Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alison McVie-Wylie is active.

Publication


Featured researches published by Alison McVie-Wylie.


Biochemical Journal | 2005

Carbohydrate-remodelled acid α-glucosidase with higher affinity for the cation-independent mannose 6-phosphate receptor demonstrates improved delivery to muscles of Pompe mice

Yunxiang Zhu; Xuemei Li; Alison McVie-Wylie; Canwen Jiang; Beth L. Thurberg; Nina Raben; Robert J. Mattaliano; Seng H. Cheng

To enhance the delivery of rhGAA (recombinant GAA, where GAA stands for acid alpha-glucosidase) to the affected muscles in Pompe disease, the carbohydrate moieties on the enzyme were remodelled to exhibit a high affinity ligand for the CI-MPR (cation-independent M6P receptor, where M6P stands for mannose 6-phosphate). This was achieved by chemically conjugating on to rhGAA, a synthetic oligosaccharide ligand bearing M6P residues in the optimal configuration for binding the receptor. The carbonyl chemistry used resulted in the conjugation of approx. six synthetic ligands on to each enzyme. The resulting modified enzyme [neo-rhGAA (modified recombinant human GAA harbouring synthetic oligosaccharide ligands)] displayed near-normal specific activity and significantly increased affinity for the CI-MPR. However, binding to the mannose receptor was unaffected despite the introduction of additional mannose residues in neo-rhGAA. Uptake studies using L6 myoblasts showed neo-rhGAA was internalized approx. 20-fold more efficiently than the unmodified enzyme. Administration of neo-rhGAA into Pompe mice also resulted in greater clearance of glycogen from all the affected muscles when compared with the unmodified rhGAA. Comparable reductions in tissue glycogen levels in the Pompe mice were realized using an approx. 8-fold lower dose of neo-rhGAA in the heart and diaphragm and an approx. 4-fold lower dose in the skeletal muscles. Treatment of older Pompe mice, which are more refractory to enzyme therapy, with 40 mg/kg neo-rhGAA resulted in near-complete clearance of glycogen from all the affected muscles as opposed to only partial correction with the unmodified rhGAA. These results demonstrate that remodelling the carbohydrate of rhGAA to improve its affinity for the CI-MPR represents a feasible approach to enhance the efficacy of enzyme replacement therapy for Pompe disease.


Molecular Genetics and Metabolism | 2008

BIOCHEMICAL AND PHARMACOLOGICAL CHARACTERIZATION OF DIFFERENT RECOMBINANT ACID α-GLUCOSIDASE PREPARATIONS EVALUATED FOR THE TREATMENT OF POMPE DISEASE

Alison McVie-Wylie; K.L. Lee; H. Qiu; X. Jin; H. Do; R. Gotschall; Beth L. Thurberg; C. Rogers; Nina Raben; Michael O’Callaghan; W. Canfield; Laura Andrews; John M. McPherson; Robert J. Mattaliano

Pompe disease results in the accumulation of lysosomal glycogen in multiple tissues due to a deficiency of acid alpha-glucosidase (GAA). Enzyme replacement therapy for Pompe disease was recently approved in Europe, the U.S., Canada, and Japan using a recombinant human GAA (Myozyme, alglucosidase alfa) produced in CHO cells (CHO-GAA). During the development of alglucosidase alfa, we examined the in vitro and in vivo properties of CHO cell-derived rhGAA, an rhGAA purified from the milk of transgenic rabbits, as well as an experimental version of rhGAA containing additional mannose-6-phosphate intended to facilitate muscle targeting. Biochemical analyses identified differences in rhGAA N-termini, glycosylation types and binding properties to several carbohydrate receptors. In a mouse model of Pompe disease, glycogen was more efficiently removed from the heart than from skeletal muscle for all enzymes, and overall, the CHO cell-derived rhGAA reduced glycogen to a greater extent than that observed with the other enzymes. The results of these preclinical studies, combined with biochemical characterization data for the three molecules described within, led to the selection of the CHO-GAA for clinical development and registration as the first approved therapy for Pompe disease.


American Journal of Pathology | 2003

Tissue-Specific Inactivation of Murine M6P/IGF2R

Andrew A. Wylie; David J. Pulford; Alison McVie-Wylie; Robert A. Waterland; Heather K. Evans; Yuan-Tsong Chen; Catherine M. Nolan; Terry C. Orton; Randy L. Jirtle

The mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R) encodes a multifunctional protein involved in lysosomal enzyme trafficking, fetal organogenesis, tumor suppression, and T cell- mediated immunity. M6P/IGF2R is an imprinted gene in mice with expression only from the maternal allele. Complete knockout of this gene causes neonatal lethality, thus preventing analysis of its multifunctional role postnatally. To help elucidate the biological functions of M6P/IGF2R in adulthood, we generated both complete and tissue-specific M6P/IGF2R knockout mice using the Cre/loxP system. We confirm that complete M6P/IGF2R knockout results in fetal overgrowth and neonatal lethality. In contrast, tissue-specific inactivation of this gene in either the liver or skeletal and cardiac muscle gives rise to viable animals with no obvious phenotype. The successful creation of viable tissue-specific M6P/IGF2R knockout mouse models will now allow for detailed analysis of receptor function in a number of cellular processes including brain development, carcinogenesis, lysosomal trafficking, and T cell-mediated immunity.


American Journal of Pathology | 2011

Inhibiting TGF-β Activity Improves Respiratory Function in mdx Mice

Carol A. Nelson; R. Bridge Hunter; Lindsay A. Quigley; Stefan Girgenrath; William Weber; Jennifer A. McCullough; Carol J. Dinardo; Lorena Ceci; Nicholas P. Clayton; Alison McVie-Wylie; Seng H. Cheng; John P. Leonard; Bruce M. Wentworth

Respiratory function is the main cause of mortality in patients with Duchenne muscular dystrophy (DMD). Elevated levels of TGF-β play a key role in the pathophysiology of DMD. To determine whether therapeutic attenuation of TGF-β signaling improves respiratory function, mdx mice were treated from 2 weeks of age to 2 months or 9 months of age with either 1D11 (a neutralizing antibody to all three isoforms of TGF-β), losartan (an angiotensin receptor antagonist), or a combination of the two agents. Respiratory function was measured in nonanesthetized mice by plethysmography. The 9-month-old mdx mice had elevated Penh values and decreased breathing frequency, due primarily to decreased inspiratory flow rate. All treatments normalized Penh values and increased peak inspiratory flow, leading to decreased inspiration times and breathing frequency. Additionally, forelimb grip strength was improved after 1D11 treatment at both 2 and 9 months of age, whereas, losartan improved grip strength only at 2 months. Decreased serum creatine kinase levels (significant improvement for all groups), increased diaphragm muscle fiber density, and decreased hydroxyproline levels (significant improvement for 1D11 only) also suggested improved muscle function after treatment. For all endpoints, 1D11 was equivalent or superior to losartan; coadministration of the two agents was not superior to 1D11 alone. In conclusion, TGF-β antagonism may be a useful therapeutic approach for treating DMD patients.


Molecular Genetics and Metabolism | 2011

Enhanced Efficacy of Enzyme Replacement Therapy in Pompe Disease Through Mannose-6-Phosphate Receptor Expression in Skeletal Muscle

Dwight D. Koeberl; Xiaoyan Luo; Baodong Sun; Alison McVie-Wylie; Jian Dai; Songtao Li; Suhrad G. Banugaria; Yuan-Tsong Chen; Deeksha Bali

Enzyme replacement therapy (ERT) with acid α-glucosidase has become available for Pompe disease; however, the response of skeletal muscle, as opposed to the heart, has been attenuated. The poor response of skeletal muscle has been attributed to the low abundance of the cation-independent mannose-6-phosphate receptor (CI-MPR) in skeletal muscle compared to heart. To further understand the role of CI-MPR in Pompe disease, muscle-specific CI-MPR conditional knockout (KO) mice were crossed with GAA-KO (Pompe disease) mice. We evaluated the impact of CI-MPR-mediated uptake of GAA by evaluating ERT in CI-MPR-KO/GAA-KO (double KO) mice. The essential role of CI-MPR was emphasized by the lack of efficacy of ERT as demonstrated by markedly reduced biochemical correction of GAA deficiency and of glycogen accumulations in double KO mice, in comparison with the administration of the same therapeutic doses in GAA-KO mice. Clenbuterol, a selective β(2)-agonist, enhanced the CI-MPR expression in skeletal tissue and also increased efficacy from GAA therapy, thereby confirming the key role of CI-MPR with regard to enzyme replacement therapy in Pompe disease. Biochemical correction improved in both muscle and non-muscle tissues, indicating that therapy could be similarly enhanced in other lysosomal storage disorders. In summary, enhanced CI-MPR expression might improve the efficacy of enzyme replacement therapy in Pompe disease through enhancing receptor-mediated uptake of GAA.


Journal of Gene Medicine | 2003

Multiple muscles in the AMD quail can be “cross-corrected” of pathologic glycogen accumulation after intravenous injection of an [E1-, polymerase-] adenovirus vector encoding human acid-α-glucosidase

Alison McVie-Wylie; Enyu Ding; T. Lawson; Delila Serra; Felicia Migone; D. Pressley; Makoto Mizutani; Tateki Kikuchi; Yuan-Tsong Chen; Andrea Amalfitano

Previously, in murine models of acid maltase deficiency (AMD), we demonstrated that intravenous administration of an improved adenovirus (Ad) vector encoding human acid alpha glucosidase (hGAA) resulted in liver transduction, followed by high‐level hepatocyte‐mediated secretion of hGAA into the plasma space. The hGAA secreted by the liver was taken up and targeted to muscle cell lysosomes. The levels of hGAA achieved by this approach resulted in clearance of lysosomal glycogen accumulations; in some muscle tissues the effect was prolonged (>6 months). We next wished to demonstrate whether this approach could be generalized across divergent species. To accomplish this goal, we determined whether a similar approach would also result in efficacy, but in a quail model of AMD.


Molecular Genetics and Metabolism | 2012

Transcriptional response to GAA deficiency (Pompe disease) in infantile-onset patients

Adam Palermo; Rachel Palmer; K.S. So; Sueli Mieko Oba-Shinjo; Mindy Zhang; Brenda Richards; S.T. Madhiwalla; Patrick Finn; A. Hasegawa; K.M. Ciociola; Mario Pescatori; Alison McVie-Wylie; Robert J. Mattaliano; Stephen L. Madden; Siem de Marie; Katherine W. Klinger; Robert Pomponio

Pompe disease is a genetic disorder resulting from a deficiency of lysosomal acid alpha-glucosidase (GAA) that manifests as a clinical spectrum with regard to symptom severity and rate of progression. In this study, we used microarrays to examine gene expression from the muscle of two cohorts of infantile-onset Pompe patients to identify transcriptional differences that may contribute to the disease phenotype. We found strong similarities among the gene expression profiles generated from biceps and quadriceps, and identified a number of signaling pathways altered in both cohorts. We also found that infantile-onset Pompe patient muscle had a gene expression pattern characteristic of immature or regenerating muscle, and exhibited many transcriptional markers of inflammation, despite having few overt signs of inflammatory infiltrate. Further, we identified genes exhibiting correlation between expression at baseline and response to therapy. This combined dataset can serve as a foundation for biological discovery and biomarker development to improve the treatment of Pompe disease.


Genetics in Medicine | 2018

An immune tolerance approach using transient low-dose methotrexate in the ERT-naïve setting of patients treated with a therapeutic protein: experience in infantile-onset Pompe disease.

Zoheb B. Kazi; Ankit K. Desai; R. Bradley Troxler; David Kronn; Seymour Packman; Marta Sabbadini; William B. Rizzo; Katalin Scherer; Omar Abdul-Rahman; Pranoot Tanpaiboon; Sheela Nampoothiri; Neerja Gupta; Annette Feigenbaum; Dmitriy Niyazov; Langston Sherry; Reeval Segel; Alison McVie-Wylie; Crystal Sung; Alexandra M. Joseph; Susan Richards; Priya S. Kishnani

PurposeTo investigate immune tolerance induction with transient low-dose methotrexate (TLD-MTX) initiated with recombinant human acid α-glucosidase (rhGAA), in treatment-naïve cross-reactive immunologic material (CRIM)-positive infantile-onset Pompe disease (IOPD) patients.MethodsNewly diagnosed IOPD patients received subcutaneous or oral 0.4 mg/kg TLD-MTX for 3 cycles (3 doses/cycle) with the first 3 rhGAA infusions. Anti-rhGAA IgG titers, classified as high-sustained (HSAT; ≥51,200, ≥2 times after 6 months), sustained intermediate (SIT; ≥12,800 and <51,200 within 12 months), or low (LT; ≤6400 within 12 months), were compared with those of 37 CRIM-positive IOPD historic comparators receiving rhGAA alone.ResultsFourteen IOPD TLD-MTX recipients at the median age of 3.8 months (range, 0.7–13.5 months) had a median last titer of 150 (range, 0–51,200) at median rhGAA duration ~83 weeks (range, 36–122 weeks). One IOPD patient (7.1%) developed titers in the SIT range and one patient (7.1%) developed titers in the HSAT range. Twelve of the 14 patients (85.7%) that received TLD-MTX remained LT, versus 5/37 HSAT (peak 51,200–409,600), 7/37 SIT (12,800–51,000), and 23/37 LT (200–12,800) among comparators.ConclusionResults of TLD-MTX coinitiated with rhGAA are encouraging and merit a larger longitudinal study.


Genetics in Medicine | 2001

Recombinant human acid alpha-glucosidase enzyme therapy for infantile glycogen storage disease type II: results of a phase I/II clinical trial.

Andrea Amalfitano; A. Resai Bengur; Richard P. Morse; Joseph M. Majure; Laura E. Case; Deborah L. Veerling; Joanne Mackey; Priya S. Kishnani; Wendy A. Smith; Alison McVie-Wylie; Jennifer A. Sullivan; George Hoganson; John A. Phillips; G. Bradley Schaefer; Joel Charrow; Russell E. Ware; Edward H. Bossen; Yuan-Tsong Chen


Genomics | 2001

Molecular cloning of a novel member of the GLUT family of transporters, SLC2A10 (GLUT10), localized on chromosome 20q13.1 : A candidate gene for NIDDM susceptibility

Alison McVie-Wylie; David R. Lamson; Yuan-Tsong Chen

Collaboration


Dive into the Alison McVie-Wylie's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Kronn

New York Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nina Raben

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge