Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 28(3): 396-406, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30281092

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by loss of dystrophin protein, leading to progressive muscle weakness and premature death due to respiratory and/or cardiac complications. Cardiac involvement is characterized by progressive dilated cardiomyopathy, decreased fractional shortening and metabolic dysfunction involving reduced metabolism of fatty acids-the major cardiac metabolic substrate. Several mouse models have been developed to study molecular and pathological consequences of dystrophin deficiency, but do not recapitulate all aspects of human disease pathology and exhibit a mild cardiac phenotype. Here we demonstrate that Cmah (cytidine monophosphate-sialic acid hydroxylase)-deficient mdx mice (Cmah-/-;mdx) have an accelerated cardiac phenotype compared to the established mdx model. Cmah-/-;mdx mice display earlier functional deterioration, specifically a reduction in right ventricle (RV) ejection fraction and stroke volume (SV) at 12 weeks of age and decreased left ventricle diastolic volume with subsequent reduced SV compared to mdx mice by 24 weeks. They further show earlier elevation of cardiac damage markers for fibrosis (Ctgf), oxidative damage (Nox4) and haemodynamic load (Nppa). Cardiac metabolic substrate requirement was assessed using hyperpolarized magnetic resonance spectroscopy indicating increased in vivo glycolytic flux in Cmah-/-;mdx mice. Early upregulation of mitochondrial genes (Ucp3 and Cpt1) and downregulation of key glycolytic genes (Pdk1, Pdk4, Ppara), also denote disturbed cardiac metabolism and shift towards glucose utilization in Cmah-/-;mdx mice. Moreover, we show long-term treatment with peptide-conjugated exon skipping antisense oligonucleotides (20-week regimen), resulted in 20% cardiac dystrophin protein restoration and significantly improved RV cardiac function. Therefore, Cmah-/-;mdx mice represent an appropriate model for evaluating cardiac benefit of novel DMD therapeutics.


Asunto(s)
Citidina Monofosfato/genética , Distrofina/deficiencia , Morfolinos/uso terapéutico , Animales , Cardiomiopatía Dilatada/genética , Carnitina O-Palmitoiltransferasa/genética , Factor de Crecimiento del Tejido Conjuntivo/análisis , Citidina Monofosfato/fisiología , Modelos Animales de Enfermedad , Distrofina/genética , Distrofina/metabolismo , Exones , Terapia Genética/métodos , Corazón/fisiopatología , Masculino , Ratones , Ratones Endogámicos mdx , Oxigenasas de Función Mixta/metabolismo , Distrofia Muscular de Duchenne/genética , Miocardio/metabolismo , NADPH Oxidasa 4/análisis , Oligonucleótidos Antisentido/genética , Péptidos/genética , Fenotipo , Volumen Sistólico , Proteína Desacopladora 3/genética , Función Ventricular Derecha
2.
Proc Natl Acad Sci U S A ; 113(39): 10962-7, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27621445

RESUMEN

The development of antisense oligonucleotide therapy is an important advance in the identification of corrective therapy for neuromuscular diseases, such as spinal muscular atrophy (SMA). Because of difficulties of delivering single-stranded oligonucleotides to the CNS, current approaches have been restricted to using invasive intrathecal single-stranded oligonucleotide delivery. Here, we report an advanced peptide-oligonucleotide, Pip6a-morpholino phosphorodiamidate oligomer (PMO), which demonstrates potent efficacy in both the CNS and peripheral tissues in severe SMA mice following systemic administration. SMA results from reduced levels of the ubiquitously expressed survival motor neuron (SMN) protein because of loss-of-function mutations in the SMN1 gene. Therapeutic splice-switching oligonucleotides (SSOs) modulate exon 7 splicing of the nearly identical SMN2 gene to generate functional SMN protein. Pip6a-PMO yields SMN expression at high efficiency in peripheral and CNS tissues, resulting in profound phenotypic correction at doses an order-of-magnitude lower than required by standard naked SSOs. Survival is dramatically extended from 12 d to a mean of 456 d, with improvement in neuromuscular junction morphology, down-regulation of transcripts related to programmed cell death in the spinal cord, and normalization of circulating insulin-like growth factor 1. The potent systemic efficacy of Pip6a-PMO, targeting both peripheral as well as CNS tissues, demonstrates the high clinical potential of peptide-PMO therapy for SMA.


Asunto(s)
Atrofia Muscular Espinal/tratamiento farmacológico , Oligonucleótidos/uso terapéutico , Péptidos/química , Envejecimiento , Alelos , Secuencia de Aminoácidos , Biomarcadores/sangre , Línea Celular , Humanos , Movimiento , Atrofia Muscular Espinal/sangre , Atrofia Muscular Espinal/patología , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Oligonucleótidos/administración & dosificación , Oligonucleótidos/farmacología , Fenotipo , Empalme del ARN/genética , Análisis de Supervivencia , Proteína 2 para la Supervivencia de la Neurona Motora/genética
3.
Hum Mol Genet ; 24(23): 6756-68, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26385637

RESUMEN

Duchenne muscular dystrophy (DMD) is a classical monogenic disorder, a model disease for genomic studies and a priority candidate for regenerative medicine and gene therapy. Although the genetic cause of DMD is well known, the molecular pathogenesis of disease and the response to therapy are incompletely understood. Here, we describe analyses of protein, mRNA and microRNA expression in the tibialis anterior of the mdx mouse model of DMD. Notably, 3272 proteins were quantifiable and 525 identified as differentially expressed in mdx muscle (P < 0.01). Therapeutic restoration of dystrophin by exon skipping induced widespread shifts in protein and mRNA expression towards wild-type expression levels, whereas the miRNome was largely unaffected. Comparison analyses between datasets showed that protein and mRNA ratios were only weakly correlated (r = 0.405), and identified a multitude of differentially affected cellular pathways, upstream regulators and predicted miRNA-target interactions. This study provides fundamental new insights into gene expression and regulation in dystrophic muscle.


Asunto(s)
Distrofina/genética , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Terapia Genética , Masculino , Ratones , Ratones Endogámicos mdx , MicroARNs/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/terapia , Mutación , Proteómica , ARN Mensajero/metabolismo
4.
Hum Mol Genet ; 24(15): 4225-37, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25935000

RESUMEN

Splice modulation therapy has shown great clinical promise in Duchenne muscular dystrophy, resulting in the production of dystrophin protein. Despite this, the relationship between restoring dystrophin to established dystrophic muscle and its ability to induce clinically relevant changes in muscle function is poorly understood. In order to robustly evaluate functional improvement, we used in situ protocols in the mdx mouse to measure muscle strength and resistance to eccentric contraction-induced damage. Here, we modelled the treatment of muscle with pre-existing dystrophic pathology using antisense oligonucleotides conjugated to a cell-penetrating peptide. We reveal that 15% homogeneous dystrophin expression is sufficient to protect against eccentric contraction-induced injury. In addition, we demonstrate a >40% increase in specific isometric force following repeated administrations. Strikingly, we show that changes in muscle strength are proportional to dystrophin expression levels. These data define the dystrophin restoration levels required to slow down or prevent disease progression and improve overall muscle function once a dystrophic environment has been established in the mdx mouse model.


Asunto(s)
Péptidos de Penetración Celular/genética , Distrofina/biosíntesis , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Oligonucleótidos Antisentido/genética , Animales , Péptidos de Penetración Celular/administración & dosificación , Modelos Animales de Enfermedad , Distrofina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética , Humanos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Oligonucleótidos Antisentido/administración & dosificación
5.
Nucleic Acids Res ; 43(1): 29-39, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25468897

RESUMEN

The potential for therapeutic application of splice-switching oligonucleotides (SSOs) to modulate pre-mRNA splicing is increasingly evident in a number of diseases. However, the primary drawback of this approach is poor cell and in vivo oligonucleotide uptake efficacy. Biological activities can be significantly enhanced through the use of synthetically conjugated cationic cell penetrating peptides (CPPs). Studies to date have focused on the delivery of a single SSO conjugated to a CPP, but here we describe the conjugation of two phosphorodiamidate morpholino oligonucleotide (PMO) SSOs to a single CPP for simultaneous delivery and pre-mRNA targeting of two separate genes, exon 23 of the Dmd gene and exon 5 of the Acvr2b gene, in a mouse model of Duchenne muscular dystrophy. Conjugations of PMOs to a single CPP were carried out through an amide bond in one case and through a triazole linkage ('click chemistry') in the other. The most active bi-specific CPP-PMOs demonstrated comparable exon skipping levels for both pre-mRNA targets when compared to individual CPP-PMO conjugates both in cell culture and in vivo in the mdx mouse model. Thus, two SSOs with different target sequences conjugated to a single CPP are biologically effective and potentially suitable for future therapeutic exploitation.


Asunto(s)
Péptidos de Penetración Celular/química , Morfolinos/química , Distrofia Muscular de Duchenne/genética , Empalme del ARN , Receptores de Activinas Tipo II/genética , Animales , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Distrofina/genética , Exones , Ratones , Ratones Endogámicos mdx , Morfolinos/síntesis química
6.
Chembiochem ; 17(14): 1312-6, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27124570

RESUMEN

Mitochondria are central to health and disease, hence there is considerable interest in developing mitochondria-targeted therapies that require the delivery of peptides or nucleic acid oligomers. However, progress has been impeded by the lack of a measure of mitochondrial import of these molecules. Here, we address this need by quantitatively detecting molecules within the mitochondrial matrix. We used a mitochondria- targeted cyclooctyne (MitoOct) that accumulates several- hundredfold in the matrix, driven by the membrane potential. There, MitoOct reacts through click chemistry with an azide on the target molecule to form a diagnostic product that can be quantified by mass spectrometry. Because the membrane potential-dependent MitoOct concentration in the matrix is essential for conjugation, we can now determine definitively whether a putative mitochondrion-targeted molecule reaches the matrix. This "ClickIn" approach will facilitate development of mitochondria-targeted therapies.


Asunto(s)
Química Clic/métodos , Sistemas de Liberación de Medicamentos/métodos , Mitocondrias/metabolismo , Azidas/análisis , Azidas/química , Azidas/farmacocinética , Ciclooctanos/química , Ciclooctanos/farmacocinética , Portadores de Fármacos/química , Humanos , Espectrometría de Masas , Membranas Mitocondriales/metabolismo , Terapia Molecular Dirigida/métodos
7.
Mol Ther ; 23(8): 1341-1348, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25959011

RESUMEN

The fatal X-linked Duchenne muscular dystrophy (DMD), characterized by progressive muscle wasting and muscle weakness, is caused by mutations within the DMD gene. The use of antisense oligonucleotides (AOs) modulating pre-mRNA splicing to restore the disrupted dystrophin reading frame, subsequently generating a shortened but functional protein has emerged as a potential strategy in DMD treatment. AO therapy has recently been applied to induce out-of-frame exon skipping of myostatin pre-mRNA, knocking-down expression of myostatin protein, and such an approach is suggested to enhance muscle hypertrophy/hyperplasia and to reduce muscle necrosis. Within this study, we investigated dual exon skipping of dystrophin and myostatin pre-mRNAs using phosphorodiamidate morpholino oligomers conjugated with an arginine-rich peptide (B-PMOs). Intraperitoneal administration of B-PMOs was performed in neonatal mdx males on the day of birth, and at weeks 3 and 6. At week 9, we observed in treated mice (as compared to age-matched, saline-injected controls) normalization of muscle mass, a recovery in dystrophin expression, and a decrease in muscle necrosis, particularly in the diaphragm. Our data provide a proof of concept for antisense therapy combining dystrophin restoration and myostatin inhibition for the treatment of DMD.


Asunto(s)
Distrofina/genética , Exones , Miostatina/genética , Oligonucleótidos Antisentido/química , Sistemas de Lectura Abierta , Empalme Alternativo , Animales , Animales Recién Nacidos , Arginina/química , Diafragma/metabolismo , Modelos Animales de Enfermedad , Distrofina/metabolismo , Terapia Genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Morfolinos/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Miostatina/metabolismo , Necrosis , Péptidos/química , Sistemas de Lectura
8.
Nucleic Acids Res ; 42(5): 3207-17, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24366877

RESUMEN

Cell-penetrating peptide-mediated delivery of phosphorodiamidate morpholino oligomers (PMOs) has shown great promise for exon-skipping therapy of Duchenne Muscular Dystrophy (DMD). Pip6a-PMO, a recently developed conjugate, is particularly efficient in a murine DMD model, although mechanisms responsible for its increased biological activity have not been studied. Here, we evaluate the cellular trafficking and the biological activity of Pip6a-PMO in skeletal muscle cells and primary cardiomyocytes. Our results indicate that Pip6a-PMO is taken up in the skeletal muscle cells by an energy- and caveolae-mediated endocytosis. Interestingly, its cellular distribution is different in undifferentiated and differentiated skeletal muscle cells (vesicular versus nuclear). Likewise, Pip6a-PMO mainly accumulates in cytoplasmic vesicles in primary cardiomyocytes, in which clathrin-mediated endocytosis seems to be the pre-dominant uptake pathway. These differences in cellular trafficking correspond well with the exon-skipping data, with higher activity in myotubes than in myoblasts or cardiomyocytes. These differences in cellular trafficking thus provide a possible mechanistic explanation for the variations in exon-skipping activity and restoration of dystrophin protein in heart muscle compared with skeletal muscle tissues in DMD models. Overall, Pip6a-PMO appears as the most efficient conjugate to date (low nanomolar EC50), even if limitations remain from endosomal escape.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Exones , Morfolinos/metabolismo , Mioblastos Esqueléticos/metabolismo , Miocitos Cardíacos/metabolismo , Péptidos/metabolismo , Animales , Células Cultivadas , Endocitosis , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/metabolismo , Empalme del ARN
9.
Nano Lett ; 15(7): 4364-73, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26042553

RESUMEN

Antisense oligonucleotides (ASOs) have the potential to revolutionize medicine due to their ability to manipulate gene function for therapeutic purposes. ASOs are chemically modified and/or incorporated within nanoparticles to enhance their stability and cellular uptake, however, a major challenge is the poor understanding of their uptake mechanisms, which would facilitate improved ASO designs with enhanced activity and reduced toxicity. Here, we study the uptake mechanism of three therapeutically relevant ASOs (peptide-conjugated phosphorodiamidate morpholino (PPMO), 2'Omethyl phosphorothioate (2'OMe), and phosphorothioated tricyclo DNA (tcDNA) that have been optimized to induce exon skipping in models of Duchenne muscular dystrophy (DMD). We show that PPMO and tcDNA have high propensity to spontaneously self-assemble into nanoparticles. PPMO forms micelles of defined size and their net charge (zeta potential) is dependent on the medium and concentration. In biomimetic conditions and at low concentrations, PPMO obtains net negative charge and its uptake is mediated by class A scavenger receptor subtypes (SCARAs) as shown by competitive inhibition and RNAi silencing experiments in vitro. In vivo, the activity of PPMO was significantly decreased in SCARA1 knockout mice compared to wild-type animals. Additionally, we show that SCARA1 is involved in the uptake of tcDNA and 2'OMe as shown by competitive inhibition and colocalization experiments. Surface plasmon resonance binding analysis to SCARA1 demonstrated that PPMO and tcDNA have higher binding profiles to the receptor compared to 2'OMe. These results demonstrate receptor-mediated uptake for a range of therapeutic ASO chemistries, a mechanism that is dependent on their self-assembly into nanoparticles.


Asunto(s)
Nanopartículas/química , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacocinética , Receptores Depuradores de Clase A/metabolismo , Animales , Secuencia de Bases , Línea Celular , Exones , Terapia Genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Micelas , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , Receptores Depuradores de Clase A/genética
10.
Nucleic Acids Res ; 40(5): 2152-67, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22070883

RESUMEN

Anti-miRs are oligonucleotide inhibitors complementary to miRNAs that have been used extensively as tools to gain understanding of specific miRNA functions and as potential therapeutics. We showed previously that peptide nucleic acid (PNA) anti-miRs containing a few attached Lys residues were potent miRNA inhibitors. Using miR-122 as an example, we report here the PNA sequence and attached amino acid requirements for efficient miRNA targeting and show that anti-miR activity is enhanced substantially by the presence of a terminal-free thiol group, such as a Cys residue, primarily due to better cellular uptake. We show that anti-miR activity of a Cys-containing PNA is achieved by cell uptake through both clathrin-dependent and independent routes. With the aid of two PNA analogues having intrinsic fluorescence, thiazole orange (TO)-PNA and [bis-o-(aminoethoxy)phenyl]pyrrolocytosine (BoPhpC)-PNA, we explored the subcellular localization of PNA anti-miRs and our data suggest that anti-miR targeting of miR-122 may take place in or associated with endosomal compartments. Our findings are valuable for further design of PNAs and other oligonucleotides as potent anti-miR agents.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Ácidos Nucleicos de Péptidos/química , Ácidos Nucleicos de Péptidos/metabolismo , Aminoácidos/química , Línea Celular , Endocitosis , Endosomas , Humanos , Ácidos Nucleicos de Péptidos/análisis , Compuestos de Sulfhidrilo/química , Regulación hacia Arriba
11.
Hum Mol Genet ; 20(3): 413-21, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21062902

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused, in most cases, by the complete absence of the 427 kDa cytoskeletal protein, dystrophin. There is no effective treatment, and affected individuals die from respiratory failure and cardiomyopathy by age 30. Here, we investigated whether cardiomyopathy could be prevented in animal models of DMD by increasing diaphragm utrophin or dystrophin expression and thereby restoring diaphragm function. In a transgenic mdx mouse, where utrophin was over expressed in the skeletal muscle and the diaphragm, but not in the heart, we found cardiac function, specifically right and left ventricular ejection fraction as measured using in vivo magnetic resonance imaging, was restored to wild-type levels. In mdx mice treated with a peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) that resulted in high levels of dystrophin restoration in the skeletal muscle and the diaphragm only, cardiac function was also restored to wild-type levels. In dystrophin/utrophin-deficient double-knockout (dKO) mice, a more severely affected animal model of DMD, treatment with a PPMO again produced high levels of dystrophin only in the skeletal muscle and the diaphragm, and once more restored cardiac function to wild-type levels. In the dKO mouse, there was no difference in heart function between treatment of the diaphragm plus the heart and treatment of the diaphragm alone. Restoration of diaphragm and other respiratory muscle function, irrespective of the method used, was sufficient to prevent cardiomyopathy in dystrophic mice. This novel mechanism of treating respiratory muscles to prevent cardiomyopathy in dystrophic mice warrants further investigation for its implications on the need to directly treat the heart in DMD.


Asunto(s)
Cardiomiopatías/prevención & control , Diafragma/fisiopatología , Distrofina/metabolismo , Morfolinas/farmacología , Distrofia Muscular Animal/tratamiento farmacológico , Utrofina/metabolismo , Animales , Proteínas del Citoesqueleto/metabolismo , Diafragma/efectos de los fármacos , Diafragma/metabolismo , Distrofina/genética , Corazón/fisiopatología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Ratones Transgénicos , Morfolinos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/fisiopatología , Volumen Sistólico , Utrofina/genética
12.
RNA ; 17(5): 933-43, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21441346

RESUMEN

MicroRNAs (miRNAs) are small non-coding RNAs involved in fine-tuning of gene regulation. Antisense oligonucleotides (ONs) are promising tools as anti-miRNA (anti-miR) agents toward therapeutic applications and to uncover miRNA function. Such anti-miR ONs include 2'-O-methyl (OMe), cationic peptide nucleic acids like K-PNA-K3, and locked nucleic acid (LNA)-based anti-miRs such as LNA/DNA or LNA/OMe. Northern blotting is a widely used and robust technique to detect miRNAs. However, miRNA quantification in the presence of anti-miR ONs has proved to be challenging, due to detection artifacts, which has led to poor understanding of miRNA fate upon anti-miR binding. Here we show that anti-miR ON bound to miR-122 can prevent the miRNA from being properly precipitated into the purified RNA fraction using the standard RNA extraction protocol (TRI-Reagent), yielding an RNA extract that does not reflect the real cellular levels of the miRNA. An increase in the numbers of equivalents of isopropanol during the precipitation step leads to full recovery of the targeted miRNA back into the purified RNA extract. Following our improved protocol, we demonstrate by Northern blotting, in conjunction with a PNA decoy strategy and use of high denaturing PAGE, that high-affinity anti-miRs (K-PNA-K3, LNA/DNA, and LNA/OMe) sequester miR-122 without causing miRNA degradation, while miR-122 targeting with a lower-affinity anti-miR (OMe) seems to promote degradation of the miRNA. The technical issues explored in this work will have relevance for other hybridization-based techniques for miRNA quantification in the presence of anti-miR ONs.


Asunto(s)
Northern Blotting/métodos , MicroARNs/análisis , Oligonucleótidos/análisis , Línea Celular Tumoral , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Oligonucleótidos/genética , Estabilidad del ARN
13.
Org Biomol Chem ; 11(43): 7621-30, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24105028

RESUMEN

A novel method for the parallel synthesis of peptide-biocargo conjugates was developed that utilizes affinity purification for fast isolation of the conjugates in order to avoid time consuming HPLC purification. The methodology was applied to create two libraries of cell-penetrating peptide (CPP)-PNA705 conjugates from parallel-synthesized peptide libraries. The conjugates were tested for their ability to induce splicing redirection in HeLa pLuc705 cells. The results demonstrate how the novel methodology can be applied for screening purposes in order to find suitable CPP-biocargo combinations and further optimization of CPPs.


Asunto(s)
Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/metabolismo , Ácidos Nucleicos de Péptidos/química , Ácidos Nucleicos de Péptidos/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , Péptidos de Penetración Celular/química , Cromatografía Líquida de Alta Presión , Células HeLa , Humanos , Conformación Molecular , Biblioteca de Péptidos , Empalme del ARN/genética , ARN Mensajero/genética
14.
Nucleic Acids Res ; 39(12): 5284-98, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21345932

RESUMEN

Numerous human genetic diseases are caused by mutations that give rise to aberrant alternative splicing. Recently, several of these debilitating disorders have been shown to be amenable for splice-correcting oligonucleotides (SCOs) that modify splicing patterns and restore the phenotype in experimental models. However, translational approaches are required to transform SCOs into usable drug products. In this study, we present a new cell-penetrating peptide, PepFect14 (PF14), which efficiently delivers SCOs to different cell models including HeLa pLuc705 and mdx mouse myotubes; a cell culture model of Duchenne's muscular dystrophy (DMD). Non-covalent PF14-SCO nanocomplexes induce splice-correction at rates higher than the commercially available lipid-based vector Lipofectamine 2000 (LF2000) and remain active in the presence of serum. Furthermore, we demonstrate the feasibility of incorporating this delivery system into solid formulations that could be suitable for several therapeutic applications. Solid dispersion technique is utilized and the formed solid formulations are as active as the freshly prepared nanocomplexes in solution even when stored at an elevated temperatures for several weeks. In contrast, LF2000 drastically loses activity after being subjected to same procedure. This shows that using PF14 is a very promising translational approach for the delivery of SCOs in different pharmaceutical forms.


Asunto(s)
Péptidos de Penetración Celular/química , Lipopéptidos/química , Oligonucleótidos Antisentido/administración & dosificación , Empalme Alternativo , Animales , Péptidos de Penetración Celular/metabolismo , Péptidos de Penetración Celular/toxicidad , Células Cultivadas , Medios de Cultivo , Medio de Cultivo Libre de Suero , Endocitosis , Células HeLa , Humanos , Cinética , Luz , Lipopéptidos/metabolismo , Lipopéptidos/toxicidad , Ratones , Fibras Musculares Esqueléticas/metabolismo , Nanoestructuras/química , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/metabolismo , Dispersión de Radiación , Soluciones , Temperatura
15.
Cells ; 12(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37759507

RESUMEN

Interleukin-6 (IL-6) is a pleiotropic cytokine that plays a crucial role in maintaining normal homeostatic processes under the pathogenesis of various inflammatory and autoimmune diseases. This context-dependent effect from a cytokine is due to two distinctive forms of signaling: cis-signaling and trans-signaling. IL-6 cis-signaling involves binding IL-6 to the membrane-bound IL-6 receptor and Glycoprotein 130 (GP130) signal-transducing subunit. By contrast, in IL-6 trans-signaling, complexes of IL-6 and the soluble form of the IL-6 receptor (sIL-6R) signal via membrane-bound GP130. Various strategies have been employed in the past decade to target the pro-inflammatory effect of IL-6 in numerous inflammatory disorders. However, their development has been hindered since these approaches generally target global IL-6 signaling, also affecting the anti-inflammatory effects of IL-6 signaling too. Therefore, novel strategies explicitly targeting the pro-inflammatory IL-6 trans-signaling without affecting the IL-6 cis-signaling are required and carry immense therapeutic potential. Here, we have developed a novel approach to specifically decoy IL-6-mediated trans-signaling by modulating alternative splicing in GP130, an IL-6 signal transducer, by employing splice switching oligonucleotides (SSO), to induce the expression of truncated soluble isoforms of the protein GP130. This isoform is devoid of signaling domains but allows for specifically sequestering the IL-6/sIL-6R receptor complex with high affinity in serum and thereby suppressing inflammation. Using the state-of-the-art Pip6a cell-penetrating peptide conjugated to PMO-based SSO targeting GP130 for efficient in vivo delivery, reduced disease phenotypes in two different inflammatory mouse models of systemic and intestinal inflammation were observed. Overall, this novel gene therapy platform holds great potential as a refined therapeutic intervention for chronic inflammatory diseases.


Asunto(s)
Citocinas , Interleucina-6 , Animales , Ratones , Receptor gp130 de Citocinas , Inflamación , Oligonucleótidos
16.
Org Biomol Chem ; 10(4): 746-54, 2012 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-22124653

RESUMEN

Chimeric 2'-O-methyl oligoribonucleotides (2'-OMe ORNs) containing internucleotide linkages which were modified with phosphonoacetate (PACE) or thiophosphonoacetate (thioPACE) were prepared by solid-phase synthesis. The modified 2'-OMe ORNs contained a central phosphate or phosphorothioate sequence with up to 4 PACE or thioPACE modifications, respectively, at either end of the ORN in a "gapmer" motif. Both PACE and thioPACE 2'-OMe ORNs formed stable duplexes with complementary RNA. The majority of these duplexes had higher thermal melting temperatures than an unmodified RNA:RNA duplex. The modified 2'-OMe ORNs were effective passenger strands with complementary, unmodified siRNAs, for inducing siRNA activity in a dual luciferase assay in the presence of a lipid transfecting agent. As single strands, thioPACE 2'-OMe ORNs were efficiently taken up by HeLa cells in the absence of a lipid transfecting agent. Furthermore, thioPACE modifications greatly improved the potency of a 2'-OMe phosphorothioate ORN as an inhibitor of microRNA-122 in Huh7 cells, without lipid transfection.


Asunto(s)
Oligorribonucleótidos/química , Oligorribonucleótidos/farmacología , Ácido Fosfonoacético/química , Ácido Fosfonoacético/farmacología , Secuencia de Bases , Línea Celular , Células HeLa , Humanos , MicroARNs/antagonistas & inhibidores , Oligorribonucleótidos/síntesis química , Oligorribonucleótidos/farmacocinética , Ácido Fosfonoacético/síntesis química , Ácido Fosfonoacético/farmacocinética , ARN Interferente Pequeño/farmacología , Técnicas de Síntesis en Fase Sólida , Compuestos de Sulfhidrilo/síntesis química , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacocinética , Compuestos de Sulfhidrilo/farmacología
17.
Mol Ther ; 19(7): 1295-303, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21505427

RESUMEN

Induced splice modulation of pre-mRNAs shows promise to correct aberrant disease transcripts and restore functional protein and thus has therapeutic potential. Duchenne muscular dystrophy (DMD) results from mutations that disrupt the DMD gene open reading frame causing an absence of dystrophin protein. Antisense oligonucleotide (AO)-mediated exon skipping has been shown to restore functional dystrophin in mdx mice and DMD patients treated intramuscularly in two recent phase 1 clinical trials. Critical to the therapeutic success of AO-based treatment will be the ability to deliver AOs systemically to all affected tissues including the heart. Here, we report identification of a series of transduction peptides (Pip5) as AO conjugates for enhanced systemic and particularly cardiac delivery. One of the lead peptide-AO conjugates, Pip5e-AO, showed highly efficient exon skipping and dystrophin production in mdx mice with complete correction of the aberrant DMD transcript in heart, leading to >50% of the normal level of dystrophin in heart. Mechanistic studies indicated that the enhanced activity of Pip5e-phosphorodiamidate morpholino (PMO) is partly explained by more efficient nuclear delivery. Pip5 series derivatives therefore have significant potential for advancing the development of exon skipping therapies for DMD and may have application for enhanced cardiac delivery of other biotherapeutics.


Asunto(s)
Exones/genética , Miocardio/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Péptidos/genética , Péptidos/metabolismo , Animales , Western Blotting , Inmunohistoquímica , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Transducción Genética
18.
Nucleic Acids Res ; 38(13): 4466-75, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20223773

RESUMEN

MicroRNAs (miRNAs) play an important role in diverse physiological processes and are potential therapeutic agents. Synthetic oligonucleotides (ONs) of different chemistries have proven successful for blocking miRNA expression. However, their specificity and efficiency have not been fully evaluated. Here, we show that peptide nucleic acids (PNAs) efficiently block a key inducible miRNA expressed in the haematopoietic system, miR-155, in cultured B cells as well as in mice. Remarkably, miR-155 inhibition by PNA in primary B cells was achieved in the absence of any transfection agent. In mice, the high efficiency of the treatment was demonstrated by a strong overlap in global gene expression between B cells isolated from anti-miR-155 PNA-treated and miR-155-deficient mice. Interestingly, PNA also induced additional changes in gene expression. Our analysis provides a useful platform to aid the design of efficient and specific anti-miRNA ONs for in vivo use.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/antagonistas & inhibidores , Ácidos Nucleicos de Péptidos , Animales , Linfocitos B/metabolismo , Sitios de Unión , Células Cultivadas , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Microondas , Ácidos Nucleicos de Péptidos/síntesis química , ARN Mensajero/química , ARN Mensajero/metabolismo
19.
Methods Mol Biol ; 2434: 3-31, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35213007

RESUMEN

This introduction charts the history of the development of the major chemical modifications that have influenced the development of nucleic acids therapeutics focusing in particular on antisense oligonucleotide analogues carrying modifications in the backbone and sugar. Brief mention is made of siRNA development and other applications that have by and large utilized the same modifications. We also point out the pitfalls of the use of nucleic acids as drugs, such as their unwanted interactions with pattern recognition receptors, which can be mitigated by chemical modification or used as immunotherapeutic agents.


Asunto(s)
Ácidos Nucleicos , Ácidos Nucleicos/genética , Ácidos Nucleicos/uso terapéutico , Oligonucleótidos/genética , Oligonucleótidos/uso terapéutico , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
20.
JCI Insight ; 7(24)2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36346674

RESUMEN

Antisense oligonucleotides (ASOs) have emerged as one of the most innovative new genetic drug modalities. However, their high molecular weight limits their bioavailability for otherwise-treatable neurological disorders. We investigated conjugation of ASOs to an antibody against the murine transferrin receptor, 8D3130, and evaluated it via systemic administration in mouse models of the neurodegenerative disease spinal muscular atrophy (SMA). SMA, like several other neurological and neuromuscular diseases, is treatable with single-stranded ASOs that modulate splicing of the survival motor neuron 2 (SMN2) gene. Administration of 8D3130-ASO conjugate resulted in elevated levels of bioavailability to the brain. Additionally, 8D3130-ASO yielded therapeutic levels of SMN2 splicing in the central nervous system of adult human SMN2-transgenic (hSMN2-transgenic) mice, which resulted in extended survival of a severely affected SMA mouse model. Systemic delivery of nucleic acid therapies with brain-targeting antibodies offers powerful translational potential for future treatments of neuromuscular and neurodegenerative diseases.


Asunto(s)
Atrofia Muscular Espinal , Enfermedades Neurodegenerativas , Ratones , Animales , Humanos , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Sistema Nervioso Central , Oligonucleótidos Antisentido/uso terapéutico , Ratones Transgénicos , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA