Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Mol Ther Methods Clin Dev ; 8: 8-20, 2018 Mar 16.
Article in English | MEDLINE | ID: mdl-29349096

ABSTRACT

Recombinant adeno-associated virus (rAAV)-mediated gene delivery shows promise to transduce the pancreas, but safety/efficacy in a neoplastic context is not well established. To identify an ideal AAV serotype, route, and vector dose and assess safety, we have investigated the use of three AAV serotypes (6, 8, and 9) expressing GFP in a self-complementary (sc) AAV vector under an EF1α promoter (scAAV.GFP) following systemic or retrograde pancreatic intraductal delivery. Systemic delivery of scAAV9.GFP transduced the pancreas with high efficiency, but gene expression did not exceed >45% with the highest dose, 5 × 1012 viral genomes (vg). Intraductal delivery of 1 × 1011 vg scAAV6.GFP transduced acini, ductal cells, and islet cells with >50%, ∼48%, and >80% efficiency, respectively, and >80% pancreatic transduction was achieved with 5 × 1011 vg. In a KrasG12D-driven pancreatic cancer mouse model, intraductal delivery of scAAV6.GFP targeted acini, epithelial, and stromal cells and exhibited persistent gene expression 5 months post-delivery. In normal mice, intraductal delivery induced a transient increase in serum amylase/lipase that resolved within a day of infusion with no sustained pancreatic inflammation or fibrosis. Similarly, in PDAC mice, intraductal delivery did not increase pancreatic intraepithelial neoplasia progression/fibrosis. Our study demonstrates that scAAV6 targets the pancreas/neoplasm efficiently and safely via retrograde pancreatic intraductal delivery.

2.
Hum Gene Ther ; 28(5): 392-402, 2017 05.
Article in English | MEDLINE | ID: mdl-28056565

ABSTRACT

Adeno-associated viral (AAV) vectors are promising vehicles for hemophilia gene therapy, with favorable clinical trial data seen in the treatment of hemophilia B. In an effort to optimize the expression of human coagulation factor VIII (hFVIII) for the treatment of hemophilia A, an extensive study was performed with numerous combinations of liver-specific promoter and enhancer elements with a codon-optimized hFVIII transgene. After generating 42 variants of three reduced-size promoters and three small enhancers, transgene cassettes were packaged within a single AAV capsid, AAVrh10, to eliminate performance differences due to the capsid type. Each hFVIII vector was administered to FVIII knockout (KO) mice at a dose of 1010 genome copies (GC) per mouse. Criteria for distinguishing the performance of the different enhancer/promoter combinations were established prior to the initiation of the studies. These criteria included prominently the level of hFVIII activity (0.12-2.12 IU/mL) and the pattern of development of anti-hFVIII antibodies. In order to evaluate the impact of capsid on hFVIII expression and antibody formation, one of the enhancer and promoter combinations that exhibited high hFVIII immunogenicity was evaluated using AAV8, AAV9, AAVrh10, AAVhu37, and AAVrh64R1 capsids. The capsids subdivided into two groups: those that generated anti-hFVIII antibodies in ≤20% of mice (AAV8 and AAV9), and those that generated anti-hFVIII antibodies in >20% of mice (AAVrh10, AAVhu37, and AAVrh64R1). The results of this study, which entailed extensive vector optimization and in vivo testing, demonstrate the significant impact that transcriptional control elements and capsid can have on vector performance.


Subject(s)
Factor VIII/genetics , Genetic Therapy , Genetic Vectors/therapeutic use , Hemophilia A/therapy , Animals , Capsid Proteins/genetics , Capsid Proteins/immunology , Dependovirus/genetics , Disease Models, Animal , Factor VIII/therapeutic use , Gene Expression Regulation , Gene Transfer Techniques , Hemophilia A/genetics , Humans , Immune Tolerance/genetics , Liver/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic , Transgenes/immunology
3.
Hum Gene Ther ; 27(12): 947-961, 2016 12.
Article in English | MEDLINE | ID: mdl-27897038

ABSTRACT

The field of adeno-associated virus (AAV) gene therapy has progressed rapidly over the past decade, with the advent of novel capsid serotype and organ-specific promoters, and an increasing understanding of the immune response to AAV administration. In particular, liver-directed therapy has made remarkable strides, with a number of clinical trials currently planned and ongoing in hemophilia A and B, as well as other liver disorders. This review focuses on liver-directed AAV gene therapy, including historic context, current challenges, and future developments.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Liver Diseases/genetics , Liver Diseases/therapy , Animals , Humans
4.
Hum Gene Ther ; 26(10): 647-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26076707

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene. It is the most common, severe childhood form of muscular dystrophy. We investigated an alternative to dystrophin replacement by overexpressing ITGA7 using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal muscle that, like the dystrophin-glycoprotein complex, links the extracellular matrix to the internal actin cytoskeleton. ITGA7 is expressed in DMD patients and overexpression does not elicit an immune response to the transgene. We delivered rAAVrh.74.MCK.ITGA7 systemically at 5-7 days of age to the mdx/utrn(-/-) mouse deficient for dystrophin and utrophin, a severe mouse model of DMD. At 8 weeks postinjection, widespread expression of ITGA7 was observed at the sarcolemma of multiple muscle groups following gene transfer. The increased expression of ITGA7 significantly extended longevity and reduced common features of the mdx/utrn(-/-) mouse, including kyphosis. Overexpression of α7 expression protected against loss of force following contraction-induced damage and increased specific force in the diaphragm and EDL muscles 8 weeks after gene transfer. Taken together, these results further support the use of α7 integrin as a potential therapy for DMD.


Subject(s)
Antigens, CD/genetics , Dystrophin/genetics , Integrin alpha Chains/genetics , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Animals , Antigens, CD/administration & dosage , Antigens, CD/biosynthesis , Dependovirus , Disease Models, Animal , Dystrophin/deficiency , Gene Expression Regulation , Genetic Therapy/methods , Humans , Integrin alpha Chains/administration & dosage , Integrin alpha Chains/biosynthesis , Mice , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology
5.
PLoS One ; 10(4): e0124411, 2015.
Article in English | MEDLINE | ID: mdl-25923107

ABSTRACT

Fibrosis refers to the accumulation of excess extracellular matrix (ECM) components and represents a key feature of many chronic inflammatory diseases. Unfortunately, no currently available treatments specifically target this important pathogenic mechanism. MicroRNAs (miRNAs) are short, non-coding RNAs that post-transcriptionally repress target gene expression and the development of miRNA-based therapeutics is being actively pursued for a diverse array of diseases. Because a single miRNA can target multiple genes, often within the same pathway, variations in the level of individual miRNAs can potently influence disease phenotypes. Members of the miR-29 family, which include miR-29a, miR-29b and miR-29c, are strong inhibitors of ECM synthesis and fibrosis-associated decreases in miR-29 have been reported in multiple organs. We observed downregulation of miR-29a/b/c in fibrotic livers of carbon tetrachloride (CCl4) treated mice as well as in isolated human hepatocytes exposed to the pro-fibrotic cytokine TGF-ß. Importantly, we demonstrate that a single systemic injection of a miR-29a expressing adeno-associated virus (AAV) can prevent and even reverse histologic and biochemical evidence of fibrosis despite continued exposure to CCl4. The observed therapeutic benefits were associated with AAV transduction of hepatocytes but not hepatic stellate cells, which are the main ECM producing cells in fibroproliferative liver diseases. Our data therefore demonstrate that delivery of miR-29 to the hepatic parenchyma using a clinically relevant gene delivery platform protects injured livers against fibrosis and, given the consistent fibrosis-associated downregulation of miR-29, suggests AAV-miR-29 based therapies may be effective in treating a variety of fibroproliferative disorders.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Dependovirus/genetics , Genetic Vectors/administration & dosage , Hepatocytes/metabolism , Liver Cirrhosis/therapy , MicroRNAs/genetics , Animals , Carbon Tetrachloride , Cell Line , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression Regulation , Genetic Vectors/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/pathology , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , MicroRNAs/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology
6.
Neurosurgery ; 76(2): 216-25; discussion 225, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25549186

ABSTRACT

BACKGROUND: After injection into muscle and peripheral nerves, a variety of viral vectors undergo retrograde transport to lower motor neurons. However, because of its attractive safety profile and durable gene expression, adeno-associated virus (AAV) remains the only vector to have been applied to the human nervous system for the treatment of neurodegenerative disease. Nonetheless, only a very small fraction of intramuscularly injected AAV vector arrives at the spinal cord. OBJECTIVE: To engineer a novel AAV vector by inserting a neuronal targeting peptide (Tet1), with binding properties similar to those of tetanus toxin, into the AAV1 capsid. METHODS: Integral to this approach was the use of structure-based design to increase the effectiveness of functional capsid engineering. This approach allowed the optimization of scaffolding regions for effective display of the foreign epitope while minimizing disruption of the native capsid structure. We also validated an approach by which low-titer tropism-modified AAV vectors can be rescued by particle mosaicism with unmodified capsid proteins. RESULTS: Importantly, our rationally engineered AAV1-based vectors exhibited markedly enhanced transduction of cultured motor neurons, diminished transduction of nontarget cells, and markedly superior retrograde delivery compared with unmodified AAV1 vector. CONCLUSION: This approach promises a significant advancement in the rational engineering of AAV vectors for diseases of the nervous system and other organs.


Subject(s)
DNA-Binding Proteins/genetics , Gene Transfer Techniques , Genetic Vectors/chemical synthesis , Proto-Oncogene Proteins/genetics , Transduction, Genetic/methods , Animals , Capsid Proteins/chemistry , Dependovirus/genetics , Genetic Therapy/methods , HEK293 Cells , HeLa Cells , Humans , Mixed Function Oxygenases , Protein Structure, Quaternary , Rats , Rats, Sprague-Dawley
7.
Hum Gene Ther ; 25(11): 977-87, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25275822

ABSTRACT

Abstract Gene therapy approaches using recombinant adeno-associated virus serotype 2 (rAAV2) and serotype 8 (rAAV8) have achieved significant clinical benefits. The generation of rAAV Reference Standard Materials (RSM) is key to providing points of reference for particle titer, vector genome titer, and infectious titer for gene transfer vectors. Following the example of the rAAV2RSM, here we have generated and characterized a novel RSM based on rAAV serotype 8. The rAAV8RSM was produced using transient transfection, and the purification was based on density gradient ultracentrifugation. The rAAV8RSM was distributed for characterization along with standard assay protocols to 16 laboratories worldwide. Mean titers and 95% confidence intervals were determined for capsid particles (mean, 5.50×10(11) pt/ml; CI, 4.26×10(11) to 6.75×10(11) pt/ml), vector genomes (mean, 5.75×10(11) vg/ml; CI, 3.05×10(11) to 1.09×10(12) vg/ml), and infectious units (mean, 1.26×10(9) IU/ml; CI, 6.46×10(8) to 2.51×10(9) IU/ml). Notably, there was a significant degree of variation between institutions for each assay despite the relatively tight correlation of assay results within an institution. This outcome emphasizes the need to use RSMs to calibrate the titers of rAAV vectors in preclinical and clinical studies at a time when the field is maturing rapidly. The rAAV8RSM has been deposited at the American Type Culture Collection (VR-1816) and is available to the scientific community.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genome, Viral , HEK293 Cells , Humans , Reference Standards , Transformation, Genetic , Virion/genetics , Virus Cultivation/standards
8.
Lancet ; 383(9923): 1129-37, 2014 Mar 29.
Article in English | MEDLINE | ID: mdl-24439297

ABSTRACT

BACKGROUND: Choroideremia is an X-linked recessive disease that leads to blindness due to mutations in the CHM gene, which encodes the Rab escort protein 1 (REP1). We assessed the effects of retinal gene therapy with an adeno-associated viral (AAV) vector encoding REP1 (AAV.REP1) in patients with this disease. METHODS: In a multicentre clinical trial, six male patients (aged 35-63 years) with choroideremia were administered AAV.REP1 (0·6-1·0×10(10) genome particles, subfoveal injection). Visual function tests included best corrected visual acuity, microperimetry, and retinal sensitivity tests for comparison of baseline values with 6 months after surgery. This study is registered with ClinicalTrials.gov, number NCT01461213. FINDINGS: Despite undergoing retinal detachment, which normally reduces vision, two patients with advanced choroideremia who had low baseline best corrected visual acuity gained 21 letters and 11 letters (more than two and four lines of vision). Four other patients with near normal best corrected visual acuity at baseline recovered to within one to three letters. Mean gain in visual acuity overall was 3·8 letters (SE 4·1). Maximal sensitivity measured with dark-adapted microperimetry increased in the treated eyes from 23·0 dB (SE 1·1) at baseline to 25·3 dB (1·3) after treatment (increase 2·3 dB [95% CI 0·8-3·8]). In all patients, over the 6 months, the increase in retinal sensitivity in the treated eyes (mean 1·7 [SE 1·0]) was correlated with the vector dose administered per mm(2) of surviving retina (r=0·82, p=0·04). By contrast, small non-significant reductions (p>0·05) were noted in the control eyes in both maximal sensitivity (-0·8 dB [1·5]) and mean sensitivity (-1·6 dB [0·9]). One patient in whom the vector was not administered to the fovea re-established variable eccentric fixation that included the ectopic island of surviving retinal pigment epithelium that had been exposed to vector. INTERPRETATION: The initial results of this retinal gene therapy trial are consistent with improved rod and cone function that overcome any negative effects of retinal detachment. These findings lend support to further assessment of gene therapy in the treatment of choroideremia and other diseases, such as age-related macular degeneration, for which intervention should ideally be applied before the onset of retinal thinning. FUNDING: UK Department of Health and Wellcome Trust.


Subject(s)
Adaptor Proteins, Signal Transducing/administration & dosage , Choroideremia/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Adaptor Proteins, Signal Transducing/genetics , Adenoviridae/genetics , Adult , Aged , Choroideremia/physiopathology , Fluorescence , Gene Transfer Techniques , Humans , Injections, Intraocular , Male , Middle Aged , Retinal Detachment/physiopathology , Retinal Detachment/therapy , Transgenes/genetics , Visual Acuity/physiology
9.
Mol Ther ; 22(4): 713-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24145553

ABSTRACT

Overexpression of GALGT2 in skeletal muscle can stimulate the glycosylation of α dystroglycan and the upregulation of normally synaptic dystroglycan-binding proteins, some of which are dystrophin and laminin α2 surrogates known to be therapeutic for several forms of muscular dystrophy. This article describes the vascular delivery of GALGT2 gene therapy in a large animal model, the rhesus macaque. Recombinant adeno-associated virus, rhesus serotype 74 (rAAVrh74), was used to deliver GALGT2 via the femoral artery to the gastrocnemius muscle using an isolated focal limb perfusion method. GALGT2 expression averaged 44 ± 4% of myofibers after treatment in macaques with low preexisting anti-rAAVrh74 serum antibodies, and expression was reduced to 9 ± 4% of myofibers in macaques with high preexisting rAAVrh74 immunity (P < 0.001; n = 12 per group). This was the case regardless of the addition of immunosuppressants, including prednisolone, tacrolimus, and mycophenolate mofetil. GALGT2-treated macaque muscles showed increased glycosylation of α dystroglycan and increased expression of dystrophin and laminin α2 surrogate proteins, including utrophin, plectin1, agrin, and laminin α5. These experiments demonstrate successful transduction of rhesus macaque muscle with rAAVrh74.MCK.GALGT2 after vascular delivery and induction of molecular changes thought to be therapeutic in several forms of muscular dystrophy.


Subject(s)
Dystrophin/biosynthesis , Gene Transfer Techniques , Genetic Therapy , Laminin/biosynthesis , Muscular Dystrophies/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Dystroglycans/genetics , Dystroglycans/metabolism , Dystrophin/genetics , Gene Expression Regulation , Glycosyltransferases/genetics , Laminin/genetics , Macaca mulatta/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Muscular Dystrophies/therapy
10.
Hum Mol Genet ; 22(24): 4929-37, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23863459

ABSTRACT

Pharmacologic strategies have provided modest improvement in the devastating muscle-wasting disease, Duchenne muscular dystrophy (DMD). Pre-clinical gene therapy studies have shown promise in the mdx mouse model; however, studies conducted after disease onset fall short of fully correcting muscle strength or protecting against contraction-induced injury. Here we examine the treatment effect on muscle physiology in aged dystrophic mice with significant disease pathology by combining two promising therapies: micro-dystrophin gene replacement and muscle enhancement with follistatin, a potent myostatin inhibitor. Individual treatments with micro-dystrophin and follistatin demonstrated marked improvement in mdx mice but were insufficient to fully restore muscle strength and response to injury to wild-type levels. Strikingly, when combined, micro-dystrophin/follistatin treatment restored force generation and conferred resistance to contraction-induced injury in aged mdx mice. Pre-clinical studies with miniature dystrophins have failed to demonstrate full correction of the physiological defects seen in mdx mice. Importantly, the addition of a muscle enhancement strategy with delivery of follistatin in combination with micro-dystrophin gene therapy completely restored resistance to eccentric contraction-induced injury and improved force. Eccentric contraction-induced injury is a pre-clinical parameter relevant to the exercise induced injury that occurs in DMD patients, and herein, we demonstrate compelling evidence for the therapeutic potential of micro-dystrophin/follistatin combinatorial therapy.


Subject(s)
Dystrophin/genetics , Follistatin/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Animals , Dependovirus/genetics , Disease Models, Animal , Dystrophin/metabolism , Follistatin/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle Strength/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/therapy
11.
Muscle Nerve ; 47(5): 731-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23553538

ABSTRACT

INTRODUCTION: Recent in vitro studies suggest that CAPN3 deficiency leads initially to accelerated myofiber formation followed by depletion of satellite cells (SC). In normal muscle, up-regulation of miR-1 and miR-206 facilitates transition from proliferating SCs to differentiating myogenic progenitors. METHODS: We examined the histopathological stages, Pax7 SC content, and muscle-specific microRNA expression in biopsy specimens from well-characterized LGMD 2A patients to gain insight into disease pathogenesis. RESULTS: Three distinct stages of pathological changes were identified that represented the continuum of the dystrophic process from prominent inflammation with necrosis and regeneration to prominent fibrosis, which correlated with age and disease duration. Pax7-positive SCs were highest in the fibrotic group and correlated with down-regulation of miR-1, miR-133a, and miR-206. CONCLUSIONS: These observations, and other published reports, are consistent with microRNA dysregulation leading to inability of Pax7-positive SCs to transit from proliferation to differentiation. This results in impaired regeneration and fibrosis.


Subject(s)
MicroRNAs/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , PAX7 Transcription Factor/metabolism , Regeneration/physiology , Satellite Cells, Skeletal Muscle/metabolism , Adolescent , Adult , Cell Differentiation , Cell Proliferation , Child , Child, Preschool , Female , Fibrosis , Humans , Male , MicroRNAs/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Muscular Dystrophies, Limb-Girdle/physiopathology , Myoblasts/metabolism , Myoblasts/pathology , PAX7 Transcription Factor/genetics , Satellite Cells, Skeletal Muscle/pathology
12.
Mol Ther Nucleic Acids ; 2: e86, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23591809

ABSTRACT

Gene therapy has historically focused on delivering protein-coding genes to target cells or tissues using a variety of vectors. In recent years, the field has expanded to include gene-silencing strategies involving delivery of noncoding inhibitory RNAs, such as short hairpin RNAs or microRNAs (miRNAs). Often called RNA interference (RNAi) triggers, these small inhibitory RNAs are difficult or impossible to visualize in living cells or tissues. To circumvent this detection problem and ensure efficient delivery in preclinical studies, vectors can be engineered to coexpress a fluorescent reporter gene to serve as a marker of transduction. In this study, we set out to optimize adeno-associated viral (AAV) vectors capable of delivering engineered miRNAs and green fluorescent protein (GFP) reporter genes to skeletal muscle. Although the more broadly utilized enhanced GFP (eGFP) gene derived from the jellyfish, Aequorea victoria was a conventional choice, we were concerned about some previous studies suggesting this protein was myotoxic. We thus opted to test vectors carrying the humanized Renilla reniformis-derived GFP (hrGFP) gene, which has not seen as extensive usage as eGFP but was purported to be a safer and less cytotoxic alternative. Employing AAV6 vector dosages typically used in preclinical gene transfer studies (3×10(10) -1 × 10(11) particles), we found that hrGFP caused dose-dependent myopathy when delivered to wild-type (wt) mouse muscle, whereas identical titers of AAV6 carrying eGFP were relatively benign. Dose de-escalation at or below 8 × 10(9) AAV particles effectively reduced or eliminated hrGFP-associated myotoxicity, but also had dampening effects on green fluorescence and miRNA-mediated gene silencing in whole muscles. We conclude that hrGFP is impractical for use as a transduction marker in preclinical, AAV-based RNA interference therapy studies where adult mouse muscle is the target organ. Moreover, our data support that eGFP is superior to hrGFP as a reporter gene in mouse muscle. These results may impact the design of future preclinical gene therapy studies targeting muscles and non-muscle tissues alike.Molecular Therapy - Nucleic Acids (2013) 2, e86; doi:10.1038/mtna.2013.16; published online 16 April 2013.

13.
Mol Ther ; 21(3): 520-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319059

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle disease caused by mutations in the DMD gene, with loss of its gene product, dystrophin. Dystrophin helps link integral membrane proteins to the actin cytoskeleton and stabilizes the sarcolemma during muscle activity. We investigated an alternative therapeutic approach to dystrophin replacement by overexpressing human α7 integrin (ITGA7) using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal and cardiac muscle that links the extracellular matrix (ECM) to the actin skeleton. It is modestly upregulated in DMD muscle and has been proposed to be an important modifier of dystrophic symptoms. We delivered rAAV8.MCK.ITGA7 to the lower limb of mdx mice through isolated limb perfusion (ILP) of the femoral artery. We demonstrated ~50% of fibers in the tibialis anterior (TA) and extensor digitorum longus (EDL) overexpressing α7 integrin at the sarcolemma following AAV gene transfer. The increase in ITGA7 in skeletal muscle significantly protected against loss of force following eccentric contraction-induced injury compared with untreated (contralateral) muscles while specific force following tetanic contraction was unchanged. Reversal of additional dystrophic features included reduced Evans blue dye (EBD) uptake and increased muscle fiber diameter. Taken together, this data shows that rAAV8.MCK.ITGA7 gene transfer stabilizes the sarcolemma potentially preserving mdx muscle from further damage. This therapeutic approach demonstrates promise as a viable treatment for DMD with further implications for other forms of muscular dystrophy.


Subject(s)
Antigens, CD/genetics , Dependovirus/genetics , Genetic Vectors , Integrin alpha Chains/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Antigens, CD/metabolism , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Extracellular Matrix/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Integrin alpha Chains/metabolism , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Mutation , Sarcolemma/genetics , Up-Regulation
14.
J Clin Invest ; 122(8): 2871-83, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22820288

ABSTRACT

miR-122, an abundant liver-specific microRNA (miRNA), regulates cholesterol metabolism and promotes hepatitis C virus (HCV) replication. Reduced miR-122 expression in hepatocellular carcinoma (HCC) correlates with metastasis and poor prognosis. Nevertheless, the consequences of sustained loss of function of miR-122 in vivo have not been determined. Here, we demonstrate that deletion of mouse Mir122 resulted in hepatosteatosis, hepatitis, and the development of tumors resembling HCC. These pathologic manifestations were associated with hyperactivity of oncogenic pathways and hepatic infiltration of inflammatory cells that produce pro-tumorigenic cytokines, including IL-6 and TNF. Moreover, delivery of miR-122 to a MYC-driven mouse model of HCC strongly inhibited tumorigenesis, further supporting the tumor suppressor activity of this miRNA. These findings reveal critical functions for miR-122 in the maintenance of liver homeostasis and have important therapeutic implications, including the potential utility of miR-122 delivery for selected patients with HCC and the need for careful monitoring of patients receiving miR-122 inhibition therapy for HCV.


Subject(s)
Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Base Sequence , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Cell Proliferation , Cell Survival/genetics , Cytokines/biosynthesis , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression , Genes, Tumor Suppressor , Genes, myc , Humans , Lipid Metabolism/genetics , Lipids/blood , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Liver Neoplasms, Experimental/etiology , Mice , Mice, 129 Strain , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/therapeutic use , Monocytes/immunology , Monocytes/pathology , Neutrophils/immunology , Neutrophils/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
15.
PLoS One ; 7(6): e39233, 2012.
Article in English | MEDLINE | ID: mdl-22720081

ABSTRACT

The dysferlinopathies comprise a group of untreatable muscle disorders including limb girdle muscular dystrophy type 2B, Miyoshi myopathy, distal anterior compartment syndrome, and rigid spine syndrome. As with other forms of muscular dystrophy, adeno-associated virus (AAV) gene transfer is a particularly auspicious treatment strategy, however the size of the DYSF cDNA (6.5 kb) negates packaging into traditional AAV serotypes known to express well in muscle (i.e. rAAV1, 2, 6, 8, 9). Potential advantages of a full cDNA versus a mini-gene include: maintaining structural-functional protein domains, evading protein misfolding, and avoiding novel epitopes that could be immunogenic. AAV5 has demonstrated unique plasticity with regards to packaging capacity and recombination of virions containing homologous regions of cDNA inserts has been implicated in the generation of full-length transcripts. Herein we show for the first time in vivo that homologous recombination following AAV5.DYSF gene transfer leads to the production of full length transcript and protein. Moreover, gene transfer of full-length dysferlin protein in dysferlin deficient mice resulted in expression levels sufficient to correct functional deficits in the diaphragm and importantly in skeletal muscle membrane repair. Intravascular regional gene transfer through the femoral artery produced high levels of transduction and enabled targeting of specific muscle groups affected by the dysferlinopathies setting the stage for potential translation to clinical trials. We provide proof of principle that AAV5 mediated delivery of dysferlin is a highly promising strategy for treatment of dysferlinopathies and has far-reaching implications for the therapeutic delivery of other large genes.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Membrane Proteins/genetics , Muscle Proteins/genetics , Recombination, Genetic , Dysferlin , Humans
16.
Neurosci Lett ; 527(2): 90-9, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22609847

ABSTRACT

Our Translational Gene Therapy Center has used small molecules for exon skipping and mutation suppression and gene transfer to replace or provide surrogate genes as tools for molecular-based approaches for the treatment of muscular dystrophies. Exon skipping is targeted at the pre-mRNA level allowing one or more exons to be omitted to restore the reading frame. In Duchenne Muscular Dystrophy (DMD), clinical trials have been performed with two different oligomers, a 2'O-methyl-ribo-oligonucleoside-phosphorothioate (2'OMe) and a phosphorodiamidate morpholino (PMO). Both have demonstrated early evidence of efficacy. A second molecular approach involves suppression of stop codons to promote readthrough of the DMD gene. We have been able to establish proof of principle for mutation suppression using the aminoglycoside, gentamicin. A safer, orally administered, alternative agent referred to as Ataluren (PTC124) has been used in clinical trials and is currently under consideration for approval by the FDA. Using a gene therapy approach, we have completed two trials and have initiated a third. For DMD, we used a mini-dystrophin transferred in adeno-associated virus (AAV). In this trial an immune response was seen directed against transgene product, a quite unexpected outcome that will help guide further studies. For limb girdle muscular dystrophy 2D (alpha-sarcoglycan deficiency), the transgene was again transferred using AAV but in this study, a muscle specific creatine kinase promoter controlled gene expression that persisted for six months. A third gene therapy trial has been initiated with transfer of the follistatin gene in AAV directly to the quadriceps muscle. Two diseases with selective quadriceps muscle weakness are undergoing gene transfer including sporadic inclusion body myositis (sIBM) and Becker muscular dystrophy (BMD). Increasing the size and strength of the muscle is the goal of this study. Most importantly, no adverse events have been encountered in any of these clinical trials.


Subject(s)
Genetic Therapy/methods , Muscular Dystrophies/therapy , Clinical Trials as Topic , Codon, Terminator , Creatine Kinase/genetics , Dependovirus/genetics , Dystrophin/genetics , Dystrophin/metabolism , Exons , Follistatin/genetics , Genetic Vectors , Humans , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutation , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/therapy , Oxadiazoles/therapeutic use , Promoter Regions, Genetic , RNA Precursors/genetics , Sarcoglycans/genetics , Sarcoglycans/metabolism
17.
Muscle Nerve ; 45(2): 163-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22246869

ABSTRACT

INTRODUCTION: In this study we address the challenging issue of potential use of muscle strength to predict function in clinical trials. This has immediate relevance to translational studies that attempt to improve quadriceps strength in sporadic inclusion-body myositis (sIBM). METHODS: Maximum voluntary isometric contraction testing as a measure of muscle strength and a battery of functional outcomes were tested in 85 ambulatory subjects with sIBM. RESULTS: Marked quadriceps weakness was noted in all patients. Strength was correlated with distance walked at 2 and 6 minutes. Additional correlations were found with time to get up from a chair, climb stairs, and step up on curbs. CONCLUSIONS: Quadriceps (knee extensor) strength correlated with performance in this large cohort of sIBM subjects, which demonstrated its potential to predict function in this disease. These data provide initial support for use of muscle strength as a surrogate for function, although validation in a clinical trial is required.


Subject(s)
Knee Joint/physiopathology , Muscle Strength/physiology , Myositis, Inclusion Body/pathology , Myositis, Inclusion Body/physiopathology , Quadriceps Muscle/physiopathology , Adult , Aged , Aged, 80 and over , Female , Functional Laterality , Humans , Isometric Contraction/physiology , Male , Middle Aged , Predictive Value of Tests , Psychomotor Performance/physiology , Walking/physiology
18.
PLoS Pathog ; 7(9): e1002281, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21980295

ABSTRACT

The liver removes quickly the great bulk of virus circulating in blood, leaving only a small fraction to infect the host, in a manner characteristic of each virus. The scavenger cells of the liver sinusoids are implicated, but the mechanism is entirely unknown. Here we show, borrowing a mouse model of adenovirus clearance, that nearly all infused adenovirus is cleared by the liver sinusoidal endothelial cell (LSEC). Using refined immunofluorescence microscopy techniques for distinguishing macrophages and endothelial cells in fixed liver, and identifying virus by two distinct physicochemical methods, we localized adenovirus 1 minute after infusion mainly to the LSEC (∼90%), finding ∼10% with Kupffer cells (KC) and none with hepatocytes. Electron microscopy confirmed our results. In contrast with much prior work claiming the main scavenger to be the KC, our results locate the clearance mechanism to the LSEC and identify this cell as a key site of antiviral activity.


Subject(s)
Adenoviridae Infections/metabolism , Adenoviridae/metabolism , Blood-Borne Pathogens , Endothelium, Vascular/metabolism , Liver/metabolism , Adenoviridae/immunology , Adenoviridae/ultrastructure , Adenoviridae Infections/immunology , Animals , Cells, Cultured , Endothelium, Vascular/immunology , Endothelium, Vascular/ultrastructure , Endothelium, Vascular/virology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Hepatocytes/virology , Humans , Kupffer Cells/immunology , Kupffer Cells/metabolism , Kupffer Cells/ultrastructure , Kupffer Cells/virology , Liver/immunology , Liver/ultrastructure , Liver/virology , Mice , Mice, Inbred BALB C
19.
Methods Mol Biol ; 807: 239-58, 2011.
Article in English | MEDLINE | ID: mdl-22034033

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors are capable of mediating long-term gene expression in a wide variety of animals, including primates. The rAAV genome is packaged into the virion as single-stranded DNA devoid of any viral genes. A proportion of the single-stranded genomes are converted into transcriptionally active double-stranded DNA (dsDNA) early after nuclear entry by second-strand synthesis mediated by host repair DNA polymerases or/and by annealing of the rAAV (-) and (+) strands. Second-generation, self-complementary vectors are packaged as single-strand hairpins and rapidly assume a dsDNA conformation independent of the action of polymerases. In both cases, linear dsDNA vector genomes circularize and can undergo concatemerization into higher order forms (McCarty et al. Annu Rev Genet 38: 819-845, 2004; Schultz and Chamberlain Mol Ther 16: 1189-1199, 2008; Duan et al. J Virol 72: 8568-8577, 1998). As a result, rAAV vector genomes are maintained mainly as circular monomeric and concatemeric episomal forms in skeletal muscle and liver (Schnepp et al. J Virol 77: 3495-3504, 2003; Penaud-Budloo et al. J Virol 82: 7875-7885, 2008; Nakai et al. J Virol 75: 6969-6976, 2001). Moreover, in nonhuman primate skeletal muscle, it has been shown that rAAV episomes assimilate into chromatin with a typical nucleosomal pattern that presumably is important for persistence and gene expression in quiescent tissues over a period of several years (Penaud-Budloo et al. J Virol 82: 7875-7885, 2008). Conversely, although rAAV is not considered as an integrative vector per se, introduction of exogenous DNA into the nuclear compartment can result in low-level vector assimilation into the host genome. One mechanism appears to involve vector insertion at sites of double-strand DNA breaks using cellular DNA repair enzymes. As rAAV gene transfer technology and applications mature, a better characterization of the genetic fate of the rAAV genome is critical to accurately evaluate the risk/benefit ratio for a particular disease indication. In this chapter, two complementary methods are detailed to enable characterization of rAAV molecular structure in a particular target tissue and estimation of its integration frequency.


Subject(s)
Dependovirus/genetics , Dependovirus/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Animals , Blotting, Southern , Cell Line , Mice , Polymerase Chain Reaction
20.
Ann Neurol ; 68(5): 629-38, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21031578

ABSTRACT

OBJECTIVE: The aim of this study was to attain long-lasting alpha-sarcoglycan gene expression in limb-girdle muscular dystrophy, type 2D (LGMD2D) subjects mediated by adeno-associated virus (AAV) gene transfer under control of a muscle specific promoter (tMCK). METHODS: rAAV1.tMCK.hSGCA (3.25 × 10¹¹ vector genomes) was delivered to the extensor digitorum brevis muscle of 3 subjects with documented SGCA mutations via a double-blind, randomized, placebo controlled trial. Control sides received saline. The blind was not broken until the study was completed at 6 months and all results were reported to the oversight committee. RESULTS: Persistent alpha-sarcoglycan gene expression was achieved for 6 months in 2 of 3 LGMD2D subjects. Markers for muscle fiber transduction other than alpha-sarcoglycan included expression of major histocompatibility complex I, increase in muscle fiber size, and restoration of the full sarcoglycan complex. Mononuclear inflammatory cells recruited to the site of gene transfer appeared to undergo programmed cell death, demonstrated by terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick-end labeling and caspase-3 staining. A patient failing gene transfer demonstrated an early rise in neutralizing antibody titers and T-cell immunity to AAV, validated by enzyme-linked immunospot on the second day after gene injection. This was in clear distinction to other participants with satisfactory gene expression. INTERPRETATION: The findings of this gene replacement study in LGMD2D subjects have important implications not previously demonstrated in muscular dystrophy. Long-term, sustainable gene expression of alpha-sarcoglycan was observed following gene transfer mediated by AAV. The merit of a muscle-specific tMCK promoter, not previously used in a clinical trial, was evident, and the potential for reversal of disease was displayed.


Subject(s)
Gene Transfer Techniques/adverse effects , Muscular Dystrophies, Limb-Girdle/therapy , Sarcoglycans/genetics , Adolescent , Adult , Apoptosis , Child , Dependovirus/genetics , Female , Gene Expression , Genetic Therapy/methods , Genetic Vectors/immunology , Humans , Leukocytes, Mononuclear/metabolism , Male , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Sarcoglycans/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...