Network


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

Hotspot


Dive into the research topics where Elizabeth M. Snella is active.

Publication


Featured researches published by Elizabeth M. Snella.


Molecular Therapy | 2011

Safe, Efficient, and Reproducible Gene Therapy of the Brain in the Dog Models of Sanfilippo and Hurler Syndromes

N. Matthew Ellinwood; Jérôme Ausseil; Nathalie Desmaris; Stéphanie Bigou; Song Liu; Jackie K. Jens; Elizabeth M. Snella; Eman Mohammed; Christopher B Thomson; Sylvie Raoul; Béatrice Joussemet; Françoise Roux; Yan Cherel; Yaouen Lajat; Monique Piraud; Rachid Benchaouir; Stephan Hermening; Harald Petry; Roseline Froissart; Marc Tardieu; Carine Ciron; Philippe Moullier; Jennifer Parkes; Karen L. Kline; Irène Maire; Marie-Thérèse Vanier; Jean-Michel Heard; Marie-Anne Colle

Recent trials in patients with neurodegenerative diseases documented the safety of gene therapy based on adeno-associated virus (AAV) vectors deposited into the brain. Inborn errors of the metabolism are the most frequent causes of neurodegeneration in pre-adulthood. In Sanfilippo syndrome, a lysosomal storage disease in which heparan sulfate oligosaccharides accumulate, the onset of clinical manifestation is before 5 years. Studies in the mouse model showed that gene therapy providing the missing enzyme α-N-acetyl-glucosaminidase to brain cells prevents neurodegeneration and improves behavior. We now document safety and efficacy in affected dogs. Animals received eight deposits of a serotype 5 AAV vector, including vector prepared in insect Sf9 cells. As shown previously in dogs with the closely related Hurler syndrome, immunosuppression was necessary to prevent neuroinflammation and elimination of transduced cells. In immunosuppressed dogs, vector was efficiently delivered throughout the brain, induced α-N-acetyl-glucosaminidase production, cleared stored compounds and storage lesions. The suitability of the procedure for clinical application was further assessed in Hurler dogs, providing information on reproducibility, tolerance, appropriate vector type and dosage, and optimal age for treatment in a total number of 25 treated dogs. Results strongly support projects of human trials aimed at assessing this treatment in Sanfilippo syndrome.


Science Translational Medicine | 2010

Replacing the Enzyme α-l-Iduronidase at Birth Ameliorates Symptoms in the Brain and Periphery of Dogs with Mucopolysaccharidosis Type I

Ashley Dierenfeld; Michael F. McEntee; Carole Vogler; Charles H. Vite; Agnes H. Chen; Merry Passage; Steven Q. Le; S. Shah; Jacqueline K. Jens; Elizabeth M. Snella; K.L. Kline; J.D. Parkes; Wendy A. Ware; L.E. Moran; A. J. Fales-Williams; J.A. Wengert; R.D. Whitley; D.M. Betts; A.M. Boal; E.A. Riedesel; William Gross; N.M. Ellinwood; Patricia Dickson

Replacing the enzyme α-l-iduronidase at birth ameliorates symptoms in the brain and periphery of dogs with mucopolysaccharidosis type I. When Dogs Really Are Man’s Best Friend For certain diseases, dogs provide an excellent large-animal model and the lysosomal storage disorder mucopolysaccharidosis type I is no exception. In this disease, a defect in the enzyme α-l-iduronidase prevents breakdown of glycosaminoglycans, resulting in their accumulation in the lysosomes of cells, leading to engorged and dysfunctional cells. A variety of serious complications ensue such as enlarged organs, skeletal defects, corneal clouding, abnormal heart valves, and cognitive deficits. Although a human recombinant form of the enzyme has proved successful in treating patients with less severe forms of the disease, only some of the symptoms are ameliorated and patients often develop antibodies to the enzyme. Dierenfeld and colleagues reasoned that starting enzyme replacement therapy at birth could halt or even reverse the more serious disease symptoms such as heart valve defects, skeletal deformities, and cognitive impairment and might prevent antibodies from forming against human recombinant α-l-iduronidase. Working in a naturally occurring dog model of mucopolysaccharidosis type I in which the animals have a defective α-l-iduronidase enzyme and show most of the symptoms of the human disease, Dierenfeld et al. administered two different doses of α-l-iduronidase (0.58 and 1.57 mg/kg) intravenously every week starting a few days after birth. The authors show that starting enzyme replacement therapy at such a young age prevented the formation of antibodies against the enzyme due to neonatal immune tolerance. Treated dogs showed marked reductions in glycosaminoglycans in a variety of different tissues including the mitral heart valve, which has been refractory to traditional enzyme treatment administered at later ages. Furthermore, treated dogs showed relatively normal skeletons and lacked the stiff gait, lax joints, upturned nose, and poor neck flexibility exhibited by untreated animals with the disease. Compellingly, glycosaminoglycan levels and cortical atrophy in the brain decreased and corneal clouding improved slightly (at the higher dose) in the treated dogs. These results show that starting intravenous administration of α-l-iduronidase at higher doses and as soon as possible after birth increases the efficacy of enzyme replacement therapy and ensures immune tolerance to the enzyme. These promising results in man’s best friend suggest that children with severe mucopolysaccharidosis type I or other early-onset lysosomal storage diseases should start enzyme replacement therapy as soon as possible after birth. Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disease caused by loss of activity of α-l-iduronidase and attendant accumulation of the glycosaminoglycans dermatan sulfate and heparan sulfate. Current treatments are suboptimal and do not address residual disease including corneal clouding, skeletal deformities, valvular heart disease, and cognitive impairment. We treated neonatal dogs with MPS I with intravenous recombinant α-l-iduronidase replacement therapy at the conventional 0.58 mg/kg or a higher 1.57 mg/kg weekly dose for 56 to 81 weeks. In contrast to previous results in animals and patients treated at a later age, the dogs failed to mount an antibody response to enzyme therapy, consistent with the induction of immune tolerance in neonates. The higher dose of enzyme led to complete normalization of lysosomal storage in the liver, spleen, lung, kidney, synovium, and myocardium, as well as in the hard-to-treat mitral valve. Cardiac biochemistry and function were restored, and there were improvements in skeletal disease as shown by clinical and radiographic assessments. Glycosaminoglycan levels in the brain were normalized after intravenous enzyme therapy, in the presence or absence of intrathecal administration of recombinant α-l-iduronidase. Histopathological evidence of glycosaminoglycan storage in the brain was ameliorated with the higher-dose intravenous therapy and was further improved by combining intravenous and intrathecal therapy. These findings argue that neonatal testing and early treatment of patients with MPS I may more effectively treat this disease.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Active site mutant transgene confers tolerance to human β-glucuronidase without affecting the phenotype of MPS VII mice

William S. Sly; Carole Vogler; Jeffrey H. Grubb; Mi Zhou; Jinxing Jiang; Xiao Yan Zhou; Shunji Tomatsu; Yanhua Bi; Elizabeth M. Snella

Mucopolysaccharidosis type VII (MPS VII; Sly syndrome) is an autosomal recessive lysosomal storage disorder due to an inherited deficiency of β-glucuronidase. A naturally occurring mouse model for this disease was discovered at The Jackson Laboratory and shown to be due to homozygosity for a 1-bp deletion in exon 10 of the gus gene. The murine model MPS VII (gusmps/mps) has been very well characterized and used extensively to evaluate experimental strategies for lysosomal storage diseases, including bone marrow transplantation, enzyme replacement therapy, and gene therapy. To enhance the value of this model for enzyme and gene therapy, we produced a transgenic mouse expressing the human β-glucuronidase cDNA with an amino acid substitution at the active site nucleophile (E540A) and bred it onto the MPS VII (gusmps/mps) background. We demonstrate here that the mutant mice bearing the active site mutant human transgene retain the clinical, morphological, biochemical, and histopathological characteristics of the original MPS VII (gusmps/mps) mouse. However, they are now tolerant to immune challenge with human β-glucuronidase. This “tolerant MPS VII mouse model” should be useful for preclinical trials evaluating the effectiveness of enzyme and/or gene therapy with the human gene products likely to be administered to human patients with MPS VII.


Molecular Genetics and Metabolism | 2010

Early versus late treatment of spinal cord compression with long-term intrathecal enzyme replacement therapy in canine mucopolysaccharidosis type I

Patricia Dickson; Stephen Hanson; Michael F. McEntee; Charles H. Vite; Carole Vogler; Anton Mlikotic; Agnes H. Chen; Katherine P. Ponder; Mark E. Haskins; Brigette L. Tippin; Steven Q. Le; Merry Passage; Catalina Guerra; Ashley Dierenfeld; Jackie K. Jens; Elizabeth M. Snella; Shih-hsin Kan; N. Matthew Ellinwood

Enzyme replacement therapy (ERT) with intravenous recombinant human alpha-l-iduronidase (IV rhIDU) is a treatment for patients with mucopolysaccharidosis I (MPS I). Spinal cord compression develops in MPS I patients due in part to dural and leptomeningeal thickening from accumulated glycosaminoglycans (GAG). We tested long-term and every 3-month intrathecal (IT) and weekly IV rhIDU in MPS I dogs age 12-15months (Adult) and MPS I pups age 2-23days (Early) to determine whether spinal cord compression could be reversed, stabilized, or prevented. Five treatment groups of MPS I dogs were evaluated (n=4 per group): IT+IV Adult, IV Adult, IT + IV Early, 0.58mg/kg IV Early and 1.57mg/kg IV Early. IT + IV rhIDU (Adult and Early) led to very high iduronidase levels in cervical, thoracic, and lumber spinal meninges (3600-29,000% of normal), while IV rhIDU alone (Adult and Early) led to levels that were 8.2-176% of normal. GAG storage was significantly reduced from untreated levels in spinal meninges of IT + IV Early (p<.001), IT+IV Adult (p=.001), 0.58mg/kg IV Early (p=.002) and 1.57mg/kg IV Early (p<.001) treatment groups. Treatment of dogs shortly after birth with IT+IV rhIDU (IT + IV Early) led to normal to near-normal GAG levels in the meninges and histologic absence of storage vacuoles. Lysosomal storage was reduced in spinal anterior horn cells in 1.57mg/kg IV Early and IT + IV Early animals. All dogs in IT + IV Adult and IV Adult groups had compression of their spinal cord at 12-15months of age determined by magnetic resonance imaging and was due to protrusion of spinal disks into the canal. Cord compression developed in 3 of 4 dogs in the 0.58mg/kg IV Early group; 2 of 3 dogs in the IT + IV Early group; and 0 of 4 dogs in the 1.57mg/kg IV Early group by 12-18months of age. IT + IV rhIDU was more effective than IV rhIDU alone for treatment of meningeal storage, and it prevented meningeal GAG accumulation when begun early. High-dose IV rhIDU from birth (1.57mg/kg weekly) appeared to prevent cord compression due to protrusion of spinal disks.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2013

Rescue of dystrophic skeletal muscle by PGC-1α involves restored expression of dystrophin associated protein complex components and satellite cell signaling

Katrin Hollinger; Delphine Gardan-Salmon; Connie Santana; Drance Rice; Elizabeth M. Snella; Joshua T. Selsby

Duchenne muscular dystrophy is typically diagnosed in the preschool years because of locomotor defects, indicative of muscle damage. Thus, effective therapies must be able to rescue muscle from further decline. We have established that peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Pgc-1α) gene transfer will prevent many aspects of dystrophic pathology, likely through upregulation of utrophin and increased oxidative capacity; however, the extent to which it will rescue muscle with disease manifestations has not been determined. Our hypothesis is that gene transfer of Pgc-1α into declining muscle will reduce muscle injury compared with control muscle. To test our hypothesis, adeno-associated virus 6 (AAV6) driving expression of Pgc-1α was injected into single hind limbs of 3-wk-old mdx mice, while the contralateral limb was given a sham injection. At 6 wk of age, treated solei had 37% less muscle injury compared with sham-treated muscles (P < 0.05). Resistance to contraction-induced injury was improved 10% (P < 0.05), likely driven by the five-fold (P < 0.05) increase in utrophin protein expression and increase in dystrophin-associated complex members. Treated muscles were more resistant to fatigue, which was likely caused by the corresponding increase in oxidative markers. Pgc-1α overexpressing limbs also exhibited increased expression of genes related to muscle repair and autophagy. These data indicate that the Pgc-1α pathway remains a good therapeutic target, as it reduced muscle injury and improved function using a rescue paradigm. Further, these data also indicate that the beneficial effects of Pgc-1α gene transfer are more complex than increased utrophin expression and oxidative gene expression.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Missense models [Gustm(E536A)Sly, Gustm(E536Q)Sly, and Gustm(L175F)Sly] of murine mucopolysaccharidosis type VII produced by targeted mutagenesis

Shunji Tomatsu; Koji O. Orii; Carole Vogler; Jeffrey H. Grubb; Elizabeth M. Snella; Monica A. Gutierrez; Tatiana Dieter; Kazuko Sukegawa; Tadao Orii; Naomi Kondo; William S. Sly

Human mucopolysaccharidosis VII (MPS VII, Sly syndrome) results from a deficiency of β-glucuronidase (GUS) and has been associated with a wide range in severity of clinical manifestations. To study missense mutant models of murine MPS VII with phenotypes of varying severity, we used targeted mutagenesis to produce E536A and E536Q, corresponding to active-site nucleophile replacements E540A and E540Q in human GUS, and L175F, corresponding to the most common human mutation, L176F. The E536A mouse had no GUS activity in any tissue and displayed a severe phenotype like that of the originally described MPS VII mice carrying a deletion mutation (gusmps/mps). E536Q and L175F mice had low levels of residual activity and milder phenotypes. All three mutant MPS models showed progressive lysosomal storage in many tissues but had different rates of accumulation. The amount of urinary glycosaminoglycan excretion paralleled the clinical severity, with urinary glycosaminoglycans remarkably higher in E536A mice than in E536Q or L175F mice. Molecular analysis showed that the Gus mRNA levels were quantitatively similar in the three mutant mouse strains and normal mice. These mouse models, which mimic different clinical phenotypes of human MPS VII, should be useful in studying pathogenesis and also provide useful models for studying enzyme replacement therapy and targeted correction of missense mutations.


Apmis | 2011

Glycosaminoglycan storage in neuroanatomical regions of mucopolysaccharidosis I dogs following intrathecal recombinant human iduronidase

Agnes H. Chen; Carole Vogler; Michael F. McEntee; Stephen Hanson; N. Matthew Ellinwood; Jackie K. Jens; Elizabeth M. Snella; Merry Passage; Steven Q. Le; Catalina Guerra; Patricia Dickson

Chen A, Vogler C, McEntee M, Hanson S, Ellinwood MN, Jens J, Snella E, Passage M, Le S, Guerra C, Dickson P. Glycosaminoglycan storage in neuroanatomical regions of mucopolysaccharidosis I dogs following intrathecal recombinant human iduronidase. APMIS 2011; 119: 513–21.


PLOS ONE | 2016

A Mutation in LTBP2 Causes Congenital Glaucoma in Domestic Cats (Felis catus)

Markus H. Kuehn; Koren Lipsett; Marilyn Menotti-Raymond; S. Scott Whitmore; Todd E. Scheetz; Victor A. David; Stephen J. O'Brien; Zhongyuan Zhao; Jackie K. Jens; Elizabeth M. Snella; N. Matthew Ellinwood; Gillian J. McLellan

The glaucomas are a group of diseases characterized by optic nerve damage that together represent a leading cause of blindness in the human population and in domestic animals. Here we report a mutation in LTBP2 that causes primary congenital glaucoma (PCG) in domestic cats. We identified a spontaneous form of PCG in cats and established a breeding colony segregating for PCG consistent with fully penetrant, autosomal recessive inheritance of the trait. Elevated intraocular pressure, globe enlargement and elongated ciliary processes were consistently observed in all affected cats by 8 weeks of age. Varying degrees of optic nerve damage resulted by 6 months of age. Although subtle lens zonular instability was a common feature in this cohort, pronounced ectopia lentis was identified in less than 10% of cats examined. Thus, glaucoma in this pedigree is attributed to histologically confirmed arrest in the early post-natal development of the aqueous humor outflow pathways in the anterior segment of the eyes of affected animals. Using a candidate gene approach, significant linkage was established on cat chromosome B3 (LOD 18.38, θ = 0.00) using tightly linked short tandem repeat (STR) loci to the candidate gene, LTBP2. A 4 base-pair insertion was identified in exon 8 of LTBP2 in affected individuals that generates a frame shift that completely alters the downstream open reading frame and eliminates functional domains. Thus, we describe the first spontaneous and highly penetrant non-rodent model of PCG identifying a valuable animal model for primary glaucoma that closely resembles the human disease, providing valuable insights into mechanisms underlying the disease and a valuable animal model for testing therapies.


Experimental Neurology | 2015

Diffusion tensor imaging and myelin composition analysis reveal abnormal myelination in corpus callosum of canine mucopolysaccharidosis I

James M. Provenzale; Igor Nestrasil; Steven Chen; Shih hsin Kan; Steven Q. Le; Jacqueline K. Jens; Elizabeth M. Snella; Kristen N. Vondrak; Jennifer K. Yee; Charles H. Vite; David Elashoff; Lewei Duan; Raymond Y. Wang; N. Matthew Ellinwood; Miguel Guzman; Elsa Shapiro; Patricia Dickson

Children with mucopolysaccharidosis I (MPS I) develop hyperintense white matter foci on T2-weighted brain magnetic resonance (MR) imaging that are associated clinically with cognitive impairment. We report here a diffusion tensor imaging (DTI) and tissue evaluation of white matter in a canine model of MPS I. We found that two DTI parameters, fractional anisotropy (a measure of white matter integrity) and radial diffusivity (which reflects degree of myelination) were abnormal in the corpus callosum of MPS I dogs compared to carrier controls. Tissue studies of the corpus callosum showed reduced expression of myelin-related genes and an abnormal composition of myelin in MPS I dogs. We treated MPS I dogs with recombinant alpha-L-iduronidase, which is the enzyme that is deficient in MPS I disease. The recombinant alpha-L-iduronidase was administered by intrathecal injection into the cisterna magna. Treated dogs showed partial correction of corpus callosum myelination. Our findings suggest that abnormal myelination occurs in the canine MPS I brain, that it may underlie clinically-relevant brain imaging findings in human MPS I patients, and that it may respond to treatment.


Molecular Genetics and Metabolism | 2012

Accelerated clinical disease and pathology in mucopolysaccharidosis type IIIB and GalNAc transferase double knockout mice.

Eman Mohammed; Elizabeth M. Snella; Michelle Rutz-Mendicino; Franklin D. Echevarria; Rafi Simon Awedikian; Elizabeth M. Whitley; N. Matthew Ellinwood

Mucopolysaccharidosis type IIIB (MPS IIIB) is a neuropathic lysosomal storage disorder (LSD) resulting from an inherited deficiency of N-acetyl-α-D-glucosaminidase (Naglu) activity, an enzyme required to degrade the glycosaminoglycan heparan sulfate (HS). A deficiency in Naglu activity leads to lysosomal accumulation of HS as a primary storage substrate, and the gangliosides GM2 and GM3 as secondary accumulation products. To test the effect on neuropathogenesis of ganglioside accumulation, we bred mice deficient in both Naglu and GalNaAcT activities. The latter is the enzyme required for synthesis of GM2 and other complex gangliosides. Contrary to our expectation and to double knockout (DKO) studies where GalNAcT was knocked out in combination with other LSDs, our DKO mice showed a drastically shortened lifespan (24.5±1.4 weeks, versus 50.5±0.9 weeks (MPS IIIB), and 38.6±1.2 weeks (GalNAcT)). To confirm that HS storage was the primary element resulting in the accelerated disease in our DKO mice, and not a locus tightly linked to the Naglu gene, we replicated our study with MPS IIIA mice, and found a virtually identical result (27.5±1.8 weeks, versus 53.8±1.6 weeks). All DKO mice showed motor signs of hind limb ataxia and hyper-extension, which were not seen in single KO or normal mice. At approximately 5 months of age, the MPS IIIB-DKO showed a unique pattern of vacuolization and nerve fiber degeneration in the corpus callosum, seen only in the DKO mice, as well as the relatively early intracytoplasmic vacuolation of many neurons and glia characteristic of the MPS IIIB mice. We analyzed motor performance on a rocking Rota-Rod beginning at 3 months of age. The MPS IIIA-DKO and MPS IIIB-DKO mice showed impaired performance and were statistically different from all parental lines. In particular, the MPS IIIB-DKO mice were significantly different from the parent MPS IIIB strains at 3, 5, and 6 months (p≤0.0245). In conclusion we identified an accelerated phenotype associated with MPS IIIB within a DKO model system which showed white matter changes, with attendant performance deficits and a drastically shortened lifespan. This was in stark contrast to our expectations of a salutary response to the elimination of GM2. Despite this, the accelerated pathology and clinical signs represent a potentially improved system to study MPS IIIB neuropathogenesis as well as the role of complex gangliosides in normal CNS function.

Collaboration


Dive into the Elizabeth M. Snella's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patricia Dickson

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Steven Q. Le

Los Angeles Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carole Vogler

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Charles H. Vite

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Gillian J. McLellan

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge