Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Eat Weight Disord ; 27(8): 3245-3256, 2022 Dec.
Article En | MEDLINE | ID: mdl-35902481

PURPOSE: Weight Bias Internalization (WBI) is pervasive and potentially damaging for health. Little is known about WBI in youth. As negative effects of WBI have been observed when controlling for BMI, measures that allow WBI to be assessed across different weight categories are needed. The Modified Weight Bias Internalization Scale (WBIS-M) is one of the most frequently used scales in this field. Our purpose was to obtain a Spanish validated version of the WBIS-M for adolescents across different weight statuses. METHODS: The data were collected from 298 secondary students (mean age 14.31; 48.32% girls; 18.8% were overweight and 6.4% had obesity). Internal structure was examined by a cross-validation analysis, using both exploratory and confirmatory factor analyses in different subsamples. RESULTS: Item 1 showed a psychometric anomalous functioning and was deleted. The one-factor structure of the 10-item version was confirmed with adequate fit ([EFA (KMO = 0.915, χ2(55) = 1075.633, p < 0.0001)]; [CFA (χ2(35) = 200.515; GFI = 0.995; PGFI = 0.992; NFI = 0.991; SRMR = 0.060)]). Internal consistency was high [Formula: see text] ω = 0.93). Significant correlations with the same set of external variables assessed in the original version (anti-fat bias, self-esteem, mood, body dissatisfaction, drive for thinness, binge eating), all of them correlates of WBI in adolescents, were found. Girls and participants with obesity obtained higher scores. CONCLUSION: The results provide support for the validity and reliability of our WBIS-M version for use with adolescents across weight categories in Spain. LEVEL OF EVIDENCE: Level III, evidence obtained from well-designed cohort studies.


Body Image , Weight Prejudice , Female , Humans , Adolescent , Male , Reproducibility of Results , Surveys and Questionnaires , Obesity , Psychometrics
2.
Mol Ther Methods Clin Dev ; 23: 370-389, 2021 Dec 10.
Article En | MEDLINE | ID: mdl-34761052

Delivery of adeno-associated viral vectors (AAVs) to cerebrospinal fluid (CSF) has emerged as a promising approach to achieve widespread transduction of the central nervous system (CNS) and peripheral nervous system (PNS), with direct applicability to the treatment of a wide range of neurological diseases, particularly lysosomal storage diseases. Although studies in small animal models have provided proof of concept and experiments in large animals demonstrated feasibility in bigger brains, there is not much information on long-term safety or durability of the effect. Here, we report a 7-year study in healthy beagle dogs after intra-CSF delivery of a single, clinically relevant dose (2 × 1013 vg/dog) of AAV9 vectors carrying the canine sulfamidase, the enzyme deficient in mucopolysaccharidosis type IIIA. Periodic monitoring of CSF and blood, clinical and neurological evaluations, and magnetic resonance and ultrasound imaging of target organs demonstrated no toxicity related to treatment. AAV9-mediated gene transfer resulted in detection of sulfamidase activity in CSF throughout the study. Analysis at tissue level showed widespread sulfamidase expression and activity in the absence of histological findings in any region of encephalon, spinal cord, or dorsal root ganglia. Altogether, these results provide proof of durability of expression and long-term safety for intra-CSF delivery of AAV-based gene transfer vectors encoding therapeutic proteins to the CNS.

3.
Nat Commun ; 12(1): 5343, 2021 09 09.
Article En | MEDLINE | ID: mdl-34504088

Mucopolysaccharidosis type IVA (MPSIVA) or Morquio A disease, a lysosomal storage disorder, is caused by N-acetylgalactosamine-6-sulfate sulfatase (GALNS) deficiency, resulting in keratan sulfate (KS) and chondroitin-6-sulfate accumulation. Patients develop severe skeletal dysplasia, early cartilage deterioration and life-threatening heart and tracheal complications. There is no cure and enzyme replacement therapy cannot correct skeletal abnormalities. Here, using CRISPR/Cas9 technology, we generate the first MPSIVA rat model recapitulating all skeletal and non-skeletal alterations experienced by patients. Treatment of MPSIVA rats with adeno-associated viral vector serotype 9 encoding Galns (AAV9-Galns) results in widespread transduction of bones, cartilage and peripheral tissues. This led to long-term (1 year) increase of GALNS activity and whole-body correction of KS levels, thus preventing body size reduction and severe alterations of bones, teeth, joints, trachea and heart. This study demonstrates the potential of AAV9-Galns gene therapy to correct the disabling MPSIVA pathology, providing strong rationale for future clinical translation to MPSIVA patients.


Chondroitinsulfatases/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy/methods , Mucopolysaccharidosis IV/therapy , Musculoskeletal System/metabolism , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/ultrastructure , Chondroitinsulfatases/deficiency , Chondroitinsulfatases/metabolism , Gene Expression Regulation, Enzymologic , Genetic Vectors/genetics , Humans , Male , Microscopy, Electron, Transmission , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/genetics , Musculoskeletal System/pathology , Musculoskeletal System/ultrastructure , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
4.
EMBO Mol Med ; 10(8)2018 08.
Article En | MEDLINE | ID: mdl-29987000

Prevalence of type 2 diabetes (T2D) and obesity is increasing worldwide. Currently available therapies are not suited for all patients in the heterogeneous obese/T2D population, hence the need for novel treatments. Fibroblast growth factor 21 (FGF21) is considered a promising therapeutic agent for T2D/obesity. Native FGF21 has, however, poor pharmacokinetic properties, making gene therapy an attractive strategy to achieve sustained circulating levels of this protein. Here, adeno-associated viral vectors (AAV) were used to genetically engineer liver, adipose tissue, or skeletal muscle to secrete FGF21. Treatment of animals under long-term high-fat diet feeding or of ob/ob mice resulted in marked reductions in body weight, adipose tissue hypertrophy and inflammation, hepatic steatosis, inflammation and fibrosis, and insulin resistance for > 1 year. This therapeutic effect was achieved in the absence of side effects despite continuously elevated serum FGF21. Furthermore, FGF21 overproduction in healthy animals fed a standard diet prevented the increase in weight and insulin resistance associated with aging. Our study underscores the potential of FGF21 gene therapy to treat obesity, insulin resistance, and T2D.


Diabetes Mellitus, Type 2/therapy , Fibroblast Growth Factors/genetics , Genetic Therapy , Insulin Resistance , Obesity/therapy , Adipocytes/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Body Weight , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat , Energy Metabolism , Fatty Liver/therapy , Fibroblast Growth Factors/metabolism , Fibrosis/therapy , Gene Transfer Techniques , Hyperplasia/therapy , Liver/metabolism , Liver/pathology , Male , Mice , Muscle, Skeletal/metabolism , Obesity/genetics , Pancreatitis/therapy
5.
Hum Mol Genet ; 26(8): 1535-1551, 2017 04 15.
Article En | MEDLINE | ID: mdl-28334745

Gene therapy is a promising therapeutic alternative for Lysosomal Storage Disorders (LSD), as it is not necessary to correct the genetic defect in all cells of an organ to achieve therapeutically significant levels of enzyme in body fluids, from which non-transduced cells can uptake the protein correcting their enzymatic deficiency. Animal models are instrumental in the development of new treatments for LSD. Here we report the generation of the first mouse model of the LSD Muccopolysaccharidosis Type IIID (MPSIIID), also known as Sanfilippo syndrome type D. This autosomic recessive, heparan sulphate storage disease is caused by deficiency in N-acetylglucosamine 6-sulfatase (GNS). Mice deficient in GNS showed lysosomal storage pathology and loss of lysosomal homeostasis in the CNS and peripheral tissues, chronic widespread neuroinflammation, reduced locomotor and exploratory activity and shortened lifespan, a phenotype that closely resembled human MPSIIID. Moreover, treatment of the GNS-deficient animals with GNS-encoding adeno-associated viral (AAV) vectors of serotype 9 delivered to the cerebrospinal fluid completely corrected pathological storage, improved lysosomal functionality in the CNS and somatic tissues, resolved neuroinflammation, restored normal behaviour and extended lifespan of treated mice. Hence, this work represents the first step towards the development of a treatment for MPSIIID.


