 resulted in dose-dependent reduction of vascular permeability in two different mouse models of bradykinin-driven vascular leakage, demonstrating that RNAi-mediated reduction of FXII can potentially mitigate excess bradykinin stimulation. Lastly, ex vivo human plasma HK cleavage assay indicated FXII-dependent bradykinin generation. Together, these data suggest that RNAi-mediated knockdown of FXII by ALN-F12 is a potentially promising approach for the prophylactic treatment of HAE.


Asunto(s)
Angioedemas Hereditarios/tratamiento farmacológico , Bradiquinina/biosíntesis , Factor XII/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Animales , Permeabilidad Capilar/efectos de los fármacos , Proteína Inhibidora del Complemento C1/genética , Factor XII/análisis , Femenino , Humanos , Quininógenos/metabolismo , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , Ratas
2.
Nucleic Acids Res ; 47(7): 3306-3320, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30820542

RESUMEN

For oligonucleotide therapeutics, chemical modifications of the sugar-phosphate backbone are frequently used to confer drug-like properties. Because 2'-deoxy-2'-fluoro (2'-F) nucleotides are not known to occur naturally, their safety profile was assessed when used in revusiran and ALN-TTRSC02, two short interfering RNAs (siRNAs), of the same sequence but different chemical modification pattern and metabolic stability, conjugated to an N-acetylgalactosamine (GalNAc) ligand for targeted delivery to hepatocytes. Exposure to 2'-F-monomer metabolites was low and transient in rats and humans. In vitro, 2'-F-nucleoside 5'-triphosphates were neither inhibitors nor preferred substrates for human polymerases, and no obligate or non-obligate chain termination was observed. Modest effects on cell viability and mitochondrial DNA were observed in vitro in a subset of cell types at high concentrations of 2'-F-nucleosides, typically not attained in vivo. No apparent functional impact on mitochondria and no significant accumulation of 2'-F-monomers were observed after weekly administration of two GalNAc-siRNA conjugates in rats for ∼2 years. Taken together, the results support the conclusion that 2'-F nucleotides can be safely applied for the design of metabolically stabilized therapeutic GalNAc-siRNAs with favorable potency and prolonged duration of activity allowing for low dose and infrequent dosing.


Asunto(s)
Acetilgalactosamina/efectos adversos , Acetilgalactosamina/química , Desoxirribonucleótidos/efectos adversos , Desoxirribonucleótidos/química , Flúor/química , ARN Interferente Pequeño/efectos adversos , ARN Interferente Pequeño/química , Animales , Femenino , Flúor/efectos adversos , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
3.
N Engl J Med ; 377(9): 819-828, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28691885

RESUMEN

BACKGROUND: Current hemophilia treatment involves frequent intravenous infusions of clotting factors, which is associated with variable hemostatic protection, a high treatment burden, and a risk of the development of inhibitory alloantibodies. Fitusiran, an investigational RNA interference (RNAi) therapy that targets antithrombin (encoded by SERPINC1), is in development to address these and other limitations. METHODS: In this phase 1 dose-escalation study, we enrolled 4 healthy volunteers and 25 participants with moderate or severe hemophilia A or B who did not have inhibitory alloantibodies. Healthy volunteers received a single subcutaneous injection of fitusiran (at a dose of 0.03 mg per kilogram of body weight) or placebo. The participants with hemophilia received three injections of fitusiran administered either once weekly (at a dose of 0.015, 0.045, or 0.075 mg per kilogram) or once monthly (at a dose of 0.225, 0.45, 0.9, or 1.8 mg per kilogram or a fixed dose of 80 mg). The study objectives were to assess the pharmacokinetic and pharmacodynamic characteristics and safety of fitusiran. RESULTS: No thromboembolic events were observed during the study. The most common adverse events were mild injection-site reactions. Plasma levels of fitusiran increased in a dose-dependent manner and showed no accumulation with repeated administration. The monthly regimen induced a dose-dependent mean maximum antithrombin reduction of 70 to 89% from baseline. A reduction in the antithrombin level of more than 75% from baseline resulted in median peak thrombin values at the lower end of the range observed in healthy participants. CONCLUSIONS: Once-monthly subcutaneous administration of fitusiran resulted in dose-dependent lowering of the antithrombin level and increased thrombin generation in participants with hemophilia A or B who did not have inhibitory alloantibodies. (Funded by Alnylam Pharmaceuticals; ClinicalTrials.gov number, NCT02035605 .).


Asunto(s)
Antitrombina III/antagonistas & inhibidores , Hemofilia A/terapia , Hemofilia B/terapia , Tratamiento con ARN de Interferencia , Adulto , Antitrombinas/sangre , Relación Dosis-Respuesta a Droga , Voluntarios Sanos , Hemofilia A/sangre , Hemofilia B/sangre , Humanos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Método Simple Ciego , Trombina/biosíntesis , Adulto Joven
4.
Nucleic Acids Res ; 45(19): 10969-10977, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28981809

RESUMEN

Covalent attachment of a synthetic triantennary N-acetylagalactosamine (GalNAc) ligand to chemically modified siRNA has enabled asialoglycoprotein (ASGPR)-mediated targeted delivery of therapeutically active siRNAs to hepatocytes in vivo. This approach has become transformative for the delivery of RNAi therapeutics as well as other classes of investigational oligonucleotide therapeutics to the liver. For efficient functional delivery of intact drug into the desired subcellular compartment, however, it is critical that the nucleic acids are stabilized against nucleolytic degradation. Here, we compared two siRNAs of the same sequence but with different modification pattern resulting in different degrees of protection against nuclease activity. In vitro stability studies in different biological matrices show that 5'-exonuclease is the most prevalent nuclease activity in endo-lysosomal compartments and that additional stabilization in the 5'-regions of both siRNA strands significantly enhances the overall metabolic stability of GalNAc-siRNA conjugates. In good agreement with in vitro findings, the enhanced stability translated into substantially improved liver exposure, gene silencing efficacy and duration of effect in mice. Follow-up studies with a second set of conjugates targeting a different transcript confirmed the previous results, provided additional insights into kinetics of RISC loading and demonstrated excellent translation to non-human primates.


Asunto(s)
Acetilgalactosamina/farmacocinética , Riñón/metabolismo , Hígado/metabolismo , ARN Interferente Pequeño/farmacocinética , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/metabolismo , Animales , Área Bajo la Curva , Sistemas de Liberación de Medicamentos/métodos , Humanos , Hígado/citología , Masculino , Tasa de Depuración Metabólica , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/metabolismo
5.
Nucleic Acids Res ; 43(16): 7984-8001, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26220182

RESUMEN

Most delivery systems for small interfering RNA therapeutics depend on endocytosis and release from endo-lysosomal compartments. One approach to improve delivery is to identify small molecules enhancing these steps. It is unclear to what extent such enhancers can be universally applied to different delivery systems and cell types. Here, we performed a compound library screen on two well-established siRNA delivery systems, lipid nanoparticles and cholesterol conjugated-siRNAs. We identified fifty-one enhancers improving gene silencing 2-5 fold. Strikingly, most enhancers displayed specificity for one delivery system only. By a combination of quantitative fluorescence and electron microscopy we found that the enhancers substantially differed in their mechanism of action, increasing either endocytic uptake or release of siRNAs from endosomes. Furthermore, they acted either on the delivery system itself or the cell, by modulating the endocytic system via distinct mechanisms. Interestingly, several compounds displayed activity on different cell types. As proof of principle, we showed that one compound enhanced siRNA delivery in primary endothelial cells in vitro and in the endocardium in the mouse heart. This study suggests that a pharmacological approach can improve the delivery of siRNAs in a system-specific fashion, by exploiting distinct mechanisms and acting upon multiple cell types.


Asunto(s)
ARN Interferente Pequeño/administración & dosificación , Animales , Células Cultivadas , Colesterol , Endosomas/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Células HeLa , Hepatocitos/metabolismo , Humanos , Lípidos , Ratones , Nanopartículas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Bibliotecas de Moléculas Pequeñas
6.
Proc Natl Acad Sci U S A ; 111(11): 3955-60, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24516150

RESUMEN

siRNA therapeutics have promise for the treatment of a wide range of genetic disorders. Motivated by lipoproteins, we report lipopeptide nanoparticles as potent and selective siRNA carriers with a wide therapeutic index. Lead material cKK-E12 showed potent silencing effects in mice (ED50 ∼ 0.002 mg/kg), rats (ED50 < 0.01 mg/kg), and nonhuman primates (over 95% silencing at 0.3 mg/kg). Apolipoprotein E plays a significant role in the potency of cKK-E12 both in vitro and in vivo. cKK-E12 was highly selective toward liver parenchymal cell in vivo, with orders of magnitude lower doses needed to silence in hepatocytes compared with endothelial cells and immune cells in different organs. Toxicity studies showed that cKK-E12 was well tolerated in rats at a dose of 1 mg/kg (over 100-fold higher than the ED50). To our knowledge, this is the most efficacious and selective nonviral siRNA delivery system for gene silencing in hepatocytes reported to date.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Lipopéptidos/química , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , Animales , Apolipoproteínas E/metabolismo , Microscopía por Crioelectrón , Silenciador del Gen , Hepatocitos/metabolismo , Macaca fascicularis , Ratones , ARN Interferente Pequeño/uso terapéutico , Ratas
7.
J Hepatol ; 64(4): 899-907, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26658687

RESUMEN

BACKGROUND & AIMS: The Hippo pathway controls organ size through a negative regulation of the transcription co-activator Yap1. The overexpression of hyperactive mutant Yap1 or deletion of key components in the Hippo pathway leads to increased organ size in different species. Analysis of interactions of this pathway with other cellular signals corroborating organ size control is limited in part due to the difficulties associated with development of rodent models. METHODS: Here, we develop a new model of reversible induction of the liver size in mice using siRNA-nanoparticles targeting two kinases of the Hippo pathway, namely, mammalian Ste20 family kinases 1 and 2 (Mst1 and Mst2), and an upstream regulator, neurofibromatosis type II (Nf2). RESULTS: The triple siRNAs nanoparticle-induced hepatomegaly in mice phenocopies one observed with Mst1(-/-)Mst2(-/-) liver-specific depletion, as shown by extensive proliferation of hepatocytes and activation of Yap1. The simultaneous co-treatment with a fourth siRNA nanoparticle against Yap1 fully blocked the liver growth. Hippo pathway-induced liver enlargement is associated with p53 activation, evidenced by its accumulation in the nuclei and upregulation of its target genes. Moreover, injections of the triple siRNAs nanoparticle in p53(LSL/LSL) mice shows that livers lacking p53 expression grow faster and exceed the size of livers in p53 wild-type animals, indicating a role of p53 in controlling Yap1-induced liver growth. CONCLUSION: Our data show that siRNA-nanoparticulate manipulation of gene expression can provide the reversible control of organ size in adult animals, which presents a new avenue for the investigation of complex regulatory networks in liver.


Asunto(s)
Genómica/métodos , Hígado/crecimiento & desarrollo , Nanopartículas , Interferencia de ARN , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas de Ciclo Celular , Expresión Génica , Genes de la Neurofibromatosis 2 , Factor de Crecimiento de Hepatocito/genética , Hepatomegalia/etiología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Fosfoproteínas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Serina-Treonina Quinasa 3 , Proteína p53 Supresora de Tumor/fisiología , Proteínas Señalizadoras YAP
8.
Chembiochem ; 17(11): 985-9, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27121751

RESUMEN

Small interfering RNA (siRNA)-mediated silencing requires siRNA loading into the RNA-induced silencing complex (RISC). Presence of 5'-phosphate (5'-P) is reported to be critical for efficient RISC loading of the antisense strand (AS) by anchoring it to the mid-domain of the Argonaute2 (Ago2) protein. Phosphorylation of exogenous duplex siRNAs is thought to be accomplished by cytosolic Clp1 kinase. However, although extensive chemical modifications are essential for siRNA-GalNAc conjugate activity, they can significantly impair Clp1 kinase activity. Here, we further elucidated the effect of 5'-P on the activity of siRNA-GalNAc conjugates. Our results demonstrate that a subset of sequences benefit from the presence of exogenous 5'-P. For those that do, incorporation of 5'-(E)-vinylphosphonate (5'-VP), a metabolically stable phosphate mimic, results in up to 20-fold improved in vitro potency and up to a threefold benefit in in vivo activity by promoting Ago2 loading and enhancing metabolic stability.


Asunto(s)
Acetilgalactosamina/química , Organofosfonatos/química , Interferencia de ARN , ARN Interferente Pequeño/química , Compuestos de Vinilo/química , Animales , Apolipoproteínas B/antagonistas & inhibidores , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Proteínas Argonautas/antagonistas & inhibidores , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Células Cultivadas , Factor IX/antagonistas & inhibidores , Factor IX/genética , Factor IX/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Lipoproteínas LDL/sangre , Ratones , Ratones Endogámicos C57BL , Organofosfonatos/farmacología , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN , Complejo Silenciador Inducido por ARN/química , Complejo Silenciador Inducido por ARN/metabolismo , Factores de Transcripción/metabolismo , Compuestos de Vinilo/farmacología
9.
J Am Chem Soc ; 136(49): 16958-61, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25434769

RESUMEN

Conjugation of small interfering RNA (siRNA) to an asialoglycoprotein receptor ligand derived from N-acetylgalactosamine (GalNAc) facilitates targeted delivery of the siRNA to hepatocytes in vitro and in vivo. The ligands derived from GalNAc are compatible with solid-phase oligonucleotide synthesis and deprotection conditions, with synthesis yields comparable to those of standard oligonucleotides. Subcutaneous (SC) administration of siRNA-GalNAc conjugates resulted in robust RNAi-mediated gene silencing in liver. Refinement of the siRNA chemistry achieved a 5-fold improvement in efficacy over the parent design in vivo with a median effective dose (ED50) of 1 mg/kg following a single dose. This enabled the SC administration of siRNA-GalNAc conjugates at therapeutically relevant doses and, importantly, at dose volumes of ≤1 mL. Chronic weekly dosing resulted in sustained dose-dependent gene silencing for over 9 months with no adverse effects in rodents. The optimally chemically modified siRNA-GalNAc conjugates are hepatotropic and long-acting and have the potential to treat a wide range of diseases involving liver-expressed genes.


Asunto(s)
Acetilgalactosamina/química , Silenciador del Gen , Hepatocitos/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Animales , Ratones , Ratones Endogámicos C57BL , Estructura Molecular
10.
RNA ; 18(3): 557-68, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22294662

RESUMEN

Since the discovery of RNA interference (RNAi), researchers have identified a variety of small interfering RNA (siRNA) structures that demonstrate the ability to silence gene expression through the classical RISC-mediated mechanism. One such structure, termed "Dicer-substrate siRNA" (dsiRNA), was proposed to have enhanced potency via RISC-mediated gene silencing, although a comprehensive comparison of canonical siRNAs and dsiRNAs remains to be described. The present study evaluates the in vitro and in vivo activities of siRNAs and dsiRNAs targeting Phosphatase and Tensin Homolog (PTEN) and Factor VII (FVII). More than 250 compounds representing both siRNA and dsiRNA structures were evaluated for silencing efficacy. Lead compounds were assessed for duration of silencing and other key parameters such as cytokine induction. We identified highly active compounds from both canonical siRNAs and 25/27 dsiRNAs. Lead compounds were comparable in potency both in vitro and in vivo as well as duration of silencing in vivo. Duplexes from both structural classes tolerated 2'-OMe chemical modifications well with respect to target silencing, although some modified dsiRNAs demonstrated reduced activity. On the other hand, dsiRNAs were more immunostimulatory as compared with the shorter siRNAs, both in vitro and in vivo. Because the dsiRNA structure does not confer any appreciable benefits in vitro or in vivo while demonstrating specific liabilities, further studies are required to support their applications in RNAi therapeutics.


Asunto(s)
Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ribonucleasa III/metabolismo , Animales , Secuencia de Bases , Factor VII/genética , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Fosfohidrolasa PTEN/genética , Complejo Silenciador Inducido por ARN/metabolismo , Ratas
11.
Blood ; 120(9): 1916-22, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22611156

RESUMEN

Anemia linked to a relative deficiency of renal erythropoietin production is a significant cause of morbidity and medical expenditures in the developed world. Recombinant erythropoietin is expensive and has been linked to excess cardiovascular events. Moreover, some patients become refractory to erythropoietin because of increased production of factors such as hepcidin. During fetal life, the liver, rather than the kidney, is the major source of erythropoietin. In the present study, we show that it is feasible to reactivate hepatic erythropoietin production and suppress hepcidin levels using systemically delivered siRNAs targeting the EglN prolyl hydroxylases specifically in the liver, leading to improved RBC production in models of anemia caused by either renal insufficiency or chronic inflammation with enhanced hepcidin production.


Asunto(s)
Eritropoyetina/deficiencia , Eritropoyetina/genética , Procolágeno-Prolina Dioxigenasa/genética , ARN Interferente Pequeño/genética , Anemia/etiología , Anemia/genética , Anemia/terapia , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Secuencia de Bases , Células Cultivadas , Eritropoyesis/genética , Eritropoyetina/metabolismo , Estudios de Factibilidad , Femenino , Hepcidinas , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Inflamación/complicaciones , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Procolágeno-Prolina Dioxigenasa/metabolismo , Interferencia de ARN , Insuficiencia Renal/complicaciones
12.
Mol Ther ; 21(8): 1570-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23799535

RESUMEN

In recent years, RNA interference (RNAi) therapeutics, most notably with lipid nanoparticle-based delivery systems, have advanced into human clinical trials. The results from these early clinical trials suggest that lipid nanoparticles (LNPs), and the novel ionizable lipids that comprise them, will be important materials in this emerging field of medicine. A persistent theme in the use of materials for biomedical applications has been the incorporation of biodegradability as a means to improve biocompatibility and/or to facilitate elimination. Therefore, the aim of this work was to further advance the LNP platform through the development of novel, next-generation lipids that combine the excellent potency of the most advanced lipids currently available with biodegradable functionality. As a representative example of this novel class of biodegradable lipids, the lipid evaluated in this work displays rapid elimination from plasma and tissues, substantially improved tolerability in preclinical studies, while maintaining in vivo potency on par with that of the most advanced lipids currently available.


Asunto(s)
Sistemas de Liberación de Medicamentos , Técnicas de Transferencia de Gen , Lípidos/química , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/genética , Animales , Línea Celular , Factor VII/genética , Factor VII/metabolismo , Silenciador del Gen , Terapia Genética , Humanos , Lípidos/farmacocinética , Macaca fascicularis , Masculino , Ratones , Nanopartículas/química , Nanopartículas/toxicidad , Interferencia de ARN , ARN Interferente Pequeño/química , Ratas
13.
Nature ; 449(7163): 745-7, 2007 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-17898712

RESUMEN

Systemic administration of synthetic small interfering RNAs (siRNAs) effectively silences hepatocyte gene expression in rodents and primates. Whether or not in vivo gene silencing by synthetic siRNA can disrupt the endogenous microRNA (miRNA) pathway remains to be addressed. Here we show that effective target-gene silencing in the mouse and hamster liver can be achieved by systemic administration of synthetic siRNA without any demonstrable effect on miRNA levels or activity. Indeed, siRNA targeting two hepatocyte-specific genes (apolipoprotein B and factor VII) that achieved efficient (approximately 80%) silencing of messenger RNA transcripts and a third irrelevant siRNA control were administered to mice without significant changes in the levels of three hepatocyte-expressed miRNAs (miR-122, miR-16 and let-7a) or an effect on miRNA activity. Moreover, multiple administrations of an siRNA targeting the hepatocyte-expressed gene Scap in hamsters achieved long-term mRNA silencing without significant changes in miR-122 levels. This study advances the use of siRNAs as safe and effective tools to silence gene transcripts in animal studies, and supports the continued advancement of RNA interference therapeutics using synthetic siRNA.


Asunto(s)
MicroARNs/genética , MicroARNs/metabolismo , Interferencia de ARN , Animales , Northern Blotting , Cricetinae , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , MicroARNs/administración & dosificación , MicroARNs/farmacología , Interferencia de ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(5): 1864-9, 2010 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-20080679

RESUMEN

Significant effort has been applied to discover and develop vehicles which can guide small interfering RNAs (siRNA) through the many barriers guarding the interior of target cells. While studies have demonstrated the potential of gene silencing in vivo, improvements in delivery efficacy are required to fulfill the broadest potential of RNA interference therapeutics. Through the combinatorial synthesis and screening of a different class of materials, a formulation has been identified that enables siRNA-directed liver gene silencing in mice at doses below 0.01 mg/kg. This formulation was also shown to specifically inhibit expression of five hepatic genes simultaneously, after a single injection. The potential of this formulation was further validated in nonhuman primates, where high levels of knockdown of the clinically relevant gene transthyretin was observed at doses as low as 0.03 mg/kg. To our knowledge, this formulation facilitates gene silencing at orders-of-magnitude lower doses than required by any previously described siRNA liver delivery system.


Asunto(s)
Materiales Biocompatibles/química , Silenciador del Gen , Lípidos/química , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Animales , Materiales Biocompatibles/síntesis química , Sistemas de Liberación de Medicamentos , Factor VII/antagonistas & inhibidores , Factor VII/genética , Células HeLa , Hepatocitos/metabolismo , Humanos , Lípidos/síntesis química , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Interferencia de ARN
15.
Nature ; 441(7089): 111-4, 2006 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-16565705

RESUMEN

The opportunity to harness the RNA interference (RNAi) pathway to silence disease-causing genes holds great promise for the development of therapeutics directed against targets that are otherwise not addressable with current medicines. Although there are numerous examples of in vivo silencing of target genes after local delivery of small interfering RNAs (siRNAs), there remain only a few reports of RNAi-mediated silencing in response to systemic delivery of siRNA, and there are no reports of systemic efficacy in non-rodent species. Here we show that siRNAs, when delivered systemically in a liposomal formulation, can silence the disease target apolipoprotein B (ApoB) in non-human primates. APOB-specific siRNAs were encapsulated in stable nucleic acid lipid particles (SNALP) and administered by intravenous injection to cynomolgus monkeys at doses of 1 or 2.5 mg kg(-1). A single siRNA injection resulted in dose-dependent silencing of APOB messenger RNA expression in the liver 48 h after administration, with maximal silencing of >90%. This silencing effect occurred as a result of APOB mRNA cleavage at precisely the site predicted for the RNAi mechanism. Significant reductions in ApoB protein, serum cholesterol and low-density lipoprotein levels were observed as early as 24 h after treatment and lasted for 11 days at the highest siRNA dose, thus demonstrating an immediate, potent and lasting biological effect of siRNA treatment. Our findings show clinically relevant RNAi-mediated gene silencing in non-human primates, supporting RNAi therapeutics as a potential new class of drugs.


Asunto(s)
Primates/genética , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Animales , Apolipoproteínas B/deficiencia , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
16.
Mol Ther ; 19(12): 2186-200, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21971424

RESUMEN

Lipid nanoparticles (LNPs) are currently the most effective in vivo delivery systems for silencing target genes in hepatocytes employing small interfering RNA. Antigen-presenting cells (APCs) are also potential targets for LNP siRNA. We examined the uptake, intracellular trafficking, and gene silencing potency in primary bone marrow macrophages (bmMΦ) and dendritic cells of siRNA formulated in LNPs containing four different ionizable cationic lipids namely DLinDAP, DLinDMA, DLinK-DMA, and DLinKC2-DMA. LNPs containing DLinKC2-DMA were the most potent formulations as determined by their ability to inhibit the production of GAPDH target protein. Also, LNPs containing DLinKC2-DMA were the most potent intracellular delivery agents as indicated by confocal studies of endosomal versus cytoplamic siRNA location using fluorescently labeled siRNA. DLinK-DMA and DLinKC2-DMA formulations exhibited improved gene silencing potencies relative to DLinDMA but were less toxic. In vivo results showed that LNP siRNA systems containing DLinKC2-DMA are effective agents for silencing GAPDH in APCs in the spleen and peritoneal cavity following systemic administration. Gene silencing in APCs was RNAi mediated and the use of larger LNPs resulted in substantially reduced hepatocyte silencing, while similar efficacy was maintained in APCs. These results are discussed with regard to the potential of LNP siRNA formulations to treat immunologically mediated diseases.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Cationes/química , Silenciador del Gen , Gliceraldehído-3-Fosfato Deshidrogenasas/antagonistas & inhibidores , Lípidos/administración & dosificación , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Western Blotting , Médula Ósea , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Endocitosis , Citometría de Flujo , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Antígenos Comunes de Leucocito/antagonistas & inhibidores , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Liposomas , Hígado/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Interferencia de ARN , ARN Interferente Pequeño/genética
17.
Angew Chem Int Ed Engl ; 51(34): 8529-33, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22782619

RESUMEN

Special (lipid) delivery: The role of the ionizable lipid pK(a) in the in vivo delivery of siRNA by lipid nanoparticles has been studied with a large number of head group modifications to the lipids. A tight correlation between the lipid pK(a) value and silencing of the mouse FVII gene (FVII ED(50) ) was found, with an optimal pK(a) range of 6.2-6.5. The most potent cationic lipid from this study has ED(50) levels around 0.005 mg kg(-1) in mice and less than 0.03 mg kg(-1) in non-human primates.


Asunto(s)
Silenciador del Gen , Lípidos/administración & dosificación , Hígado/fisiología , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Aminas/química , Animales , Femenino , Terapia Genética/métodos , Humanos , Cinética , Lípidos/química , Liposomas/administración & dosificación , Liposomas/química , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , ARN Interferente Pequeño/química
18.
Mol Ther ; 18(7): 1357-64, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20461061

RESUMEN

Lipid nanoparticles (LNPs) have proven to be highly efficient carriers of short-interfering RNAs (siRNAs) to hepatocytes in vivo; however, the precise mechanism by which this efficient delivery occurs has yet to be elucidated. We found that apolipoprotein E (apoE), which plays a major role in the clearance and hepatocellular uptake of physiological lipoproteins, also acts as an endogenous targeting ligand for ionizable LNPs (iLNPs), but not cationic LNPs (cLNPs). The role of apoE was investigated using both in vitro studies employing recombinant apoE and in vivo studies in wild-type and apoE(-/-) mice. Receptor dependence was explored in vitro and in vivo using low-density lipoprotein receptor (LDLR(-/-))-deficient mice. As an alternative to endogenous apoE-based targeting, we developed a targeting approach using an exogenous ligand containing a multivalent N-acetylgalactosamine (GalNAc)-cluster, which binds with high affinity to the asialoglycoprotein receptor (ASGPR) expressed on hepatocytes. Both apoE-based endogenous and GalNAc-based exogenous targeting appear to be highly effective strategies for the delivery of iLNPs to liver.


Asunto(s)
Interferencia de ARN/fisiología , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Receptor de Asialoglicoproteína/metabolismo , Femenino , Células HeLa , Hepatocitos/metabolismo , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Receptores de LDL/genética , Receptores de LDL/metabolismo
19.
Proc Natl Acad Sci U S A ; 105(33): 11915-20, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18695239

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates low density lipoprotein receptor (LDLR) protein levels and function. Loss of PCSK9 increases LDLR levels in liver and reduces plasma LDL cholesterol (LDLc), whereas excess PCSK9 activity decreases liver LDLR levels and increases plasma LDLc. Here, we have developed active, cross-species, small interfering RNAs (siRNAs) capable of targeting murine, rat, nonhuman primate (NHP), and human PCSK9. For in vivo studies, PCSK9 and control siRNAs were formulated in a lipidoid nanoparticle (LNP). Liver-specific siRNA silencing of PCSK9 in mice and rats reduced PCSK9 mRNA levels by 50-70%. The reduction in PCSK9 transcript was associated with up to a 60% reduction in plasma cholesterol concentrations. These effects were shown to be mediated by an RNAi mechanism, using 5'-RACE. In transgenic mice expressing human PCSK9, siRNAs silenced the human PCSK9 transcript by >70% and significantly reduced PCSK9 plasma protein levels. In NHP, a single dose of siRNA targeting PCSK9 resulted in a rapid, durable, and reversible lowering of plasma PCSK9, apolipoprotein B, and LDLc, without measurable effects on either HDL cholesterol (HDLc) or triglycerides (TGs). The effects of PCSK9 silencing lasted for 3 weeks after a single bolus i.v. administration. These results validate PCSK9 targeting with RNAi therapeutics as an approach to specifically lower LDLc, paving the way for the development of PCSK9-lowering agents as a future strategy for treatment of hypercholesterolemia.


Asunto(s)
LDL-Colesterol/sangre , Primates/metabolismo , ARN Interferente Pequeño/genética , Serina Endopeptidasas/metabolismo , Animales , Humanos , Hígado/enzimología , Ratones , Ratones Noqueados , Estructura Molecular , Primates/genética , ARN Mensajero/genética , Ratas , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Factores de Tiempo
20.
J Neurochem ; 115(6): 1434-44, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21044072

RESUMEN

Brain injury caused by ischemia is a major cause of human mortality and physical/cognitive disability worldwide. Experimentally, brain ischemia can be induced surgically by permanent middle cerebral artery occlusion. Using this model, we studied the influence of transthyretin in ischemic stroke. Transthyretin (TTR) is normally responsible for the transport of thyroid hormones and retinol in the blood and CSF. We found that TTR null mice (TTR(-/-) ) did not show significant differences in cortical infarction 24 h after permanent middle cerebral artery occlusion compared with TTR(+/+) control littermates. However, TTR null mice, heterozygous for the heat-shock transcription factor 1 (TTR(-/-) HSF1(+/-) mice), which compromised the stress response, showed a significant increase in cortical infarction, cerebral edema and the microglial-leukocyte response compared with TTR(+/+) HSF1(+/-) mice. Unexpectedly, we observed novel TTR distribution throughout the infarct, localized to disintegrated ß-tubulin III(+) neurons and cell debris. Specific elimination of TTR synthesis in the liver by RNAi had no effect on TTR distribution in the infarct, indicating that the observed TTR infiltration derived from CSF and not from the serum. This finding is corroborated by results from 'in situ' hybridization and real time PCR that excluded the presence of transthyretin mRNA in the infarct and peri-infarct areas. Our data suggest that in conditions of a compromised heat-shock response, CSF TTR contributes to control neuronal cell death, edema and inflammation, thereby influencing the survival of endangered neurons in cerebral ischemia.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/líquido cefalorraquídeo , Prealbúmina/líquido cefalorraquídeo , Animales , Biomarcadores/líquido cefalorraquídeo , Isquemia Encefálica/líquido cefalorraquídeo , Muerte Celular/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA