Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Pharmacol Exp Ther ; 382(3): 277-286, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35717448

RESUMEN

Mucopolysaccharidosis type IIIB (MPS IIIB; Sanfilippo syndrome B; OMIM #252920) is a lethal, pediatric, neuropathic, autosomal recessive, and lysosomal storage disease with no approved therapy. Patients are deficient in the activity of N-acetyl-alpha-glucosaminidase (NAGLU; EC 3.2.150), necessary for normal lysosomal degradation of the glycosaminoglycan heparan sulfate (HS). Tralesinidase alfa (TA), a fusion protein comprised of recombinant human NAGLU and a modified human insulin-like growth factor 2, is in development as an enzyme replacement therapy that is administered via intracerebroventricular (ICV) infusion, thus circumventing the blood brain barrier. Previous studies have confirmed ICV infusion results in widespread distribution of TA throughout the brains of mice and nonhuman primates. We assessed the long-term tolerability, pharmacology, and clinical efficacy of TA in a canine model of MPS IIIB over a 20-month study. Long-term administration of TA was well tolerated as compared with administration of vehicle. TA was widely distributed across brain regions, which was confirmed in a follow-up 8-week pharmacokinetic/pharmacodynamic study. MPS IIIB dogs treated for up to 20 months had near-normal levels of HS and nonreducing ends of HS in cerebrospinal fluid and central nervous system (CNS) tissues. TA-treated MPS IIIB dogs performed better on cognitive tests and had improved CNS pathology and decreased cerebellar volume loss relative to vehicle-treated MPS IIIB dogs. These findings demonstrate the ability of TA to prevent or limit the biochemical, pathologic, and cognitive manifestations of canine MPS IIIB disease, thus providing support of its potential long-term tolerability and efficacy in MPS IIIB subjects. SIGNIFICANCE STATEMENT: This work illustrates the efficacy and tolerability of tralesinidase alfa as a potential therapeutic for patients with mucopolysaccharidosis type IIIB (MPS IIIB) by documenting that administration to the central nervous system of MPS IIIB dogs prevents the accumulation of disease-associated glycosaminoglycans in lysosomes, hepatomegaly, cerebellar atrophy, and cognitive decline.


Asunto(s)
Mucopolisacaridosis III , Animales , Encéfalo/metabolismo , Niño , Modelos Animales de Enfermedad , Perros , Terapia de Reemplazo Enzimático , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/líquido cefalorraquídeo , Heparitina Sulfato/uso terapéutico , Humanos , Mucopolisacaridosis III/tratamiento farmacológico , Mucopolisacaridosis III/patología
2.
Sci Rep ; 10(1): 20365, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230178

RESUMEN

Mucopolysaccharidosis type IIIB (MPS IIIB; Sanfilippo syndrome B) is an autosomal recessive lysosomal storage disorder caused by the deficiency of alpha-N-acetylglucosaminidase activity, leading to increased levels of nondegraded heparan sulfate (HS). A mouse model has been useful to evaluate novel treatments for MPS IIIB, but has limitations. In this study, we evaluated the naturally occurring canine model of MPS IIIB for the onset and progression of biochemical and neuropathological changes during the preclinical stages (onset approximately 24-30 months of age) of canine MPS IIIB disease. Even by 1 month of age, MPS IIIB dogs had elevated HS levels in brain and cerebrospinal fluid. Analysis of histopathology of several disease-relevant regions of the forebrain demonstrated progressive lysosomal storage and microglial activation despite a lack of cerebrocortical atrophy in the oldest animals studied. More pronounced histopathology changes were detected in the cerebellum, where progressive lysosomal storage, astrocytosis and microglial activation were observed. Microglial activation was particularly prominent in cerebellar white matter and within the deep cerebellar nuclei, where neuron loss also occurred. The findings in this study will form the basis of future assessments of therapeutic efficacy in this large animal disease model.


Asunto(s)
Acetilglucosaminidasa/deficiencia , Cerebelo/patología , Corteza Cerebral/patología , Enfermedades de los Perros/patología , Mucopolisacaridosis III/patología , Prosencéfalo/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enfermedades de los Perros/metabolismo , Perros , Femenino , Heparitina Sulfato/metabolismo , Histocitoquímica , Humanos , Lisosomas/metabolismo , Lisosomas/patología , Masculino , Microglía/metabolismo , Microglía/patología , Mucopolisacaridosis III/metabolismo , Neuronas/metabolismo , Neuronas/patología , Prosencéfalo/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
3.
Front Immunol ; 11: 100, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117254

RESUMEN

Pigs with severe combined immunodeficiency (SCID) are an emerging biomedical animal model. Swine are anatomically and physiologically more similar to humans than mice, making them an invaluable tool for preclinical regenerative medicine and cancer research. One essential step in further developing this model is the immunological humanization of SCID pigs. In this work we have generated T- B- NK- SCID pigs through site directed CRISPR/Cas9 mutagenesis of IL2RG within a naturally occurring DCLRE1C (ARTEMIS)-/- genetic background. We confirmed ART-/-IL2RG-/Y pigs lacked T, B, and NK cells in both peripheral blood and lymphoid tissues. Additionally, we successfully performed a bone marrow transplant on one ART-/-IL2RG-/Y male SCID pig with bone marrow from a complete swine leukocyte antigen (SLA) matched donor without conditioning to reconstitute porcine T and NK cells. Next, we performed in utero injections of cultured human CD34+ selected cord blood cells into the fetal ART-/-IL2RG-/Y SCID pigs. At birth, human CD45+ CD3ε+ cells were detected in cord and peripheral blood of in utero injected SCID piglets. Human leukocytes were also detected within the bone marrow, spleen, liver, thymus, and mesenteric lymph nodes of these animals. Taken together, we describe critical steps forwards the development of an immunologically humanized SCID pig model.


Asunto(s)
Trasplante de Médula Ósea , Subunidad gamma Común de Receptores de Interleucina/genética , Inmunodeficiencia Combinada Grave/genética , Animales , Animales Modificados Genéticamente , Antígenos CD34 , Sistemas CRISPR-Cas , Diferenciación Celular , Quimera , Proteínas de Unión al ADN/deficiencia , Modelos Animales de Enfermedad , Marcación de Gen , Ingeniería Genética , Supervivencia de Injerto , Reacción Huésped-Injerto , Humanos , Células Asesinas Naturales , Modelos Animales , Porcinos , Linfocitos T/metabolismo , Trasplante Heterólogo
5.
PLoS One ; 11(5): e0154412, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27149523

RESUMEN

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.


Asunto(s)
Enfermedades de los Gatos/genética , Glaucoma/veterinaria , Proteínas de Unión a TGF-beta Latente/genética , Animales , Humor Acuoso/fisiología , Enfermedades de los Gatos/congénito , Gatos , Femenino , Ligamiento Genético , Glaucoma/congénito , Glaucoma/genética , Presión Intraocular/genética , Proteínas de Unión a TGF-beta Latente/fisiología , Masculino , Mutación/genética , Linaje , Fenotipo , Células Ganglionares de la Retina/patología
6.
Exp Neurol ; 273: 1-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26222335

RESUMEN

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.


Asunto(s)
Cuerpo Calloso/patología , Mucopolisacaridosis I/patología , Proteínas de la Mielina/metabolismo , Vaina de Mielina/patología , Animales , Imagen de Difusión Tensora , Modelos Animales de Enfermedad , Perros , Femenino , Humanos , Iduronidasa/uso terapéutico , Procesamiento de Imagen Asistido por Computador , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Masculino , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/genética , Proteínas de la Mielina/genética , Evaluación de Resultado en la Atención de Salud , ARN Mensajero/metabolismo
7.
Comp Med ; 63(2): 163-73, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23582423

RESUMEN

The mucopolysaccharidosis type I (MPS I) dog model has been important in the development of therapies for human patients. We treated dogs with enzyme replacement therapy (ERT) by various approaches. Dogs assessed included untreated MPS I dogs, heterozygous carrier dogs, and MPS I dogs treated with intravenous ERT as adults (beginning at age 13 to 16 mo), intrathecal and intravenous ERT as adults (beginning at age 13 to 16 mo), or intrathecal ERT as juveniles (beginning at age 4 mo). We then characterized the neuroimaging findings of 32 of these dogs (age, 12 to 30 mo). Whole and midsagittal volumes of the corpus callosum, measured from brain MRI, were significantly smaller in affected dogs compared with unaffected heterozygotes. Corpus callosum volumes in dogs that were treated with intrathecal ERT from 4 mo until 21 mo of age were indistinguishable from those of age-matched carrier controls. Dogs with MPS I showed cerebral ventricular enlargement and cortical atrophy as early as 12 mo of age. Ventricular enlargement was greater in untreated MPS I dogs than in age-matched dogs treated with intrathecal ERT as juveniles or adults. However, treated dogs still showed some ventricular enlargement or cortical atrophy (or both). Understanding the progression of neuroimaging findings in dogs with MPS I and their response to brain-directed therapy may improve preclinical studies for new human-directed therapies. In particular, corpus callosum volumes may be useful quantitative neuroimaging markers for MPS-related brain disease and its response to therapy.


Asunto(s)
Modelos Animales de Enfermedad , Perros , Terapia de Reemplazo Enzimático/métodos , Mucopolisacaridosis I/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Femenino , Iduronidasa/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Mucopolisacaridosis I/patología , Neuroimagen , Proteínas Recombinantes/uso terapéutico
8.
Mol Genet Metab ; 107(1-2): 129-35, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22867887

RESUMEN

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.


Asunto(s)
Mucopolisacaridosis III/genética , Mucopolisacaridosis III/patología , N-Acetilgalactosaminiltransferasas/genética , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glicoesfingolípidos/metabolismo , Masculino , Ratones , Ratones Noqueados , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/mortalidad , Prueba de Desempeño de Rotación con Aceleración Constante
9.
Comp Med ; 61(3): 251-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21819695

RESUMEN

Feline breeding colonies face genetic constraints involving founder effects. A Siamese-founded colony used to study primary congenital glaucoma displayed coat colors additional to the Siamese coat. Genes affecting pigment can exhibit pleiotropy on ocular development and function. To remove potentially confounding phenotypes from our colony, we documented the source and frequency of the Siamese allele at the gene for tyrosinase (TYR), the dilution allele at melanophilin (MLPH), and the brown allele at tyrosinase-related protein 1 (TYRP1). We used PCR-RFLP diagnostics to genotype cats in our colony for the published alleles. A commercially acquired phenotypically normal tom was the source of the dilute allele. A founding Siamese queen was the source of the brown allele. Founders also were blood-typed and screened for disease-associated alleles segregating in Siamese cats at 3 loci (ASB, GLB1, and CEP290). Siamese founders were normal at all loci except ASB, at which both animals carried the hypomorpic allele. Current stock is being managed to limit production of glaucomatous cats with brown, dilute, or Siamese phenotypes or homozygosity for the ASB hypomorphic allele. Genotyping will aid in the elimination of these alleles. The clinical effect of these phenotypes and alleles on the glaucoma phenotype is uncertain, but their elimination will remove potentially confounding effects. In conclusion, when founding a colony, stock should be selected or screened to limit potentially confounding phenotypes. When studying the immune, nervous, and visual systems, screening stock for alleles known to be associated with coat color may be warranted.


Asunto(s)
Cruzamiento , Gatos/genética , Modelos Animales de Enfermedad , Glaucoma/genética , Ciencia de los Animales de Laboratorio , Proteínas Adaptadoras Transductoras de Señales/genética , Alelos , Animales , Femenino , Efecto Fundador , Genotipo , Glaucoma/patología , Masculino , Glicoproteínas de Membrana/genética , Monofenol Monooxigenasa/genética , Linaje , Fenotipo
10.
Mol Ther ; 19(2): 251-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21139569

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Terapia Genética/métodos , Mucopolisacaridosis III/terapia , Mucopolisacaridosis I/terapia , Acetilglucosaminidasa/genética , Animales , Encéfalo/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Reacción en Cadena de la Polimerasa
11.
Sci Transl Med ; 2(60): 60ra89, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21123810

RESUMEN

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.


Asunto(s)
Terapia Enzimática , Iduronidasa/administración & dosificación , Iduronidasa/uso terapéutico , Mucopolisacaridosis I/terapia , Animales , Animales Recién Nacidos , Huesos/patología , Encéfalo/metabolismo , Encéfalo/patología , Perros , Glicosaminoglicanos/metabolismo , Humanos , Iduronidasa/genética , Articulaciones/patología , Lisosomas/metabolismo , Mucopolisacaridosis I/patología , Mucopolisacaridosis I/fisiopatología , Distribución Tisular
12.
Hum Mol Genet ; 12(9): 961-73, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12700165

RESUMEN

Mucopolysaccharidosis VII (MPS VII, Sly syndrome) is an autosomal recessive lysosomal storage disease caused by beta-glucuronidase (GUS) deficiency. A naturally occurring mouse model of that disease has been very useful for studying experimental approaches to therapy. However, immune responses can complicate evaluation of the long-term benefits of enzyme replacement or gene therapy delivered to adult MPS VII mice. To make this model useful for studying the long-term effectiveness and side effects of experimental therapies delivered to adult mice, we developed a new MPS VII mouse model, which is tolerant to both human and murine GUS. To achieve this, we used homologous recombination to introduce simultaneously a human cDNA transgene expressing inactive human GUS into intron 9 of the murine Gus gene and a targeted active site mutation (E536A) into the adjacent exon 10. When the heterozygote products of germline transmission were bred to homozygosity, the homozygous mice expressed no GUS enzyme activity but expressed inactive human GUS protein highly and were tolerant to immune challenge with human enzyme. Expression of the mutant murine Gus gene was reduced to about 10% of normal levels, but the inactive murine GUS enzyme also conferred tolerance to murine GUS. This MPS VII mouse model should be useful to evaluate therapeutic responses in adult mice receiving repetitive doses of enzyme or mice receiving gene therapy as adults. Heterozygotes expressed only 9.5-26% of wild-type levels of murine GUS instead of the expected 50%, indicating a dominant-negative effect of the mutant enzyme monomers on the activity of GUS tetramers in different tissues. Corrective gene therapy in this model should provide high enough levels of expression of normal GUS monomers to overcome the dominant negative effect of mutant monomers on newly synthesized GUS tetramers in most tissues.


Asunto(s)
Modelos Animales de Enfermedad , Glucuronidasa/genética , Glucuronidasa/inmunología , Mucopolisacaridosis VII/genética , Animales , Glucuronidasa/deficiencia , Humanos , Ratones , Ratones Transgénicos , Mucopolisacaridosis VII/metabolismo , Mucopolisacaridosis VII/patología , Mutación Missense
13.
Proc Natl Acad Sci U S A ; 99(23): 14982-7, 2002 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-12403825

RESUMEN

Human mucopolysaccharidosis VII (MPS VII, Sly syndrome) results from a deficiency of beta-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 (gus(mps/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.


Asunto(s)
Glucuronidasa/genética , Mucopolisacaridosis VII/genética , Mutación Missense , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Hígado/patología , Lisosomas/enzimología , Ratones , Mucopolisacaridosis VII/enzimología , Mucopolisacaridosis VII/patología , Mutagénesis Sitio-Dirigida , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA