Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Inherit Metab Dis ; 41(6): 965-976, 2018 11.
Article in English | MEDLINE | ID: mdl-30043186

ABSTRACT

BACKGROUND: Glycogen storage disease type Ia (GSD Ia) in dogs closely resembles human GSD Ia. Untreated patients with GSD Ia develop complications associated with glucose-6-phosphatase (G6Pase) deficiency. Survival of human patients on intensive nutritional management has improved; however, long-term complications persist including renal failure, nephrolithiasis, hepatocellular adenomas (HCA), and a high risk for hepatocellular carcinoma (HCC). Affected dogs fail to thrive with dietary therapy alone. Treatment with gene replacement therapy using adeno-associated viral vectors (AAV) expressing G6Pase has greatly prolonged life and prevented hypoglycemia in affected dogs. However, long-term complications have not been described to date. METHODS: Five GSD Ia-affected dogs treated with AAV-G6Pase were evaluated. Dogs were euthanized due to reaching humane endpoints related to liver and/or kidney involvement, at 4 to 8 years of life. Necropsies were performed and tissues were analyzed. RESULTS: Four dogs had liver tumors consistent with HCA and HCC. Three dogs developed renal failure, but all dogs exhibited progressive kidney disease histologically. Urolithiasis was detected in two dogs; uroliths were composed of calcium oxalate and calcium phosphate. One affected and one carrier dog had polycystic ovarian disease. Bone mineral density was not significantly affected. CONCLUSIONS: Here, we show that the canine GSD Ia model demonstrates similar long-term complications as GSD Ia patients in spite of gene replacement therapy. Further development of gene therapy is needed to develop a more effective treatment to prevent long-term complications of GSD Ia.


Subject(s)
Carcinoma, Hepatocellular/etiology , Genetic Therapy , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/therapy , Liver Neoplasms/etiology , Animals , Dependovirus/genetics , Disease Models, Animal , Dogs , Female , Genetic Vectors , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hypoglycemia/genetics , Hypoglycemia/metabolism , Liver/pathology , Male
2.
Hum Gene Ther ; 23(4): 407-18, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22185325

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) is the inherited deficiency of glucose-6-phosphatase (G6Pase), primarily found in liver and kidney, which causes life-threatening hypoglycemia. Dogs with GSD-Ia were treated with double-stranded adeno-associated virus (AAV) vectors encoding human G6Pase. Administration of an AAV9 pseudotyped (AAV2/9) vector to seven consecutive GSD-Ia neonates prevented hypoglycemia during fasting for up to 8 hr; however, efficacy eventually waned between 2 and 30 months of age, and readministration of a new pseudotype was eventually required to maintain control of hypoglycemia. Three of these dogs succumbed to acute hypoglycemia between 7 and 9 weeks of age; however, this demise could have been prevented by earlier readministration an AAV vector, as demonstrated by successful prevention of mortality of three dogs treated earlier in life. Over the course of this study, six out of nine dogs survived after readministration of an AAV vector. Of these, each dog required readministration on average every 9 months. However, two were not retreated until >34 months of age, while one with preexisting antibodies was re-treated three times in 10 months. Glycogen content was normalized in the liver following vector administration, and G6Pase activity was increased in the liver of vector-treated dogs in comparison with GSD-Ia dogs that received only with dietary treatment. G6Pase activity reached approximately 40% of normal in two female dogs following AAV2/9 vector administration. Elevated aspartate transaminase in absence of inflammation indicated that hepatocellular turnover in the liver might drive the loss of vector genomes. Survival was prolonged for up to 60 months in dogs treated by readministration, and all dogs treated by readministration continue to thrive despite the demonstrated risk for recurrent hypoglycemia and mortality from waning efficacy of the AAV2/9 vector. These preclinical data support the further translation of AAV vector-mediated gene therapy in GSD-Ia.


Subject(s)
Dependovirus/genetics , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Animals , Dogs , Genetic Therapy , Genetic Vectors , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/metabolism , Hypoglycemia/genetics , Hypoglycemia/metabolism , Hypoglycemia/therapy , Liver/metabolism
3.
Mol Ther ; 19(11): 1961-70, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21730973

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) is caused by the deficiency of glucose-6-phosphatase (G6Pase). Long-term complications of GSD-Ia include life-threatening hypoglycemia and proteinuria progressing to renal failure. A double-stranded (ds) adeno-associated virus serotype 2 (AAV2) vector encoding human G6Pase was pseudotyped with four serotypes, AAV2, AAV7, AAV8, and AAV9, and we evaluated efficacy in 12-day-old G6pase (-/-) mice. Hypoglycemia during fasting (plasma glucose <100 mg/dl) was prevented for >6 months by the dsAAV2/7, dsAAV2/8, and dsAAV2/9 vectors. Prolonged fasting for 8 hours revealed normalization of blood glucose following dsAAV2/9 vector administration at the higher dose. The glycogen content of kidney was reduced by >65% with both the dsAAV2/7 and dsAAV2/9 vectors, and renal glycogen content was stably reduced between 7 and 12 months of age for the dsAAV2/9 vector-treated mice. Every vector-treated group had significantly reduced glycogen content in the liver, in comparison with untreated G6pase (-/-) mice. G6Pase was expressed in many renal epithelial cells of with the dsAAV2/9 vector for up to 12 months. Albuminuria and renal fibrosis were reduced by the dsAAV2/9 vector. Hepatorenal correction in G6pase (-/-) mice demonstrates the potential of AAV vectors for the correction of inherited diseases of metabolism.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Animals , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Vectors/administration & dosage , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/mortality , Humans , Hypoglycemia/genetics , Hypoglycemia/therapy , Kaplan-Meier Estimate , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout
4.
ILAR J ; 50(2): 122-7, 2009.
Article in English | MEDLINE | ID: mdl-19293457

ABSTRACT

Scientists first described inborn errors of metabolism, also termed inherited disorders of metabolism, early in the 20th century and since then have determined the biochemical and genetic bases of a great number of these disorders both in humans and in an increasing number of companion animals. The availability of metabolic screening tests has advanced the biochemical and genetic characterization in affected breeds of companion animals of inherited metabolic disorders involving amino acid, carbohydrate, fatty acid, and metal metabolism. Advances in gene therapy have led to the development of new treatments for inherited disorders of metabolism, and animal models have played a critical role in this research. For example, glycogen storage disease type Ia in dogs was highly responsive to adeno-associated viral vectormediated gene therapy, which prolonged survival and for more than a year prevented hypoglycemia during fasting. Gene therapy for other glycogen storage diseases and metabolic disorders will also be feasible. The establishment of a breeding colony and the ability to sustain affected animals are critical steps toward evaluating the safety and efficacy of gene therapy with clinically relevant endpoints. The further development of gene therapy for inherited disorders of metabolism could lead to curative therapy for affected humans and animals alike.


Subject(s)
Disease Models, Animal , Genetic Therapy/methods , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/therapy , Animals , Cats , Dogs
5.
Mol Ther ; 16(8): 1366-71, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18560415

ABSTRACT

Glycogen storage disease type II (Pompe disease; MIM 232300) stems from the deficiency of acid alpha-glucosidase (GAA; acid maltase; EC 3.2.1.20), which primarily involves cardiac and skeletal muscles. An adeno-associated virus 2/8 (AAV2/8) vector containing the muscle creatine kinase (MCK) (CK1) reduced glycogen content by approximately 50% in the heart and quadriceps in GAA-knockout (GAA-KO) mice; furthermore, an AAV2/8 vector containing the hybrid alpha-myosin heavy chain enhancer-/MCK enhancer-promoter (MHCK7) cassette reduced glycogen content by >95% in heart and >75% in the diaphragm and quadriceps. Transduction with an AAV2/8 vector was higher in the quadriceps than in the gastrocnemius. An AAV2/9 vector containing the MHCK7 cassette corrected GAA deficiency in the distal hindlimb, and glycogen accumulations were substantially cleared by human GAA (hGAA) expression therein; however, the analogous AAV2/7 vector achieved much lower efficacy. Administration of the MHCK7-containing vectors significantly increased striated muscle function as assessed by increased Rotarod times at 18 weeks after injection, whereas the CK1-containing vector did not increase Rotarod performance. Importantly, type IIb myofibers in the extensor digitalis longus (EDL) were transduced, thereby correcting a myofiber type that is unresponsive to enzyme replacement therapy. In summary, AAV8 and AAV9-pseudotyped vectors containing the MHCK7 regulatory cassette achieved enhanced efficacy in Pompe disease mice.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Muscle, Striated/metabolism , Animals , Creatine Kinase, MM Form/genetics , Creatine Kinase, MM Form/metabolism , Enhancer Elements, Genetic/genetics , Female , Genetic Vectors/genetics , Glycogen/metabolism , Glycogen Storage Disease Type II/genetics , Hindlimb/metabolism , Hindlimb/pathology , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Striated/enzymology , Muscle, Striated/pathology , Myocardium/metabolism , Myosin Heavy Chains/genetics , Promoter Regions, Genetic/genetics , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Transduction, Genetic , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
6.
Mol Ther ; 16(4): 665-72, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18362924

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) profoundly impairs glucose release by the liver due to glucose-6-phosphatase (G6Pase) deficiency. An adeno-associated virus (AAV) containing a small human G6Pase transgene was pseudotyped with AAV8 (AAV2/8) to optimize liver tropism. Survival was prolonged in 2-week-old G6Pase (-/-) mice by 600-fold fewer AAV2/8 vector particles (vp), in comparison to previous experiments involving this model (2 x 10(9) vp; 3 x 10(11) vp/kg). When the vector was pseudotyped with AAV1, survival was prolonged only at a higher dose (3 x 10(13) vp/kg). The AAV2/8 vector uniquely prevented hypoglycemia during fasting and fully corrected liver G6Pase deficiency in GSD-Ia mice and dogs. The AAV2/8 vector has prolonged survival in three GSD-Ia dogs to >11 months, which validated this strategy in the large animal model for GSD-Ia. Urinary biomarkers, including lactate and 3-hydroxybutyrate, were corrected by G6Pase expression solely in the liver. Glycogen accumulation in the liver was reduced almost to the normal level in vector-treated GSD-Ia mice and dogs, as was the hepatocyte growth factor (HGF) in GSD-Ia mice. These preclinical data demonstrated the efficacy of correcting hepatic G6Pase deficiency, and support the further preclinical development of AAV vector-mediated gene therapy for GSD-Ia.


Subject(s)
Dependovirus/genetics , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Hypoglycemia/therapy , Animals , Disease Models, Animal , Dogs , Genetic Therapy , Genetic Vectors , Glucose-6-Phosphatase/biosynthesis , Glycogen Storage Disease Type I/enzymology , Humans , Hypoglycemia/enzymology , Liver Glycogen/metabolism , Mice , Mice, Knockout
7.
Mol Ther ; 14(6): 822-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16987711

ABSTRACT

Glycogen storage disease type II (GSD-II; Pompe disease; MIM 232300) is an inherited muscular dystrophy caused by deficiency in the activity of the lysosomal enzyme acid alpha-glucosidase (GAA). We hypothesized that chimeric GAA containing an alternative signal peptide could increase the secretion of GAA from transduced cells and enhance the receptor-mediated uptake of GAA in striated muscle. The relative secretion of chimeric GAA from transfected 293 cells increased up to 26-fold. Receptor-mediated uptake of secreted, chimeric GAA corrected cultured GSD-II patient cells. High-level hGAA was sustained in the plasma of GSD-II mice for 24 weeks following administration of an AAV2/8 vector encoding chimeric GAA; furthermore, GAA activity was increased and glycogen content was significantly reduced in striated muscle and in the brain. Administration of only 1 x 10(10) vector particles increased GAA activity in the heart and diaphragm for >18 weeks, whereas 3 x 10(10) vector particles increased GAA activity and reduced glycogen content in the heart, diaphragm, and quadriceps. Furthermore, an AAV2/2 vector encoding chimeric GAA produced secreted hGAA for >12 weeks in the majority of treated GSD-II mice. Thus, chimeric, highly secreted GAA enhanced the efficacy of AAV vector-mediated gene therapy in GSD-II mice.


Subject(s)
Dependovirus/genetics , Glucan 1,4-alpha-Glucosidase/genetics , Glycogen Storage Disease Type II/therapy , Animals , Blotting, Western , Cell Line , Cells, Cultured , Fibroblasts/metabolism , Genetic Therapy/methods , Genetic Vectors/genetics , Glucan 1,4-alpha-Glucosidase/metabolism , Glycogen/metabolism , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Humans , Mice , Mice, Knockout , Protein Sorting Signals/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Treatment Outcome
8.
Mol Ther ; 12(5): 876-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16005263

ABSTRACT

Glycogen storage disease type II (GSD-II; Pompe disease) is caused by a deficiency of acid alpha-glucosidase (GAA; acid maltase) and manifests as muscle weakness, hypertrophic cardiomyopathy, and respiratory failure. Adeno-associated virus vectors containing either a liver-specific promoter (LSP) (AAV-LSPhGAApA) or a hybrid CB promoter (AAV-CBhGAApA) to drive human GAA expression were pseudotyped as AAV8 and administered to immunocompetent GAA-knockout mice. Secreted hGAA was detectable in plasma between 1 day and 12 weeks postadministration with AAV-LSPhGAApA and only from 1 to 8 days postadministration for AAV-CBGAApA. No anti-GAA antibodies were detected in response to AAV-LSPhGAApA (<1:200), whereas AAV-CBhGAApA provoked an escalating antibody response starting 2 weeks postadministration. The LSP drove approximately 60-fold higher GAA expression than the CB promoter in the liver by 12 weeks following vector administration. Furthermore, the detected cellular immunity was provoked by AAV-CBhGAApA, as detected by ELISpot and CD4+/CD8+ lymphocyte immunodetection. GAA activity was increased to higher than normal and glycogen content was reduced to essentially normal levels in the heart and skeletal muscle following administration of AAV-LSPhGAApA. Therefore, liver-restricted GAA expression with an AAV vector evaded immunity and enhanced efficacy in GSD-II mice.


Subject(s)
Creatine Kinase, MM Form/metabolism , Genetic Vectors , Glycogen Storage Disease Type II/metabolism , Liver/metabolism , alpha-Glucosidases/biosynthesis , Animals , Antibody Formation , Creatine Kinase , DNA, Viral , Dependovirus/genetics , Enhancer Elements, Genetic , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glycogen/metabolism , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/therapy , Humans , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Plasmids/genetics , Promoter Regions, Genetic , alpha-Glucosidases/immunology
9.
J Am Vet Med Assoc ; 226(12): 2016-9, 2001, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15989184

ABSTRACT

A 7-month-old sexually intact male Cocker Spaniel was admitted to the North Carolina State University Veterinary Teaching Hospital for evaluation of lethargy, panting, and excessive salivation that had become progressively severe during a 5-hour period. Despite intensive medical care, the dog died within the first 24 hours of hospitalization, and death was attributed to acute, severe, necrotizing pneumonia. Lung tissue collected at necropsy by use of swabs was cultured and yielded an isolate of Escherichia coli; because of the rapid progression of illness in an otherwise healthy dog, the isolate underwent virulence typing and was determined to be a necrotoxigenic E. coli. Necrotoxigenic E. coli produce a toxin called cytotoxic necrotizing factor and are known to be involved in extraintestinal infections, including urinary tract infection, in humans and animals. Virulence typing of E. coli isolates from dogs with peracute pneumonia is recommended to further characterize the epidemiologic characteristics and public health importance of necrotoxigenic E. coli.


Subject(s)
Dog Diseases/microbiology , Escherichia coli Infections/veterinary , Escherichia coli/pathogenicity , Hemorrhage/veterinary , Pneumonia, Bacterial/veterinary , Animals , Bacterial Typing Techniques/veterinary , Dog Diseases/diagnosis , Dogs , Escherichia coli/isolation & purification , Escherichia coli Infections/diagnosis , Escherichia coli Infections/microbiology , Fatal Outcome , Hemorrhage/diagnosis , Hemorrhage/microbiology , Lung/microbiology , Lung/pathology , Male , Pneumonia, Bacterial/diagnosis , Pneumonia, Bacterial/microbiology , Virulence
10.
Mol Ther ; 11(6): 889-98, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15922959

ABSTRACT

Glycogen storage disease type II (Pompe disease) causes death in infancy from cardiorespiratory failure due to acid alpha-glucosidase (GAA; acid maltase) deficiency. An AAV2 vector pseudotyped as AAV6 (AAV2/6 vector) transiently expressed high-level human GAA in GAA-knockout (GAA-KO) mice without reducing glycogen storage; however, in immunodeficient GAA-KO/SCID mice the AAV2/6 vector expressed high-level GAA and reduced the glycogen content of the injected muscle for 24 weeks. A CD4+/CD8+ lymphocytic infiltrate was observed in response to the AAV2/6 vector in immunocompetent GAA-KO mice. When a muscle-specific creatine kinase promoter was substituted for the CB promoter (AAV-MCKhGAApA), that AAV2/6 vector expressed high-level GAA and reduced glycogen content in immunocompetent GAA-KO mice. Muscle-restricted expression of hGAA provoked only a humoral (not cellular) immune response. Intravenous administration of a high number of particles of AAV-MCKhGAApA as AAV2/7 reduced the glycogen content of the heart and skeletal muscle and corrected individual myofibers in immunocompetent GAA-KO mice 24 weeks postinjection. In summary, persistent correction of muscle glycogen content was achieved with an AAV vector containing a muscle-specific promoter in GAA-KO mice, and this approach should be considered for muscle-targeted gene therapy in Pompe disease.


Subject(s)
Creatine Kinase/genetics , Dependovirus/genetics , Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Muscle, Skeletal/enzymology , Promoter Regions, Genetic/genetics , alpha-Glucosidases/genetics , Animals , Antibodies/blood , Antibody Formation , Creatine Kinase, MM Form , DNA, Viral/analysis , Enhancer Elements, Genetic/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glycogen/analysis , Glycogen Storage Disease Type II/genetics , Humans , Injections, Intramuscular , Isoenzymes/genetics , Mice , Mice, Knockout , Muscle, Skeletal/chemistry , Myocardium/chemistry , Myocardium/enzymology , alpha-Glucosidases/analysis , alpha-Glucosidases/immunology
11.
Vet Radiol Ultrasound ; 45(4): 336-9, 2004.
Article in English | MEDLINE | ID: mdl-15373261

ABSTRACT

Congenital cerebellar disorders can be caused by in utero or neonatal infections, genetic aberrations causing malformations, and neurodegenerative processes. Most congenital cerebellar disorders are diagnosed definitively with histopathology. However, antemortem diagnosis of cerebellar malformations can be made by imaging the brain. This paper describes the antemortem appearance of a congenital cerebellar malformation in a Boston Terrier puppy on ultrasound images. This appearance was compared with the postmortem findings that were comparable to Dandy Walker Syndrome in humans. A previous report of this syndrome in Boston Terriers suggests the problem may be inherited in this breed.


Subject(s)
Dandy-Walker Syndrome/veterinary , Dog Diseases/diagnostic imaging , Animals , Animals, Newborn , Dandy-Walker Syndrome/diagnostic imaging , Diagnosis, Differential , Dog Diseases/pathology , Dogs , Male , Pedigree , Ultrasonography/veterinary
12.
J Vet Intern Med ; 18(2): 201-8, 2004.
Article in English | MEDLINE | ID: mdl-15058771

ABSTRACT

Adult-onset cerebellar cortical degeneration recently has been reported in American Staffordshire Terriers. We describe the clinical and histopathologic features of this disease and examine its mode of inheritance in 63 affected dogs. The age at which neurologic deficits 1st were recognized varied from 18 months to 9 years, with the majority of dogs presented to veterinarians between 4 and 6 years of age. Time from onset of clinical signs to euthanasia varied from 6 months to 6.5 years, with the majority of affected dogs surviving from 2 to 4 years. Initial neurologic findings included stumbling, truncal sway, and ataxia exacerbated by lifting the head up and negotiating stairs. Signs progressed to obvious ataxia characterized by dysmetria, nystagmus, coarse intention tremor, variable loss of menace reaction, marked truncal sway, and falling with transient opisthotonus. With continued progression, dogs became unable to walk without falling repeatedly. Cerebellar atrophy was visible on magnetic resonance images and on gross pathology. Histopathologic findings included marked loss of Purkinje neurons with thinning of the molecular and granular layers and increased cellularity of the cerebellar nuclei. The closest common ancestor of the dogs was born in the 1950s and inheritance was most consistent with an autosomal recessive mode of transmission with a prevalence estimated at 1 in 400 dogs. This inherited disease is comparable to the group of diseases known as spinocerebellar ataxias in humans. Many spinocerebellar ataxias in humans are caused by nucleotide repeats, and this genetic aberration merits investigation as a potential cause of the disease in American Staffordshire Terriers.


Subject(s)
Cerebellar Ataxia/veterinary , Dog Diseases/epidemiology , Dog Diseases/pathology , Animals , Cerebellar Ataxia/epidemiology , Cerebellar Ataxia/pathology , Cerebellum/pathology , Dog Diseases/diagnostic imaging , Dog Diseases/etiology , Dog Diseases/genetics , Dogs , Europe/epidemiology , Female , Male , Pedigree , Purkinje Cells/pathology , Radiography , United States/epidemiology
13.
Vet Clin Pathol ; 26(4): 182-186, 1997.
Article in English | MEDLINE | ID: mdl-12658582

ABSTRACT

A 5-year old female Boxer with a 1-week history of progressive paresis and paraplegia had a T10-13 subarachnoid filling defect on myelography. Exploratory hemilaminectomy revealed an intramedullary spinal cord tumor which was subsequently diagnosed as a poorly differentiated glioma, most likely an anaplastic ependymoma. The cytologic, histologic, and immunocytochemical staining characteristics of this neoplasm are described. Differential diagnoses, including primary and secondary tumors involving the central nervous system are discussed.

SELECTION OF CITATIONS
SEARCH DETAIL
...