Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Gene Ther ; 30(7-8): 641-648, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36977769

RESUMO

Adeno-associated virus (AAV) vector-based gene therapy is potentially curative for various genetic diseases; however, the development of a scalable purification method for full-genome AAV vectors remains crucial to increase productivity and reduce cost of GMP production. In this study, we developed a large-scale short-term purification method for functional full-genome AAV particles by using 2-step cesium chloride (CsCl) density-gradient ultracentrifugation with a zonal rotor. The 2-step CsCl method with a zonal rotor improves separation between empty and full-genome AAV particles, reducing the ultracentrifugation time (4-5 h) and increasing the AAV volume for purification. The highly purified full-genome AAV particles were confirmed by analytical ultracentrifugation (AUC), droplet digital PCR (ddPCR) in the whole region of the AAV vector genome, transduction efficiency in target cells, and transmission electronic microscopy (TEM). The high-purity AAV9 particles were obtained using culture supernatant during vector preparation rather than cell lysate. CsCl could be simply removed by a hydroxyapatite column. Interestingly, ddPCR analysis revealed that "empty" AAV particles contain small fragments of the inverted terminal repeat (ITR), probably due to unexpected packaging of Rep-mediated ITR fragments. This large-scale functional AAV vector purification with ultracentrifugation would be effective for gene therapy.


Assuntos
Dependovirus , Vetores Genéticos , Ultracentrifugação , Dependovirus/genética
2.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35163475

RESUMO

Duchenne muscular dystrophy (DMD) is caused by loss-of-function mutations in the dystrophin gene on chromosome Xp21. Disruption of the dystrophin-glycoprotein complex (DGC) on the cell membrane causes cytosolic Ca2+ influx, resulting in protease activation, mitochondrial dysfunction, and progressive myofiber degeneration, leading to muscle wasting and fragility. In addition to the function of dystrophin in the structural integrity of myofibers, a novel function of asymmetric cell division in muscular stem cells (satellite cells) has been reported. Therefore, it has been suggested that myofiber instability may not be the only cause of dystrophic degeneration, but rather that the phenotype might be caused by multiple factors, including stem cell and myofiber functions. Furthermore, it has been focused functional regulation of satellite cells by intracellular communication of extracellular vesicles (EVs) in DMD pathology. Recently, a novel molecular mechanism of DMD pathogenesis-circulating RNA molecules-has been revealed through the study of target pathways modulated by the Neutral sphingomyelinase2/Neutral sphingomyelinase3 (nSMase2/Smpd3) protein. In addition, adeno-associated virus (AAV) has been clinically applied for DMD therapy owing to the safety and long-term expression of transduction genes. Furthermore, the EV-capsulated AAV vector (EV-AAV) has been shown to be a useful tool for the intervention of DMD, because of the high efficacy of the transgene and avoidance of neutralizing antibodies. Thus, we review application of AAV and EV-AAV vectors for DMD as novel therapeutic strategy.


Assuntos
Vesículas Extracelulares/virologia , Distrofia Muscular de Duchenne/terapia , Células Satélites de Músculo Esquelético/metabolismo , Esfingomielina Fosfodiesterase/genética , Animais , Ácidos Nucleicos Livres/genética , Dependovirus/genética , Vesículas Extracelulares/genética , Vesículas Extracelulares/transplante , Terapia Genética , Vetores Genéticos , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Transdução Genética
3.
Calcif Tissue Int ; 106(6): 665-677, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32076747

RESUMO

Hypophosphatasia (HPP) is a systemic skeletal disease caused by mutations in the gene encoding tissue-nonspecific alkaline phosphatase (TNALP). We recently reported that survival of HPP model mice can be prolonged using an adeno-associated virus (AAV) vector expressing bone-targeted TNALP with deca-aspartate at the C terminus (TNALP-D10); however, abnormal bone structure and hypomineralization remained in the treated mice. Here, to develop a more effective and clinically applicable approach, we assessed whether transfection with TNALP-D10 expressing virus vector at a higher dose than previously used would ameliorate bone structure defects. We constructed a self-complementary AAV8 vector expressing TNALP driven by the chicken beta-actin (CBA) promoter (scAAV8-CB-TNALP-D10). The vector was injected into both quadriceps femoris muscles of newborn HPP mice at a dose of 4.5 × 1012 vector genome (v.g.)/body, resulting in 20 U/mL of serum ALP activity. The 4.5 × 1012 v.g./body-treated HPP mice grew normally and displayed improved bone structure at the knee joints in X-ray images. Micro-CT analysis showed normal trabecular bone structure and mineralization. The mechanical properties of the femur were also recovered. Histological analysis of the femurs demonstrated that ALP replacement levels were sufficient to promote normal, growth plate cartilage arrangement. These results suggest that AAV vector-mediated high-dose TNALP-D10 therapy is a promising option for improving the quality of life (QOL) of patients with the infantile form of HPP.


Assuntos
Fosfatase Alcalina/genética , Osso Esponjoso/patologia , Hipofosfatasia/terapia , Animais , Dependovirus , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos , Camundongos , Qualidade de Vida
4.
Mol Vis ; 22: 816-26, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27440998

RESUMO

PURPOSE: We examined the neuroprotective effects of exogenous brain-derived neurotrophic factor (BDNF), which provides protection to retinal ganglion cells (RGCs) in rodents, in a model of transient intraocular pressure (IOP) elevation using a mutant (triple Y-F) self-complementary adeno-associated virus type 2 vector encoding BDNF (tm-scAAV2-BDNF). METHODS: The tm-scAAV2-BDNF or control vector encoding green fluorescent protein (GFP; tm-scAAV2-GFP) was intravitreally administered to rats, which were then divided into four groups: control, ischemia/reperfusion (I/R) injury only, I/R injury with tm-scAAV2-GFP, and tm-scAAV2-BDNF. I/R injury was then induced by transiently increasing IOP, after which the rats were euthanized to measure the inner retinal thickness and cell counts in the RGC layer. RESULTS: Intravitreous injection of tm-scAAV2-BDNF resulted in high levels of BDNF expression in the neural retina. Histological analysis showed that the inner retinal thickness and cell numbers in the RGC layer were preserved after transient IOP elevation in eyes treated with tm-scAAV2-BDNF but not in the other I/R groups. Significantly reduced glial fibrillary acidic protein (GFAP) immunostaining after I/R injury in the rats that received tm-scAAV2-BDNF indicated reduced retinal stress, and electroretinogram (ERG) analysis confirmed preservation of retinal function in the tm-scAAV2-BDNF group. CONCLUSIONS: These results demonstrate the feasibility and effectiveness of neuroprotective gene therapy using tm-scAAV2-BDNF to protect the inner retina from transiently high intraocular pressure. An in vivo gene therapeutic approach to the clinical management of retinal diseases in conditions such as glaucoma, retinal artery occlusion, hypertensive retinopathy, and diabetic retinopathy thus appears feasible.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Dependovirus/metabolismo , Pressão Intraocular , Mutação/genética , Tirosina/genética , Animais , Contagem de Células , Modelos Animais de Doenças , Eletrorretinografia , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Ratos Sprague-Dawley , Retina/lesões , Retina/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transdução Genética
5.
Mol Ther ; 22(2): 409-419, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24322332

RESUMO

Gene therapy for neuropathic pain requires efficient gene delivery to both central and peripheral nervous systems. We previously showed that an adenoassociated virus serotype 9 (AAV9) vector expressing short-hairpin RNA (shRNA) could suppress target molecule expression in the dorsal root ganglia (DRG) and spinal cord upon intrathecal injection. To evaluate the therapeutic potential of this approach, we constructed an AAV9 vector encoding shRNA against vanilloid receptor 1 (TRPV1), which is an important target gene for acute pain, but its role in chronic neuropathic pain remains unclear. We intrathecally injected it into the subarachnoid space at the upper lumbar spine of mice 3 weeks after spared nerve injury (SNI). Delivered shTRPV1 effectively suppressed mRNA and protein expression of TRPV1 in the DRG and spinal cord, and it attenuated nerve injury-induced thermal allodynia 10-28 days after treatment. Our study provides important evidence for the contribution of TRPV1 to thermal hypersensitivity in neuropathic pain and thus establishes intrathecal AAV9-mediated gene delivery as an investigative and potentially therapeutic platform for the nervous system.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Hiperalgesia/genética , RNA Interferente Pequeno/genética , Canais de Cátion TRPV/genética , Animais , Sequência de Bases , Dependovirus/imunologia , Modelos Animais de Doenças , Feminino , Gânglios Espinais/metabolismo , Expressão Gênica , Ordem dos Genes , Inativação Gênica , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Hiperalgesia/terapia , Injeções Espinhais , Camundongos , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/terapia , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/química , RNA Interferente Pequeno/metabolismo , Medula Espinal/metabolismo , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo
6.
Mol Pain ; 9: 36, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23866078

RESUMO

BACKGROUND: There is considerable interest in inducing RNA interference (RNAi) in neurons to study gene function and identify new targets for disease intervention. Although short interfering RNAs (siRNAs) have been used to silence genes in neurons, in vivo delivery of RNAi remains a major challenge, especially by systemic administration. We have developed a highly efficient method for in vivo gene silencing in dorsal root ganglia (DRG) by using short hairpin RNA-expressing single-stranded adeno-associated virus 9 (ssAAV9-shRNA). RESULTS: Intraperitoneal administration of ssAAV9-shRNA to neonatal mice resulted in highly effective and specific silencing of a target gene in DRG. We observed an approximately 80% reduction in target mRNA in the DRG, and 74.7% suppression of the protein was confirmed by Western blot analysis. There were no major side effects, and the suppression effect lasted for more than three months after the injection of ssAAV9-shRNA. CONCLUSIONS: Although we previously showed substantial inhibition of target gene expression in DRG via intrathecal ssAAV9-shRNA administration, here we succeeded in inhibiting target gene expression in DRG neurons via intraperitoneal injection of ssAAV9-shRNA. AAV9-mediated delivery of shRNA will pave the way for creating animal models for investigating the molecular biology of the mechanisms of pain and sensory ganglionopathies.


Assuntos
Dependovirus/genética , Gânglios Espinais/metabolismo , RNA Interferente Pequeno/genética , Animais , Linhagem Celular , Dependovirus/metabolismo , Expressão Gênica , Inativação Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos ICR , Modelos Animais , Neurônios/metabolismo , Dor/genética , Dor/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
7.
Brain ; 135(Pt 3): 833-46, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22252998

RESUMO

Amyotrophic lateral sclerosis is a fatal neurodegenerative disease characterized by progressive motoneuron loss. Redistribution of transactive response deoxyribonucleic acid-binding protein 43 from the nucleus to the cytoplasm and the presence of cystatin C-positive Bunina bodies are considered pathological hallmarks of amyotrophic lateral sclerosis, but their significance has not been fully elucidated. Since all reported rodent transgenic models using wild-type transactive response deoxyribonucleic acid-binding protein 43 failed to recapitulate these features, we expected a species difference and aimed to make a non-human primate model of amyotrophic lateral sclerosis. We overexpressed wild-type human transactive response deoxyribonucleic acid-binding protein 43 in spinal cords of cynomolgus monkeys and rats by injecting adeno-associated virus vector into the cervical cord, and examined the phenotype using behavioural, electrophysiological, neuropathological and biochemical analyses. These monkeys developed progressive motor weakness and muscle atrophy with fasciculation in distal hand muscles first. They also showed regional cytoplasmic transactive response deoxyribonucleic acid-binding protein 43 mislocalization with loss of nuclear transactive response deoxyribonucleic acid-binding protein 43 staining in the lateral nuclear group of spinal cord innervating distal hand muscles and cystatin C-positive cytoplasmic aggregates, reminiscent of the spinal cord pathology of patients with amyotrophic lateral sclerosis. Transactive response deoxyribonucleic acid-binding protein 43 mislocalization was an early or presymptomatic event and was later associated with neuron loss. These findings suggest that the transactive response deoxyribonucleic acid-binding protein 43 mislocalization leads to α-motoneuron degeneration. Furthermore, truncation of transactive response deoxyribonucleic acid-binding protein 43 was not a prerequisite for motoneuronal degeneration, and phosphorylation of transactive response deoxyribonucleic acid-binding protein 43 occurred after degeneration had begun. In contrast, similarly prepared rat models expressed transactive response deoxyribonucleic acid-binding protein 43 only in the nucleus of motoneurons. There is thus a species difference in transactive response deoxyribonucleic acid-binding protein 43 pathology, and our monkey model recapitulates amyotrophic lateral sclerosis pathology to a greater extent than rodent models, providing a valuable tool for studying the pathogenesis of sporadic amyotrophic lateral sclerosis.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Animais , Atrofia , Comportamento Animal/fisiologia , Western Blotting , Cistatina C/metabolismo , Dependovirus , Modelos Animais de Doenças , Eletromiografia , Fenômenos Eletrofisiológicos , Vetores Genéticos , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/metabolismo , Debilidade Muscular/genética , Debilidade Muscular/patologia , Neuritos/patologia , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Especificidade da Espécie , Medula Espinal/metabolismo , Técnicas Estereotáxicas
8.
Biochem Biophys Res Commun ; 405(2): 204-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21219850

RESUMO

Systemic injections of AAV vectors generally transduce to the liver more effectively than to cardiac and skeletal muscles. The short hairpin RNA (shRNA)-expressing AAV9 (shRNA-AAV9) can also reduce target gene expression in the liver, but not enough in cardiac or skeletal muscles. Higher doses of shRNA-AAV9 required for inhibiting target genes in cardiac and skeletal muscles often results in shRNA-related toxicity including microRNA oversaturation that can induce fetal liver failure. In this study, we injected high-dose shRNA-AAV9 to neonates and efficiently silenced genes in cardiac and skeletal muscles without inducing liver toxicity. This is because AAV is most likely diluted or degraded in the liver than in cardiac or skeletal muscle during cell division after birth. We report that this systemically injected shRNA-AAV method does not induce any major side effects, such as liver dysfunction, and the dose of shRNA-AAV is sufficient for gene silencing in skeletal and cardiac muscle tissues. This novel method may be useful for generating gene knockdown in skeletal and cardiac mouse tissues, thus providing mouse models useful for analyzing diseases caused by loss-of-function of target genes.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Animais , Dependovirus , Vetores Genéticos/administração & dosagem , Células HEK293 , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos ICR , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase/genética , Superóxido Dismutase-1
9.
Cancer Invest ; 29(5): 353-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21599511

RESUMO

To determine the adeno-associated virus (AAV) serotype that most efficiently mediates muscle expression of antiangiogenic proteins, we injected four different serotype (1, 2, 7, and 8) AAV vectors encoding mouse endostatin (mEnd) or human soluble FLK-1 (hsFLK-1) into a quadriceps muscle of C57BL/6 mice. The highest plasma levels of therapeutic protein were observed in AAV8-injected mice (8 > 7 > 1 > 2). Sustained expression of mEnd was detected for 6 months, whereas concentrations of hsFLK-1 declined to the background level within 2 weeks caused by neutralizing anti-hsFLK-1 antibody. These data demonstrate that AAV8 (mEnd) serotype is the most efficient mediator for protein expression.


Assuntos
Dependovirus/metabolismo , Endostatinas/biossíntese , Técnicas de Transferência de Genes , Vetores Genéticos , Músculo Quadríceps/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Animais , Anticorpos/sangue , Creatina Quinase Forma MM/genética , Dependovirus/classificação , Dependovirus/genética , Endostatinas/sangue , Endostatinas/genética , Ensaio de Imunoadsorção Enzimática , Células HeLa , Humanos , Injeções Intramusculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Regiões Promotoras Genéticas , Sorotipagem , Fatores de Tempo , Transfecção , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
10.
Mol Genet Metab ; 96(3): 91-6, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19091614

RESUMO

Fabry disease is caused by the deficiency of lysosomal alpha-galactosidase A (alpha-gal A) and usually develops clinical manifestations during childhood/adolescence. Adult Fabry model mice have been successfully treated by various viral vectors. Here, in order to examine the feasibility of preventive gene therapy, we compared AAV vector-mediated gene transfer into neonatal and adult model mice. AAV serotype 1 vector (AAV1) carrying human alpha-gal A cDNA driven by the CAG promoter was intravenously injected into adult (12 weeks old) and neonatal (2 days old) Fabry model mice, and were sacrificed for detailed examination 25 weeks after vector injection. AAV1 vector preferentially transduced the liver in male adult and sustained high concentration of alpha-gal A was detected in the liver, heart and plasma. In contrast, AAV1-mediated gene expression was suppressed in similarly treated female adult mice. When the vector was systemically injected into neonates, moderate increase in plasma alpha-gal A and cardiac-specific expression of alpha-gal A were observed independently of mouse sex. The high levels of alpha-gal A activity in the heart appear to be due to the strong activity of the CAG promoter in the heart. Globotriaosylceramide (Gb3) accumulation was efficiently inhibited in the liver and heart by a single injection into both adult and neonatal animals. The biodistribution of the AAV1 vector and levels of alpha-gal A expression are markedly different between adult and neonatal mice. Neonatal injection is effective to inhibit Gb3 accumulation and therefore, might help prevent failure of major organs during adulthood.


Assuntos
Dependovirus/genética , Doença de Fabry/genética , Doença de Fabry/terapia , Terapia Genética , Glicoesfingolipídeos/metabolismo , alfa-Galactosidase/administração & dosagem , Animais , Animais Recém-Nascidos , Dependovirus/metabolismo , Doença de Fabry/metabolismo , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Triexosilceramidas/metabolismo , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
11.
Hum Gene Ther Methods ; 30(4): 137-143, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31317781

RESUMO

Recombinant adeno-associated virus (rAAV) is a promising gene delivery vehicle that has been approved as a gene therapy drug for some genetic disorders, and is being evaluated in clinical trials. To further promote clinical research under the Food and Drug Administration Investigational New Drug application, the stability of rAAV must be assessed under various conditions. However, there is scant data concerning the stability of a variety of rAAV serotypes. We hypothesized that the difference of capsid structure causes differences in stability. To investigate this hypothesis, rAAV serotypes (rAAV1, rAAV2, rAAV8, and rAAV9) were exposed to diluents and various environmental conditions, including ultraviolet (UV) irradiation, 0.1 M sodium hydroxide (NaOH), 0.06% sodium hypochlorite (NaClO), tap water, and 70% ethanol (EtOH). The changes of the infectivity of the treated samples were assessed by transduction in HeLaRC32 cells as a criterion of stability. The infectivity between recombinant and wild-type AAV (wtAAV2) was also analyzed. The activity of all rAAV serotypes was weakened by UV irradiation and NaOH and NaClO exposure. Treatment for 10 days with tap water or 70% EtOH did not appreciably inactivate rAAV1, rAAV8, and rAAV9, but did affect the activity of rAAV2. Furthermore, the infectivity of rAAV2 did not surpass wtAAV2 infectivity. The results will be important for clinical studies for gene therapy using rAAV.


Assuntos
Dependovirus , Vetores Genéticos , Dependovirus/efeitos dos fármacos , Dependovirus/genética , Dependovirus/patogenicidade , Dependovirus/efeitos da radiação , Terapia Genética , Células HEK293 , Humanos , Hidróxido de Sódio/farmacologia , Hipoclorito de Sódio/farmacologia , Raios Ultravioleta , Inativação de Vírus/efeitos dos fármacos , Inativação de Vírus/efeitos da radiação , Água/farmacologia
12.
Mol Ther ; 15(1): 38-43, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164773

RESUMO

Metachromatic leukodystrophy (MLD) is a lysosomal storage disorder caused by a deficiency of arylsulfatase A (ASA) and is characterized by deposition of sulfatide in all organs, particularly the nervous system. Recently, formylglycine-generating enzyme (FGE) was found to be essential for activation of sulfatases. This study examined the utility of FGE co-expression in AAV type 1 vector (AAV1)-mediated gene therapy of ASA knockout (MLD) mice. AAV1-ASA alone or AAV1-ASA and AAV1-FGE were co-injected into a single site of the hippocampus. Enzyme assay and immunohistochemical analysis showed that ASA was detected not only in the injected hemisphere but also in the non-injected hemisphere by 7 months after injection. Level of ASA activity and extent of ASA distribution were significantly enhanced by co-introduction of AAV1-FGE. Marked reductions in sulfatide levels were observed throughout the entire brain. The unexpectedly widespread distribution of ASA may be due to a combination of diffusion in extracellular spaces, transport through axons, and circulation in cerebrospinal fluid. The rotarod test revealed improvement of neurological functions. These results demonstrate that direct injection of AAV1 vectors expressing ASA and FGE represents a highly promising approach with significant implications for the development of clinical protocols for MLD gene therapy.


Assuntos
Arilsulfatases/metabolismo , Dependovirus/genética , Regulação Enzimológica da Expressão Gênica , Terapia Genética , Glicina/biossíntese , Leucodistrofia Metacromática/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Animais , Arilsulfatases/genética , Modelos Animais de Doenças , Vetores Genéticos/genética , Glicina/análogos & derivados , Humanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/patologia , Leucodistrofia Metacromática/terapia , Camundongos
13.
Mol Ther Methods Clin Dev ; 11: 180-190, 2018 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-30533449

RESUMO

Recombinant adeno-associated virus serotype 9 (rAAV9) can specifically transduce muscle and neuronal tissues; thus, rAAV9 can potentially be used in gene therapy. However, rAAV9 is the most challenging rAAV serotype to purify. Traditionally, rAAV9 has been purified by ultracentrifugation, which is not scalable. We recently described a chromatographic purification protocol for rAAV1; this protocol can achieve scalable purifications. In this study, we attempted to optimize this protocol for purifying rAAV9 preparations, and we developed a novel, effective method for high-yield purification of rAAV9 using quaternary ammonium anion exchangers and size-exclusion chromatography. The final purified rAAV9 contained mainly three capsid proteins, as observed by SDS-PAGE. Furthermore, negative-stain electron microscopy demonstrated that 96.1% ± 1.1% of rAAV9 particles carried the viral genome containing the EGFP transgene, indicating that impurities and empty capsids can be eliminated with our purification protocol. The final rAAV9 titer obtained by our protocol totaled 2.5 ± 0.4 × 1015 viral genomes produced from ∼3.2 × 109 HEK293EB cells. We confirmed that our protocol can also be applied to purify other varied AAV genome constructs. Our protocol can scale up production of pure rAAV9, in compliance with current good manufacturing practice, for clinical applications in human gene therapy.

14.
Mol Ther Methods Clin Dev ; 3: 15058, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26913289

RESUMO

Recombinant adeno-associated virus (rAAV) is an attractive tool for gene transfer and shows potential for use in human gene therapies. The current methods for the production and purification of rAAV from the transfected cell lysate are mainly based on cesium chloride and iodixanol density ultracentrifugation, although those are not scalable. Meanwhile, chromatography-based systems are more scalable. Therefore, in this study, we developed a novel method for the production and purification of rAAV serotype 1 (rAAV1) from serum-free culture supernatant based on ion-exchange and gel-filtration chromatography to obtain highly purified products with an ultracentrifugation-free technique towards Good Manufacturing Practice (GMP) production. The purified rAAV1 displayed three clear and sharp bands (VP1, VP2, and VP3) following sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and more than 90% of rAAV1 particles contained fully packaged viral genomes according to negative-stain electron micrographic analysis. Consequently, the resultant genomic titer of the purified rAAV1 was 3.63 × 10(13) v.g./ml (the total titer was 4.17 × 10(13) v.g.) from the 4 × 10(9) HEK293 cells. This novel chromatography-based method will facilitate scale-up of manufacturing for clinical applications in gene therapy.

15.
Mol Ther Methods Clin Dev ; 3: 15059, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26904710

RESUMO

Hypophosphatasia (HPP) is an inherited disease caused by genetic mutations in the gene encoding tissue-nonspecific alkaline phosphatase (TNALP). This results in defects in bone and tooth mineralization. We recently demonstrated that TNALP-deficient (Akp2 (-/-) ) mice, which mimic the phenotype of the severe infantile form of HPP, can be treated by intravenous injection of a recombinant adeno-associated virus (rAAV) expressing bone-targeted TNALP with deca-aspartates at the C-terminus (TNALP-D10) driven by the tissue-nonspecific CAG promoter. To develop a safer and more clinically applicable transduction strategy for HPP gene therapy, we constructed a self-complementary type 8 AAV (scAAV8) vector that expresses TNALP-D10 via the muscle creatine kinase (MCK) promoter (scAAV8-MCK-TNALP-D10) and examined the efficacy of muscle-directed gene therapy. When scAAV8-MCK-TNALP-D10 was injected into the bilateral quadriceps of neonatal Akp2 (-/-) mice, the treated mice grew well and survived for more than 3 months, with a healthy appearance and normal locomotion. Improved bone architecture, but limited elongation of the long bone, was demonstrated on X-ray images. Micro-CT analysis showed hypomineralization and abnormal architecture of the trabecular bone in the epiphysis. These results suggest that rAAV-mediated, muscle-specific expression of TNALP-D10 represents a safe and practical option to treat the severe infantile form of HPP.

16.
Hum Gene Ther ; 16(8): 929-36, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16076251

RESUMO

Metachromatic leukodystrophy (MLD) is a lysosomal storage disorder involving inherited deficiency of arylsulfatase A (ASA). The disease is characterized by progressive demyelination and widespread deposition of sulfatide in both the central and peripheral nervous systems. Direct injection of viral vector through the blood-brain barrier is a possible gene therapy approach to MLD. However, to treat all brain cells, it is essential to secrete a sufficient amount of functional ASA from limited numbers of transduced cells. In the present study, we tested the utility of formylglycine-generating enzyme (FGE) for overexpression of functional ASA. FGE is a posttranslational modifying enzyme essential for activating multiple forms of sulfatases including ASA. COS-7 cells were transfected with ASA- and FGE-expressing plasmids. ASA activity was increased up to 20-fold in cell lysates and 70-fold in conditioned medium by coexpression of FGE. Intravenous injection of the expression plasmids into MLD knockout mice by a hydrodynamics-based procedure resulted in a significant synergistic increase in ASA activity both in liver and serum. Blot hybridization analysis of FGE mRNA demonstrated that the expression of endogenous FGE was particularly low in human brain. Our results suggest, on the basis of cross-correction of ASA deficiency, that coexpression of FGE is essential for gene therapy of MLD.


Assuntos
Alanina/análogos & derivados , Cerebrosídeo Sulfatase/biossíntese , Cerebrosídeo Sulfatase/genética , Terapia Genética/métodos , Glicina/análogos & derivados , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/terapia , Sulfatases/metabolismo , Alanina/biossíntese , Animais , Barreira Hematoencefálica , Células COS , Cerebrosídeo Sulfatase/deficiência , Chlorocebus aethiops , Técnicas de Transferência de Genes , Glicina/biossíntese , Injeções Intravenosas , Leucodistrofia Metacromática/fisiopatologia , Leucodistrofia Metacromática/veterinária , Camundongos , Camundongos Knockout , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Plasmídeos , Sulfatases/genética , Transdução Genética , Transfecção
17.
J Nippon Med Sch ; 72(5): 254-61, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16247224

RESUMO

Fabry disease is an inherited lysosomal storage disorder characterized by a pathological intracellular glycosphingolipid deposition. The disease is caused by a deficit in the lysosomal enzyme alpha-galatosidase A, the gene for which is located in the X chrosomal region Xq 22. Globotriaosylceramide (Gb3) accumulate progressively in multi-organ vulnerable cells throughout the body, including cardiovascular, renal, and cerebrovascular systems. The present manuscript is to review cardiovascular and renal manifestations of Fabry disease and the new diagnostic procedures for earlier detection and the therapeutic assessments of this disease. We are applying noninvasive cardiovascular and microcirculation analysis methods and novel cardiac biomarkers. Novel therapeutic strategies for this disease have been developing in recent years, which include the clinically introduced enzyme infusion replacement therapy and experimentally developing gene-transfer therapy. We have reported that AAV-mediated muscule-directed gene transfer is very effective for long-term systemic delivery of alpha-gal A (25% of normal mice enzyme activity), resulting in complete clearance of multi-organs Gb3 accumulation. Echocardiographic and immunohistochemical examination demonstrated structural improvement of cardiac hypertrophy. When and to whom the novel therapeutic strategies should be applied to obtain the maximum efficacy and safety remain to be established.


Assuntos
Doença de Fabry/complicações , Doença de Fabry/terapia , Técnicas de Transferência de Genes , Hipertrofia Ventricular Esquerda/etiologia , Nefropatias/etiologia , Adolescente , Adulto , Humanos , Pessoa de Meia-Idade , alfa-Galactosidase/uso terapêutico
18.
Sci Rep ; 5: 13104, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26283284

RESUMO

Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by a functional deficiency in human arylsulfatase A (hASA). We recently reported that ependymal cells and the choroid plexus are selectively transduced by intracerebroventricular (ICV) injection of adeno-associated virus serotype 1 (AAV1) vector and serve as a biological reservoir for the secretion of lysosomal enzymes into the cerebrospinal fluid (CSF). In the present study, we examined the feasibility of this AAV-mediated gene therapy to treat MLD model mice. Preliminary experiments showed that the hASA level in the CSF after ICV injection of self-complementary (sc) AAV1 was much higher than in mice injected with single-stranded AAV1 or scAAV9. However, when 18-week-old MLD mice were treated with ICV injection of scAAV1, the concentration of hASA in the CSF gradually decreased and was not detectable at 12 weeks after injection, probably due to the development of anti-hASA antibodies. As a result, the sulfatide levels in brain tissues of treated MLD mice were only slightly reduced compared with those of untreated MLD mice. These results suggest that this approach is potentially promising for treating MLD, but that controlling the immune response appears to be crucial for long-term expression of therapeutic proteins in the CSF.


Assuntos
Adenoviridae/genética , Cerebrosídeo Sulfatase/administração & dosagem , Líquido Cefalorraquidiano/metabolismo , Terapia Genética/métodos , Leucodistrofia Metacromática/enzimologia , Leucodistrofia Metacromática/terapia , Animais , Cerebrosídeo Sulfatase/genética , Terapia de Reposição de Enzimas/métodos , Vetores Genéticos/genética , Injeções Intraventriculares , Leucodistrofia Metacromática/líquido cefalorraquidiano , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Resultado do Tratamento
19.
Sci Rep ; 4: 5506, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24981028

RESUMO

Enzyme replacement via the cerebrospinal fluid (CSF) has been shown to ameliorate neurological symptoms in model animals with neuropathic metabolic disorders. Gene therapy via the CSF offers a means to achieve a long-term sustainable supply of therapeutic proteins within the central nervous system (CNS) by setting up a continuous source of transgenic products. In the present study, a serotype 1 adeno-associated virus (AAV1) vector was injected into a lateral cerebral ventricle in adult mice to transduce the gene encoding human lysosomal enzyme arylsulfatase A (hASA) into the cells of the CNS. Widespread transduction and stable expression of hASA in the choroid plexus and ependymal cells was observed throughout the ventricles for more than 1 year after vector injection. Although humoral immunity to hASA developed after 6 weeks, which diminished the hASA levels detected in CSF from AAV1-injected mice, hASA levels in CSF were maintained for at least 12 weeks when the mice were tolerized to hASA prior of vector injection. Our results suggest that the cells lining the ventricles could potentially serve as a biological reservoir for long-term continuous secretion of lysosomal enzymes into the CSF following intracerebroventricular injection of an AAV1 vector.


Assuntos
Adenoviridae/genética , Cerebrosídeo Sulfatase/líquido cefalorraquidiano , Cerebrosídeo Sulfatase/genética , Epêndima/fisiologia , Marcação de Genes/métodos , Vetores Genéticos/genética , Animais , Epêndima/citologia , Injeções Intraventriculares , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos C57BL , Transfecção/métodos
20.
J Nippon Med Sch ; 79(6): 394-402, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23291836

RESUMO

A variety of gene transfer strategies have been developed to treat inherited, degenerative, and acquired diseases. Among the different vector systems developed so far, recombinant adeno-associated viral (AAV) vectors have shown notable benefits, including prolonged gene expression, transduction of both dividing and nondividing cells, and a lack of pathogenicity caused by wild-type infections. Thanks to these features, the use of AAV vectors as a gene transfer tool has increased dramatically during the past several years, and several recent clinical trials have used AAV vectors. However, AAV vectors are more complicated to produce than are other viral vectors. With steady advances toward clinical application, much effort has been made to isolate novel AAV serotypes and to develop methods for their efficient, scalable, and versatile production and purification. Here we review state of the art methods for AAV vector production and purification, which we have refined in our laboratory.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Transdução Genética/métodos , Animais , Dependovirus/classificação , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Sorotipagem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa