115 results on '"Venditti CP"'
Search Results
2. Defects in muscle branched-chain amino acid oxidation contribute to impaired lipid metabolism
- Author
-
Lerin C, Goldfine AB, Boes T, Liu M, Kasif S, Dreyfuss JM, De Sousa-Coelho AL, Daher G, Manoli I, Sysol JR, Isganaitis E, Jessen N, Goodyear LJ, Beebe K, Gall W, Venditti CP, and Patti ME
- Published
- 2016
3. Enamel defects and salivary methylmalonate in methylmalonic acidemia
- Author
-
Bassim, CW, primary, Wright, JT, additional, Guadagnini, JP, additional, Muralidharan, R, additional, Sloan, J, additional, Domingo, DL, additional, Venditti, CP, additional, and Hart, TC, additional
- Published
- 2009
- Full Text
- View/download PDF
4. Optimization of systemic AAV9 gene therapy in Niemann-Pick disease type C1 mice.
- Author
-
Mylvara AV, Gibson AL, Gu T, Davidson CD, Incao AA, Melnyk K, Pierre-Jacques D, Cologna SM, Venditti CP, Porter FD, and Pavan WJ
- Abstract
Niemann-Pick disease, type C1 (NPC1) is a rare, fatal neurodegenerative disorder caused by pathological variants in NPC1 , which encodes a lysosomal cholesterol transport protein. There are no FDA approved treatments for this disorder. Both systemic and central nervous system delivery of AAV9- hNPC1 have shown significant disease amelioration in NPC1 murine models. To assess the impact of dose and window of therapeutic efficacy in Npc1
m1N mice, we systemically administered three different doses of AAV9- hNPC1 at 4 weeks old and the medium dose at pre-, early, and post-symptomatic timepoints. Higher vector doses and treatment earlier in life were associated with enhanced transduction in the nervous system and resulted in significantly increased lifespan. Similar beneficial effects were noted after gene therapy in Npc1I1061T mice, a model that recapitulates a common human hypomorphic variant. Our findings help define dose ranges, treatment ages, and efficacy in severe and hypomorphic models of NPC1 deficiency and suggest that earlier delivery of AAV9- hNPC1 in a pre-symptomatic disease state is likely to yield optimal outcomes in individuals with NPC1., Competing Interests: Conflict of Interest Statement CPV and WJP have NIH patents filed on work related to NPC1 genes and the AAV gene therapy treatment of NPC1 (US Patent Publication Numbers 20180104289, 20210113635).- Published
- 2024
- Full Text
- View/download PDF
5. Severe heart failure in a unique case of cobalamin-C-deficiency resolved with LVAD implantation and subsequent heart transplantation.
- Author
-
Hjalmarsson C, Backelin C, Thoren A, Bergh N, Sloan JL, Manoli I, Venditti CP, and Dellgren G
- Abstract
Introduction Cobalamin c deficiency (cblC), an inborn error of vitamin B12 metabolism, is caused by mutations of the MMACHC gene. It usually leads to a multisystemic disease; 50% of all patients with cblC have various structural heart defects. Severe congestive heart failure (HF) may also occur and its prognosis is poorly documented. Case report We present the case of a young man who had been diagnosed with cblC due to C331T mutation in the MMACHC gene at the age of 3 days and had been treated with substitution therapy (OH-Cbl, mecobalamine, carnitine, betaine, and calcium folinate) since then. He had mildly impaired cognitive function; an ectopic hypophysis/pituitary insufficiency, with adequate hormone replacement therapy; obstructive sleep apnea syndrome, treated with CPAP, bronchial asthma, and obesity (BMI of 30). The liver and kidney functions were normal. He developed severe dilated cardiomyopathy and HF at the age of 12y. With medical treatment, his condition improved and he was stable (NYHA class II) for several years. Six years later, his status deteriorated rapidly, as he developed advanced HF, INTERMACS 3. The cardiac ultrasound revealed dilated ventricles with severely depressed ejection fraction (EF), increased filling pressures, and pulmonary hypertension (sPAP 60 mmHg). Cardiac MRI showed extremely dilated chambers (LVedv 609 mL, RVedv 398 mL) with pronounced non-compaction, and a left ventricle EF of 13%. A primary prophylactic ICD and a left ventricular assist device (LVAD/HM3) were implanted, and the patient was subsequently listed for heart transplantation (HTx). After 25 months on the waiting list, he underwent an uncomplicated HTx. However postoperatively, he got two episodes of cardiac tamponade, as well as mediastinitis, treated with antibiotics and vaccum assisted closure. He developed severe kidney failure, which fully recovered after two months, and was treated successfully for an early moderate allograft rejection (ISHT 2). At the latest outward visit, twelve months after HTx, the patient was doing excellent. Summary To the best of our knowledge, this is the first ever reported case of a patient with CblC undergoing an LVAD implantation and subsequently a HTx. Although both interventions were complicated with bleeding events, this seems to be a treatment option for advanced HF in patients with CblC., Competing Interests: The authors report no relationships that could be construed as a conflict of interest. There are no relevant financial disclosures from any of the authors., (© 2024 The Authors. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
6. Intellectual disability and autism in propionic acidemia: a biomarker-behavioral investigation implicating dysregulated mitochondrial biology.
- Author
-
Shchelochkov OA, Farmer CA, Chlebowski C, Adedipe D, Ferry S, Manoli I, Pass A, McCoy S, Van Ryzin C, Sloan J, Thurm A, and Venditti CP
- Subjects
- Humans, Male, Female, Child, Child, Preschool, Adolescent, Autistic Disorder metabolism, Autistic Disorder genetics, Adult, Methylmalonyl-CoA Decarboxylase genetics, Methylmalonyl-CoA Decarboxylase metabolism, Young Adult, Carnitine analogs & derivatives, Carnitine metabolism, Carnitine blood, Citrates, Propionic Acidemia genetics, Biomarkers blood, Intellectual Disability genetics, Mitochondria metabolism, Autism Spectrum Disorder metabolism, Autism Spectrum Disorder genetics
- Abstract
Propionic acidemia (PA) is an autosomal recessive condition (OMIM #606054), wherein pathogenic variants in PCCA and PCCB impair the activity of propionyl-CoA carboxylase. PA is associated with neurodevelopmental disorders, including intellectual disability (ID) and autism spectrum disorder (ASD); however, the correlates and mechanisms of these outcomes remain unknown. Using data from a subset of participants with PA enrolled in a dedicated natural history study (n = 33), we explored associations between neurodevelopmental phenotypes and laboratory parameters. Twenty (61%) participants received an ID diagnosis, and 12 of the 31 (39%) who were fully evaluated received the diagnosis of ASD. A diagnosis of ID, lower full-scale IQ (sample mean = 65 ± 26), and lower adaptive behavior composite scores (sample mean = 67 ± 23) were associated with several biomarkers. Higher concentrations of plasma propionylcarnitine, plasma total 2-methylcitrate, serum erythropoietin, and mitochondrial biomarkers plasma FGF21 and GDF15 were associated with a more severe ID profile. Reduced 1-
13 C-propionate oxidative capacity and decreased levels of plasma and urinary glutamine were also associated with a more severe ID profile. Only two parameters, increased serum erythropoietin and decreased plasma glutamine, were associated with ASD. Plasma glycine, one of the defining features of PA, was not meaningfully associated with either ID or ASD. Thus, while both ID and ASD were commonly observed in our PA cohort, only ID was robustly associated with metabolic parameters. Our results suggest that disease severity and associated mitochondrial dysfunction may play a role in CNS complications of PA and identify potential biomarkers and candidate surrogate endpoints., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2024
- Full Text
- View/download PDF
7. Lipodystrophy in methylmalonic acidemia associated with elevated FGF21 and abnormal methylmalonylation.
- Author
-
Manoli I, Sysol JR, Head PE, Epping MW, Gavrilova O, Crocker MK, Sloan JL, Koutsoukos SA, Wang C, Ktena YP, Mendelson S, Pass AR, Zerfas PM, Hoffmann V, Vernon HJ, Fletcher LA, Reynolds JC, Tsokos MG, Stratakis CA, Voss SD, Chen KY, Brown RJ, Hamosh A, Berry GT, Chen XS, Yanovski JA, and Venditti CP
- Subjects
- Animals, Humans, Mice, Mice, Transgenic, Amino Acid Metabolism, Inborn Errors complications, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors metabolism, Fibroblast Growth Factors, Lipodystrophy
- Abstract
A distinct adipose tissue distribution pattern was observed in patients with methylmalonyl-CoA mutase deficiency, an inborn error of branched-chain amino acid (BCAA) metabolism, characterized by centripetal obesity with proximal upper and lower extremity fat deposition and paucity of visceral fat, that resembles familial multiple lipomatosis syndrome. To explore brown and white fat physiology in methylmalonic acidemia (MMA), body composition, adipokines, and inflammatory markers were assessed in 46 patients with MMA and 99 matched controls. Fibroblast growth factor 21 levels were associated with acyl-CoA accretion, aberrant methylmalonylation in adipose tissue, and an attenuated inflammatory cytokine profile. In parallel, brown and white fat were examined in a liver-specific transgenic MMA mouse model (Mmut-/- TgINS-Alb-Mmut). The MMA mice exhibited abnormal nonshivering thermogenesis with whitened brown fat and had an ineffective transcriptional response to cold stress. Treatment of the MMA mice with bezafibrates led to clinical improvement with beiging of subcutaneous fat depots, which resembled the distribution seen in the patients. These studies defined what we believe to be a novel lipodystrophy phenotype in patients with defects in the terminal steps of BCAA oxidation and demonstrated that beiging of subcutaneous adipose tissue in MMA could readily be induced with small molecules.
- Published
- 2024
- Full Text
- View/download PDF
8. Mission possible: Gene therapy for inherited metabolic diseases.
- Author
-
Baruteau J, Keshavan N, and Venditti CP
- Subjects
- Humans, Genetic Therapy, Metabolic Diseases genetics, Metabolic Diseases therapy
- 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. Biomarkers to predict disease progression and therapeutic response in isolated methylmalonic acidemia.
- Author
-
Manoli I, Gebremariam A, McCoy S, Pass AR, Gagné J, Hall C, Ferry S, Van Ryzin C, Sloan JL, Sacchetti E, Catesini G, Rizzo C, Martinelli D, Spada M, Dionisi-Vici C, and Venditti CP
- Subjects
- Humans, Mutation, Biomarkers, Disease Progression, Methylmalonic Acid, Methylmalonyl-CoA Mutase genetics, Methylmalonyl-CoA Mutase metabolism, Amino Acid Metabolism, Inborn Errors diagnosis, Amino Acid Metabolism, Inborn Errors therapy, Amino Acid Metabolism, Inborn Errors complications
- Abstract
Methylmalonic Acidemia (MMA) is a heterogenous group of inborn errors of metabolism caused by a defect in the methylmalonyl-CoA mutase (MMUT) enzyme or the synthesis and transport of its cofactor, 5'-deoxy-adenosylcobalamin. It is characterized by life-threatening episodes of ketoacidosis, chronic kidney disease, and other multiorgan complications. Liver transplantation can improve patient stability and survival and thus provides clinical and biochemical benchmarks for the development of hepatocyte-targeted genomic therapies. Data are presented from a US natural history protocol that evaluated subjects with different types of MMA including mut-type (N = 91), cblB-type (15), and cblA-type MMA (17), as well as from an Italian cohort of mut-type (N = 19) and cblB-type MMA (N = 2) subjects, including data before and after organ transplantation in both cohorts. Canonical metabolic markers, such as serum methylmalonic acid and propionylcarnitine, are variable and affected by dietary intake and renal function. We have therefore explored the use of the 1-
13 C-propionate oxidation breath test (POBT) to measure metabolic capacity and the changes in circulating proteins to assess mitochondrial dysfunction (fibroblast growth factor 21 [FGF21] and growth differentiation factor 15 [GDF15]) and kidney injury (lipocalin-2 [LCN2]). Biomarker concentrations are higher in patients with the severe mut0 -type and cblB-type MMA, correlate with a decreased POBT, and show a significant response postliver transplant. Additional circulating and imaging markers to assess disease burden are necessary to monitor disease progression. A combination of biomarkers reflecting disease severity and multisystem involvement will be needed to help stratify patients for clinical trials and assess the efficacy of new therapies for MMA., (© 2023 SSIEM. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)- 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. New insights into the pathophysiology of methylmalonic acidemia.
- Author
-
Head PE, Meier JL, and Venditti CP
- Subjects
- Humans, Mitochondria metabolism, Methylmalonyl-CoA Mutase metabolism, Methylmalonic Acid, Amino Acid Metabolism, Inborn Errors therapy, Propionic Acidemia
- Abstract
Methylmalonic acidemia (MMA) is a severe inborn error of metabolism that is characterized by pleiotropic metabolic perturbations and multiorgan pathology. Treatment options are limited and non-curative as the underlying causative molecular mechanisms remain unknown. While earlier studies have focused on the potential direct toxicity of metabolites such as methylmalonic and propionic acid as a mechanism to explain disease pathophysiology, new observations have revealed that aberrant acylation, specifically methylmalonylation, is a characteristic feature of MMA. The mitochondrial sirtuin enzyme SIRT5 is capable of recognizing and removing this PTM, however, reduced protein levels of SIRT5 along with other mitochondrial SIRTs 3 and 4 in MMA and potentially reduced function of all three indicates aberrant acylation may require clinical intervention. Therefore, targeting posttranslational modifications may represent a new therapeutic approach to treat MMA and related organic acidemias., (Published 2023. This article is a U.S. Government work and is in the public domain in the USA.)
- 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. Anaplerosis in action.
- Author
-
Head PE and Venditti CP
- Subjects
- Myocardium metabolism, Citric Acid Cycle
- Published
- 2023
- Full Text
- View/download PDF
15. 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
16. Treatment of metabolic disorders using genomic technologies: Lessons from methylmalonic acidemia.
- Author
-
Venturoni LE and Venditti CP
- Subjects
- Dependovirus genetics, Genomics, Humans, Methylmalonic Acid, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors therapy, Methylmalonyl-CoA Mutase
- Abstract
Hereditary methylmalonic acidemia (MMA) caused by deficiency of the enzyme methylmalonyl-CoA mutase (MMUT) is a relatively common and severe organic acidemia. The recalcitrant nature of the condition to conventional dietary and medical management has led to the use of elective liver and combined liver-kidney transplantation in some patients. However, liver transplantation is intrinsically limited by organ availability, the risks of surgery, procedural and life-long management costs, transplant comorbidities, and a remaining underlying risk of complications related to MMA despite transplantation. Here, we review pre-clinical studies that present alternative approaches to solid organ transplantation as a treatment for MMUT MMA, including adeno-associated viral gene addition therapy, mRNA therapy, and genome editing, with and without nuclease enhancement., (Published 2022. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2022
- Full Text
- View/download PDF
17. 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
18. ZBTB11 dysfunction: spectrum of brain abnormalities, biochemical signature and cellular consequences.
- Author
-
Sumathipala D, Strømme P, Fattahi Z, Lüders T, Sheng Y, Kahrizi K, Einarsen IH, Sloan JL, Najmabadi H, van den Heuvel L, Wevers RA, Guerrero-Castillo S, Mørkrid L, Valayannopoulos V, Backe PH, Venditti CP, van Karnebeek CD, Nilsen H, Frengen E, and Misceo D
- Subjects
- Brain, Humans, Amino Acid Metabolism, Inborn Errors genetics, Metabolism, Inborn Errors genetics, Nervous System Malformations
- Abstract
Bi-allelic pathogenic variants in ZBTB11 have been associated with intellectual developmental disorder, autosomal recessive 69 (MRT69; OMIM 618383). We report five patients from three families with novel, bi-allelic variants in ZBTB11. We have expanded the clinical phenotype of MRT69, documenting varied severity of atrophy affecting different brain regions and described combined malonic and methylmalonic aciduria as a biochemical manifestation. As ZBTB11 encodes for a transcriptional regulator, we performeded chromatin immunoprecipitation-sequencing targeting ZBTB11 in fibroblasts from patients and controls. Chromatin immunoprecipitation-sequencing revealed binding of wild-type ZBTB11 to promoters in 238 genes, among which genes encoding proteins involved in mitochondrial functions and RNA processing are over-represented. Mutated ZBTB11 showed reduced binding to 61 of the targeted genes, indicating that the variants act as loss of function. Most of these genes are related to mitochondrial functions. Transcriptome analysis of the patient fibroblasts revealed dysregulation of mitochondrial functions. In addition, we uncovered that reduced binding of the mutated ZBTB11 to ACSF3 leads to decreased ACSF3 transcript level, explaining combined malonic and methylmalonic aciduria. Collectively, these results expand the clinical spectrum of ZBTB11-related neurological disease and give insight into the pathophysiology in which the dysfunctional ZBTB11 affect mitochondrial functions and RNA processing contributing to the neurological and biochemical phenotypes., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2022
- Full Text
- View/download PDF
19. Aberrant methylmalonylation underlies methylmalonic acidemia and is attenuated by an engineered sirtuin.
- Author
-
Head PE, Myung S, Chen Y, Schneller JL, Wang C, Duncan N, Hoffman P, Chang D, Gebremariam A, Gucek M, Manoli I, and Venditti CP
- Subjects
- Animals, Genetic Therapy, Humans, Methylmalonyl-CoA Mutase genetics, Methylmalonyl-CoA Mutase metabolism, Mice, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors metabolism, Amino Acid Metabolism, Inborn Errors therapy, Sirtuins genetics
- Abstract
Organic acidemias such as methylmalonic acidemia (MMA) are a group of inborn errors of metabolism that typically arise from defects in the catabolism of amino and fatty acids. Accretion of acyl-CoA species is postulated to underlie disease pathophysiology, but the mechanism(s) remain unknown. Here, we surveyed hepatic explants from patients with MMA and unaffected donors, in parallel with samples from various mouse models of methylmalonyl-CoA mutase deficiency. We found a widespread posttranslational modification, methylmalonylation, that inhibited enzymes in the urea cycle and glycine cleavage pathway in MMA. Biochemical studies and mouse genetics established that sirtuin 5 (SIRT5) controlled the metabolism of MMA-related posttranslational modifications. SIRT5 was engineered to resist acylation-driven inhibition via lysine to arginine mutagenesis. The modified SIRT5 was used to create an adeno-associated viral 8 (AAV8) vector and systemically delivered to mutant and control mice. Gene therapy ameliorated hyperammonemia and reduced global methylmalonylation in the MMA mice.
- Published
- 2022
- Full Text
- View/download PDF
20. Probing the functional consequence and clinical relevance of CD320 p.E88del, a variant in the transcobalamin receptor gene.
- Author
-
Pangilinan F, Watkins D, Bernard D, Chen Y, Dong N, Wu Q, Ozel-Abaan H, Kaur M, Caggana M, Morrissey M, Browne ML, Mills JL, Van Ryzin C, Shchelochkov O, Sloan J, Venditti CP, Sarafoglou K, Rosenblatt DS, Kay DM, and Brody LC
- Subjects
- Antigens, CD, Genetic Association Studies, Humans, Infant, Infant, Newborn, Vitamin B 12 metabolism, Receptors, Cell Surface genetics, Transcobalamins genetics, Transcobalamins metabolism
- Abstract
The biological and clinical significance of the p.E88del variant in the transcobalamin receptor, CD320, is unknown. This allele is annotated in ClinVar as likely benign, pathogenic, and of uncertain significance. To determine functional consequence and clinical relevance of this allele, we employed cell culture and genetic association studies. Fibroblasts from 16 CD320 p.E88del homozygotes exhibited reduced binding and uptake of cobalamin. Complete ascertainment of newborns with transiently elevated C3 (propionylcarnitine) in New York State demonstrated that homozygosity for CD320 p.E88del was over-represented (7/348, p < 6 × 10
-5 ). Using population data, we estimate that ~85% of the p.E88del homozygotes born in the same period did not have elevated C3, suggesting that cobalamin metabolism in the majority of these infants with this genotype is unaffected. Clinical follow-up of 4/9 homozygous individuals uncovered neuropsychological findings, mostly in speech and language development. None of these nine individuals exhibited perturbation of cobalamin metabolism beyond the newborn stage even during periods of acute illness. Newborns homozygous for this allele in the absence of other factors are at low risk of requiring clinical intervention, although more studies are required to clarify the natural history of various CD320 variants across patient populations., (© 2022 Wiley Periodicals LLC. This article has been contributed to by US Government employees and their work is in the public domain in the USA.)- Published
- 2022
- Full Text
- View/download PDF
21. 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
22. Correction to: 1- 13 C-propionate breath testing as a surrogate endpoint to assess efficacy of liver-directed therapies in methylmalonic acidemia (MMA).
- Author
-
Manoli I, Pass AR, Harrington EA, Sloan JL, Gagné J, McCoy S, Bell SL, Hattenbach JD, Leitner BP, Duckworth CJ, Fletcher LA, Cassimatis TM, Galarreta CI, Thurm A, Snow J, Van Ryzin C, Ferry S, Mew NA, Shchelochkov OA, Chen KY, and Venditti CP
- Published
- 2021
- Full Text
- View/download PDF
23. 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
24. Severity modeling of propionic acidemia using clinical and laboratory biomarkers.
- Author
-
Shchelochkov OA, Manoli I, Juneau P, Sloan JL, Ferry S, Myles J, Schoenfeld M, Pass A, McCoy S, Van Ryzin C, Wenger O, Levin M, Zein W, Huryn L, Snow J, Chlebowski C, Thurm A, Kopp JB, Chen KY, and Venditti CP
- Subjects
- Biomarkers, Humans, Laboratories, Liver Transplantation, Propionic Acidemia diagnosis, Propionic Acidemia genetics
- Abstract
Purpose: To conduct a proof-of-principle study to identify subtypes of propionic acidemia (PA) and associated biomarkers., Methods: Data from a clinically diverse PA patient population ( https://clinicaltrials.gov/ct2/show/NCT02890342 ) were used to train and test machine learning models, identify PA-relevant biomarkers, and perform validation analysis using data from liver-transplanted participants. k-Means clustering was used to test for the existence of PA subtypes. Expert knowledge was used to define PA subtypes (mild and severe). Given expert classification, supervised machine learning (support vector machine with a polynomial kernel, svmPoly) performed dimensional reduction to define relevant features of each PA subtype., Results: Forty participants enrolled in the study; five underwent liver transplant. Analysis with k-means clustering indicated that several PA subtypes may exist on the biochemical continuum. The conventional PA biomarkers, plasma total 2-methylctirate and propionylcarnitine, were not statistically significantly different between nontransplanted and transplanted participants motivating us to search for other biomarkers. Unbiased dimensional reduction using svmPoly revealed that plasma transthyretin, alanine:serine ratio, GDF15, FGF21, and in vivo 1-
13 C-propionate oxidation, play roles in defining PA subtypes., Conclusion: Support vector machine prioritized biomarkers that helped classify propionic acidemia patients according to severity subtypes, with important ramifications for future clinical trials and management of PA., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2021
- Full Text
- View/download PDF
25. 1- 13 C-propionate breath testing as a surrogate endpoint to assess efficacy of liver-directed therapies in methylmalonic acidemia (MMA).
- Author
-
Manoli I, Pass AR, Harrington EA, Sloan JL, Gagné J, McCoy S, Bell SL, Hattenbach JD, Leitner BP, Duckworth CJ, Fletcher LA, Cassimatis TM, Galarreta CI, Thurm A, Snow J, Van Ryzin C, Ferry S, Mew NA, Shchelochkov OA, Chen KY, and Venditti CP
- Subjects
- Biomarkers, Breath Tests, Humans, Liver, Methylmalonic Acid, Amino Acid Metabolism, Inborn Errors diagnosis, Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors therapy, Propionates
- Abstract
Purpose: To develop a safe and noninvasive in vivo assay of hepatic propionate oxidative capacity., Methods: A modified 1-
13 C-propionate breath test was administered to 57 methylmalonic acidemia (MMA) subjects, including 19 transplant recipients, and 16 healthy volunteers. Isotopomer enrichment (13 CO2 /12 CO2 ) was measured in exhaled breath after an enteral bolus of sodium-1-13 C-propionate, and normalized for CO2 production. 1-13 C-propionate oxidation was then correlated with clinical, laboratory, and imaging parameters collected via a dedicated natural history protocol., Results: Lower propionate oxidation was observed in patients with the severe mut0 and cblB subtypes of MMA, but was near normal in those with the cblA and mut- forms of the disorder. Liver transplant recipients demonstrated complete restoration of 1-13 C-propionate oxidation to control levels. 1-13 C-propionate oxidation correlated with cognitive test result, growth indices, bone mineral density, renal function, and serum biomarkers. Test repeatability was robust in controls and in MMA subjects (mean coefficient of variation 6.9% and 12.8%, respectively), despite widely variable serum methylmalonic acid concentrations in the patients., Conclusion: Propionate oxidative capacity, as measured with 1-13 C-propionate breath testing, predicts disease severity and clinical outcomes, and could be used to assess the therapeutic effects of liver-targeted genomic therapies for MMA and related disorders of propionate metabolism., Trial Registration: This clinical study is registered in www.clinicaltrials.gov with the ID: NCT00078078. Study URL: http://clinicaltrials.gov/ct2/show/NCT00078078., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2021
- Full Text
- View/download PDF
26. 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
27. 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
28. 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
29. Considerations of expanded carrier screening: Lessons learned from combined malonic and methylmalonic aciduria.
- Author
-
Gabriel MC, Rice SM, Sloan JL, Mossayebi MH, Venditti CP, and Al-Kouatly HB
- Subjects
- Adult, Amino Acid Metabolism, Inborn Errors pathology, Amino Acid Metabolism, Inborn Errors psychology, Amniocentesis psychology, Carboxy-Lyases genetics, Female, Genetic Counseling psychology, Heterozygote, Humans, Male, Malonyl Coenzyme A genetics, Metabolism, Inborn Errors pathology, Metabolism, Inborn Errors psychology, Methylmalonic Acid, Mutation, Pregnancy, Truth Disclosure, Amino Acid Metabolism, Inborn Errors genetics, Carboxy-Lyases deficiency, Coenzyme A Ligases genetics, Genetic Carrier Screening, Metabolism, Inborn Errors genetics
- Abstract
Background: Expanded carrier screening (ECS) utilizes high-throughput next-generation sequencing to evaluate an individual's carrier status for multiple conditions. Combined malonic and methylmalonic aciduria (CMAMMA) due to ACSF3 deficiency is a rare inherited disease included in such screening panels. Some cases have been reported with metabolic symptoms in childhood yet other cases describe a benign clinical course, suggesting the clinical phenotype is not well defined., Methods/case Report: Clinical and laboratory findings during the prenatal period were obtained retrospectively from medical records., Results: A 37-year-old nulliparous woman and her partner were each identified as carriers of ACSF3 variants and presented at 9 weeks gestation for prenatal genetic consultation. The couple received extensive genetic counseling and proceeded with chorionic villus sampling at 11 weeks gestation. Subsequent analysis confirmed that the fetus inherited both parental ACSF variants. The couple was devastated by the results and after reviewing options of pregnancy continuation and termination, they decided to terminate the pregnancy. Following this decision, the patient was diagnosed with acute stress disorder., Conclusion: This case highlights how expanded carrier screening adds complexity to reproductive decision-making. Stronger guidelines and additional research are needed to direct and evaluate the timing, composition, and implementation of ECS panels., (© 2021 The Authors. Molecular Genetics & Genomic Medicine published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
30. 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
31. Safety questions for AAV gene therapy.
- Author
-
Venditti CP
- Subjects
- Animals, Dependovirus genetics, Dogs, Genetic Therapy, Liver, Longitudinal Studies, Hemophilia A
- Published
- 2021
- Full Text
- View/download PDF
32. 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
33. 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
34. The vitamin B12 processing enzyme, mmachc, is essential for zebrafish survival, growth and retinal morphology.
- Author
-
Sloan JL, Achilly NP, Arnold ML, Catlett JL, Blake T, Bishop K, Jones M, Harper U, English MA, Anderson S, Trivedi NS, Elkahloun A, Hoffmann V, Brooks BP, Sood R, and Venditti CP
- Subjects
- Animals, Homocystinuria genetics, Homocystinuria pathology, Humans, Mice, Mutation genetics, Optic Nerve growth & development, Optic Nerve pathology, Oxidoreductases genetics, Retina growth & development, Retina metabolism, Vitamin B 12 analogs & derivatives, Vitamin B 12 metabolism, Vitamin B 12 Deficiency metabolism, Vitamin B 12 Deficiency pathology, Zebrafish genetics, Zebrafish growth & development, Carrier Proteins genetics, Morphogenesis genetics, Vitamin B 12 genetics, Vitamin B 12 Deficiency genetics, Zebrafish Proteins genetics
- Abstract
Cobalamin C (cblC) deficiency, the most common inborn error of intracellular cobalamin metabolism, is caused by mutations in MMACHC, a gene responsible for the processing and intracellular trafficking of vitamin B12. This recessive disorder is characterized by a failure to metabolize cobalamin into adenosyl- and methylcobalamin, which results in the biochemical perturbations of methylmalonic acidemia, hyperhomocysteinemia and hypomethioninemia caused by the impaired activity of the downstream enzymes, methylmalonyl-CoA mutase and methionine synthase. Cobalamin C deficiency can be accompanied by a wide spectrum of clinical manifestations, including progressive blindness, and, in mice, manifests with very early embryonic lethality. Because zebrafish harbor a full complement of cobalamin metabolic enzymes, we used genome editing to study the loss of mmachc function and to develop the first viable animal model of cblC deficiency. mmachc mutants survived the embryonic period but perished in early juvenile life. The mutants displayed the metabolic and clinical features of cblC deficiency including methylmalonic acidemia, severe growth retardation and lethality. Morphologic and metabolic parameters improved when the mutants were raised in water supplemented with small molecules used to treat patients, including hydroxocobalamin, methylcobalamin, methionine and betaine. Furthermore, mmachc mutants bred to express rod and/or cone fluorescent reporters, manifested a retinopathy and thin optic nerves (ON). Expression analysis using whole eye mRNA revealed the dysregulation of genes involved in phototransduction and cholesterol metabolism. Zebrafish with mmachc deficiency recapitulate the several of the phenotypic and biochemical features of the human disorder, including ocular pathology, and show a response to established treatments., (Published by Oxford University Press 2020.)
- Published
- 2020
- Full Text
- View/download PDF
35. High-dose hydroxocobalamin achieves biochemical correction and improvement of neuropsychiatric deficits in adults with late onset cobalamin C deficiency.
- Author
-
Higashimoto T, Kim AY, Ogawa JT, Sloan JL, Almuqbil MA, Carlson JM, Manoli I, Venditti CP, Gunay-Aygun M, and Wang T
- Abstract
Cobalamin C ( cblC ) deficiency is the most common inborn error of intracellular cobalamin metabolism caused by pathogenic variant(s) in MMACHC and manifests with methylmalonic acidemia, hyperhomocysteinemia, and hypomethioninemia with a variable age of presentation. Individuals with late-onset cblC may be asymptomatic until manifesting neuropsychiatric symptoms, thromboembolic events, and renal disease. Although hydroxocobalamin provides a foundation for therapy, optimal dose regimen for adult patients has not been systematically evaluated. We report three adult siblings with late-onset cblC disease, and their biochemical and clinical responses to high-dose hydroxocobalamin. The 28-year-old proband presented with severe psychosis, progressive neurological deterioration, and deep venous thrombosis complicated by a pulmonary embolism. MRI studies identified lesions in the spinal cord, periventricular white matter, and basal ganglia. Serum homocysteine and methylmalonic acid levels were markedly elevated. Hydroxocobalamin at standard dose (1 mg/day) initially resulted in partial metabolic correction. A regimen of high-dose hydroxocobalamin (25 mg/day) together with betaine and folic acid resulted in rapid and sustainable biochemical correction, resolution of psychosis, improvement of neurological functions, and amelioration of brain and spinal cord lesions. Two siblings who did not manifest neuropsychiatric symptoms or thromboembolism achieved a satisfactory metabolic control with the same high-dose regimen. Hydroxocobalamin injection was then spaced out to 25 mg weekly with good and sustainable metabolic control. All three patients are compound heterozygotes for c.271dupA p.Arg91LysfsX14 and c.389A > G p.Tyr130Cys. This study highlights the importance of evaluating intracellular cobalamin metabolism in adults with neuropsychiatric manifestations and/or thromboembolic events, and demonstrates that high-dose hydroxocobalamin achieves rapid and sustainable metabolic control and improvement in neuropsychiatric outcomes in adults with late-onset cblC disease., Competing Interests: The authors declare that they have no conflict of interest., (© 2019 The Authors. Journal of Inherited Metabolic Disease published by John Wiley & Sons Ltd on behalf of SSIEM.)
- Published
- 2019
- Full Text
- View/download PDF
36. Chronic kidney disease in propionic acidemia.
- Author
-
Shchelochkov OA, Manoli I, Sloan JL, Ferry S, Pass A, Van Ryzin C, Myles J, Schoenfeld M, McGuire P, Rosing DR, Levin MD, Kopp JB, and Venditti CP
- Subjects
- Adolescent, Adult, Biomarkers, Child, Child, Preschool, Creatinine blood, Cross-Sectional Studies, Cystatin C analysis, Cystatin C blood, Female, Glomerular Filtration Rate, Humans, Kidney, Lipocalin-2 analysis, Lipocalin-2 blood, Male, Middle Aged, Prevalence, Propionic Acidemia epidemiology, Uric Acid analysis, Uric Acid blood, Propionic Acidemia complications, Renal Insufficiency, Chronic epidemiology
- Abstract
Purpose: Propionic acidemia (PA) is a severe metabolic disorder characterized by multiorgan pathology, including renal disease. The prevalence of chronic kidney disease (CKD) in PA patients and factors associated with CKD in PA are not known., Methods: Thirty-one subjects diagnosed with PA underwent laboratory and clinical evaluations through a dedicated natural history study at the National Institutes of Health (ClinicalTrials.gov identifier: NCT02890342)., Results: Cross-sectional analysis of the creatinine-based estimated glomerular filtration rate (eGFR) in subjects with native kidneys revealed an age-dependent decline in renal function (P < 0.002). Among adults with PA, 4/8 (50%) had eGFR <60 mL/min/1.73 m
2 . There was a significant discrepancy between eGFRs calculated using estimating equations based on serum creatinine compared with serum cystatin C (P < 0.0001). The tubular injury marker, plasma lipocalin-2, and plasma uric acid were strongly associated with CKD (P < 0.0001). The measured 24-hour creatinine excretion was below normal, even after adjusting for age, height, and sex., Conclusion: CKD is common in adults with PA and is associated with age. The poor predictive performance of standard eGFR estimating equations, likely due to reduced creatine synthesis in kidney and liver, could delay the recognition of CKD and management of ensuing complications in this population.- Published
- 2019
- Full Text
- View/download PDF
37. 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
38. 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
39. Macrophage derived TNFα promotes hepatic reprogramming to Warburg-like metabolism.
- Author
-
Tarasenko TN, Jestin M, Matsumoto S, Saito K, Hwang S, Gavrilova O, Trivedi N, Zerfas PM, Barca E, DiMauro S, Senac J, Venditti CP, Cherukuri M, and McGuire PJ
- Subjects
- Animals, Biological Products pharmacology, Cell Line, Tumor, Hepatocytes drug effects, Homeostasis drug effects, Homeostasis physiology, Humans, Immunity, Innate drug effects, Inflammation drug therapy, Inflammation metabolism, Liver drug effects, Macrophages drug effects, Mice, Mice, Inbred C57BL, Hepatocytes metabolism, Liver metabolism, Macrophages metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
During infection, hepatocytes must undergo a reprioritization of metabolism, termed metabolic reprogramming. Hepatic metabolic reprogramming in response to infection begins within hours of infection, suggesting a mechanism closely linked to pathogen recognition. Following injection with polyinosinic:polycytidylic acid, a mimic of viral infection, a robust hepatic innate immune response could be seen involving the TNFα pathway at 2 h. Repeated doses led to the adoption of Warburg-like metabolism in the liver as determined by in vivo metabolic imaging, expression analyses, and metabolomics. Hepatic macrophages, Kupffer cells, were able to induce Warburg-like metabolism in hepatocytes in vitro via TNFα. Eliminating macrophages in vivo or blocking TNFα in vitro or in vivo resulted in abrogation of the metabolic phenotype, establishing an immune-metabolic axis in hepatic metabolic reprogramming. Overall, we suggest that macrophages, as early sensors of pathogens, instruct hepatocytes via TNFα to undergo metabolic reprogramming to cope with challenges to homeostasis initiated by infection. This work not only addresses a key component of end-organ physiology, but also raises questions about the side effects of biologics in the treatment of inflammatory diseases. KEY MESSAGES: • Hepatocytes develop Warburg-like metabolism in vivo during viral infection. • Macrophage TNFα promotes expression of glycolytic enzymes in hepatocytes. • Blocking this immune-metabolic axis abrogates Warburg-like metabolism in the liver. • Implications for patients being treated for inflammatory diseases with biologics.
- Published
- 2019
- Full Text
- View/download PDF
40. Noninvasive monitoring of chronic kidney disease using pH and perfusion imaging.
- Author
-
Pavuluri K, Manoli I, Pass A, Li Y, Vernon HJ, Venditti CP, and McMahon MT
- Subjects
- Amino Acid Metabolism, Inborn Errors diagnosis, Animals, Disease Models, Animal, Glomerular Filtration Rate physiology, Kidney pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Perfusion Imaging methods, Renal Insufficiency, Chronic diagnosis, Kidney diagnostic imaging, Magnetic Resonance Imaging methods, Renal Insufficiency, Chronic diagnostic imaging, Renal Insufficiency, Chronic pathology
- Abstract
Chronic Kidney Disease (CKD) is a cardinal feature of methylmalonic acidemia (MMA), a prototypic organic acidemia. Impaired growth, low activity, and protein restriction affect muscle mass and lower serum creatinine, which can delay diagnosis and management of renal disease. We have designed an alternative strategy for monitoring renal function based on administration of a pH sensitive MRI agent and assessed this in a mouse model. This protocol produced three metrics: kidney contrast, ~4% for severe renal disease mice compared to ~13% and ~25% for moderate renal disease and healthy controls, filtration fraction (FF), ~15% for severe renal disease mice compared to ~79% and 100% for moderate renal disease and healthy controls, and variation in pH, ~0.45 units for severe disease mice compared to 0.06 and 0.01 for moderate disease and healthy controls. Our results demonstrate that MRI can be used for early detection and monitoring of CKD.
- Published
- 2019
- Full Text
- View/download PDF
41. 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
42. 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
43. Effects of medical food leucine content in the management of methylmalonic and propionic acidemias.
- Author
-
Myles JG, Manoli I, and Venditti CP
- Subjects
- Amino Acid Metabolism, Inborn Errors blood, Animals, Deficiency Diseases blood, Deficiency Diseases etiology, Deficiency Diseases prevention & control, Homocystinuria blood, Homocystinuria diet therapy, Humans, Isoleucine blood, Isoleucine deficiency, Leucine adverse effects, Propionic Acidemia blood, Valine blood, Valine deficiency, Vitamin B 12 Deficiency blood, Vitamin B 12 Deficiency congenital, Vitamin B 12 Deficiency diet therapy, Amino Acid Metabolism, Inborn Errors diet therapy, Diet, Protein-Restricted adverse effects, Foods, Specialized adverse effects, Leucine therapeutic use, Propionic Acidemia diet therapy
- Abstract
Purpose of Review: The current review highlights the varied effects of medical foods high in leucine (Leu) and devoid of valine (Val) and isoleucine (Ile) in the management of methylmalonic acidemia (MMA) and propionic acidemia and cobalamin C (cblC) deficiency, aiming to advance dietary practices., Recent Findings: Leu is a key metabolic regulator with a multitude of effects on different organ systems. Recent observational studies have demonstrated that these effects can have unintended consequences in patients with MMA as a result of liberal use of medical foods. The combination of protein restriction and medical food use in MMA and propionic acidemia results in an imbalanced branched-chain amino acid (BCAA) dietary content with a high Leu-to-Val and/or Ile ratio. This leads to decreased plasma levels of Val and Ile and predicts impaired brain uptake of multiple essential amino acids. Decreased transport of methionine (Met) across the blood-brain barrier due to high circulating Leu levels is of particular concern in cblC deficiency in which endogenous Met synthesis is impaired., Summary: Investigations into the optimal composition of medical foods for MMA and propionic acidemia, and potential scenarios in which Leu supplementation may be beneficial are needed. Until then, MMA/propionic acidemia medical foods should be used judiciously in the dietary management of these patients and avoided altogether in cblC deficiency.
- Published
- 2018
- Full Text
- View/download PDF
44. 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
45. 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
46. Genome editing for inborn errors of metabolism: advancing towards the clinic.
- Author
-
Schneller JL, Lee CM, Bao G, and Venditti CP
- Subjects
- Humans, Gene Editing methods, Genetic Therapy methods, Metabolism, Inborn Errors genetics
- Abstract
Inborn errors of metabolism (IEM) include many disorders for which current treatments aim to ameliorate disease manifestations, but are not curative. Advances in the field of genome editing have recently resulted in the in vivo correction of murine models of IEM. Site-specific endonucleases, such as zinc-finger nucleases and the CRISPR/Cas9 system, in combination with delivery vectors engineered to target disease tissue, have enabled correction of mutations in disease models of hemophilia B, hereditary tyrosinemia type I, ornithine transcarbamylase deficiency, and lysosomal storage disorders. These in vivo gene correction studies, as well as an overview of genome editing and future directions for the field, are reviewed and discussed herein.
- Published
- 2017
- Full Text
- View/download PDF
47. 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
48. Methylmalonic and propionic acidemias: clinical management update.
- Author
-
Fraser JL and Venditti CP
- Subjects
- Brain Injury, Chronic etiology, Brain Injury, Chronic prevention & control, Food, Formulated, Humans, Liver Transplantation, Amino Acid Metabolism, Inborn Errors complications, Amino Acid Metabolism, Inborn Errors diagnosis, Amino Acid Metabolism, Inborn Errors diet therapy, Amino Acid Metabolism, Inborn Errors surgery, Propionic Acidemia complications, Propionic Acidemia diagnosis, Propionic Acidemia diet therapy, Propionic Acidemia surgery
- Abstract
Purpose of Review: Recent clinical studies and management guidelines for the treatment of the organic acidopathies methylmalonic acidemia (MMA) and propionic acidemia address the scope of interventions to maximize health and quality of life. Unfortunately, these disorders continue to cause significant morbidity and mortality due to acute and chronic systemic and end-organ injury., Recent Findings: Dietary management with medical foods has been a mainstay of therapy for decades, yet well controlled patients can manifest growth, development, cardiac, ophthalmological, renal, and neurological complications. Patients with organic acidopathies suffer metabolic brain injury that targets specific regions of the basal ganglia in a distinctive pattern, and these injuries may occur even with optimal management during metabolic stress. Liver transplantation has improved quality of life and metabolic stability, yet transplantation in this population does not entirely prevent brain injury or the development of optic neuropathy and cardiac disease., Summary: Management guidelines should identify necessary screening for patients with methylmalonic acidemia and propionic acidemia, and improve anticipatory management of progressive end-organ disease. Liver transplantation improves overall metabolic control, but injury to nonregenerative tissues may not be mitigated. Continued use of medical foods in these patients requires prospective studies to demonstrate evidence of benefit in a controlled manner.
- Published
- 2016
- Full Text
- View/download PDF
49. Disorders of branched chain amino acid metabolism.
- Author
-
Manoli I and Venditti CP
- Abstract
The three essential branched-chain amino acids (BCAAs), leucine, isoleucine and valine, share the first enzymatic steps in their metabolic pathways, including a reversible transamination followed by an irreversible oxidative decarboxylation to coenzyme-A derivatives. The respective oxidative pathways subsequently diverge and at the final steps yield acetyl- and/or propionyl-CoA that enter the Krebs cycle. Many disorders in these pathways are diagnosed through expanded newborn screening by tandem mass spectrometry. Maple syrup urine disease (MSUD) is the only disorder of the group that is associated with elevated body fluid levels of the BCAAs. Due to the irreversible oxidative decarboxylation step distal enzymatic blocks in the pathways do not result in the accumulation of amino acids, but rather to CoA-activated small carboxylic acids identified by gas chromatography mass spectrometry analysis of urine and are therefore classified as organic acidurias. Disorders in these pathways can present with a neonatal onset severe-, or chronic intermittent- or progressive forms. Metabolic instability and increased morbidity and mortality are shared between inborn errors in the BCAA pathways, while treatment options remain limited, comprised mainly of dietary management and in some cases solid organ transplantation.
- Published
- 2016
- Full Text
- View/download PDF
50. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.