Genetic Therapy , Lysosomal Storage Diseases/therapy , Mucopolysaccharidosis III/therapy , Sulfatases/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Mice , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Phenotype , Sulfatases/administration & dosage
6.
JCI Insight ; 1(9): e86696, 2016 06 16.
Article En | MEDLINE | ID: mdl-27699273

Mucopolysaccharidosis type II (MPSII) is an X-linked lysosomal storage disease characterized by severe neurologic and somatic disease caused by deficiency of iduronate-2-sulfatase (IDS), an enzyme that catabolizes the glycosaminoglycans heparan and dermatan sulphate. Intravenous enzyme replacement therapy (ERT) currently constitutes the only approved therapeutic option for MPSII. However, the inability of recombinant IDS to efficiently cross the blood-brain barrier (BBB) limits ERT efficacy in treating neurological symptoms. Here, we report a gene therapy approach for MPSII through direct delivery of vectors to the CNS. Through a minimally invasive procedure, we administered adeno-associated virus vectors encoding IDS (AAV9-Ids) to the cerebrospinal fluid of MPSII mice with already established disease. Treated mice showed a significant increase in IDS activity throughout the encephalon, with full resolution of lysosomal storage lesions, reversal of lysosomal dysfunction, normalization of brain transcriptomic signature, and disappearance of neuroinflammation. Moreover, our vector also transduced the liver, providing a peripheral source of therapeutic protein that corrected storage pathology in visceral organs, with evidence of cross-correction of nontransduced organs by circulating enzyme. Importantly, AAV9-Ids-treated MPSII mice showed normalization of behavioral deficits and considerably prolonged survival. These results provide a strong proof of concept for the clinical translation of our approach for the treatment of Hunter syndrome patients with cognitive impairment.


Genetic Therapy , Iduronate Sulfatase/genetics , Mucopolysaccharidosis II/therapy , Animals , Dependovirus , Disease Models, Animal , Female , Genetic Vectors , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL
7.
Dis Model Mech ; 9(9): 999-1013, 2016 09 01.
Article En | MEDLINE | ID: mdl-27491071

Mucopolysaccharidosis type IIIC (MPSIIIC) is a severe lysosomal storage disease caused by deficiency in activity of the transmembrane enzyme heparan-α-glucosaminide N-acetyltransferase (HGSNAT) that catalyses the N-acetylation of α-glucosamine residues of heparan sulfate. Enzyme deficiency causes abnormal substrate accumulation in lysosomes, leading to progressive and severe neurodegeneration, somatic pathology and early death. There is no cure for MPSIIIC, and development of new therapies is challenging because of the unfeasibility of cross-correction. In this study, we generated a new mouse model of MPSIIIC by targeted disruption of the Hgsnat gene. Successful targeting left LacZ expression under control of the Hgsnat promoter, allowing investigation into sites of endogenous expression, which was particularly prominent in the CNS, but was also detectable in peripheral organs. Signs of CNS storage pathology, including glycosaminoglycan accumulation, lysosomal distension, lysosomal dysfunction and neuroinflammation were detected in 2-month-old animals and progressed with age. Glycosaminoglycan accumulation and ultrastructural changes were also observed in most somatic organs, but lysosomal pathology seemed most severe in liver. Furthermore, HGSNAT-deficient mice had altered locomotor and exploratory activity and shortened lifespan. Hence, this animal model recapitulates human MPSIIIC and provides a useful tool for the study of disease physiopathology and the development of new therapeutic approaches.


Disease Progression , Mucopolysaccharidosis III/pathology , Acetyltransferases/deficiency , Acetyltransferases/metabolism , Animals , Behavior, Animal , Brain/enzymology , Brain/pathology , Disease Models, Animal , Glycosaminoglycans/metabolism , Homeostasis , Humans , Inflammation/pathology , Longevity , Lysosomes/metabolism , Lysosomes/pathology , Lysosomes/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Mucopolysaccharidosis III/enzymology , Organ Specificity , Survival Analysis
8.
Hum Mol Genet ; 24(7): 2078-95, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25524704

Gene therapy is an attractive tool for the treatment of monogenic disorders, in particular for lysosomal storage diseases (LSD) caused by deficiencies in secretable lysosomal enzymes in which neither full restoration of normal enzymatic activity nor transduction of all affected cells are necessary. However, some LSD such as Mucopolysaccharidosis Type IIIB (MPSIIIB) are challenging because the disease's main target organ is the brain and enzymes do not efficiently cross the blood-brain barrier even if present at very high concentration in circulation. To overcome these limitations, we delivered AAV9 vectors encoding for α-N-acetylglucosaminidase (NAGLU) to the Cerebrospinal Fluid (CSF) of MPSIIIB mice with the disease already detectable at biochemical, histological and functional level. Restoration of enzymatic activity in Central Nervous System (CNS) resulted in normalization of glycosaminoglycan content and lysosomal physiology, resolved neuroinflammation and restored the pattern of gene expression in brain similar to that of healthy animals. Additionally, transduction of the liver due to passage of vectors to the circulation led to whole-body disease correction. Treated animals also showed reversal of behavioural deficits and extended lifespan. Importantly, when the levels of enzymatic activity were monitored in the CSF of dogs following administration of canine NAGLU-coding vectors to animals that were either naïve or had pre-existing immunity against AAV9, similar levels of activity were achieved, suggesting that CNS efficacy would not be compromised in patients seropositive for AAV9. Our studies provide a strong rationale for the clinical development of this novel therapeutic approach as the treatment for MPSIIIB.


Acetylglucosaminidase/genetics , Genetic Therapy/methods , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/therapy , Acetylglucosaminidase/cerebrospinal fluid , Animals , Brain/metabolism , Brain/pathology , Dependovirus/genetics , Dependovirus/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis III/cerebrospinal fluid , Mucopolysaccharidosis III/enzymology
9.
Mol Ther Methods Clin Dev ; 1: 14039, 2014.
Article En | MEDLINE | ID: mdl-26015978

Nonalcoholic fatty liver disease (NAFLD) is the most common hepatic disease worldwide, and evidence suggests that it promotes insulin resistance and type 2 diabetes. Caloric restriction (CR) is the only available strategy for NAFLD treatment. The protein deacetylase Sirtuin1 (SIRT1), which is activated by CR, increases catabolic metabolism and decreases lipogenesis and inflammation, both involved in the development of NAFLD. Here we show that adeno-associated viral vectors of serotype 8 (AAV8)-mediated liver-specific Sirt1 gene transfer prevents the development of NAFLD induced by a high carbohydrate (HC) diet. Long-term hepatic SIRT1 overexpression led to upregulation of key hepatic genes involved in ß-oxidation, prevented HC diet-induced lipid accumulation and reduced liver inflammation. AAV8-Sirt1-treated mice showed improved insulin sensitivity, increased oxidative capacity in skeletal muscle and reduced white adipose tissue inflammation. Moreover, HC feeding induced leptin resistance, which was also attenuated in AAV8-Sirt1-treated mice. Therefore, AAV-mediated gene transfer to overexpress SIRT1 specifically in the liver may represent a new gene therapy strategy to counteract NAFLD and related diseases such as type 2 diabetes.

10.
Diabetes ; 62(12): 4012-22, 2013 Dec.
Article En | MEDLINE | ID: mdl-24043756

Adipose tissue is pivotal in the regulation of energy homeostasis through the balance of energy storage and expenditure and as an endocrine organ. An inadequate mass and/or alterations in the metabolic and endocrine functions of adipose tissue underlie the development of obesity, insulin resistance, and type 2 diabetes. To fully understand the metabolic and molecular mechanism(s) involved in adipose dysfunction, in vivo genetic modification of adipocytes holds great potential. Here, we demonstrate that adeno-associated viral (AAV) vectors, especially serotypes 8 and 9, mediated efficient transduction of white (WAT) and brown adipose tissue (BAT) in adult lean and obese diabetic mice. The use of short versions of the adipocyte protein 2 or uncoupling protein-1 promoters or micro-RNA target sequences enabled highly specific, long-term AAV-mediated transgene expression in white or brown adipocytes. As proof of concept, delivery of AAV vectors encoding for hexokinase or vascular endothelial growth factor to WAT or BAT resulted in increased glucose uptake or increased vessel density in targeted depots. This method of gene transfer also enabled the secretion of stable high levels of the alkaline phosphatase marker protein into the bloodstream by transduced WAT. Therefore, AAV-mediated genetic engineering of adipose tissue represents a useful tool for the study of adipose pathophysiology and, likely, for the future development of new therapeutic strategies for obesity and diabetes.


Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2/metabolism , Hyperglycemia/metabolism , Mitochondria/metabolism , Animals , Dependovirus , Diabetes Mellitus, Type 2/genetics , Energy Metabolism/genetics , Genetic Engineering , Hyperglycemia/genetics , Male , Mice , Mice, Inbred ICR , Mice, Inbred NOD , Mice, Obese , Mitochondria/genetics
11.
J Clin Invest ; 123(8): 3254-3271, 2013 08 01.
Article En | MEDLINE | ID: mdl-23863627

For most lysosomal storage diseases (LSDs) affecting the CNS, there is currently no cure. The BBB, which limits the bioavailability of drugs administered systemically, and the short half-life of lysosomal enzymes, hamper the development of effective therapies. Mucopolysaccharidosis type IIIA (MPS IIIA) is an autosomic recessive LSD caused by a deficiency in sulfamidase, a sulfatase involved in the stepwise degradation of glycosaminoglycan (GAG) heparan sulfate. Here, we demonstrate that intracerebrospinal fluid (intra-CSF) administration of serotype 9 adenoassociated viral vectors (AAV9s) encoding sulfamidase corrects both CNS and somatic pathology in MPS IIIA mice. Following vector administration, enzymatic activity increased throughout the brain and in serum, leading to whole body correction of GAG accumulation and lysosomal pathology, normalization of behavioral deficits, and prolonged survival. To test this strategy in a larger animal, we treated beagle dogs using intracisternal or intracerebroventricular delivery. Administration of sulfamidase-encoding AAV9 resulted in transgenic expression throughout the CNS and liver and increased sulfamidase activity in CSF. High-titer serum antibodies against AAV9 only partially blocked CSF-mediated gene transfer to the brains of dogs. Consistently, anti-AAV antibody titers were lower in CSF than in serum collected from healthy and MPS IIIA-affected children. These results support the clinical translation of this approach for the treatment of MPS IIIA and other LSDs with CNS involvement.

12.
J Biol Chem ; 288(24): 17631-42, 2013 Jun 14.
Article En | MEDLINE | ID: mdl-23620587

Insulin-like growth factor I (IGF-I) exerts multiple effects on different retinal cell types in both physiological and pathological conditions. Despite the growth factor's extensively described neuroprotective actions, transgenic mice with increased intraocular levels of IGF-I showed progressive impairment of electroretinographic amplitudes up to complete loss of response, with loss of photoreceptors and bipolar, ganglion, and amacrine neurons. Neurodegeneration was preceded by the overexpression of genes related to retinal stress, acute-phase response, and gliosis, suggesting that IGF-I altered normal retinal homeostasis. Indeed, gliosis and microgliosis were present from an early age in transgenic mice, before other alterations occurred, and were accompanied by signs of oxidative stress and impaired glutamate recycling. Older mice also showed overproduction of pro-inflammatory cytokines. Our results suggest that, when chronically increased, intraocular IGF-I is responsible for the induction of deleterious cellular processes that can lead to neurodegeneration, and they highlight the importance that this growth factor may have in the pathogenesis of conditions such as ischemic or diabetic retinopathy.


Gliosis/metabolism , Insulin-Like Growth Factor I/metabolism , Neurodegenerative Diseases/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Diseases/metabolism , Amacrine Cells/metabolism , Amacrine Cells/pathology , Animals , Apoptosis , Cytokines/metabolism , Disease Models, Animal , Electroretinography , Glutamic Acid/metabolism , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Neurodegenerative Diseases/pathology , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Photoreceptor Cells, Vertebrate/pathology , Retina/metabolism , Retina/pathology , Retina/physiopathology , Signal Transduction , Tissue Culture Techniques , Transcriptome
13.
Diabetes ; 62(2): 551-60, 2013 Feb.
Article En | MEDLINE | ID: mdl-23099863

In type 1 diabetes, loss of tolerance to ß-cell antigens results in T-cell-dependent autoimmune destruction of ß cells. The abrogation of autoreactive T-cell responses is a prerequisite to achieve long-lasting correction of the disease. The liver has unique immunomodulatory properties and hepatic gene transfer results in tolerance induction and suppression of autoimmune diseases, in part by regulatory T-cell (Treg) activation. Hence, the liver could be manipulated to treat or prevent diabetes onset through expression of key genes. IGF-I may be an immunomodulatory candidate because it prevents autoimmune diabetes when expressed in ß cells or subcutaneously injected. Here, we demonstrate that transient, plasmid-derived IGF-I expression in mouse liver suppressed autoimmune diabetes progression. Suppression was associated with decreased islet inflammation and ß-cell apoptosis, increased ß-cell replication, and normalized ß-cell mass. Permanent protection depended on exogenous IGF-I expression in liver nonparenchymal cells and was associated with increased percentage of intrapancreatic Tregs. Importantly, Treg depletion completely abolished IGF-I-mediated protection confirming the therapeutic potential of these cells in autoimmune diabetes. This study demonstrates that a nonviral gene therapy combining the immunological properties of the liver and IGF-I could be beneficial in the treatment of the disease.


Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Genetic Therapy , Insulin-Like Growth Factor I/genetics , Liver/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Division/genetics , Cell Division/immunology , Humans , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Liver/immunology , Mice , Mice, Transgenic , Pancreatitis/genetics , Pancreatitis/immunology , Plasmids/genetics
14.
Hum Gene Ther ; 23(12): 1237-46, 2012 Dec.
Article En | MEDLINE | ID: mdl-22909060

Mucopolysaccharidosis type IIIA (MPSIIIA) is a rare lysosomal storage disorder caused by mutations in the sulfamidase gene. Accumulation of glycosaminoglycan (GAG) inside the lysosomes is associated with severe neurodegeneration as well as peripheral organ pathological changes leading to death of affected individuals during adolescence. There is no cure for MPSIIIA. Due to the limitation of the blood-brain barrier, enzyme replacement therapy and gene therapy strategies attempted thus far have not achieved whole-body correction of the disease. After the systemic administration of an adeno-associated virus 9 (AAV9) vector encoding for sulfamidase under the control of a ubiquitous promoter, we were able to obtain widespread expression of the therapeutic transgene in brain and in peripheral organs, and sulfamidase activity in serum of both male and female MPSIIIA mice. This was accompanied by the normalization of GAG storage levels in most peripheral organs. In brain, decrease in GAG tissue content following AAV9 gene transfer of sulfamidase was associated with the resolution of neuroinflammation. Finally, correction of disease phenotype resulted in a remarkable prolongation of survival of both male and female AAV-treated MPSIIIA mice. This proof-of-concept study will be relevant to the future development of therapies for MPSIIIA.


Genetic Therapy/methods , Genetic Vectors/pharmacology , Glycosaminoglycans/metabolism , Hydrolases/genetics , Mucopolysaccharidosis III/therapy , Animals , Central Nervous System/metabolism , Dependovirus/genetics , Disease Models, Animal , Female , Gene Expression , Genetic Vectors/administration & dosage , Liver/metabolism , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Transgenes
15.
PLoS One ; 7(7): e41511, 2012.
Article En | MEDLINE | ID: mdl-22911805

Neovascularization associated with diabetic retinopathy (DR) and other ocular disorders is a leading cause of visual impairment and adult-onset blindness. Currently available treatments are merely palliative and offer temporary solutions. Here, we tested the efficacy of antiangiogenic gene transfer in an animal model that mimics the chronic progression of human DR. Adeno-associated viral (AAV) vectors of serotype 2 coding for antiangiogenic Pigment Epithelium Derived Factor (PEDF) were injected in the vitreous of a 1.5 month-old transgenic model of retinopathy that develops progressive neovascularization. A single intravitreal injection led to long-term production of PEDF and to a striking inhibition of intravitreal neovascularization, normalization of retinal capillary density, and prevention of retinal detachment. This was parallel to a reduction in the intraocular levels of Vascular Endothelial Growth Factor (VEGF). Normalization of VEGF was consistent with a downregulation of downstream effectors of angiogenesis, such as the activity of Matrix Metalloproteinases (MMP) 2 and 9 and the content of Connective Tissue Growth Factor (CTGF). These results demonstrate long-term efficacy of AAV-mediated PEDF overexpression in counteracting retinal neovascularization in a relevant animal model, and provides evidence towards the use of this strategy to treat angiogenesis in DR and other chronic proliferative retinal disorders.


Aging/pathology , Diabetic Retinopathy/pathology , Diabetic Retinopathy/prevention & control , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Retina/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/prevention & control , Serpins/metabolism , Animals , Cell Hypoxia , Dependovirus/genetics , Diabetic Retinopathy/metabolism , Disease Models, Animal , Down-Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin-Like Growth Factor I/metabolism , Intravitreal Injections , Mice , Mice, Transgenic , Retina/pathology , Retinal Detachment/metabolism , Retinal Detachment/pathology , Retinal Neovascularization/metabolism , Time Factors , Transduction, Genetic , Vascular Endothelial Growth Factor A/metabolism
16.
Mol Ther ; 20(2): 254-66, 2012 Feb.
Article En | MEDLINE | ID: mdl-22008915

Mucopolysaccharidosis type IIIA (MPSIIIA) is an inherited lysosomal storage disease caused by deficiency of sulfamidase, resulting in accumulation of the glycosaminoglycan (GAG) heparan sulfate. It is characterized by severe progressive neurodegeneration, together with somatic alterations, which lead to death during adolescence. Here, we tested the ability of adeno-associated virus (AAV) vector-mediated genetic modification of either skeletal muscle or liver to revert the already established disease phenotype of 2-month-old MPSIIIA males and females. Intramuscular administration of AAV-Sulfamidase failed to achieve significant therapeutic benefit in either gender. In contrast, AAV8-mediated liver-directed gene transfer achieved high and sustained levels of circulating active sulfamidase, which reached normal levels in females and was fourfold higher in males, and completely corrected lysosomal GAG accumulation in most somatic tissues. Remarkably, a 50% reduction of GAG accumulation was achieved throughout the entire brain of males, which correlated with a partial improvement of the pathology of cerebellum and cortex. Liver-directed gene transfer expanded the lifespan of MPSIIIA males, underscoring the importance of reaching supraphysiological plasma levels of enzyme for maximal therapeutic benefit. These results show how liver-directed gene transfer can reverse somatic and ameliorate neurological pathology in MPSIIIA.


Central Nervous System/pathology , Genetic Therapy , Hydrolases/genetics , Liver/metabolism , Mucopolysaccharidosis III/therapy , Animals , Cerebellum/ultrastructure , Dependovirus/genetics , Disease Models, Animal , Female , Gene Order , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/pharmacokinetics , Hydrolases/metabolism , Injections, Intramuscular , Injections, Intravenous , Liver/ultrastructure , Lysosomes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/mortality , Muscle, Skeletal/metabolism , Survival Analysis , Transduction, Genetic , Visual Cortex/pathology , Visual Cortex/ultrastructure
17.
J Biol Chem ; 284(34): 22961-9, 2009 Aug 21.
Article En | MEDLINE | ID: mdl-19473988

Blood-retinal barrier (BRB) breakdown is a key event in diabetic retinopathy and other ocular disorders that leads to increased retinal vascular permeability. This causes edema and tissue damage resulting in visual impairment. Insulin-like growth factor-I (IGF-I) is involved in these processes, although the relative contribution of increased systemic versus intraocular IGF-I remains controversial. Here, to elucidate the role of this factor in BRB breakdown, transgenic mice with either local or systemic elevations of IGF-I have been examined. High intraocular IGF-I, resulting from overexpression of IGF-I in the retina, increased IGF-I receptor content and signaling and led to accumulation of vascular endothelial growth factor. This was parallel to up-regulation of vascular Intercellular adhesion molecule I and retinal infiltration by bone marrow-derived microglial cells. These alterations resulted in increased vessel paracellular permeability to both low and high molecular weight compounds in IGF-I-overexpressing retinas and agreed with the loss of vascular tight junction integrity observed by electron microscopy and the altered junctional protein content. In contrast, mice with chronically elevated serum IGF-I did not show alterations in the retinal vasculature structure and permeability, indicating that circulating IGF-I cannot initiate BRB breakdown. Consistent with a key role of IGF-I signaling in retinal diseases, a strong up-regulation of the IGF-I receptor in human retinas with marked gliosis was also observed. Thus, this study demonstrates that intraocular IGF-I, but not systemic IGF-I, is sufficient to trigger processes leading to BRB breakdown and increased retinal vascular permeability. Therefore, therapeutic interventions designed to counteract local IGF-I effects may prove successful to prevent BRB disruption.


Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/physiopathology , Insulin-Like Growth Factor I/physiology , Retina/metabolism , Aged, 80 and over , Animals , Blood-Retinal Barrier/pathology , Blotting, Western , Cattle , Female , Gliosis/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Retina/pathology
...