Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Biomed Pharmacother ; 175: 116737, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38749176

RESUMO

Antisense oligonucleotide (ASO) has emerged as a promising therapeutic approach for treating central nervous system (CNS) disorders by modulating gene expression with high selectivity and specificity. However, the poor permeability of ASO across the blood-brain barrier (BBB) diminishes its therapeutic success. Here, we designed and synthesized a series of BBB-penetrating peptides (BPP) derived from either the receptor-binding domain of apolipoprotein E (ApoE) or a transferrin receptor-binding peptide (THR). The BPPs were conjugated to phosphorodiamidate morpholino oligomers (PMO) that are chemically analogous to the 2'-O-(2-methoxyethyl) (MOE)-modified ASO approved by the FDA for treating spinal muscular atrophy (SMA). The BPP-PMO conjugates significantly increased the level of full-length SMN2 in the patient-derived SMA fibroblasts in a concentration-dependent manner with minimal to no toxicity. Furthermore, the systemic administration of the most potent BPP-PMO conjugates significantly increased the expression of full-length SMN2 in the brain and spinal cord of SMN2 transgenic adult mice. Notably, BPP8-PMO conjugate showed a 1.25-fold increase in the expression of full-length functional SMN2 in the brain. Fluorescence imaging studies confirmed that 78% of the fluorescently (Cy7)-labelled BPP8-PMO reached brain parenchyma, with 11% uptake in neuronal cells. Additionally, the BPP-PMO conjugates containing retro-inverso (RI) D-BPPs were found to possess extended half-lives compared to their L-counterparts, indicating increased stability against protease degradation while preserving the bioactivity. This delivery platform based on BPP enhances the CNS bioavailability of PMO targeting the SMN2 gene, paving the way for the development of systemically administered neurotherapeutics for CNS disorders.


Assuntos
Apolipoproteínas E , Barreira Hematoencefálica , Camundongos Transgênicos , Oligonucleotídeos Antissenso , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/farmacocinética , Humanos , Apolipoproteínas E/metabolismo , Camundongos , Morfolinos/administração & dosagem , Morfolinos/farmacocinética , Morfolinos/farmacologia , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo , Atrofia Muscular Espinal/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Peptídeos/administração & dosagem , Peptídeos/farmacologia , Peptídeos/química , Peptídeos/farmacocinética , Peptídeos Penetradores de Células/química
2.
Nucleic Acids Res ; 49(11): 6100-6113, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-34107015

RESUMO

Pulmonary diseases offer many targets for oligonucleotide therapeutics. However, effective delivery of oligonucleotides to the lung is challenging. For example, splicing mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect a significant cohort of Cystic Fibrosis (CF) patients. These individuals could potentially benefit from treatment with splice switching oligonucleotides (SSOs) that can modulate splicing of CFTR and restore its activity. However, previous studies in cell culture used oligonucleotide transfection methods that cannot be safely translated in vivo. In this report, we demonstrate effective correction of a splicing mutation in the lung of a mouse model using SSOs. Moreover, we also demonstrate effective correction of a CFTR splicing mutation in a pre-clinical CF patient-derived cell model. We utilized a highly effective delivery strategy for oligonucleotides by combining peptide-morpholino (PPMO) SSOs with small molecules termed OECs. PPMOs distribute broadly into the lung and other tissues while OECs potentiate the effects of oligonucleotides by releasing them from endosomal entrapment. The combined PPMO plus OEC approach proved to be effective both in CF patient cells and in vivo in the mouse lung and thus may offer a path to the development of novel therapeutics for splicing mutations in CF and other lung diseases.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/terapia , Pulmão/metabolismo , Morfolinos/administração & dosagem , Splicing de RNA , Animais , Células Cultivadas , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Camundongos , Mutação , Peptídeos , Mucosa Respiratória/metabolismo , Transfecção
3.
Bull Exp Biol Med ; 170(1): 58-63, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33237527

RESUMO

Mutations in pank2 gene encoding pantothenate kinase 2 determine a pantothenate kinase-associated neurodegeneration, a rare disorder characterized by iron deposition in the globus pallidus. To extend our previous work, we performed microinjections of a new pank2-specific morpholino to zebrafish embryos and thoroughly analyzed vasculature development. Vessels development was severely perturbed in the head, trunk, and tail, where blood accumulation was remarkable and associated with dilation of the posterior cardinal vein. This phenotype was specific as confirmed by p53 expression analysis and injection of the same morpholino in pank2-mutant embryos. We can conclude that pank2 gene is involved in vasculature development in zebrafish embryos. The comprehension of the underlining mechanisms could be of relevance for understanding of pantothenate kinase-associated neurodegeneration.


Assuntos
Vasos Sanguíneos/metabolismo , Coenzima A/farmacologia , Globo Pálido/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/prevenção & controle , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Modelos Animais de Doenças , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Globo Pálido/irrigação sanguínea , Globo Pálido/efeitos dos fármacos , Globo Pálido/patologia , Cabeça/irrigação sanguínea , Cabeça/crescimento & desenvolvimento , Humanos , Morfolinos/administração & dosagem , Morfolinos/genética , Morfolinos/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Cauda/irrigação sanguínea , Cauda/crescimento & desenvolvimento , Cauda/metabolismo , Tronco/irrigação sanguínea , Tronco/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra
4.
Bioconjug Chem ; 31(10): 2367-2382, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-32986398

RESUMO

A nontoxic delivery vehicle is essential for the therapeutic applications of antisense phosphorodiamidate morpholino oligonucleotides (PMOs). Though guanidinium-rich or arginine-rich cellular transporter conjugated Vivo-PMO or PPMO has been developed for in vivo application, however, either their toxicity or stability has become an issue. Previously, we reported nonpeptidic internal guanidinium transporter (IGT) mediated delivery of PMO for gene silencing and got encouraging results. In this paper, we report the synthesis of IGT using a Hg-free method for scale up and N-terminal modification of IGT with a suitable hydrophobic or lipophilic group to improve the cell permeability, endosomal escape, and mitochondrial localization and to reduce toxicity in the MTT assay. For the delivery of PMO, IGT-PMO conjugate was synthesized to target NANOG in cells, a transcription factor required for cancer stem cell proliferation and embryonic development and is involved in many cancers. Our data shows IGT-PMO-facilitated NANOG inhibition, and thereby the prevention of EpCAM-N-Cadherin-Vimentin axis mediated epithelial to mesenchymal transition (EMT) in MCF-7 cells. Moreover, unlike taxol, NANOG inhibition influences the expression of stemness factor c-Myc, Hh-Gli signaling proteins, other cancer related factors, and their respective phenotypes in cancer cells. To the best of our knowledge, this is the first report to illustrate that the IGT-PMO-mediated NANOG inhibition increases the therapeutic potential of taxol and induces G0-G1 arrest in cancer cells to prevent cancer progression. However, it warrants further investigation in other cancer cells and preclinical platforms.


Assuntos
Antineoplásicos/administração & dosagem , Morfolinos/administração & dosagem , Proteína Homeobox Nanog/antagonistas & inibidores , Paclitaxel/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Portadores de Fármacos/química , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Morfolinos/farmacocinética , Morfolinos/farmacologia , Proteína Homeobox Nanog/genética , Paclitaxel/farmacocinética , Paclitaxel/farmacologia
5.
Hepatology ; 72(5): 1786-1799, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32060934

RESUMO

BACKGROUND AND AIMS: During liver development, bipotent progenitor cells differentiate into hepatocytes and biliary epithelial cells to ensure a functional liver required to maintain organismal homeostasis. The developmental cues controlling the differentiation of committed progenitors into these cell types, however, are incompletely understood. Here, we discover an essential role for estrogenic regulation in vertebrate liver development to affect hepatobiliary fate decisions. APPROACH AND RESULTS: Exposure of zebrafish embryos to 17ß-estradiol (E2) during liver development significantly decreased hepatocyte-specific gene expression, liver size, and hepatocyte number. In contrast, pharmacological blockade of estrogen synthesis or nuclear estrogen receptor (ESR) signaling enhanced liver size and hepatocyte marker expression. Transgenic reporter fish demonstrated nuclear ESR activity in the developing liver. Chemical inhibition and morpholino knockdown of nuclear estrogen receptor 2b (esr2b) increased hepatocyte gene expression and blocked the effects of E2 exposure. esr2b-/- mutant zebrafish exhibited significantly increased expression of hepatocyte markers with no impact on liver progenitors, other endodermal lineages, or vasculature. Significantly, E2-stimulated Esr2b activity promoted biliary epithelial differentiation at the expense of hepatocyte fate, whereas loss of esr2b impaired biliary lineage commitment. Chemical and genetic epistasis studies identified bone morphogenetic protein (BMP) signaling as a mediator of the estrogen effects. The divergent impact of estrogen on hepatobiliary fate was confirmed in a human hepatoblast cell line, indicating the relevance of this pathway for human liver development. CONCLUSIONS: Our studies identify E2, esr2b, and downstream BMP activity as important regulators of hepatobiliary fate decisions during vertebrate liver development. These results have significant clinical implications for liver development in infants exposed to abnormal estrogen levels or estrogenic compounds during pregnancy.


Assuntos
Sistema Biliar/embriologia , Estradiol/metabolismo , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fígado/embriologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Sistema Biliar/citologia , Sistema Biliar/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Embrião não Mamífero , Estradiol/administração & dosagem , Receptor beta de Estrogênio/genética , Feminino , Técnicas de Silenciamento de Genes , Hepatócitos/fisiologia , Fígado/citologia , Fígado/metabolismo , Masculino , Modelos Animais , Morfolinos/administração & dosagem , Morfolinos/genética , Transdução de Sinais/genética , Células-Tronco/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
6.
ACS Infect Dis ; 5(8): 1446-1455, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31119935

RESUMO

Overexpression of bacterial efflux pumps is a driver of increasing antibiotic resistance in Gram-negative pathogens. The AcrAB-TolC efflux pump has been implicated in resistance to a number of important antibiotic classes including fluoroquinolones, macrolides, and ß-lactams. Antisense technology, such as peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs), can be utilized to inhibit expression of efflux pumps and restore susceptibility to antibiotics. Targeting of the AcrAB-TolC components with PPMOs revealed a sequence for acrA, which was the most effective at reducing antibiotic efflux. This acrA-PPMO enhances the antimicrobial effects of the levofloxacin and azithromycin in a panel of clinical Enterobacteriaceae strains. Additionally, acrA-PPMO enhanced azithromycin in vivo in a K. pneumoniae septicemia model. PPMOs targeting the homologous resistance-nodulation-division (RND)-efflux system in P. aeruginosa, MexAB-OprM, also enhanced potency to several classes of antibiotics in a panel of strains and in a cell culture infection model. These data suggest that PPMOs can be used as an adjuvant in antibiotic therapy to increase the efficacy or extend the spectrum of useful antibiotics against a variety of Gram-negative infections.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Morfolinos/farmacologia , Peptídeos/farmacologia , Animais , Antibacterianos/administração & dosagem , Azitromicina/administração & dosagem , Brônquios/citologia , Proteínas de Transporte/antagonistas & inibidores , Técnicas de Cultura de Células , Fibrose Cística , Células Epiteliais/microbiologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/genética , Feminino , Humanos , Injeções Intraperitoneais , Lipoproteínas/genética , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Morfolinos/administração & dosagem , Peptídeos/administração & dosagem
8.
J Drug Target ; 27(5-6): 566-572, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30198798

RESUMO

Drug-free macromolecular therapeutics (DFMT) have been recently developed to treat non-Hodgkin lymphoma (NHL). It is a consecutive delivery of two nanoconjugates: (1) bispecific engager that pretargets surface CD20, and (2) multivalent effector polymer that hybridises with CD20-bound engagers. Without the need of low molecular weight drug, the hybridisation of morpholino oligonucleotide containing DFMT at NHL cell surface triggers CD20 crosslinking and subsequent apoptosis. We have previously determined various factors that affect the efficacy of DFMT regarding the synthetic structures. Here, we show that DFMT-mediated apoptosis is also influenced by the state of cells. Compared with other cell cycle states, cells arrested at G2/M phase exhibit enhanced CD20 expression, and have more sustainable CD20 binding by DFMT, resulting in a higher degree of DFMT-mediated CD20 crosslinking. Moreover, the anti-apoptotic Bcl-2 protein was phosphorylated in G2/M phase, thereby increasing the cell susceptibility to DFMT. As a result, DFMT mediated augmented apoptosis in G2/M phase cells. When DFMT was combined with a polymer-docetaxel conjugate that triggered G2/M blockage, a combinatorial apoptotic effect was achieved to induce programmed cell death. Our findings suggest the co-delivery of DFMT and G2/M inhibiting drug combinations may present a therapeutic advantage in NHL treatment.


Assuntos
Apoptose/efeitos dos fármacos , Linfoma de Burkitt/tratamento farmacológico , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Morfolinos/farmacologia , Nanoconjugados/química , Antígenos CD20/genética , Antígenos CD20/imunologia , Linfoma de Burkitt/imunologia , Linfoma de Burkitt/patologia , Linhagem Celular Tumoral , Humanos , Morfolinos/administração & dosagem , Ligação Proteica
9.
Ann Neurol ; 84(6): 814-828, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30427554

RESUMO

OBJECTIVE: Galloway-Mowat syndrome (GAMOS) is a neural and renal disorder, characterized by microcephaly, brain anomalies, and early onset nephrotic syndrome. Biallelic mutations in WDR73 and the 4 subunit genes of the KEOPS complex are reported to cause GAMOS. Furthermore, an identical homozygous NUP107 (nucleoporin 107kDa) mutation was identified in 4 GAMOS-like families, although biallelic NUP107 mutations were originally identified in steroid-resistant nephrotic syndrome. NUP107 and NUP133 (nucleoporin 133kDa) are interacting subunits of the nuclear pore complex in the nuclear envelope during interphase, and these proteins are also involved in centrosome positioning and spindle assembly during mitosis. METHODS: Linkage analysis and whole exome sequencing were performed in a previously reported GAMOS family with brain atrophy and steroid-resistant nephrotic syndrome. RESULTS: We identified a homozygous NUP133 mutation, c.3335-11T>A, which results in the insertion of 9bp of intronic sequence between exons 25 and 26 in the mutant transcript. NUP133 and NUP107 interaction was impaired by the NUP133 mutation based on an immunoprecipitation assay. Importantly, focal cortical dysplasia type IIa was recognized in the brain of an autopsied patient and focal segmental glomerulosclerosis was confirmed in the kidneys of the 3 examined patients. A nup133-knockdown zebrafish model exhibited microcephaly, fewer neuronal cells, underdeveloped glomeruli, and fusion of the foot processes of the podocytes, which mimicked human GAMOS features. nup133 morphants could be rescued by human wild-type NUP133 mRNA but not by mutant mRNA. INTERPRETATION: These data indicate that the biallelic NUP133 loss-of-function mutation causes GAMOS. Ann Neurol 2018;84:814-828.


Assuntos
Predisposição Genética para Doença/genética , Hérnia Hiatal/genética , Microcefalia/genética , Antígenos de Histocompatibilidade Menor/genética , Mutação/genética , Nefrose/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Pré-Escolar , Saúde da Família , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hérnia Hiatal/diagnóstico por imagem , Hérnia Hiatal/patologia , Humanos , Lactente , Japão , Rim/metabolismo , Rim/patologia , Rim/ultraestrutura , Linfócitos/metabolismo , Linfócitos/ultraestrutura , Masculino , Microcefalia/diagnóstico por imagem , Microcefalia/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Antígenos de Histocompatibilidade Menor/ultraestrutura , Morfolinos/administração & dosagem , Mutagênese Sítio-Dirigida , Nefrose/diagnóstico por imagem , Nefrose/patologia , Complexo de Proteínas Formadoras de Poros Nucleares/ultraestrutura , Fosfopiruvato Hidratase/metabolismo , Adulto Jovem , Peixe-Zebra
10.
Methods Mol Biol ; 1828: 151-163, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171540

RESUMO

Antisense oligonucleotide induced exon skipping emerges as a promising therapeutic strategy for patients suffering from a devastating muscle disorder Duchenne muscular dystrophy (DMD). Systemic administration of antisense phosphorodiamidate morpholino oligomers (PMOs) targeting exons 6 and 8 in dystrophin mRNA of the canine X-linked muscular dystrophy model in Japan (CXMDJ) that lacks exon 7, restored dystrophin expression throughout skeletal muscle and ameliorated skeletal muscle pathology and function. However, the antisense PMO regime used in CXMDJ could not be considered for a direct application to DMD patients so far, because this type of mutation is quite rare. We have identified a DMD patient with an exon 7 deletion; and tried a direct translation of the antisense PMOs used in dog models to the DMD patient's cells. We converted fibroblasts obtained from CXMDJ dogs and from the DMD patient to myotubes by MyoD transduction using fluorescence-activated cell sorting (FACS). We subsequently designed antisense PMOs targeting identical regions of dog and human dystrophin exons 6 and 8 and administered them as a cocktail to the in vitro generated dog or human myotubes. In both cases, we observed comparable skipping efficacy of exons 6 and 8 and restoration of dystrophin protein. The accompanying skipping of exon 9, which does not alter the reading frame, varied according to the cell origin. The antisense PMOs originally administered to the CXMDJ dog model were capable of inducing multi-exon skipping of the dystrophin gene on the FACS-aided MyoD-transduced fibroblasts derived from an exon 7-deleted DMD patient. These data support the suitability of dog as a laboratory model for DMD because the similarity of dystrophin sequences allowed a successful translation of the dog's PMOs to DMD patients cells.


Assuntos
Distrofina/genética , Éxons , Morfolinos/genética , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/genética , Splicing de RNA , Animais , Células Cultivadas , Cães , Fibroblastos/metabolismo , Terapia Genética , Vetores Genéticos/genética , Humanos , Morfolinos/administração & dosagem , Distrofia Muscular de Duchenne/terapia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , Retroviridae/genética , Transdução Genética
11.
Methods Mol Biol ; 1828: 355-363, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171553

RESUMO

Efficient intracellular delivery is critical to the successful application of synthetic antisense oligonucleotides (ASOs) to modulate gene expression. The conjugation of cell-penetrating peptides (CPPs) to ASOs has been shown to significantly improve their intracellular delivery. It is important, however, that formation of the covalent linkage between the peptide and oligonucleotide is efficient and orthogonal, to ensure high yields and a homogeneous product. Described herein are efficient and facile methodologies for the conjugation of peptides to ASOs, and their subsequent labeling with various moieties such as fluorescent dyes for intracellular tracking studies.


Assuntos
Corantes Fluorescentes , Técnicas de Transferência de Genes , Oligonucleotídeos , Peptídeos , Corantes Fluorescentes/química , Humanos , Estrutura Molecular , Morfolinos/administração & dosagem , Morfolinos/química , Morfolinos/genética , Oligonucleotídeos/química , Oligonucleotídeos Antissenso , Peptídeos/química , Coloração e Rotulagem
12.
Methods Mol Biol ; 1828: 327-342, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171551

RESUMO

Exon-skipping antisense oligonucleotides (AOs) are promising treatments for muscle-related genetic ailments including Duchenne muscular dystrophy (DMD), but clinical translation is unfortunately hampered by insufficient systemic delivery. Here we describe that how one can employ a glucose-fructose injection mixture to improve muscle uptake and functional outcomes of DMD AOs in energy-deficient peripheral muscles of mdx mice. The potentiating effect of glucose-fructose on AOs in energy-deficient muscles offers a simple and economical method for enhancing AO potency, reducing screening costs for researchers and accelerating the translation of nucleic acid-based therapeutics in DMD and other muscular dystrophies.


Assuntos
Éxons , Frutose/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Splicing de RNA , Trifosfato de Adenosina/metabolismo , Animais , Clatrina/metabolismo , Distrofina/genética , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/genética , Complexo de Proteínas Associadas Distrofina/metabolismo , Endocitose , Metabolismo Energético , Frutose/metabolismo , Glucose/metabolismo , Humanos , Imuno-Histoquímica , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos mdx , Morfolinos/administração & dosagem , Morfolinos/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/genética
13.
Methods Mol Biol ; 1828: 365-379, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171554

RESUMO

Exon skipping is an emerging approach to treating Duchenne muscular dystrophy (DMD), one of the most common lethal genetic disorders. Exon skipping uses synthetic antisense oligonucleotides (AONs) to splice out frame-disrupting exon(s) of DMD mRNA to restore the reading frame of the gene products and produce truncated yet functional proteins. The FDA conditionally approved the first exon-skipping AON, called eteplirsen (brand name ExonDys51), targeting exon 51 of the DMD gene, in late 2016. Using a cocktail of AONs, multiple exons can be skipped, which can theoretically treat 80-90% of patients with DMD. Although the success of multiple exon skipping in a DMD dog model has made a significant impact on the development of therapeutics for DMD, unmodified AONs such as phosphorodiamidate morpholino oligomers (PMOs) have little efficacy in cardiac muscles. Here, we describe our technique of intravenous injection of a cocktail of peptide-conjugated PMOs (PPMOs) to skip multiple exons, exons 6 and 8, in both skeletal and cardiac muscles in dystrophic dogs and the evaluation of the efficacy and toxicity.


Assuntos
Distrofina/genética , Éxons , Morfolinos/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Miocárdio/metabolismo , Peptídeos , Splicing de RNA , Administração Intravenosa , Animais , Modelos Animais de Doenças , Cães , Distrofina/metabolismo , Expressão Gênica , Imuno-Histoquímica , Morfolinos/administração & dosagem , Morfolinos/química , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Peptídeos/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Methods Mol Biol ; 1828: 467-477, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171560

RESUMO

The application of antisense oligonucleotides (AONs) to modify pre-messenger RNA splicing has great potential for treating genetic diseases. The strategies used to redirect splicing for therapeutic purpose involve the use of AONs complementary to splice motifs, enhancer or silencer sequences. AONs to block intronic splicing silencer motifs can efficiently augment exon 7 inclusion in survival motor neuron 2 (SMN2) gene and have demonstrated robust therapeutic effects in both preclinical studies and clinical trials in spinal muscular atrophy (SMA), which has led to a recently approved drug. AONs with phosphorodiamidate morpholino oligomer (PMO) backbone have shown target engagement with restoration of the defective protein in Duchenne muscular dystrophy (DMD) and their safety profile lead to a recent conditional approval for one DMD PMO drug. PMO AONs are also effective in correcting SMN2 exon 7 splicing and rescuing SMA transgenic mice. Here we provide the details of methods that our lab has used to evaluate PMO-mediated SMN2 exon 7 inclusion in the in vivo studies conducted in SMA transgenic mice. The methods comprise mouse experiment procedures, assessment of PMOs on exon 7 inclusion at RNA levels by reverse transcription (RT-) PCR and quantitative real-time PCR. In addition, we present methodology for protein quantification using western blot in mouse tissues, on neuropathology assessment of skeletal muscle (muscle pathology and neuromuscular junction staining) as well as behaviour test in the SMA mice (righting reflex).


Assuntos
Éxons , Regulação da Expressão Gênica , Morfolinos/genética , Atrofia Muscular Espinal/genética , Splicing de RNA , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Processamento Alternativo , Animais , Animais Recém-Nascidos , Biópsia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Morfolinos/administração & dosagem , Neurônios Motores/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Junção Neuromuscular/metabolismo , Reflexo/genética
15.
Sci Transl Med ; 10(437)2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29669851

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal hereditary muscle disease caused by mutations in the gene encoding the muscle protein dystrophin. These mutations result in a shift in the open reading frame leading to loss of the dystrophin protein. Antisense oligonucleotides (ASOs) that induce exon skipping correct this frame shift during pre-mRNA splicing and partially restore dystrophin expression in mouse and dog models. We conducted a phase 1, open-label, dose-escalation clinical trial to determine the safety, pharmacokinetics, and activity of NS-065/NCNP-01, a morpholino ASO that enables skipping of exon 53. Ten patients with DMD (6 to 16 years old), carrying mutations in the dystrophin gene whose reading frame would be restored by exon 53 skipping, were administered NS-065/NCNP-01 at doses of 1.25, 5, or 20 mg/kg weekly for 12 weeks. The primary endpoint was safety; the secondary endpoints were pharmacokinetics and successful exon skipping. No severe adverse drug reactions were observed, and no treatment discontinuation occurred. Muscle biopsy samples were taken before and after treatment and compared by reverse transcription polymerase chain reaction (RT-PCR), immunofluorescence, and Western blotting to assess the amount of exon 53 skipping and dystrophin expression. NS-065/NCNP-01 induced exon 53 skipping in dystrophin-encoding mRNA in a dose-dependent manner and increased the dystrophin/spectrin ratio in 7 of 10 patients. Furthermore, the amount of exon skipping correlated with the maximum drug concentration in plasma (Cmax) and the area under the concentration-time curve in plasma (AUC0-t ). These results indicate that NS-065/NCNP-01 has a favorable safety profile and promising pharmacokinetics warranting further study in a phase 2 clinical trial.


Assuntos
Éxons/genética , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/uso terapêutico , Administração Intravenosa , Adolescente , Criança , Pré-Escolar , Distrofina/genética , Humanos , Masculino , Morfolinos/administração & dosagem , Morfolinos/uso terapêutico , Oligonucleotídeos Antissenso/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
ACS Infect Dis ; 4(5): 806-814, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29461800

RESUMO

The Burkholderia cepacia complex is a group of Gram-negative bacteria that are opportunistic pathogens in immunocompromised individuals, such as those with cystic fibrosis (CF) or chronic granulomatous disease (CGD). Burkholderia are intrinsically resistant to many antibiotics and the lack of antibiotic development necessitates novel therapeutics. Peptide-conjugated phosphorodiamidate morpholino oligomers are antisense molecules that inhibit bacterial mRNA translation. Targeting of PPMOs to the gene acpP, which is essential for membrane synthesis, lead to defects in the membrane and ultimately bactericidal activity. Exploration of additional PPMO sequences identified the ATG and Shine-Dalgarno sites as the most efficacious for targeting acpP. The CF lung is a complex microenvironment, but PPMO inhibition was still efficacious in an artificial model of CF sputum. PPMOs had low toxicity in human CF cells at doses that were antibacterial. PPMOs also reduced the bacterial burden in the lungs of immunocompromised CyBB mice, a model of CGD. Finally, the use of multiple PPMOs was efficacious in inhibiting the growth of both Burkholderia and Pseudomonas in an in vitro model of coinfection. Due to the intrinsic resistance of Burkholderia to traditional antibiotics, PPMOs represent a novel and viable approach to the treatment of Burkholderia infections.


Assuntos
Infecções por Burkholderia/microbiologia , Burkholderia/genética , Oligonucleotídeos Antissenso/genética , Pneumonia Bacteriana/microbiologia , Animais , Antibacterianos/administração & dosagem , Infecções por Burkholderia/terapia , Complexo Burkholderia cepacia/genética , Fibrose Cística/complicações , Modelos Animais de Doenças , Camundongos , Testes de Sensibilidade Microbiana , Morfolinos/administração & dosagem , Morfolinos/química , Morfolinos/genética , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/química , Pneumonia Bacteriana/terapia
17.
Int J Pharm ; 532(1): 21-28, 2017 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-28864392

RESUMO

Splice-switching antisense oligonucleotides (SSOs) are emerging therapeutics with two SSOs recently approved by the FDA for Duchenne muscular dystrophy and spinal muscular atrophy. SSOs are administered without any delivery vector and require large doses to achieve the therapeutic benefit, primarily due to their poor cellular uptake. Although cell-penetrating peptides (CPP) have shown great potential in delivering SSOs into cells, their capacity as delivery vector is limited. Here we have studied the effect of lipid conjugation on the cell permeability of a known CPP (ApoE). Myristic acid was coupled at the N-terminus of ApoE to a C-terminal cysteine residue. The myristoylated ApoE (Myr-ApoE) was conjugated to a maleimide functionalised phosphorodiamidate morpholino oligonucleotide (PMO). The Myr-ApoE-PMO conjugate showed no cytoxicity and had significantly higher efficiency in cell permeability with 30% higher splice-switching activity compared to ApoE-PMO. The self-assembly properties of this amphiphilic lipopeptide-PMO conjugate was assessed. Transmission electron microscopy showed formation of nanoparticles with amphiphile behaviour and spherical structure. The self-assembly of Myr-ApoE-PMO into nanoparticles enabled it to better bind to cell membranes and to be more efficiently taken up by fibroblast cells. These results showed that modification of physico-chemical properties of peptides to produce peptide amphiphiles enhances cellular uptake and can be used as an efficient delivery vector for therapeutic SSOs.


Assuntos
Apolipoproteínas E , Lipopeptídeos , Morfolinos , Ácido Mirístico , Nanopartículas , Apolipoproteínas E/administração & dosagem , Apolipoproteínas E/química , Transporte Biológico , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Humanos , Lipopeptídeos/administração & dosagem , Lipopeptídeos/química , Morfolinos/administração & dosagem , Morfolinos/química , Atrofia Muscular Espinal , Ácido Mirístico/administração & dosagem , Ácido Mirístico/química , Nanopartículas/administração & dosagem , Nanopartículas/química , Proteína 2 de Sobrevivência do Neurônio Motor/genética
18.
Cancer Gene Ther ; 24(8): 348-357, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28752860

RESUMO

The DNA repair enzyme O6-methylguanine DNA methyltransferase (MGMT) is epigenetically silenced in some tumors by MGMT gene promoter methylation. MGMT-hypermethylated solid tumors have enhanced susceptibility to the cytotoxic effects of alkylating chemotherapy such as temozolomide, compared with non-methylated tumors. In glioblastoma, subjects with MGMT hypermethylation have significantly longer survival rates after chemoradiotherapy. We report the first successful use of a non-ablative dose of ionizing radiation to prime human cancer cells to enhance the uptake of unmodified anti-MGMT morpholino oligonucleotide (AMON) sequences. We demonstrate >40% reduction in the in vitro proliferation index and cell viability in radiation-primed MGMT-expressing human solid tumor cells treated with a single dose of AMONs and temozolomide. We further demonstrate the feasibility of using a non-ablative dose of radiation in vivo to guide and enhance the delivery of intravenously administered AMONs to achieve 50% MGMT knockdown only at radiation-primed tumor sites in a subcutaneous tumor model. Local upregulation of physiological endocytosis after radiation may have a role in radiation-guided uptake of AMONs. This approach holds direct translational significance in glioblastoma and brain metastases where radiation is part of the standard of care; our approach to silence MGMT could overcome the significant problem of MGMT-mediated chemoresistance.


Assuntos
Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Neoplasias/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Proteínas Supressoras de Tumor/genética , Células A549 , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Quimiorradioterapia , Metilases de Modificação do DNA/biossíntese , Enzimas Reparadoras do DNA/biossíntese , Feminino , Humanos , Imuno-Histoquímica , Morfolinos/administração & dosagem , Morfolinos/genética , Morfolinos/farmacocinética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/radioterapia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacocinética , Ratos , Ratos Nus , Transfecção , Proteínas Supressoras de Tumor/biossíntese
19.
Methods Mol Biol ; 1565: 115-122, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28364238

RESUMO

Morpholino oligomers (MOs) are antisense molecules designed for sequence-specific binding of target mRNA. In bacteria, inhibition is hypothesized to occur by preventing translation initiation. Cell-penetrating peptides may be conjugated to the 5'- or 3'-termini of an MO to enhance cellular entry and therefore inhibition. Here we describe the three standard microbiological assays to assess in vitro antibacterial MO efficacy.


Assuntos
Bactérias/crescimento & desenvolvimento , Bactérias/genética , Morfolinos/química , Morfolinos/genética , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/genética , Peptídeos/química , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Peptídeos Penetradores de Células/química , Sinergismo Farmacológico , Testes de Sensibilidade Microbiana , Morfolinos/administração & dosagem
20.
Methods Mol Biol ; 1565: 241-250, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28364248

RESUMO

In vivo delivery of morpholino antisense oligonucleotides (MAO) directly into the uterine lumen of a peri-implantation period pregnant sheep is an effective technique for evaluation of gene products for conceptus development. The highly phagocytic conceptus is undergoing rapid morphological change, thereby the available MAO are readily consumed and delivered to developing cells. Here, we describe the method for preparation and surgical delivery of MAO-Endo-Porter complex to developing ovine conceptus on day 8 postmating. Also outlined are methods for posttreatment sample recovery on day 16 postmating.


Assuntos
Desenvolvimento Embrionário/genética , Técnicas de Transferência de Genes , Morfolinos/administração & dosagem , Morfolinos/genética , Oligonucleotídeos Antissenso , Útero , Animais , Feminino , Peptídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA