60 results on '"CHANDLER, RJ"'
Search Results
2. The behaviour of layered clays within a framework for the structure-related behaviour of clays
- Author
-
Cotecchia, F, Coop, Mr, and Chandler, Rj
- Published
- 1999
3. Discussion on “The Behavior at the Shrinkage Limit of Clay Undergoing Drying” by David J. Williams and John W. Sibley
- Author
-
Pincus, HJ, primary, Marinho, FAM, additional, and Chandler, RJ, additional
- Published
- 1993
- Full Text
- View/download PDF
4. A LOW-COST METHOD OF ASSESSING CLAY DESICCATION FOR LOW-RISE BUILDINGS.
- Author
-
SMITH, MG, CRILLY, MS, and CHANDLER, RJ
- Published
- 1992
- Full Text
- View/download PDF
5. 8827-9 DISCUSSION. THE EMPINGHAM RESERVOIR PROJECT.
- Author
-
HUGHES, F, MCKENNA, JM, TAYLOR, HR, JONES, HN, BRIDLE, RC, SMITH, EJ, BOWYER, GE, COLE, RG, RAY, WJF, VAUGHAN, PR, CHANDLER, RJ, WINDER, AJM, DICKENS, DG, BIGGART, A, MAXWELL, BA, CHARLES, JA, and HORSWILL, P
- Published
- 1986
- Full Text
- View/download PDF
6. Discussion on “The Behavior at the Shrinkage Limit of Clay Undergoing Drying” by David J. Williams and John W. Sibley
- Author
-
Marinho, FAM and Chandler, RJ
- Published
- 1993
- Full Text
- View/download PDF
7. 8827-9 DISCUSSION. THE EMPINGHAM RESERVOIR PROJECT.
- Author
-
WINDER, AJM, primary, COLE, RG, additional, BOWYER, GE, additional, BRIDLE, RC, additional, VAUGHAN, PR, additional, JONES, HN, additional, MCKENNA, JM, additional, HORSWILL, P, additional, SMITH, EJ, additional, CHARLES, JA, additional, MAXWELL, BA, additional, TAYLOR, HR, additional, BIGGART, A, additional, DICKENS, DG, additional, CHANDLER, RJ, additional, RAY, WJF, additional, and HUGHES, F, additional
- Published
- 1986
- Full Text
- View/download PDF
8. Systemic Treatment for Postnatal, Juvenile, and Runted Adult Mice by Retrobulbar Sinus Injection.
- Author
-
Romero D and Chandler RJ
- Subjects
- Animals, Mice, Injections methods
- Abstract
While tail vein injections are frequently used as a systemic route of delivery in adult mice, retrobulbar injections are an alternative method for systemic delivery with fewer limitations. First, tail vein injections (TVIs) are limited to adult mice where the size of the tail vein is suitable for access. Being restricted to treating adult mice can be problematic when dealing with mouse models that do not survive to adulthood. Second, TVIs are not feasible for mouse models with growth retardation phenotypes where the mice never achieve the size of adult wildtype mice. Therefore, retrobulbar injections can be successfully used to treat both young and small adult mice. Lastly, retrobulbar injections are performed under anesthesia, which is less stressful on the mice than TVIs that are commonly performed without anesthesia. This article presents a protocol and detailed instructions for retrobulbar injections that can be used for systemic delivery to small and young mice.
- Published
- 2024
- Full Text
- View/download PDF
9. Gene therapy for organic acidemias: Lessons learned from methylmalonic and propionic acidemia.
- Author
-
Chandler RJ and Venditti CP
- Subjects
- Animals, Mice, Humans, Genetic Therapy, Disease Models, Animal, Methylmalonic Acid, Propionic Acidemia genetics, Propionic Acidemia therapy, Propionic Acidemia complications, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors therapy, Amino Acid Metabolism, Inborn Errors complications, Liver Transplantation adverse effects
- Abstract
Organic acidemias (OA) are a group of rare autosomal recessive disorders of intermediary metabolism that result in a systemic elevation of organic acid. Despite optimal dietary and cofactor therapy, OA patients still suffer from potentially lethal metabolic instability and experience long-term multisystemic complications. Severely affected patients can benefit from elective liver transplantation, which restores hepatic enzymatic activity, improves metabolic stability, and provides the theoretical basis for the pursuit of gene therapy as a new treatment for patients. Because of the poor outcomes reported in those with OA, especially methylmalonic and propionic acidemia, multiple gene therapy approaches have been explored in relevant animal models. Here, we review the results of gene therapy experiments performed using MMA and PA mouse models to illustrate experimental paradigms that could be applicable for all forms of OA., (Published 2023. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2024
- Full Text
- View/download PDF
10. Natural selection has driven the recurrent loss of an immunity gene that protects Drosophila against a major natural parasite.
- Author
-
Arunkumar R, Zhou SO, Day JP, Bakare S, Pitton S, Zhang Y, Hsing CY, O'Boyle S, Pascual-Gil J, Clark B, Chandler RJ, Leitão AB, and Jiggins FM
- Subjects
- Animals, Drosophila genetics, Drosophila melanogaster genetics, Host-Parasite Interactions, Lectins genetics, Selection, Genetic, Parasites, Wasps physiology
- Abstract
Polymorphisms in immunity genes can have large effects on susceptibility to infection. To understand the origins of this variation, we have investigated the genetic basis of resistance to the parasitoid wasp Leptopilina boulardi in Drosophila melanogaster. We found that increased expression of the gene lectin-24A after infection by parasitic wasps was associated with a faster cellular immune response and greatly increased rates of killing the parasite. lectin-24A encodes a protein that is strongly up-regulated in the fat body after infection and localizes to the surface of the parasite egg. In certain susceptible lines, a deletion upstream of the lectin-24A has largely abolished expression. Other mutations predicted to abolish the function of this gene have arisen recurrently in this gene, with multiple loss-of-expression alleles and premature stop codons segregating in natural populations. The frequency of these alleles varies greatly geographically, and in some southern African populations, natural selection has driven them near to fixation. We conclude that natural selection has favored the repeated loss of an important component of the immune system, suggesting that in some populations, a pleiotropic cost to lectin-24A expression outweighs the benefits of resistance.
- Published
- 2023
- Full Text
- View/download PDF
11. Systemic gene therapy using an AAV44.9 vector rescues a neonatal lethal mouse model of propionic acidemia.
- Author
-
Chandler RJ, Di Pasquale G, Choi EY, Chang D, Smith SN, Sloan JL, Hoffmann V, Li L, Chiorini JA, and Venditti CP
- Abstract
Propionic acidemia (PA) is rare autosomal recessive metabolic disorder caused by defects in the mitochondrially localized enzyme propionyl-coenzyme A (CoA) carboxylase. Patients with PA can suffer from lethal metabolic decompensation and cardiomyopathy despite current medical management, which has led to the pursuit of gene therapy as a new treatment option for patients. Here we assess the therapeutic efficacy of a recently described adeno-associated virus (AAV) capsid, AAV44.9, to deliver a therapeutic PCCA transgene in a new mouse model of propionyl-CoA carboxylase α (PCCA) deficiency generated by genome editing. Pcca
-/- mice recapitulate the severe neonatal presentation of PA and manifest uniform neonatal lethality, absent PCCA expression, and increased 2-methylcitrate. A single injection of the AAV44.9 PCCA vector in the immediate newborn period, systemically delivered at a dose of 1e11 vector genome (vg)/pup but not 1e10 vg/pup, increased survival, reduced plasma methylcitrate, and resulted in high levels of transgene expression in the liver and heart in treated Pcca-/- mice. Our studies not only establish a versatile and accurate new mouse model of PA but further demonstrate that the AAV44.9 vectors may be suitable for treatment of many metabolic disorders where hepato-cardiac transduction following systemic delivery is desired, such as PA, and, by extension, fatty acid oxidation defects and glycogen storage disorders., Competing Interests: R.J.C., G.D.P., J.A.C., and C.P.V. are inventors on a patent application filed by the NIH on their behalf on use of AAV44.9 as a gene therapy vector to treat methylmalonic acidemia.- Published
- 2023
- Full Text
- View/download PDF
12. From Puppies to adults: In vivo editing of hepatocytes in a canine model of glycogen storage disease type Ia.
- Author
-
Chandler RJ
- Abstract
Competing Interests: The author declares no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
13. Successfully Navigating Food and Drug Administration Orphan Drug and Rare Pediatric Disease Designations for AAV9-hPCCA Gene Therapy: The National Institutes of Health Platform Vector Gene Therapy Experience.
- Author
-
Lomash RM, Shchelochkov O, Chandler RJ, Venditti CP, Pariser AR, and Ottinger EA
- Subjects
- Humans, United States, Child, United States Food and Drug Administration, Drug Approval, Rare Diseases genetics, Rare Diseases therapy, Genetic Therapy, National Institutes of Health (U.S.), Orphan Drug Production, Propionic Acidemia
- Abstract
Orphan drug designation (ODD) is an important program intended to facilitate the development of orphan drugs in the United States. An orphan drug benefiting pediatric patients can qualify as a drug for a Rare Pediatric Disease Designation (RPDD) as well. The ODD and RPDD programs provide financial incentives for development of diagnostic drugs, preventive measures, and treatment of diseases affecting small patient populations (adult and pediatric) for which commercial development would otherwise be very challenging. In 2019, a multidisciplinary group of collaborators at National Institutes of Health (NIH) embarked upon a gene therapy platform program called Platform Vector Gene Therapy (PaVe-GT) intended to develop gene therapies for four such rare disorders. An important part of PaVe-GT is to publicly share scientific and regulatory experience gained at different stages during the implementation of the PaVe-GT platform utilizing illustrative examples. The PaVe-GT team recently obtained ODD and RPDD for an adeno-associated virus gene therapy to treat propionic acidemia. Given an increasing interest in obtaining ODD for gene therapy, especially by small companies, research investigators, and patient groups, we overview the submission process and subsequently provide examples of our ODD and RPDD applications. Our ODD and RPDD applications and templates can also be found on the PaVe-GT website. Shared reference documents will have great utility to assist parties who may have limited experience with the preparation of similar applications for their orphan product.
- Published
- 2023
- Full Text
- View/download PDF
14. Systemic gene therapy for methylmalonic acidemia using the novel adeno-associated viral vector 44.9.
- Author
-
Chandler RJ, Di Pasquale G, Sloan JL, McCoy S, Hubbard BT, Kilts TM, Manoli I, Chiorini JA, and Venditti CP
- Abstract
Methylmalonic acidemia (MMA) is a severe and potentially lethal autosomal recessive inborn error of metabolism most frequently caused by mutations in the methylmalonyl-CoA mutase ( MMUT ) gene. Proof-of-concept adeno-associated virus (AAV) gene therapy studies using mouse models of MMA have demonstrated promise for this therapeutic approach but translation to the clinic could be limited by preexisting capsid immunity and vector potency. Here we explore the efficacy of a novel clade E capsid, 44.9, as a serotype for systemic AAV gene therapy for MMA. An anti-AAV44.9 neutralizing antibody (NAb) survey in adult volunteers (n = 19) and a large cohort of MMA patients (n = 48) revealed a seroprevalence rate of ∼26% and 13%, respectively. The efficacy of AAV44.9 gene delivery was examined in two murine models of MMA, representing neonatal lethal and juvenile phenotypes of MMA. Systemic delivery of the AAV44.9- Mmut vector prevented lethality and lowered disease-related metabolites in MMA mice. Tissue biodistribution and transgene expression studies in treated MMA mice showed that AAV44.9 was efficient at transducing the liver and heart. In summary, we establish that AAV44.9 exhibits a low prevalence of preexisting NAb in humans, is highly efficacious in the treatment of clinically severe MMA mouse models and is therefore a promising vector for clinical translation., Competing Interests: R.J.C., G.D., J.A.C., and C.P.V. are inventors on a patent application filed by NIH on their behalf on the use of AAV44.9 as a gene therapy vector to treat MMA., (© 2022.)
- Published
- 2022
- Full Text
- View/download PDF
15. Growth advantage of corrected hepatocytes in a juvenile model of methylmalonic acidemia following liver directed adeno-associated viral mediated nuclease-free genome editing.
- Author
-
Venturoni LE, Chandler RJ, Liao J, Hoffmann V, Ramesh N, Gordo S, Chau N, and Venditti CP
- Subjects
- Mice, Animals, Gene Editing, Dependovirus genetics, Mice, Knockout, Liver metabolism, Hepatocytes metabolism, Albumins genetics, Albumins metabolism, Methylmalonic Acid metabolism, Methylmalonyl-CoA Mutase genetics, Methylmalonyl-CoA Mutase metabolism, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors therapy, Amino Acid Metabolism, Inborn Errors metabolism
- Abstract
Methylmalonic acidemia (MMA) is a rare and severe inherited metabolic disease typically caused by mutations of the methylmalonyl-CoA mutase (MMUT) gene. Despite medical management, patients with MMA experience frequent episodes of metabolic instability, severe morbidity, and early mortality. In several preclinical studies, systemic gene therapy has demonstrated impressive improvement in biochemical and clinical phenotypes of MMA murine models. One approach uses a promoterless adeno-associated viral (AAV) vector that relies upon homologous recombination to achieve site-specific in vivo gene addition of MMUT into the last coding exon of albumin (Alb), generating a fused Alb-MMUT transcript after successful editing. We have previously demonstrated that nuclease-free AAV mediated Alb editing could effectively treat MMA mice in the neonatal period and noted that hepatocytes had a growth advantage after correction. Here, we use a transgenic knock-out mouse model of MMA that recapitulates severe clinical and biochemical symptoms to assess the benefits of Alb editing in juvenile animals. As was first noted in the neonatal gene therapy studies, we observe that gene edited hepatocytes in the MMA mice treated as juveniles exhibit a growth advantage, which allows them to repopulate the liver slowly but dramatically by 8-10 months post treatment, and subsequently manifest a biochemical and enzymatic response. In conclusion, our results suggest that the benefit of AAV mediated nuclease-free gene editing of the Alb locus to treat MMA could potentially be therapeutic for older patients., Competing Interests: Declaration of Competing Interest N.R. and J.L. are employees of LogicBio Therapeutics and receive salary support and stock options from LogicBio Therapeutics. S.G. and N.C. are former employees and shareholders of LogicBio Therapeutics., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
16. Evaluating the state of the science for adeno-associated virus integration: An integrated perspective.
- Author
-
Sabatino DE, Bushman FD, Chandler RJ, Crystal RG, Davidson BL, Dolmetsch R, Eggan KC, Gao G, Gil-Farina I, Kay MA, McCarty DM, Montini E, Ndu A, and Yuan J
- Subjects
- Animals, Humans, Mice, Mutagenesis, Insertional, Plasmids, Transgenes, Virus Integration, Dependovirus genetics, Genetic Vectors genetics
- Abstract
On August 18, 2021, the American Society of Gene and Cell Therapy (ASGCT) hosted a virtual roundtable on adeno-associated virus (AAV) integration, featuring leading experts in preclinical and clinical AAV gene therapy, to further contextualize and understand this phenomenon. Recombinant AAV (rAAV) vectors are used to develop therapies for many conditions given their ability to transduce multiple cell types, resulting in long-term expression of transgenes. Although most rAAV DNA typically remains episomal, some rAAV DNA becomes integrated into genomic DNA at a low frequency, and rAAV insertional mutagenesis has been shown to lead to tumorigenesis in neonatal mice. Currently, the risk of rAAV-mediated oncogenesis in humans is theoretical because no confirmed genotoxic events have been reported to date. However, because insertional mutagenesis has been reported in a small number of murine studies, there is a need to characterize this genotoxicity to inform research, regulatory needs, and patient care. The purpose of this white paper is to review the evidence of rAAV-related host genome integration in animal models and possible risks of insertional mutagenesis in patients. In addition, technical considerations, regulatory guidance, and bioethics are discussed., Competing Interests: Declaration of interests F.D.B. is a scientific cofounder of Biocept; has intellectual property licensed to Novartis; and is a consultant for SANA, Poseida, Encoded, and Johnson and Johnson. R.G.C. has equity and consults for LEXEO Therapeutics, an AAV gene-therapy-based company. B.L.D. is on the SAB and/or receives sponsored research support for the laboratory from Homology Medicines, Saliogen Therapeutics, Patch Bio, Moment Bio, Panorama Medicines, Resilience, Spirovant Sciences, Novartis Institute for Biomedical Research, Roche, and Sanofi. R.D. is an employee of uniQure. K.E. is an employee of BioMarin Pharmaceuticals, Inc. G.G. is supported by grants from the Cystic Fibrosis Foundation. G.G. is a scientific cofounder of Voyager Therapeutics and Aspa Therapeutics, and holds equity in these companies. G.G. is an inventor on patents with potential royalties licensed to Voyager Therapeutics, Aspa Therapeutics, and other biopharmaceutical companies. I.G.-F. is an employee of ProtaGene CGT GmbH. M.A.K. is a cofounder, serves on the BOD and SAB, and is a consultant for LogicBio Therapeutics. D.M.M. has consulting agreements with BioMarin Pharmaceuticals and Biogen Inc., and patent/licensing agreements with the University of North Carolina and Nationwide Children’s Hospital, and is an employee of NeuroGT. E.M. is an inventor on a patent for methods for vector integration retrieval. A.N. is an employee and has stock in BridgeBio Pharma, Inc. D.E.S. has intellectual property licensed to Spark Therapeutics, receives sponsored research support from Poseida Therapeutics, and is a consultant for Poseida Therapeutics and Biomarin Pharmaceuticals. J.Y. is an employee and has stock in Pfizer, Inc. The remaining authors declare no competing interests. The content of this article represents the authors’ opinions and may not necessarily represent the views of their employers., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. In vivo genome editing at the albumin locus to treat methylmalonic acidemia.
- Author
-
Schneller JL, Lee CM, Venturoni LE, Chandler RJ, Li A, Myung S, Cradick TJ, Hurley AE, Lagor WR, Bao G, and Venditti CP
- Abstract
Methylmalonic acidemia (MMA) is a metabolic disorder most commonly caused by mutations in the methylmalonyl-CoA mutase ( MMUT ) gene. Although adeno-associated viral (AAV) gene therapy has been effective at correcting the disease phenotype in MMA mouse models, clinical translation may be impaired by loss of episomal transgene expression and magnified by the need to treat patients early in life. To achieve permanent correction, we developed a dual AAV strategy to express a codon-optimized MMUT transgene from Alb and tested various CRISPR-Cas9 genome-editing vectors in newly developed knockin mouse models of MMA. For one target site in intron 1 of Alb , we designed rescue cassettes expressing MMUT behind a 2A-peptide or an internal ribosomal entry site sequence. A second guide RNA targeted the initiator codon, and the donor cassette encompassed the proximal albumin promoter in the 5' homology arm. Although all editing approaches were therapeutic, targeting the start codon of albumin allowed the use of a donor cassette that also functioned as an episome and after homologous recombination, even without the expression of Cas9, as an integrant. Targeting the albumin locus using these strategies would be effective for other metabolic disorders where early treatment and permanent long-term correction are needed., Competing Interests: The authors declare no competing interests.
- Published
- 2021
- Full Text
- View/download PDF
18. Modelling the functional genomics of Parkinson's disease in Caenorhabditis elegans: LRRK2 and beyond.
- Author
-
Chandler RJ, Cogo S, Lewis PA, and Kevei E
- Subjects
- Animals, Animals, Genetically Modified, Caenorhabditis elegans enzymology, Caenorhabditis elegans Proteins metabolism, Disease Models, Animal, Genetic Predisposition to Disease, Genome-Wide Association Study, Heredity, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Nerve Degeneration, Neurons pathology, Parkinson Disease enzymology, Parkinson Disease pathology, Phenotype, Risk Factors, Caenorhabditis elegans genetics, Caenorhabditis elegans Proteins genetics, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Mutation, Neurons enzymology, Parkinson Disease genetics
- Abstract
For decades, Parkinson's disease (PD) cases have been genetically categorised into familial, when caused by mutations in single genes with a clear inheritance pattern in affected families, or idiopathic, in the absence of an evident monogenic determinant. Recently, genome-wide association studies (GWAS) have revealed how common genetic variability can explain up to 36% of PD heritability and that PD manifestation is often determined by multiple variants at different genetic loci. Thus, one of the current challenges in PD research stands in modelling the complex genetic architecture of this condition and translating this into functional studies. Caenorhabditis elegans provide a profound advantage as a reductionist, economical model for PD research, with a short lifecycle, straightforward genome engineering and high conservation of PD relevant neural, cellular and molecular pathways. Functional models of PD genes utilising C. elegans show many phenotypes recapitulating pathologies observed in PD. When contrasted with mammalian in vivo and in vitro models, these are frequently validated, suggesting relevance of C. elegans in the development of novel PD functional models. This review will discuss how the nematode C. elegans PD models have contributed to the uncovering of molecular and cellular mechanisms of disease, with a focus on the genes most commonly found as causative in familial PD and risk factors in idiopathic PD. Specifically, we will examine the current knowledge on a central player in both familial and idiopathic PD, Leucine-rich repeat kinase 2 (LRRK2) and how it connects to multiple PD associated GWAS candidates and Mendelian disease-causing genes., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
19. Improved systemic AAV gene therapy with a neurotrophic capsid in Niemann-Pick disease type C1 mice.
- Author
-
Davidson CD, Gibson AL, Gu T, Baxter LL, Deverman BE, Beadle K, Incao AA, Rodriguez-Gil JL, Fujiwara H, Jiang X, Chandler RJ, Ory DS, Gradinaru V, Venditti CP, and Pavan WJ
- Subjects
- Animals, Disease Models, Animal, Female, Gene Expression, Genes, Reporter, Genetic Vectors administration & dosage, Male, Mice, Mice, Transgenic, Niemann-Pick C1 Protein genetics, Phenotype, Tissue Distribution, Transgenes, Treatment Outcome, Dependovirus genetics, Gene Transfer Techniques, Genetic Vectors genetics, Niemann-Pick Disease, Type C genetics, Niemann-Pick Disease, Type C therapy, Transduction, Genetic
- Abstract
Niemann-Pick C1 disease (NPC1) is a rare, fatal neurodegenerative disease caused by mutations in NPC1 , which encodes the lysosomal cholesterol transport protein NPC1. Disease pathology involves lysosomal accumulation of cholesterol and lipids, leading to neurological and visceral complications. Targeting the central nervous system (CNS) from systemic circulation complicates treatment of neurological diseases with gene transfer techniques. Selected and engineered capsids, for example, adeno-associated virus (AAV)-PHP.B facilitate peripheral-to-CNS transfer and hence greater CNS transduction than parental predecessors. We report that systemic delivery to Npc1
m1N/m1N mice using an AAV-PHP.B vector ubiquitously expressing NPC1 led to greater disease amelioration than an otherwise identical AAV9 vector. In addition, viral copy number and biodistribution of GFP-expressing reporters showed that AAV-PHP.B achieved more efficient, albeit variable, CNS transduction than AAV9 in Npc1m1N/m1N mice. This variability was associated with segregation of two alleles of the putative AAV-PHP.B receptor Ly6a in Npc1m1N/m1N mice. Our data suggest that robust improvements in NPC1 disease phenotypes occur even with modest CNS transduction and that improved neurotrophic capsids have the potential for superior NPC1 AAV gene therapy vectors., (© 2021 Davidson et al.)- Published
- 2021
- Full Text
- View/download PDF
20. ImmTOR nanoparticles enhance AAV transgene expression after initial and repeat dosing in a mouse model of methylmalonic acidemia.
- Author
-
Ilyinskii PO, Michaud AM, Rizzo GL, Roy CJ, Leung SS, Elkins SL, Capela T, Chowdhury A, Li L, Chandler RJ, Manoli I, Andres-Mateos E, Johnston LPM, Vandenberghe LH, Venditti CP, and Kishimoto TK
- Abstract
A major barrier to adeno-associated virus (AAV) gene therapy is the inability to re-dose patients due to formation of vector-induced neutralizing antibodies (Nabs). Tolerogenic nanoparticles encapsulating rapamycin (ImmTOR) provide long-term and specific suppression of adaptive immune responses, allowing for vector re-dosing. Moreover, co-administration of hepatotropic AAV vectors and ImmTOR leads to an increase of transgene expression even after the first dose. ImmTOR and AAV Anc80 encoding the methylmalonyl-coenzyme A (CoA) mutase (MMUT) combination was tested in a mouse model of methylmalonic acidemia, a disease caused by mutations in the MMUT gene. Repeated co-administration of Anc80 and ImmTOR was well tolerated and led to nearly complete inhibition of immunoglobulin (Ig)G antibodies to the Anc80 capsid. A more profound decrease of plasma levels of the key toxic metabolite, plasma methylmalonic acid (pMMA), and disease biomarker, fibroblast growth factor 21 (FGF21), was observed after treatment with the ImmTOR and Anc80-MMUT combination. In addition, there were higher numbers of viral genomes per cell (vg/cell) and increased transgene expression when ImmTOR was co-administered with Anc80-MMUT. These effects were dose-dependent, with the higher doses of ImmTOR providing higher vg/cell and mRNA levels, and an improved biomarker response. Combining of ImmTOR and AAV can not only block the IgG response against capsid, but it also appears to potentiate transduction and enhance therapeutic transgene expression in the mouse model., Competing Interests: P.O.I., A.M.M., G.L.R., C.J.R., S.S.L., S.L.E., T.C., A.C., L.P.M.J., and T.K.K. are employees and shareholders of Selecta Biosciences. E.A.-M is an employee and holds stocks of Akouos, Inc. L.H.V. received consulting fees and research funding from Selecta Biosciences and holds equity in and serves on the Scientific Advisory Board of Akouos. He is an inventor of Anc80L65, licensed to biopharmaceutical companies, including Selecta Biosciences, from which he receives royalties. C.P.V. received research funding from Selecta Biosciences. R.J.C., L.L., I.M., and C.P.V. are co-inventors on patents and patent applications filed by the NIH on their behalf.
- Published
- 2021
- Full Text
- View/download PDF
21. Central nervous system-targeted adeno-associated virus gene therapy in methylmalonic acidemia.
- Author
-
May FJ, Head PE, Venturoni LE, Chandler RJ, and Venditti CP
- Abstract
Methylmalonic acidemia (MMA) is a severe metabolic disorder most commonly caused by a mutation in the methylmalonyl-CoA mutase ( MMUT ) gene. Patients with MMA experience multisystemic disease manifestations and remain at risk for neurological disease progression, even after liver transplantation. Therefore, delivery of MMUT to the central nervous system (CNS) may provide patients with neuroprotection and, perhaps, therapeutic benefits. To specifically target the brain, we developed a neurotropic PHP.eB vector that used a CaMKII neuro-specific promoter to restrict the expression of the MMUT transgene in the neuraxis and delivered the adeno-associated virus (AAV) to mice with MMA. The PHP.eB vector transduced cells in multiple brain regions, including the striatum, and enabled high levels of expression of MMUT in the basal ganglia. Following the CNS-specific correction of MMUT expression, disease-related metabolites methylmalonic acid and 2-methylcitrate were significantly (p < 0.02) decreased in serum of treated MMA mice. Our results show that targeting MMUT expression to the CNS using a neurotropic capsid can decrease the circulating metabolite load in MMA and further highlight the benefit of extrahepatic correction for disorders of organic acid metabolism., Competing Interests: The NIH has filed patents related to MMUT AAV gene therapy vectors on behalf of R.J.C. and C.P.V.
- Published
- 2021
- Full Text
- View/download PDF
22. Promoterless, Nuclease-Free Genome Editing Confers a Growth Advantage for Corrected Hepatocytes in Mice With Methylmalonic Acidemia.
- Author
-
Chandler RJ, Venturoni LE, Liao J, Hubbard BT, Schneller JL, Hoffmann V, Gordo S, Zang S, Ko CW, Chau N, Chiang K, Kay MA, Barzel A, and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors metabolism, Animals, Animals, Newborn, Biomarkers blood, Carcinoma, Hepatocellular pathology, Disease Models, Animal, Fibroblast Growth Factors blood, Hepatocytes, Liver Neoplasms pathology, Liver Transplantation, Malonates blood, Methylmalonyl-CoA Mutase genetics, Mice, Mice, Inbred C57BL, Amino Acid Metabolism, Inborn Errors therapy, Dependovirus genetics, Gene Editing methods, Genetic Therapy methods, Methylmalonyl-CoA Mutase metabolism
- Abstract
Background and Aims: Adeno-associated viral (AAV) gene therapy has shown great promise as an alternative treatment for metabolic disorders managed using liver transplantation, but remains limited by transgene loss and genotoxicity. Our study aims to test an AAV vector with a promoterless integrating cassette, designed to provide sustained hepatic transgene expression and reduced toxicity in comparison to canonical AAV therapy., Approach and Results: Our AAV vector was designed to insert a methylmalonyl-CoA mutase (MMUT) transgene into the 3' end of the albumin locus and tested in mouse models of methylmalonic acidemia (MMA). After neonatal delivery, we longitudinally evaluated hepatic transgene expression, plasma levels of methylmalonate, and the MMA biomarker, fibroblast growth factor 21 (Fgf21), as well as integration of MMUT in the albumin locus. At necropsy, we surveyed for AAV-related hepatocellular carcinoma (HCC) in all treated MMA mice and control littermates. AAV-mediated genome editing of MMUT into the albumin locus resulted in permanent hepatic correction in MMA mouse models, which was accompanied by decreased levels of methylmalonate and Fgf21, and improved survival without HCC. With time, levels of transgene expression increased and methylmalonate progressively decreased, whereas the number of albumin-MMUT integrations and corrected hepatocytes in MMA mice increased, but not in similarly treated wild-type animals. Additionally, expression of MMUT in the setting of MMA conferred a selective growth advantage upon edited cells, which potentiates the therapeutic response., Conclusions: In conclusion, our findings demonstrate that AAV-mediated, promoterless, nuclease-free genome editing at the albumin locus provides safe and durable therapeutic benefit in neonatally treated MMA mice., (© 2020 The Authors. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2021
- Full Text
- View/download PDF
23. Enhanced Efficacy and Increased Long-Term Toxicity of CNS-Directed, AAV-Based Combination Therapy for Krabbe Disease.
- Author
-
Li Y, Miller CA, Shea LK, Jiang X, Guzman MA, Chandler RJ, Ramakrishnan SM, Smith SN, Venditti CP, Vogler CA, Ory DS, Ley TJ, and Sands MS
- Subjects
- Animals, Bone Marrow Transplantation methods, Carcinoma, Hepatocellular etiology, Combined Modality Therapy, Disease Models, Animal, Genetic Therapy methods, Genetic Vectors administration & dosage, Liver Neoplasms etiology, Mice, Dependovirus genetics, Genetic Therapy adverse effects, Genetic Vectors genetics, Leukodystrophy, Globoid Cell complications, Leukodystrophy, Globoid Cell therapy
- Abstract
Infantile globoid cell leukodystrophy (GLD, Krabbe disease) is a demyelinating disease caused by the deficiency of the lysosomal enzyme galactosylceramidase (GALC) and the progressive accumulation of the toxic metabolite psychosine. We showed previously that central nervous system (CNS)-directed, adeno-associated virus (AAV)2/5-mediated gene therapy synergized with bone marrow transplantation and substrate reduction therapy (SRT) to greatly increase therapeutic efficacy in the murine model of Krabbe disease (Twitcher). However, motor deficits remained largely refractory to treatment. In the current study, we replaced AAV2/5 with an AAV2/9 vector. This single change significantly improved several endpoints primarily associated with motor function. However, nearly all (14/16) of the combination-treated Twitcher mice and all (19/19) of the combination-treated wild-type mice developed hepatocellular carcinoma (HCC). 10 out of 10 tumors analyzed had AAV integrations within the Rian locus. Several animals had additional integrations within or near genes that regulate cell growth or death, are known or potential tumor suppressors, or are associated with poor prognosis in human HCC. Finally, the substrate reduction drug L-cycloserine significantly decreased the level of the pro-apoptotic ceramide 18:0. These data demonstrate the value of AAV-based combination therapy for Krabbe disease. However, they also suggest that other therapies or co-morbidities must be taken into account before AAV-mediated gene therapy is considered for human therapeutic trials., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The American Society of Gene and Cell Therapy. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Low incidence of hepatocellular carcinoma in mice and cats treated with systemic adeno-associated viral vectors.
- Author
-
Ferla R, Alliegro M, Dell'Anno M, Nusco E, Cullen JM, Smith SN, Wolfsberg TG, O'Donnell P, Wang P, Nguyen AD, Chandler RJ, Chen Z, Burgess SM, Vite CH, Haskins ME, Venditti CP, and Auricchio A
- Abstract
Adeno-associated viral (AAV) vectors have emerged as the preferred platform for in vivo gene transfer because of their combined efficacy and safety. However, insertional mutagenesis with the subsequent development of hepatocellular carcinomas (HCCs) has been recurrently noted in newborn mice treated with high doses of AAV, and more recently, the association of wild-type AAV integrations in a subset of human HCCs has been documented. Here, we address, in a comprehensive, prospective study, the long-term risk of tumorigenicity in young adult mice following delivery of single-stranded AAVs targeting liver. HCC incidence in mice treated with therapeutic and reporter AAVs was low, in contrast to what has been previously documented in mice treated as newborns with higher doses of AAV. Specifically, HCCs developed in 6 out 76 of AAV-treated mice, and a pathogenic integration of AAV was found in only one tumor. Also, no evidence of liver tumorigenesis was found in juvenile AAV-treated mucopolysaccharidosis type VI (MPS VI) cats followed as long as 8 years after vector administration. Together, our results support the low risk of tumorigenesis associated with AAV-mediated gene transfer targeting juvenile/young adult livers, although constant monitoring of subjects enrolled in AAV clinical trial is advisable., Competing Interests: A.A. is founder and consultant of InnovaVector srl. The other authors have no competing interests to declare., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
25. The Platform Vector Gene Therapies Project: Increasing the Efficiency of Adeno-Associated Virus Gene Therapy Clinical Trial Startup.
- Author
-
Brooks PJ, Ottinger EA, Portero D, Lomash RM, Alimardanov A, Terse P, Xu X, Chandler RJ, Geist Hauserman J, Esposito E, Bönnemann CG, Venditti CP, Austin CP, Pariser A, and Lo DC
- Subjects
- Genetic Vectors genetics, Humans, Rare Diseases genetics, Clinical Trials as Topic standards, Dependovirus genetics, Genetic Therapy methods, Genetic Therapy standards, Genetic Vectors administration & dosage, Rare Diseases therapy
- Published
- 2020
- Full Text
- View/download PDF
26. Pathogenesis of Hepatic Tumors following Gene Therapy in Murine and Canine Models of Glycogen Storage Disease.
- Author
-
Kang HR, Gjorgjieva M, Smith SN, Brooks ED, Chen Z, Burgess SM, Chandler RJ, Waskowicz LR, Grady KM, Li S, Mithieux G, Venditti CP, Rajas F, and Koeberl DD
- Abstract
Glycogen storage disease type Ia (GSD Ia) is caused by mutations in the glucose-6-phosphatase (G6Pase) catalytic subunit gene ( G6PC ). GSD Ia complications include hepatocellular adenomas (HCA) with a risk for hepatocellular carcinoma (HCC) formation. Genome editing with adeno-associated virus (AAV) vectors containing a zinc-finger nuclease (ZFN) and a G6PC donor transgene was evaluated in adult mice with GSD Ia. Although mouse livers expressed G6Pase, HCA and HCC occurred following AAV vector administration. Interestingly, vector genomes were almost undetectable in the tumors but remained relatively high in adjacent liver (p < 0.01). G6Pase activity was decreased in tumors, in comparison with adjacent liver (p < 0.01). Furthermore, AAV-G6Pase vector-treated dogs with GSD Ia developed HCC with lower G6Pase activity (p < 0.01) in comparison with adjacent liver. AAV integration and tumor marker analysis in mice revealed that tumors arose from the underlying disorder, not from vector administration. Similarly to human GSD Ia-related HCA and HCC, mouse and dog tumors did not express elevated α-fetoprotein. Taken together, these results suggest that AAV-mediated gene therapy not only corrects hepatic G6Pase deficiency, but also has potential to suppress HCA and HCC in the GSD Ia liver., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
27. Messenger RNA therapy as an option for treating metabolic disorders.
- Author
-
Chandler RJ
- Subjects
- Arginase, Humans, Lipids, RNA, Messenger, Hyperargininemia, Metabolic Diseases, Nanoparticles
- Abstract
Competing Interests: The author declares no competing interest.
- Published
- 2019
- Full Text
- View/download PDF
28. Gene Therapy for Methylmalonic Acidemia: Past, Present, and Future.
- Author
-
Chandler RJ and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors enzymology, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors pathology, Animals, Dependovirus metabolism, Disease Models, Animal, Gene Editing methods, Gene Transfer Techniques, Genetic Vectors chemistry, Genetic Vectors metabolism, Humans, Lentivirus metabolism, Methylmalonyl-CoA Mutase deficiency, Mice, Mitochondria enzymology, Mitochondria pathology, Nanoparticles administration & dosage, Nanoparticles metabolism, Proof of Concept Study, RNA, Messenger metabolism, Amino Acid Metabolism, Inborn Errors therapy, Dependovirus genetics, Genetic Therapy methods, Lentivirus genetics, Methylmalonyl-CoA Mutase genetics, RNA, Messenger genetics
- Abstract
Methylmalonic acidemia (MMA) is a severe, and sometimes lethal, monogenic metabolic disorder in need of improved treatments. A number of new genomic therapies, which include canonical adeno-associated virus gene addition, genome editing, and systemic mRNA therapy, have shown great promise in murine models of MMA. Each approach has unique advantages and disadvantages for treating genetic disorders like MMA. This article reviews traditional viral gene therapy experiments that have provided enabling proof of concept studies in animal models, and newer approaches that may emerge as effective treatments for MMA and related disorders of organic acid metabolism.
- Published
- 2019
- Full Text
- View/download PDF
29. Compliance in autism: Self-report in action.
- Author
-
Chandler RJ, Russell A, and Maras KL
- Subjects
- Adult, Anxiety psychology, Case-Control Studies, Female, Humans, Male, Self Report, Young Adult, Autistic Disorder psychology, Bullying psychology, Coercion, Cooperative Behavior, Crime Victims psychology, Self Concept
- Abstract
Previous research indicates that autistic individuals are more likely to be bullied, and that they experience heightened anxiety and diminished self-esteem. These factors are known to predict heightened compliance, which is the tendency to agree with or carry out the requests and demands of others. This has a range of potentially serious consequences, particularly for an autistic person. This study utilised self-report (the Gudjonsson Compliance Scale) and behavioural measures of compliance (the door-in-the-face task) with 26 autistic and 26 typically developing adults. Participants also completed measures of early life bullying experiences, anxiety and self-esteem. Autistic participants were more compliant on both self-report and experimental tasks, and they reported more bullying experiences, higher anxiety and reduced self-esteem. Looking at both groups, bullying, anxiety and self-esteem were all correlated with self-reported compliance on the Gudjonsson Compliance Scale, yet only self-esteem was a unique predictor. None of these predictor variables related to behavioural compliance on the door in the face; nor did Gudjonsson Compliance Scale scores predict door-in-the-face performance, which may be better explained by situational and motivational factors. Findings have important implications for a range of real-life settings including requests made in the context of research, schools, the criminal justice system and the workplace.
- Published
- 2019
- Full Text
- View/download PDF
30. FGF21 underlies a hormetic response to metabolic stress in methylmalonic acidemia.
- Author
-
Manoli I, Sysol JR, Epping MW, Li L, Wang C, Sloan JL, Pass A, Gagné J, Ktena YP, Li L, Trivedi NS, Ouattara B, Zerfas PM, Hoffmann V, Abu-Asab M, Tsokos MG, Kleiner DE, Garone C, Cusmano-Ozog K, Enns GM, Vernon HJ, Andersson HC, Grunewald S, Elkahloun AG, Girard CL, Schnermann J, DiMauro S, Andres-Mateos E, Vandenberghe LH, Chandler RJ, and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors pathology, Animals, Biomarkers blood, Disease Models, Animal, Female, Fibroblast Growth Factors blood, Genetic Therapy, Humans, Kidney Diseases metabolism, Liver metabolism, Liver pathology, Liver Transplantation, Male, Methylmalonyl-CoA Mutase genetics, Mice, Mice, Knockout, Mice, Transgenic, Mitochondria metabolism, Mitochondria pathology, Phenotype, Transcriptome, Amino Acid Metabolism, Inborn Errors metabolism, Fibroblast Growth Factors metabolism, Hormesis, Methylmalonyl-CoA Mutase metabolism, Stress, Physiological
- Abstract
Methylmalonic acidemia (MMA), an organic acidemia characterized by metabolic instability and multiorgan complications, is most frequently caused by mutations in methylmalonyl-CoA mutase (MUT). To define the metabolic adaptations in MMA in acute and chronic settings, we studied a mouse model generated by transgenic expression of Mut in the muscle. Mut-/-;TgINS-MCK-Mut mice accurately replicate the hepatorenal mitochondriopathy and growth failure seen in severely affected patients and were used to characterize the response to fasting. The hepatic transcriptome in MMA mice was characterized by the chronic activation of stress-related pathways and an aberrant fasting response when compared with controls. A key metabolic regulator, Fgf21, emerged as a significantly dysregulated transcript in mice and was subsequently studied in a large patient cohort. The concentration of plasma FGF21 in MMA patients correlated with disease subtype, growth indices, and markers of mitochondrial dysfunction but was not affected by renal disease. Restoration of liver Mut activity, by transgenesis and liver-directed gene therapy in mice or liver transplantation in patients, drastically reduced plasma FGF21 and was associated with improved outcomes. Our studies identify mitocellular hormesis as a hepatic adaptation to metabolic stress in MMA and define FGF21 as a highly predictive disease biomarker.
- Published
- 2018
- Full Text
- View/download PDF
31. Systemic Messenger RNA Therapy as a Treatment for Methylmalonic Acidemia.
- Author
-
An D, Schneller JL, Frassetto A, Liang S, Zhu X, Park JS, Theisen M, Hong SJ, Zhou J, Rajendran R, Levy B, Howell R, Besin G, Presnyak V, Sabnis S, Murphy-Benenato KE, Kumarasinghe ES, Salerno T, Mihai C, Lukacs CM, Chandler RJ, Guey LT, Venditti CP, and Martini PGV
- Published
- 2018
- Full Text
- View/download PDF
32. Systemic Messenger RNA Therapy as a Treatment for Methylmalonic Acidemia.
- Author
-
An D, Schneller JL, Frassetto A, Liang S, Zhu X, Park JS, Theisen M, Hong SJ, Zhou J, Rajendran R, Levy B, Howell R, Besin G, Presnyak V, Sabnis S, Murphy-Benenato KE, Kumarasinghe ES, Salerno T, Mihai C, Lukacs CM, Chandler RJ, Guey LT, Venditti CP, and Martini PGV
- Subjects
- Administration, Intravenous, Animals, Female, Humans, Lipids chemistry, Liver metabolism, Male, Methylmalonyl-CoA Mutase metabolism, Mice, Nanoparticles chemistry, RNA, Messenger metabolism, Amino Acid Metabolism, Inborn Errors therapy, Genetic Therapy methods, Methylmalonyl-CoA Mutase genetics, Nanoparticles administration & dosage, RNA, Messenger genetics
- Abstract
Isolated methylmalonic acidemia/aciduria (MMA) is a devastating metabolic disorder with poor outcomes despite current medical treatments. Like other mitochondrial enzymopathies, enzyme replacement therapy (ERT) is not available, and although promising, AAV gene therapy can be limited by pre-existing immunity and has been associated with genotoxicity in mice. To develop a new class of therapy for MMA, we generated a pseudoU-modified codon-optimized mRNA encoding human methylmalonyl-CoA mutase (hMUT), the enzyme most frequently mutated in MMA, and encapsulated it into biodegradable lipid nanoparticles (LNPs). Intravenous (i.v.) administration of hMUT mRNA in two different mouse models of MMA resulted in a 75%-85% reduction in plasma methylmalonic acid and was associated with increased hMUT protein expression and activity in liver. Repeat dosing of hMUT mRNA reduced circulating metabolites and dramatically improved survival and weight gain. Additionally, repeat i.v. dosing did not increase markers of liver toxicity or inflammation in heterozygote MMA mice., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
33. Recombinant Adeno-Associated Viral Integration and Genotoxicity: Insights from Animal Models.
- Author
-
Chandler RJ, Sands MS, and Venditti CP
- Subjects
- Animals, Genetic Therapy adverse effects, Humans, Mice, Dependovirus genetics, Gene Transfer Techniques adverse effects, Genetic Therapy methods
- Abstract
Currently, clinical gene therapy is experiencing a renaissance, with new products for clinical use approved in Europe and clinical trials for multiple diseases reporting positive results, especially those using recombinant adeno-associated viral (rAAV) vectors. Amid this new success, it is prudent to recall that the field of gene therapy experienced tragic setbacks in 1999 and 2002 because of the serious adverse events related to retroviral and adenoviral gene delivery in two clinical trials that resulted in the death of two patients. In both cases, the toxicity observed in humans had been documented to occur in animal models. However, these toxicities were either undetected or underappreciated before they arose in humans. rAAVs have been tested extensively in animals and animal models of disease, largely without adverse events, except for transient elevation in liver enzymes in some patients. However, a small but growing number of murine studies have documented that adeno-associated viral gene delivery can result in insertional mutagenesis. Herein, the aggregate data are reviewed from multiple murine studies where genotoxicity associated with rAAV treatment has been observed. The data emphasize the need for a proactive position to evaluate the potential risks and possible solutions associated with AAV-mediated gene therapy.
- Published
- 2017
- Full Text
- View/download PDF
34. Systemic AAV9 gene therapy improves the lifespan of mice with Niemann-Pick disease, type C1.
- Author
-
Chandler RJ, Williams IM, Gibson AL, Davidson CD, Incao AA, Hubbard BT, Porter FD, Pavan WJ, and Venditti CP
- Subjects
- Animals, Cerebellum metabolism, Cerebellum pathology, Cholesterol metabolism, Disease Models, Animal, Female, Intracellular Signaling Peptides and Proteins, Male, Mice, Mice, Inbred BALB C, Mice, Knockout, Neurons metabolism, Neurons pathology, Niemann-Pick C1 Protein, Niemann-Pick Disease, Type C genetics, Purkinje Cells metabolism, Purkinje Cells pathology, Dependovirus genetics, Genetic Therapy, Genetic Vectors administration & dosage, Longevity genetics, Niemann-Pick Disease, Type C therapy, Proteins genetics
- Abstract
Niemann-Pick disease, type C1 (NPC1) is a heritable lysosomal storage disease characterized by a progressive neurological degeneration that causes disability and premature death. A murine model of NPC1 disease (Npc1-/-) displays a rapidly progressing form of NPC1 disease which is characterized by weight loss, ataxia, increased cholesterol storage, loss of cerebellar Purkinje neurons and early lethality. To test the potential efficacy of gene therapy for NPC1, we constructed adeno-associated virus serotype 9 (AAV9) vectors to deliver the NPC1 gene under the transcriptional control of the neuronal-specific (CamKII) or a ubiquitous (EF1a) promoter. The Npc1-/- mice that received a single dose of AAV9-CamKII-NPC1 as neonates (2.6 × 1011GC) or at weaning (1.3 × 1012GC), and the mice that received a single dose of AAV9-EF1a-NPC1 at weaning (1.2 × 1012GC), exhibited an increased life span, characterized by delayed weight loss and diminished motor decline. Cholesterol storage and Purkinje neuron loss were also reduced in the central nervous system of AAV9 treated Npc1-/- mice. Treatment with AAV9-EF1a-NPC1, as compared to AAV9-CamKII-NPC1, resulted in significantly increased survival (mean survival increased from 69 days to 166 and 97 days, respectively) and growth, and reduced hepatic-cholesterol accumulation. Our results provide the first demonstration that gene therapy may represent a therapeutic option for NPC1 patients and suggest that extraneuronal NPC1 expression can further augment the lifespan of the Npc1-/- mice after systemic AAV gene delivery., (Published by Oxford University Press 2016. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2017
- Full Text
- View/download PDF
35. Neutralizing Antibodies Against Adeno-Associated Viral Capsids in Patients with mut Methylmalonic Acidemia.
- Author
-
Harrington EA, Sloan JL, Manoli I, Chandler RJ, Schneider M, McGuire PJ, Calcedo R, Wilson JM, and Venditti CP
- Subjects
- Adolescent, Adult, Alleles, Amino Acid Metabolism, Inborn Errors enzymology, Amino Acid Metabolism, Inborn Errors therapy, Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Cell Line, Child, Child, Preschool, Cross Reactions immunology, Dependovirus classification, Dependovirus genetics, Female, Genetic Therapy methods, Genetic Vectors genetics, Genotype, Humans, Male, Methylmalonyl-CoA Mutase metabolism, Mice, Young Adult, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Capsid immunology, Dependovirus immunology, Genetic Therapy adverse effects, Genetic Vectors immunology, Methylmalonyl-CoA Mutase genetics
- Abstract
Isolated methylmalonic acidemia (MMA), a group of autosomal recessive inborn errors of metabolism, is most commonly caused by complete (mut(0)) or partial (mut(-)) deficiency of the enzyme methylmalonyl-CoA mutase (MUT). The severe metabolic instability and increased mortality experienced by many affected individuals, especially those with mut(0) MMA, has led centers to use elective liver transplantation as a treatment for these patients. We have previously demonstrated the efficacy of systemic adeno-associated viral (AAV) gene delivery as a treatment for MMA in a murine model and therefore sought to survey AAV antibody titers against serotypes 2, 8, and 9 in a group of well-characterized MMA patients, accrued via a dedicated natural history study ( clinicaltrials.gov ID: NCT00078078). Plasma samples provided by 42 patients (8 mut(-) and 34 mut(0); 10 had received organ transplantation), who ranged in age between 2 and 31 years, were analyzed to examine AAV2 (n = 35), AAV8 (n = 41), and AAV9 (n = 42) antibody titers. In total, the seroprevalence of antibodies against AAV2, AAV8, or AAV9 was 20%, 22%, and 24%, respectively. We observed a lower-than-expected seropositivity rate (titers ≥1:20) in the pediatric MMA patients (2-18 years) for both AAV2 (p < 0.05) and AAV8 (p < 0.01) neutralizing antibodies (NAbs) compared with historical controls. Those with positive NAb titers were typically older than 18 years (p < 0.05 all serotypes) or had received solid organ transplantation (p < 0.01 AAV8, AAV9). The mut(0) patients who had not been transplanted (n = 24)-that is, the subset with the greatest need for improved treatments-represented the seronegative majority, with 21 out of 24 patients lacking Abs against all AAV capsids tested. The unexpected lack of NAbs against AAV in this patient population has encouraging implications for systemic gene delivery as a treatment for mut MMA.
- Published
- 2016
- Full Text
- View/download PDF
36. The proteome of methylmalonic acidemia (MMA): the elucidation of altered pathways in patient livers.
- Author
-
Caterino M, Chandler RJ, Sloan JL, Dorko K, Cusmano-Ozog K, Ingenito L, Strom SC, Imperlini E, Scolamiero E, Venditti CP, and Ruoppolo M
- Subjects
- Amino Acid Metabolism, Inborn Errors surgery, Case-Control Studies, Child, Preschool, Female, Humans, Infant, Liver Transplantation, Male, Metabolic Networks and Pathways, Two-Dimensional Difference Gel Electrophoresis, Amino Acid Metabolism, Inborn Errors metabolism, Liver metabolism, Proteome metabolism
- Abstract
Methylmalonic acidemia (MMA) is a heterogeneous and severe autosomal recessive inborn error of metabolism most commonly caused by the deficient activity of the vitamin B12 dependent enzyme, methylmalonyl-CoA mutase (MUT). The main treatment for MMA patients is the dietary restriction of propiogenic amino acids and carnitine supplementation. Despite treatment, the prognosis for vitamin B12 non-responsive patients remains poor and is associated with neonatal lethality, persistent morbidity and decreased life expectancy. While multi-organ pathology is a feature of MMA, the liver is severely impacted by mitochondrial dysfunction which likely underlies the metabolic instability experienced by the patients. Liver and/or combined liver/kidney transplantation is therefore sometimes performed in severely affected patients. Using liver specimens from donors and MMA patients undergoing elective liver transplantation collected under a dedicated natural history protocol (clinicaltrials.gov: NCT00078078), we employed proteomics to characterize the liver pathology and impaired hepatic metabolism observed in the patients. Pathway analysis revealed perturbations of enzymes involved in energy metabolism, gluconeogenesis and Krebs cycle anaplerosis. Our findings identify new pathophysiologic and therapeutic targets that could be valuable for designing alternative therapies to alleviate clinical manifestations seen in this disorder., Competing Interests: Marianna Caterino declares that she has no conflict of interest. Randy J. Chandler declares that she has no conflict of interest. Jennifer L. Sloan declares that she has no conflict of interest. Kenneth Dorko declares that she has no conflict of interest. Kristina Cusmano-Ozog declares that she has no conflict of interest. Laura Ingenito declares that she has no conflict of interest. Stephen C. Strom declares that she has no conflict of interest. Esther Imperlini declares that she has no conflict of interest. Emanuela Scolamiero declares that he has no conflict of interest. Charles P. Venditti declares that he has no conflict of interest. Margherita Ruoppolo declares that she has no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
37. Genotoxicity in Mice Following AAV Gene Delivery: A Safety Concern for Human Gene Therapy?
- Author
-
Chandler RJ, LaFave MC, Varshney GK, Burgess SM, and Venditti CP
- Subjects
- Animals, Dependovirus immunology, Drug Evaluation, Preclinical, Humans, Immunity, Cellular, Immunity, Humoral, Mice, DNA Damage, Dependovirus physiology, Genetic Therapy adverse effects
- Published
- 2016
- Full Text
- View/download PDF
38. Gene Therapy for Metabolic Diseases.
- Author
-
Chandler RJ and Venditti CP
- Abstract
Gene therapy has recently shown great promise as an effective treatment for a number of metabolic diseases caused by genetic defects in both animal models and human clinical trials. Most of the current success has been achieved using a viral mediated gene addition approach, but gene-editing technology has progressed rapidly and gene modification is being actively pursued in clinical trials. This review focuses on viral mediated gene addition approaches, because most of the current clinical trials utilize this approach to treat metabolic diseases.
- Published
- 2016
- Full Text
- View/download PDF
39. Vector design influences hepatic genotoxicity after adeno-associated virus gene therapy.
- Author
-
Chandler RJ, LaFave MC, Varshney GK, Trivedi NS, Carrillo-Carrasco N, Senac JS, Wu W, Hoffmann V, Elkahloun AG, Burgess SM, and Venditti CP
- Subjects
- Animals, Mice, Mice, Mutant Strains, Pregnancy Proteins genetics, Pregnancy Proteins metabolism, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular therapy, Dependovirus, Genetic Therapy methods, Genetic Vectors, Liver Neoplasms genetics, Liver Neoplasms metabolism, Liver Neoplasms therapy, Transduction, Genetic
- Abstract
The use of adeno-associated virus (AAV) as a gene therapy vector has been approved recently for clinical use and has demonstrated efficacy in a growing number of clinical trials. However, the safety of AAV as a vector has been challenged by a single study that documented hepatocellular carcinoma (HCC) after AAV gene delivery in mice. Most studies have not noted genotoxicity following AAV-mediated gene delivery; therefore, the possibility that there is an association between AAV and HCC is controversial. Here, we performed a comprehensive study of HCC in a large number of mice following therapeutic AAV gene delivery. Using a sensitive high-throughput integration site-capture technique and global expressional analysis, we found that AAV integration into the RNA imprinted and accumulated in nucleus (Rian) locus, and the resulting overexpression of proximal microRNAs and retrotransposon-like 1 (Rtl1) were associated with HCC. In addition, we demonstrated that the AAV vector dose, enhancer/promoter selection, and the timing of gene delivery are all critical factors for determining HCC incidence after AAV gene delivery. Together, our results define aspects of AAV-mediated gene therapy that influence genotoxicity and suggest that these features should be considered for design of both safer AAV vectors and gene therapy studies.
- Published
- 2015
- Full Text
- View/download PDF
40. Liver-directed adeno-associated virus serotype 8 gene transfer rescues a lethal murine model of citrullinemia type 1.
- Author
-
Chandler RJ, Tarasenko TN, Cusmano-Ozog K, Sun Q, Sutton VR, Venditti CP, and McGuire PJ
- Subjects
- Ammonia blood, Animals, Argininosuccinate Synthase deficiency, Argininosuccinate Synthase metabolism, Citrulline blood, Dependovirus classification, Disease Models, Animal, Genetic Vectors, Humans, Liver virology, Mice, Organ Specificity, Phenotype, Thyroxine-Binding Globulin genetics, Argininosuccinate Synthase genetics, Citrullinemia genetics, Citrullinemia therapy, Dependovirus genetics, Genetic Therapy, Liver enzymology
- Abstract
Citrullinemia type 1 (CTLN1) is an autosomal recessive disorder of metabolism caused by a deficiency of argininosuccinate synthetase. Despite optimal management, CTLN1 patients still suffer from lethal metabolic instability and experience life-threatening episodes of acute hyperammonemia. A murine model of CTLN1 (fold/fold) that displays lethality within the first 21 days of life was used to determine the efficacy of adeno-associated viral (AAV) gene transfer as a potential therapy. An AAV serotype 8 (AAV8) vector was engineered to express the human ASS1 cDNA under the control of a liver-specific promoter (thyroxine-binding globulin, TBG), AAV8-TBG-hASS1, and delivered to 7-10 days old mice via intraperitoneal injection. Greater than 95% of the mice were rescued from lethality and survival was extended beyond 100 days after receiving a single dose of vector. AAV8-TBG-hASS1 treatment resulted in liver-specific expression of hASS1, increased ASS1 enzyme activity, reduction in plasma ammonia and citrulline concentrations and significant phenotypic improvement of the fold/fold growth and skin phenotypes. These experiments highlight a gene transfer approach using AAV8 vector for liver-targeted gene therapy that could serve as a treatment for CTLN1.
- Published
- 2013
- Full Text
- View/download PDF
41. Targeting proximal tubule mitochondrial dysfunction attenuates the renal disease of methylmalonic acidemia.
- Author
-
Manoli I, Sysol JR, Li L, Houillier P, Garone C, Wang C, Zerfas PM, Cusmano-Ozog K, Young S, Trivedi NS, Cheng J, Sloan JL, Chandler RJ, Abu-Asab M, Tsokos M, Elkahloun AG, Rosen S, Enns GM, Berry GT, Hoffmann V, DiMauro S, Schnermann J, and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors pathology, Animals, Antioxidants therapeutic use, Biomarkers metabolism, Blotting, Western, DNA Primers genetics, Enzyme-Linked Immunosorbent Assay, Fluorescein-5-isothiocyanate, Genotype, Glomerular Filtration Rate genetics, Humans, Immunohistochemistry, Methylmalonyl-CoA Mutase genetics, Methylmalonyl-CoA Mutase metabolism, Mice, Mice, Knockout, Microarray Analysis, Microscopy, Electron, Transmission, Nephritis, Interstitial genetics, Real-Time Polymerase Chain Reaction, Transgenes genetics, Ubiquinone pharmacology, Amino Acid Metabolism, Inborn Errors drug therapy, Amino Acid Metabolism, Inborn Errors enzymology, Antioxidants pharmacology, Disease Models, Animal, Kidney Tubules, Proximal physiopathology, Methylmalonyl-CoA Mutase deficiency
- Abstract
Isolated methylmalonic acidemia (MMA), caused by deficiency of the mitochondrial enzyme methylmalonyl-CoA mutase (MUT), is often complicated by end stage renal disease that is resistant to conventional therapies, including liver transplantation. To establish a viable model of MMA renal disease, Mut was expressed in the liver of Mut(-/-) mice as a stable transgene under the control of an albumin (INS-Alb-Mut) promoter. Mut(-/-);Tg(INS-Alb-Mut) mice, although completely rescued from neonatal lethality that was displayed by Mut(-/-) mice, manifested a decreased glomerular filtration rate (GFR), chronic tubulointerstitial nephritis and ultrastructural changes in the proximal tubule mitochondria associated with aberrant tubular function, as demonstrated by single-nephron GFR studies. Microarray analysis of Mut(-/-);Tg(INS-Alb-Mut) kidneys identified numerous biomarkers, including lipocalin-2, which was then used to monitor the response of the GFR to antioxidant therapy in the mouse model. Renal biopsies and biomarker analysis from a large and diverse patient cohort (ClinicalTrials.gov identifier: NCT00078078) precisely replicated the findings in the animals, establishing Mut(-/-);Tg(INS-Alb-Mut) mice as a unique model of MMA renal disease. Our studies suggest proximal tubular mitochondrial dysfunction is a key pathogenic mechanism of MMA-associated kidney disease, identify lipocalin-2 as a biomarker of increased oxidative stress in the renal tubule, and demonstrate that antioxidants can attenuate the renal disease of MMA.
- Published
- 2013
- Full Text
- View/download PDF
42. Pre-clinical efficacy and dosing of an AAV8 vector expressing human methylmalonyl-CoA mutase in a murine model of methylmalonic acidemia (MMA).
- Author
-
Chandler RJ and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors mortality, Amino Acid Metabolism, Inborn Errors pathology, Amino Acid Sequence, Animals, Disease Models, Animal, Gene Transfer Techniques, Genetic Vectors, Humans, Methylmalonyl-CoA Mutase metabolism, Mice, Mice, Knockout, Molecular Sequence Data, Phenotype, Sequence Alignment, Survival Rate, Amino Acid Metabolism, Inborn Errors therapy, Dependovirus genetics, Genetic Therapy, Methylmalonyl-CoA Mutase genetics
- Abstract
We demonstrate that human methylmalonyl-CoA mutase (MUT), delivered using an AAV serotype 8 vector, rescues the lethal phenotype displayed by mice with MMA and provides long-term phenotypic correction. In addition to defining a lower limit of effective dosing, our studies establish that neither a species barrier to mitochondrial processing nor an apparent immune response to MUT limits the murine model as an experimental platform to test the efficacy of human gene therapy vectors for MMA., (Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
43. Gene therapy in a murine model of methylmalonic acidemia using rAAV9-mediated gene delivery.
- Author
-
Sénac JS, Chandler RJ, Sysol JR, Li L, and Venditti CP
- Subjects
- Animals, Animals, Newborn, Disease Models, Animal, Gene Transfer Techniques, Genetic Vectors, Kidney pathology, Kidney physiology, Liver enzymology, Methylmalonyl-CoA Mutase genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation, Retreatment, Transgenes, Amino Acid Metabolism, Inborn Errors therapy, Dependovirus genetics, Genetic Therapy methods
- Abstract
Methylmalonic acidemia (MMA), an inherited metabolic disorder caused by deficient activity of methylmalonyl-CoA mutase, carries a poor prognosis for long-term survival. While administration of a recombinant adeno-associated virus serotype 8 vector (rAAV8) can rescue Mut(-/-) mice from neonatal lethality and provide sustained phenotypic correction, translation of gene therapy to human subjects will likely require multiple rounds of systemic administration and, ideally, the use of a vector that transduces the kidney. To examine the effectiveness of alternative rAAVs in the treatment of MMA, a serotype 9 rAAV expressing the Mut cDNA was constructed and delivered to newborn Mut(-/-) mice (n=11). rAAV9 gene therapy directed hepatic transgene expression within 24 h and effectively rescued the Mut(-/-) mice from lethality, conferred long-term survival, markedly improved metabolism and resulted in striking preservation of renal function and histology. Systemic readministration of the vector at a dose similar to that used in human clinical trials (2.5 × 10(9) GC of rAAV9 per gram) to older, treated Mut(-/-) mice (n=5) lowered circulating metabolites, increased in vivo propionate oxidative capacity and produced transgene expression in the kidney and liver. Our data support the use of an rAAV9 vector in the acute and chronic treatment of MMA, and highlight the renal tropism afforded by this novel serotype.
- Published
- 2012
- Full Text
- View/download PDF
44. Combined methylmalonic acidemia and homocystinuria, cblC type. I. Clinical presentations, diagnosis and management.
- Author
-
Carrillo-Carrasco N, Chandler RJ, and Venditti CP
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, Infant, Infant, Newborn, Male, Methionine metabolism, Methylmalonic Acid metabolism, Models, Biological, Models, Genetic, Neonatal Screening methods, Oxidoreductases, Pregnancy, Prenatal Diagnosis, Vitamin B 12 Deficiency congenital, Amino Acid Metabolism, Inborn Errors diagnosis, Amino Acid Metabolism, Inborn Errors genetics, Carrier Proteins genetics, Homocystinuria diagnosis, Homocystinuria genetics, Mutation, Vitamin B 12 metabolism
- Abstract
Combined methylmalonic acidemia and homocystinuria, cblC type, is an inborn error of intracellular cobalamin metabolism with a wide spectrum of clinical manifestations that is stated to be the most common inherited disorder of cobalamin metabolism. This metabolic disease is caused by mutations in the MMACHC gene and results in impaired intracellular synthesis of adenosylcobalamin and methylcobalamin, cofactors for the methylmalonyl-CoA mutase and methionine synthase enzymes. Elevated methylmalonic acid and homocysteine with decreased methionine production are the biochemical hallmarks of this disorder. Awareness of the diverse clinical presentations associated with cblC disease is necessary to provide a timely diagnosis, to guide management of affected individuals and to establish a framework for the future treatment of individuals detected through expanded newborn screening. This article reviews the biochemistry, clinical presentations, genotype-phenotype correlations, diagnosis and management of cblC disease.
- Published
- 2012
- Full Text
- View/download PDF
45. Exome sequencing identifies ACSF3 as a cause of combined malonic and methylmalonic aciduria.
- Author
-
Sloan JL, Johnston JJ, Manoli I, Chandler RJ, Krause C, Carrillo-Carrasco N, Chandrasekaran SD, Sysol JR, O'Brien K, Hauser NS, Sapp JC, Dorward HM, Huizing M, Barshop BA, Berry SA, James PM, Champaigne NL, de Lonlay P, Valayannopoulos V, Geschwind MD, Gavrilov DK, Nyhan WL, Biesecker LG, and Venditti CP
- Subjects
- Adolescent, Aged, Amino Acid Sequence, Carboxy-Lyases deficiency, Carboxy-Lyases genetics, Child, Preschool, Coenzyme A Ligases chemistry, Female, Humans, Male, Malonyl Coenzyme A, Methylmalonic Acid, Middle Aged, Molecular Sequence Data, Mutation, Missense, Coenzyme A Ligases genetics, Exons, Metabolism, Inborn Errors genetics
- Abstract
We used exome sequencing to identify the genetic basis of combined malonic and methylmalonic aciduria (CMAMMA). We sequenced the exome of an individual with CMAMMA and followed up with sequencing of eight additional affected individuals (cases). This included one individual who was identified and diagnosed by searching an exome database. We identify mutations in ACSF3, encoding a putative methylmalonyl-CoA and malonyl-CoA synthetase as a cause of CMAMMA. We also examined a canine model of CMAMMA, which showed pathogenic mutations in a predicted ACSF3 ortholog. ACSF3 mutant alleles occur with a minor allele frequency of 0.0058 in ∼1,000 control individuals, predicting a CMAMMA population incidence of ∼1:30,000. ACSF3 deficiency is the first human disorder identified as caused by mutations in a gene encoding a member of the acyl-CoA synthetase family, a diverse group of evolutionarily conserved proteins, and may emerge as one of the more common human metabolic disorders.
- Published
- 2011
- Full Text
- View/download PDF
46. A barrel of monkeys: scAAV8 gene therapy for hemophilia in nonhuman primates.
- Author
-
Chandler RJ and Venditti CP
- Subjects
- Animals, Humans, Capsid Proteins metabolism, Dependovirus genetics, Factor IX metabolism, Genetic Therapy methods, Hemophilia B therapy
- Published
- 2011
- Full Text
- View/download PDF
47. Adeno-associated virus serotype 8 gene transfer rescues a neonatal lethal murine model of propionic acidemia.
- Author
-
Chandler RJ, Chandrasekaran S, Carrillo-Carrasco N, Senac JS, Hofherr SE, Barry MA, and Venditti CP
- Subjects
- Animals, Citrates blood, Disease Models, Animal, Gene Transfer Techniques, Methylmalonyl-CoA Decarboxylase genetics, Methylmalonyl-CoA Decarboxylase metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Survival Analysis, Dependovirus genetics, Genetic Therapy, Genetic Vectors genetics, Propionic Acidemia therapy
- Abstract
Propionic acidemia (PA) is an autosomal recessive disorder of metabolism caused by a deficiency of propionyl-coenzyme A carboxylase (PCC). Despite optimal dietary and cofactor therapy, PA patients still suffer from lethal metabolic instability and experience multisystemic complications. A murine model of PA (Pcca(-/-)) of animals that uniformly die within the first 48 hr of life was used to determine the efficacy of adeno-associated viral (AAV) gene transfer as a potential therapy for PA. An AAV serotype 8 (AAV8) vector was engineered to express the human PCCA cDNA and delivered to newborn mice via an intrahepatic injection. Greater than 64% of the Pcca(-/-) mice were rescued after AAV8-mediated gene transfer and survived until day of life 16 or beyond. Western analysis of liver extracts showed that PCC was completely absent from Pcca(-/-) mice but was restored to greater than wild-type levels after AAV gene therapy. The treated Pcca(-/-) mice also exhibited markedly reduced plasma levels of 2-methylcitrate compared with the untreated Pcca(-/-) mice, which indicates significant PCC enzymatic activity was provided by gene transfer. At the time of this report, the oldest treated Pcca(-/-) mice are over 6 months of age. In summary, AAV gene delivery of PCCA effectively rescues Pcca(-/-) mice from neonatal lethality and substantially ameliorates metabolic markers of the disease. These experiments demonstrate a gene transfer approach using AAV8 that might be used as a treatment for PA, a devastating and often lethal disorder desperately in need of new therapeutic options.
- Published
- 2011
- Full Text
- View/download PDF
48. Liver-directed recombinant adeno-associated viral gene delivery rescues a lethal mouse model of methylmalonic acidemia and provides long-term phenotypic correction.
- Author
-
Carrillo-Carrasco N, Chandler RJ, Chandrasekaran S, and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors enzymology, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors pathology, Amino Acid Metabolism, Inborn Errors therapy, Animals, Disease Models, Animal, Injections, Methylmalonyl-CoA Mutase genetics, Methylmalonyl-CoA Mutase therapeutic use, Mice, Organ Specificity, Phenotype, Survival Analysis, Thyroxine-Binding Proteins metabolism, Time Factors, Dependovirus genetics, Gene Transfer Techniques, Genetic Therapy, Liver metabolism, Recombination, Genetic genetics
- Abstract
Methylmalonic acidemia is a severe metabolic disorder caused by a deficiency of the ubiquitously expressed mitochondrial enzyme, methylmalonyl-CoA mutase (MUT). Liver transplantation has been used to treat a small number of patients with variable success, and whether liver-directed gene therapy might be employed in such a pleiotropic metabolic disorder is uncertain. In this study, we examined the therapeutic effects of hepatocyte-directed delivery of the Mut gene to mice with a severe form of methylmalonic acidemia. We show that a single intrahepatic injection of recombinant adeno-associated virus serotype 8 expressing the Mut gene under the control of the liver-specific thyroxine-binding globulin (TBG) promoter is sufficient to rescue Mut(-/-) mice from neonatal lethality and provide long-term phenotypic correction. Treated Mut(-/-) mice lived beyond 1 year of age, had improved growth, lower plasma methylmalonic acid levels, and an increased capacity to oxidize [1-(13)C]propionate in vivo. The older treated mice showed increased Mut transcription, presumably mediated by upregulation of the TBG promoter during senescence. The results indicate that the stable transduction of a small number of hepatocytes with the Mut gene can be efficacious in the phenotypic correction of an inborn error of organic acid metabolism and support the rapid translation of liver-directed gene therapy vectors already optimized for human subjects to patients with methylmalonic acidemia.
- Published
- 2010
- Full Text
- View/download PDF
49. Ion-abrasion scanning electron microscopy reveals distorted liver mitochondrial morphology in murine methylmalonic acidemia.
- Author
-
Murphy GE, Lowekamp BC, Zerfas PM, Chandler RJ, Narasimha R, Venditti CP, and Subramaniam S
- Subjects
- Amino Acid Metabolism, Inborn Errors pathology, Animals, Blotting, Western, Mice, Microscopy, Confocal, Mitochondria, Liver pathology, Models, Biological, Oxidative Phosphorylation, Amino Acid Metabolism, Inborn Errors metabolism, Methylmalonic Acid metabolism, Microscopy, Electron, Scanning, Mitochondria, Liver ultrastructure
- Abstract
Methylmalonic acidemia is a lethal inborn error of metabolism that causes mitochondrial impairment, multi-organ dysfunction and a shortened lifespan. Previous transmission electron microscope studies of thin sections from normal (Mut(+/+)) and diseased (Mut(-/-)) tissue found that the mitochondria appear to occupy a progressively larger volume of mutant cells with age, becoming megamitochondria. To assess changes in shape and volume of mitochondria resulting from the mutation, we carried out ion-abrasion scanning electron microscopy (IA-SEM), a method for 3D imaging that involves the iterative use of a focused gallium ion beam to abrade the surface of the specimen, and a scanning electron beam to image the newly exposed surface. Using IA-SEM, we show that mitochondria are more convoluted and have a broader distribution of sizes in the mutant tissue. Compared to normal cells, mitochondria from mutant cells have a larger surface-area-to-volume ratio, which can be attributed to their convoluted shape and not to their elongation or reduced volume. The 3D imaging approach and image analysis described here could therefore be useful as a diagnostic tool for the evaluation of disease progression in aberrant cells at resolutions higher than that currently achieved using confocal light microscopy., (Published by Elsevier Inc.)
- Published
- 2010
- Full Text
- View/download PDF
50. Long-term rescue of a lethal murine model of methylmalonic acidemia using adeno-associated viral gene therapy.
- Author
-
Chandler RJ and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors blood, Amino Acid Metabolism, Inborn Errors genetics, Animals, Blotting, Western, Dependovirus genetics, Genetic Vectors genetics, Humans, Methylmalonic Acid blood, Methylmalonyl-CoA Mutase genetics, Methylmalonyl-CoA Mutase metabolism, Mice, Mice, Mutant Strains, Polymerase Chain Reaction, Amino Acid Metabolism, Inborn Errors therapy, Genetic Therapy methods, Methylmalonyl-CoA Mutase physiology
- Abstract
Methylmalonic acidemia (MMA) is an organic acidemia caused by deficient activity of the mitochondrial enzyme methylmalonyl-CoA mutase (MUT). This disorder is associated with lethal metabolic instability and carries a poor prognosis for long-term survival. A murine model of MMA that replicates a severe clinical phenotype was used to examine the efficacy of recombinant adeno-associated virus (rAAV) serotype 8 gene therapy as a treatment for MMA. Lifespan extension, body weight, circulating metabolites, transgene expression, and whole animal propionate oxidation were examined as outcome parameters after gene therapy. One-hundred percent of the untreated Mut(-/-) mice (n = 58) died by day of life (DOL) 72, whereas >95% of the adeno-associated virus-treated Mut(-/-) mice (n = 27) have survived for > or = 1 year. Despite a gradual loss of transgene expression and elevated circulating metabolites in the treated Mut(-/-) mice, the animals are indistinguishable from unaffected control littermates in size and activity levels. These experiments provide the first definitive evidence that gene therapy will have clinical utility in the treatment of MMA and support the development of gene therapy for other organic acidemias.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.