Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Am Chem Soc ; 143(9): 3416-3429, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33626278

RESUMO

The extra hepatic delivery of antisense oligonucleotides (ASOs) remains a challenge and hampers the widespread application of this powerful class of therapeutic agents. In that regard, pancreatic beta cells are a particularly attractive but challenging cell type because of their pivotal role in diabetes and the fact that they are refractory to uptake of unconjugated ASOs. To circumvent this, we have expanded our understanding of the structure activity relationship of ASOs conjugated to Glucagon Like Peptide 1 Receptor (GLP1R) agonist peptide ligands. We demonstrate the key role of the linker chemistry and its optimization to design maleimide based conjugates with improved in vivo efficacy. In addition, truncation studies and scoping of a diverse set of GLP1R agonists proved fruitful to identify additional targeting ligands efficacious in vivo including native hGLP1(7-36)NH2. Variation of the carrier peptide also shed some light on the dramatic impact of subtle sequence differences on the corresponding ASO conjugate performance in vivo, an area which clearly warrant further investigations. We have confirmed the remarkable potential of GLP1R agonist conjugation for the delivery of ASOs to pancreatic beta cell by effectively knocking down islet amyloid polypeptide (IAPP) mRNA, a potential proapoptotic target, in mice.


Assuntos
Portadores de Fármacos/química , Receptor do Peptídeo Semelhante ao Glucagon 1/química , Células Secretoras de Insulina/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Peptídeos/química , Sequência de Aminoácidos , Animais , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Células HEK293 , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Camundongos Endogâmicos C57BL , Estrutura Molecular , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade
2.
Nucleic Acids Res ; 47(3): 1070-1081, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30535404

RESUMO

Efficient delivery of therapeutic RNA beyond the liver is the fundamental obstacle preventing its clinical utility. Lipid conjugation increases plasma half-life and enhances tissue accumulation and cellular uptake of small interfering RNAs (siRNAs). However, the mechanism relating lipid hydrophobicity, structure, and siRNA pharmacokinetics is unclear. Here, using a diverse panel of biologically occurring lipids, we show that lipid conjugation directly modulates siRNA hydrophobicity. When administered in vivo, highly hydrophobic lipid-siRNAs preferentially and spontaneously associate with circulating low-density lipoprotein (LDL), while less lipophilic lipid-siRNAs bind to high-density lipoprotein (HDL). Lipid-siRNAs are targeted to lipoprotein receptor-enriched tissues, eliciting significant mRNA silencing in liver (65%), adrenal gland (37%), ovary (35%), and kidney (78%). Interestingly, siRNA internalization may not be completely driven by lipoprotein endocytosis, but the extent of siRNA phosphorothioate modifications may also be a factor. Although biomimetic lipoprotein nanoparticles have been explored for the enhancement of siRNA delivery, our findings suggest that hydrophobic modifications can be leveraged to incorporate therapeutic siRNA into endogenous lipid transport pathways without the requirement for synthetic formulation.


Assuntos
Lipídeos/química , RNA Interferente Pequeno/farmacocinética , Animais , Proteínas Sanguíneas/metabolismo , Feminino , Células HeLa , Hepatócitos/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Rim/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Interferência de RNA , RNA Interferente Pequeno/síntese química , RNA Interferente Pequeno/química , Receptores de LDL/metabolismo , Distribuição Tecidual
3.
Nucleic Acids Res ; 46(5): 2185-2196, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29432571

RESUMO

Small interfering RNA (siRNA)-based drugs require chemical modifications or formulation to promote stability, minimize innate immunity, and enable delivery to target tissues. Partially modified siRNAs (up to 70% of the nucleotides) provide significant stabilization in vitro and are commercially available; thus are commonly used to evaluate efficacy of bio-conjugates for in vivo delivery. In contrast, most clinically-advanced non-formulated compounds, using conjugation as a delivery strategy, are fully chemically modified (100% of nucleotides). Here, we compare partially and fully chemically modified siRNAs in conjugate mediated delivery. We show that fully modified siRNAs are retained at 100x greater levels in various tissues, independently of the nature of the conjugate or siRNA sequence, and support productive mRNA silencing. Thus, fully chemically stabilized siRNAs may provide a better platform to identify novel moieties (peptides, aptamers, small molecules) for targeted RNAi delivery.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Interferência de RNA , Processamento Pós-Transcricional do RNA , RNA Interferente Pequeno/genética , Animais , Aptâmeros de Nucleotídeos/química , Células Cultivadas , Feminino , Vetores Genéticos/genética , Células HeLa , Humanos , Lipídeos/química , Camundongos Endogâmicos C57BL , Peptídeos/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética , Distribuição Tecidual
4.
Mol Ther ; 26(6): 1520-1528, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29699940

RESUMO

Small extracellular vesicles (sEVs) show promise as natural nano-devices for delivery of therapeutic RNA, but efficient loading of therapeutic RNA remains a challenge. We have recently shown that the attachment of cholesterol to small interfering RNAs (siRNAs) enables efficient and productive loading into sEVs. Here, we systematically explore the ability of lipid conjugates-fatty acids, sterols, and vitamins-to load siRNAs into sEVs and support gene silencing in primary neurons. Hydrophobicity of the conjugated siRNAs defined loading efficiency and the silencing activity of siRNA-sEVs complexes. Vitamin-E-conjugated siRNA supported the best loading into sEVs and productive RNA delivery to neurons.


Assuntos
Vesículas Extracelulares/química , Lipídeos/química , RNA Interferente Pequeno/química , Células Cultivadas , Inativação Gênica/fisiologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Interferência de RNA
5.
Mol Ther ; 26(11): 2580-2591, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30143435

RESUMO

Effective transvascular delivery of therapeutic oligonucleotides to the brain presents a major hurdle to the development of gene silencing technologies for treatment of genetically defined neurological disorders. Distribution to the brain after systemic administrations is hampered by the low permeability of the blood-brain barrier (BBB) and the rapid clearance kinetics of these drugs from the blood. Here we show that transient osmotic disruption of the BBB enables transvascular delivery of hydrophobically modified small interfering RNA (hsiRNA) to the rat brain. Intracarotid administration of 25% mannitol and hsiRNA conjugated to phosphocholine-docosahexanoic acid (PC-DHA) resulted in broad ipsilateral distribution of PC-DHA-hsiRNAs in the brain. PC-DHA conjugation enables hsiRNA retention in the parenchyma proximal to the brain vasculature and enabled active internalization by neurons and astrocytes. Moreover, transvascular delivery of PC-DHA-hsiRNAs effected Htt mRNA silencing in the striatum (55%), hippocampus (51%), somatosensory cortex (52%), motor cortex (37%), and thalamus (33%) 1 week after administration. Aside from mild gliosis induced by osmotic disruption of the BBB, transvascular delivery of PC-DHA-hsiRNAs was not associated with neurotoxicity. Together, these findings provide proof-of-concept that temporary disruption of the BBB is an effective strategy for the delivery of therapeutic oligonucleotides to the brain.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Proteína Huntingtina/genética , Neurônios/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Barreira Hematoencefálica/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Artérias Carótidas/fisiologia , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/química , Inativação Gênica , Terapia Genética/métodos , Humanos , Proteína Huntingtina/antagonistas & inibidores , Interações Hidrofóbicas e Hidrofílicas , Manitol/administração & dosagem , Neurônios/patologia , Fosforilcolina/administração & dosagem , Fosforilcolina/química , RNA Interferente Pequeno/química , Ratos
6.
Molecules ; 24(2)2019 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-30634486

RESUMO

We recently reported that (E)-5'-vinylphosphonate (5'-VP) is a metabolically-stable phosphate mimic for siRNA and demonstrated that 5'-VP improves the potency of the fully modified siRNAs in vivo. Here, we report an alternative synthesis of 5'-VP modified guide strand using S-pivaloyl-2-thioethyl (tBu-SATE) protecting group. The tBu-SATE group is readily removed during the final cleavage of the oligonucleotide from the solid support and providing a more convenient route for the synthesis of siRNA guide strand carrying a 5'-vinylphosphonate.


Assuntos
Organofosfonatos/química , RNA Interferente Pequeno/síntese química , Compostos de Vinila/química , Estrutura Molecular , RNA Interferente Pequeno/química
7.
Bioconjug Chem ; 28(6): 1758-1766, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28462988

RESUMO

Ligand-conjugated siRNAs have the potential to achieve targeted delivery and efficient silencing in neurons following local administration in the central nervous system (CNS). We recently described the activity and safety profile of a docosahexaenoic acid (DHA)-conjugated, hydrophobic siRNA (DHA-hsiRNA) targeting Huntingtin (Htt) mRNA in mouse brain. Here, we report the synthesis of an amide-modified, phosphocholine-containing DHA-hsiRNA conjugate (PC-DHA-hsiRNA), which closely resembles the endogenously esterified biological structure of DHA. We hypothesized that this modification may enhance neuronal delivery in vivo. We demonstrate that PC-DHA-hsiRNA silences Htt in mouse primary cortical neurons and astrocytes. After intrastriatal delivery, Htt-targeting PC-DHA-hsiRNA induces ∼80% mRNA silencing and 71% protein silencing after 1 week. However, PC-DHA-hsiRNA did not substantially outperform DHA-hsiRNA under the conditions tested. Moreover, at the highest locally administered dose (4 nmol, 50 µg), we observe evidence of PC-DHA-hsiRNA-mediated reactive astrogliosis. Lipophilic ligand conjugation enables siRNA delivery to neural tissues, but rational design of functional, nontoxic siRNA conjugates for CNS delivery remains challenging.


Assuntos
Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Tecido Parenquimatoso/metabolismo , RNA Interferente Pequeno/síntese química , Animais , Encéfalo/patologia , Ácidos Docosa-Hexaenoicos/química , Estabilidade de Medicamentos , Inativação Gênica , Proteína Huntingtina/genética , Camundongos , Fosforilcolina/química , Interferência de RNA , RNA Mensageiro , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Serina/química , Resultado do Tratamento
8.
Nucleic Acids Res ; 43(18): 8664-72, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26400165

RESUMO

One of the major obstacles to the pharmaceutical success of oligonucleotide therapeutics (ONTs) is efficient delivery from the point of injection to the intracellular setting where functional gene silencing occurs. In particular, a significant fraction of internalized ONTs are nonproductively sequestered in endo-lysosomal compartments. Here, we describe a two-step, robust assay for high-throughput de novo detection of small bioactive molecules that enhance cellular uptake, endosomal escape, and efficacy of ONTs. Using this assay, we screened the LOPAC (Sigma-Aldrich) Library of Pharmacologically Active Compounds and discovered that Guanabenz acetate (Wytensin™), an FDA-approved drug formerly used as an antihypertensive agent, is capable of markedly increasing the cellular internalization and target mRNA silencing of hydrophobically modified siRNAs (hsiRNAs), yielding a ∼100-fold decrease in hsiRNA IC50 (from 132 nM to 2.4 nM). This is one of the first descriptions of a high-throughput small-molecule screen to identify novel chemistries that specifically enhance siRNA intracellular efficacy, and can be applied toward expansion of the chemical diversity of ONTs.


Assuntos
Guanabenzo/farmacologia , Interferência de RNA , RNA Interferente Pequeno/química , RNA Interferente Pequeno/metabolismo , Transporte Biológico , Guanabenzo/química , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Interações Hidrofóbicas e Hidrofílicas , RNA Interferente Pequeno/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
9.
Nat Chem Biol ; 8(3): 301-10, 2012 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-22306580

RESUMO

Guanine-rich DNA sequences that can adopt non-Watson-Crick structures in vitro are prevalent in the human genome. Whether such structures normally exist in mammalian cells has, however, been the subject of active research for decades. Here we show that the G-quadruplex-interacting drug pyridostatin promotes growth arrest in human cancer cells by inducing replication- and transcription-dependent DNA damage. A chromatin immunoprecipitation sequencing analysis of the DNA damage marker γH2AX provided the genome-wide distribution of pyridostatin-induced sites of damage and revealed that pyridostatin targets gene bodies containing clusters of sequences with a propensity for G-quadruplex formation. As a result, pyridostatin modulated the expression of these genes, including the proto-oncogene SRC. We observed that pyridostatin reduced SRC protein abundance and SRC-dependent cellular motility in human breast cancer cells, validating SRC as a target of this drug. Our unbiased approach to define genomic sites of action for a drug establishes a framework for discovering functional DNA-drug interactions.


Assuntos
Aminoquinolinas/farmacologia , Antineoplásicos/farmacologia , Dano ao DNA , DNA/química , DNA/efeitos dos fármacos , Ácidos Picolínicos/farmacologia , Aminoquinolinas/síntese química , Aminoquinolinas/química , Antineoplásicos/síntese química , Antineoplásicos/química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA/genética , Ensaios de Seleção de Medicamentos Antitumorais , Quadruplex G/efeitos dos fármacos , Humanos , Peso Molecular , Ácidos Picolínicos/síntese química , Ácidos Picolínicos/química , Proto-Oncogene Mas , Relação Estrutura-Atividade , Células Tumorais Cultivadas
10.
Nucleic Acids Res ; 40(4): 1499-508, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22021377

RESUMO

SP1 is a ubiquitous transcription factor that is involved in the regulation of various house-keeping genes. It is known that it acts by binding to a double-stranded consensus motif. Here, we have discovered that SP1 binds also to a non-canonical DNA structure, a G-quadruplex, with high affinity. In particular, we have studied the SP1 binding site within the promoter region of the c-KIT oncogene and found that this site can fold into an anti-parallel two-tetrad G-quadruplex. SP1 pull-down experiments from cellular extracts, together with biophysical binding assays revealed that SP1 has a comparable binding affinity for this G-quadruplex structure and the canonical SP1 duplex sequence. Using SP1 ChIP-on-chip data sets, we have also found that 87% of SP1 binding sites overlap with G-quadruplex forming sequences. Furthermore, while many of these immuoprecipitated sequences (36%) even lack the minimal SP1 consensus motif, 5'-GGGCGG-3', we have shown that 77% of them are putative G-quadruplexes. Collectively, these data suggest that SP1 is able to bind both, canonical SP1 duplex DNA as well as G-quadruplex structures in vitro and we hypothesize that both types of interactions may occur in cells.


Assuntos
Quadruplex G , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/metabolismo , Sítios de Ligação , Genoma , Células HeLa , Humanos , Motivos de Nucleotídeos , Proteínas Proto-Oncogênicas c-kit/genética
11.
Elife ; 132024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38319152

RESUMO

A self-cleaving ribozyme that maps to an intron of the cytoplasmic polyadenylation element-binding protein 3 (Cpeb3) gene is thought to play a role in human episodic memory, but the underlying mechanisms mediating this effect are not known. We tested the activity of the murine sequence and found that the ribozyme's self-scission half-life matches the time it takes an RNA polymerase to reach the immediate downstream exon, suggesting that the ribozyme-dependent intron cleavage is tuned to co-transcriptional splicing of the Cpeb3 mRNA. Our studies also reveal that the murine ribozyme modulates maturation of its harboring mRNA in both cultured cortical neurons and the hippocampus: inhibition of the ribozyme using an antisense oligonucleotide leads to increased CPEB3 protein expression, which enhances polyadenylation and translation of localized plasticity-related target mRNAs, and subsequently strengthens hippocampal-dependent long-term memory. These findings reveal a previously unknown role for self-cleaving ribozyme activity in regulating experience-induced co-transcriptional and local translational processes required for learning and memory.


Stored within DNA are the instructions cells need to make proteins. In order for proteins to get made, the region of DNA that codes for the desired protein (known as the gene) must first be copied into a molecule called messenger RNA (or mRNA for short). Once transcribed, the mRNA undergoes further modifications, including removing redundant segments known as introns. It then travels to molecular machines that translate its genetic sequence into the building blocks of the protein. Following transcription, some RNAs can fold into catalytic segments known as self-cleaving ribozymes which promote the scission of their own genetic sequence. One such ribozyme resides in the intron of a gene for CPEB3, a protein which adds a poly(A) tail to various mRNAs, including some involved in learning and memory. Although this ribozyme is found in most mammals, its biological role is poorly understood. Previous studies suggested that the ribozyme cleaves itself at the same time as the mRNA for CPEB3 is transcribed. This led Chen et al. to hypothesize that the rate at which these two events occur impacts the amount of CPEB3 produced, resulting in changes in memory and learning. If the ribozyme cleaves quickly, the intron is disrupted and may not be properly removed, leading to less CPEB3 being made. However, if the ribozyme is inhibited, the intron remains intact and is efficiently excised, resulting in higher levels of CPEB3 protein. To test how the ribozyme impacts CPEB3 production, Chen et al. inhibited the enzyme from cutting itself with antisense oligonucleotides (ASOs). The ASOs were applied to in vitro transcription systems, neurons cultured in the laboratory and the brains of living mice in an area called the hippocampus. The in vitro and cell culture experiments led to higher levels of CPEB3 protein and the addition of more poly(A) tails to mRNAs involved in neuron communication. Injection of the ASOs into the brains of mice had the same effect, and also improved their memory and learning. The findings of Chen et al. show a new mechanism for controlling protein production, and suggest that ASOs could be used to increase the levels of CPEB3 and modulate neuronal activity. This is the first time a biological role for a self-cleaving ribozyme in mammals has been identified, and the approach used could be applied to investigate the function of two other self-cleaving ribozymes located in introns in humans.


Assuntos
RNA Catalítico , Camundongos , Humanos , Animais , RNA Catalítico/genética , RNA Catalítico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Poliadenilação , Memória de Longo Prazo , Neurônios/metabolismo , Proteínas de Ligação a RNA/metabolismo
12.
bioRxiv ; 2023 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-37333407

RESUMO

A self-cleaving ribozyme that maps to an intron of the cytoplasmic polyadenylation element binding protein 3 (CPEB3) gene is thought to play a role in human episodic memory, but the underlying mechanisms mediating this effect are not known. We tested the activity of the murine sequence and found that the ribozyme's self-scission half-life matches the time it takes an RNA polymerase to reach the immediate downstream exon, suggesting that the ribozyme-dependent intron cleavage is tuned to co-transcriptional splicing of the CPEB3 mRNA. Our studies also reveal that the murine ribozyme modulates maturation of its harboring mRNA in both cultured cortical neurons and the hippocampus: inhibition of the ribozyme using an antisense oligonucleotide leads to increased CPEB3 protein expression, which enhances polyadenylation and translation of localized plasticity-related target mRNAs, and subsequently strengthens hippocampal-dependent long-term memory. These findings reveal a previously unknown role for self-cleaving ribozyme activity in regulating experience-induced co-transcriptional and local translational processes required for learning and memory.

13.
Nucleic Acid Ther ; 32(4): 300-311, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35612431

RESUMO

We evaluated the potential of AGTR1, the principal receptor for angiotensin II (Ang II) and a member of the G protein-coupled receptor family, for targeted delivery of antisense oligonucleotides (ASOs) in cells and tissues with abundant AGTR1 expression. Ang II peptide ASO conjugates maintained robust AGTR1 signaling and receptor internalization when ASO was placed at the N-terminus of the peptide, but not at C-terminus. Conjugation of Ang II peptide improved ASO potency up to 12- to 17-fold in AGTR1-expressing cells. Additionally, evaluation of Ang II conjugates in cells lacking AGTR1 revealed no enhancement of ASO potency. Ang II peptide conjugation improves potency of ASO in mouse heart, adrenal, and adipose tissues. The data presented in this report add to a growing list of approaches for improving ASO potency in extrahepatic tissues.


Assuntos
Oligonucleotídeos Antissenso , Receptor Tipo 1 de Angiotensina , Animais , Camundongos , Oligonucleotídeos Antissenso/farmacologia , Receptor Tipo 1 de Angiotensina/genética , Transdução de Sinais
14.
Trends Pharmacol Sci ; 42(7): 588-604, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34020790

RESUMO

Synthetic therapeutic oligonucleotides (STO) represent the third bonafide platform for drug discovery in the pharmaceutical industry after small molecule and protein therapeutics. So far, thirteen STOs have been approved by regulatory agencies and over one hundred of them are in different stages of clinical trials. STOs hybridize to their target RNA or DNA in cells via Watson-Crick base pairing to exert their pharmacological effects. This unique class of therapeutic agents has the potential to target genes and gene products that are considered undruggable by other therapeutic platforms. However, STOs must overcome several extracellular and intracellular obstacles to interact with their biological RNA targets inside cells. These obstacles include degradation by extracellular nucleases, scavenging by the reticuloendothelial system, filtration by the kidney, traversing the capillary endothelium to access the tissue interstitium, cell-surface receptor-mediated endocytic uptake, and escape from endolysosomal compartments to access the nuclear and/or cytoplasmic compartments where their targets reside. In this review, we present the recent advances in this field with a specific focus on antisense oligonucleotides (ASOs) and siRNA therapeutics.


Assuntos
Oligonucleotídeos Antissenso , Oligonucleotídeos , Descoberta de Drogas , RNA Interferente Pequeno
15.
J Med Chem ; 63(15): 8471-8484, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32677436

RESUMO

Despite recent advances, targeted delivery of therapeutic oligonucleotide to extra-hepatic tissues continues to be a challenging endeavor and efficient ligand-receptor systems need to be identified. To determine the feasibility of using neurotensin to improve the productive uptake of antisense oligonucleotides (ASO), we synthesized neurotensin-ASO conjugates and evaluated their cellular uptake and activity in cells and in mice. We performed a comprehensive structure-activity relationship study of the conjugates and determined the influence of ASO charge, ASO length, peptide charge, linker chemistry and ligand identity on receptor binding and internalization. We identified a modified neurotensin peptide capable of improving the cellular uptake and activity of gapmer ASOs in sortilin expressing cells (sixfold) and in spinal cord in mice (twofold). Neurotensin conjugation also improved the potency of morpholino ASO designed to correct splicing of survival motor neuron pre-mRNA in the cortex and striatum after intracerebroventricular injection. Neurotensin-mediated targeted delivery represents a possible approach for enhancing the potency of ASOs with diverse nucleic acid modifications.


Assuntos
Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Neurotensina/química , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacocinética , Animais , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Morfolinos/administração & dosagem , Morfolinos/química , Morfolinos/farmacocinética , Oligonucleotídeos Antissenso/química
16.
J Org Chem ; 74(15): 5211-8, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19518105

RESUMO

We report the preparation and solution study of a set of template-assembled synthetic G-quartets (TASQs) bound to different cations. These G-quartet baskets effectively extract cations of different sizes and valencies. They form isolated G-quartets with small cations such as Na+ and Sr(2+), and dimeric assemblies with larger cations such as Cs+. Their structures were determined by using (1)H NMR spectroscopy, and their sizes were evaluated by using a series of pulsed-field gradient NMR experiments. The effect of anion has been studied, and the cation selectivities have been investigated by a series of competition experiments.


Assuntos
Césio/química , Quadruplex G , Sódio/química , Estrôncio/química , Cátions/química , Espectroscopia de Ressonância Magnética , Tamanho da Partícula
19.
Nucleic Acid Ther ; 27(6): 323-334, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29022758

RESUMO

Therapeutic oligonucleotides, such as small interfering RNAs (siRNAs), hold great promise for the treatment of incurable genetically defined disorders by targeting cognate toxic gene products for degradation. To achieve meaningful tissue distribution and efficacy in vivo, siRNAs must be conjugated or formulated. Clear understanding of the pharmacokinetic (PK)/pharmacodynamic behavior of these compounds is necessary to optimize and characterize the performance of therapeutic oligonucleotides in vivo. In this study, we describe a simple and reproducible methodology for the evaluation of in vivo blood/plasma PK profiles and tissue distribution of oligonucleotides. The method is based on serial blood microsampling from the saphenous vein, coupled to peptide nucleic acid hybridization assay for quantification of guide strands. Performed with minimal number of animals, this method allowed unequivocal detection and sensitive quantification without the need for amplification, or further modification of the oligonucleotides. Using this methodology, we compared plasma clearances and tissue distribution profiles of two different hydrophobically modified siRNAs (hsiRNAs). Notably, cholesterol-hsiRNA presented slow plasma clearances and mainly accumulated in the liver, whereas, phosphocholine-docosahexaenoic acid-hsiRNA was rapidly cleared from the plasma and preferably accumulated in the kidney. These data suggest that the PK/biodistribution profiles of modified hsiRNAs are determined by the chemical nature of the conjugate. Importantly, the method described in this study constitutes a simple platform to conduct pilot assessments of the basic clearance and tissue distribution profiles, which can be broadly applied for evaluation of new chemical variants of siRNAs and micro-RNAs.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Hibridização de Ácido Nucleico , Oligonucleotídeos/farmacocinética , RNA Interferente Pequeno/farmacocinética , Animais , Colesterol/sangue , Colesterol/química , Ácidos Docosa-Hexaenoicos/sangue , Ácidos Docosa-Hexaenoicos/química , Feminino , Rim/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/sangue , Ácidos Nucleicos Peptídicos/análise , Fosforilcolina/sangue , Fosforilcolina/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/sangue , Distribuição Tecidual
20.
Mol Ther Nucleic Acids ; 5(8): e344, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27504598

RESUMO

The use of siRNA-based therapies for the treatment of neurodegenerative disease requires efficient, nontoxic distribution to the affected brain parenchyma, notably the striatum and cortex. Here, we describe the synthesis and activity of a fully chemically modified siRNA that is directly conjugated to docosahexaenoic acid (DHA), the most abundant polyunsaturated fatty acid in the mammalian brain. DHA conjugation enables enhanced siRNA retention throughout both the ipsilateral striatum and cortex following a single, intrastriatal injection (ranging from 6-60 µg). Within these tissues, DHA conjugation promotes internalization by both neurons and astrocytes. We demonstrate efficient and specific silencing of Huntingtin mRNA expression in both the ipsilateral striatum (up to 73%) and cortex (up to 51%) after 1 week. Moreover, following a bilateral intrastriatal injection (60 µg), we achieve up to 80% silencing of a secondary target, Cyclophilin B, at both the mRNA and protein level. Importantly, DHA-hsiRNAs do not induce neural cell death or measurable innate immune activation following administration of concentrations over 20 times above the efficacious dose. Thus, DHA conjugation is a novel strategy for improving siRNA activity in mouse brain, with potential to act as a new therapeutic platform for the treatment of neurodegenerative disorders.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA