Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Hum Mol Genet ; 26(8): 1535-1551, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28334745

RESUMO

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.


Assuntos
Terapia Genética , Doenças por Armazenamento dos Lisossomos/terapia , Mucopolissacaridose III/terapia , Sulfatases/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Humanos , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/patologia , Camundongos , Mucopolissacaridose III/genética , Mucopolissacaridose III/patologia , Fenótipo , Sulfatases/administração & dosagem
2.
Hum Mol Genet ; 24(7): 2078-95, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25524704

RESUMO

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.


Assuntos
Acetilglucosaminidase/genética , Terapia Genética/métodos , Mucopolissacaridose III/genética , Mucopolissacaridose III/terapia , Acetilglucosaminidase/líquido cefalorraquidiano , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Dependovirus/genética , Dependovirus/metabolismo , Feminino , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mucopolissacaridose III/líquido cefalorraquidiano , Mucopolissacaridose III/enzimologia
3.
Nature ; 475(7355): 217-21, 2011 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-21706032

RESUMO

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Assuntos
Reparo do DNA/genética , Modelos Animais de Doenças , Marcação de Genes/métodos , Terapia Genética/métodos , Genoma/genética , Hemofilia B/genética , Hemostasia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Endonucleases/química , Endonucleases/genética , Endonucleases/metabolismo , Éxons/genética , Fator IX/análise , Fator IX/genética , Vetores Genéticos/genética , Células HEK293 , Hemofilia B/fisiopatologia , Humanos , Íntrons/genética , Fígado/metabolismo , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Fenótipo , Homologia de Sequência , Dedos de Zinco
4.
J Biol Chem ; 290(27): 16772-85, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25971976

RESUMO

The human insulin-like growth factor 2 (IGF2) and insulin genes are located within the same genomic region. Although human genomic studies have demonstrated associations between diabetes and the insulin/IGF2 locus or the IGF2 mRNA-binding protein 2 (IGF2BP2), the role of IGF2 in diabetes pathogenesis is not fully understood. We previously described that transgenic mice overexpressing IGF2 specifically in ß-cells (Tg-IGF2) develop a pre-diabetic state. Here, we characterized the effects of IGF2 on ß-cell functionality. Overexpression of IGF2 led to ß-cell dedifferentiation and endoplasmic reticulum stress causing islet dysfunction in vivo. Both adenovirus-mediated overexpression of IGF2 and treatment of adult wild-type islets with recombinant IGF2 in vitro further confirmed the direct implication of IGF2 on ß-cell dysfunction. Treatment of Tg-IGF2 mice with subdiabetogenic doses of streptozotocin or crossing these mice with a transgenic model of islet lymphocytic infiltration promoted the development of overt diabetes, suggesting that IGF2 makes islets more susceptible to ß-cell damage and immune attack. These results indicate that increased local levels of IGF2 in pancreatic islets may predispose to the onset of diabetes. This study unravels an unprecedented role of IGF2 on ß-cells function.


Assuntos
Diabetes Mellitus/genética , Fator de Crescimento Insulin-Like II/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Animais , Desdiferenciação Celular , Linhagem Celular Tumoral , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Humanos , Insulina/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos
5.
Blood ; 122(19): 3283-7, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24085764

RESUMO

Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.


Assuntos
Endonucleases/genética , Fator IX/biossíntese , Terapia Genética/métodos , Vetores Genéticos , Genoma , Hemofilia B/terapia , Dedos de Zinco/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Endonucleases/metabolismo , Fator IX/genética , Fator IX/metabolismo , Hemofilia B/genética , Hemofilia B/patologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Multimerização Proteica
6.
J Biol Chem ; 288(24): 17631-42, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23620587

RESUMO

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.


Assuntos
Gliose/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Doenças Neurodegenerativas/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Doenças Retinianas/metabolismo , Células Amácrinas/metabolismo , Células Amácrinas/patologia , Animais , Apoptose , Citocinas/metabolismo , Modelos Animais de Doenças , Eletrorretinografia , Ácido Glutâmico/metabolismo , Fator de Crescimento Insulin-Like I/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Doenças Neurodegenerativas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo , Células Fotorreceptoras de Vertebrados/patologia , Retina/metabolismo , Retina/patologia , Retina/fisiopatologia , Transdução de Sinais , Técnicas de Cultura de Tecidos , Transcriptoma
7.
Mol Ther ; 21(9): 1727-37, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23857231

RESUMO

Immune responses directed against viral capsid proteins constitute a main safety concern in the use of adeno-associated virus (AAV) as gene transfer vectors in humans. Pharmacological immunosuppression has been proposed as a solution to the problem; however, the approach suffers from several potential limitations. Using MHC class II epitopes initially identified within human IgG, named Tregitopes, we showed that it is possible to modulate CD8+ T cell responses to several viral antigens in vitro. We showed that incubation of peripheral blood mononuclear cells with these epitopes triggers proliferation of CD4+CD25+FoxP3+ T cells that suppress killing of target cells loaded with MHC class I antigens in an antigen-specific fashion, through a mechanism that seems to require cell-to-cell contact. Expression of a construct encoding for the AAV capsid structural protein fused to Tregitopes resulted in reduction of CD8+ T cell reactivity against the AAV capsid following immunization with an adenoviral vector expressing capsid. This was accompanied by an increase in frequency of CD4+CD25+FoxP3+ T cells in spleens and lower levels of inflammatory infiltrates in injected tissues. This proof-of-concept study demonstrates modulation of CD8+ T cell reactivity to an antigen using regulatory T cell epitopes is possible.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos , Imunoglobulina G/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Capsídeo/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Células Cultivadas , Dependovirus/genética , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Terapia Genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulina G/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Linfócitos T Reguladores/imunologia
8.
Mol Ther ; 20(2): 254-66, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22008915

RESUMO

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.


Assuntos
Sistema Nervoso Central/patologia , Terapia Genética , Hidrolases/genética , Fígado/metabolismo , Mucopolissacaridose III/terapia , Animais , Cerebelo/ultraestrutura , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/farmacocinética , Hidrolases/metabolismo , Injeções Intramusculares , Injeções Intravenosas , Fígado/ultraestrutura , Lisossomos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucopolissacaridose III/genética , Mucopolissacaridose III/mortalidade , Músculo Esquelético/metabolismo , Análise de Sobrevida , Transdução Genética , Córtex Visual/patologia , Córtex Visual/ultraestrutura
9.
Blood ; 115(23): 4678-88, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20335222

RESUMO

Muscle represents an important tissue target for adeno-associated viral (AAV) vector-mediated gene transfer of the factor IX (FIX) gene in hemophilia B (HB) subjects with advanced liver disease. Previous studies of direct intramuscular administration of an AAV-FIX vector in humans showed limited efficacy. Here we adapted an intravascular delivery system of AAV vectors encoding the FIX transgene to skeletal muscle of HB dogs. The procedure, performed under transient immunosuppression (IS), resulted in widespread transduction of muscle and sustained, dose-dependent therapeutic levels of canine FIX transgene up to 10-fold higher than those obtained by intramuscular delivery. Correction of bleeding time correlated clinically with a dramatic reduction of spontaneous bleeding episodes. None of the dogs (n = 14) receiving the AAV vector under transient IS developed inhibitory antibodies to canine FIX; transient inhibitor was detected after vector delivery without IS. The use of AAV serotypes with high tropism for muscle and low susceptibility to anti-AAV2 antibodies allowed for efficient vector administration in naive dogs and in the presence of low- but not high-titer anti-AAV2 antibodies. Collectively, these results demonstrate the feasibility of this approach for treatment of HB and highlight the importance of IS to prevent immune responses to the FIX transgene product.


Assuntos
Dependovirus , Fator IX/biossíntese , Terapia Genética , Vetores Genéticos , Hemofilia B/terapia , Terapia de Imunossupressão , Músculo Esquelético , Animais , Anticorpos Antivirais/sangue , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Cães , Fator IX/genética , Hemofilia B/sangue , Hemofilia B/genética , Hemorragia/sangue , Hemorragia/genética , Hemorragia/terapia , Humanos , Injeções Intramusculares , Transdução Genética
10.
Hepatology ; 52(6): 1877-87, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20931557

RESUMO

UNLABELLED: RNA interference (RNAi) is being evaluated as an alternative therapeutic strategy for hepatitis C virus (HCV) infection. The use of viral vectors encoding short hairpin RNAs (shRNAs) has been the most common strategy employed to provide sustained expression of RNAi effectors. However, overexpression and incomplete processing of shRNAs has led to saturation of the endogenous miRNA pathway, resulting in toxicity. The use of endogenous microRNAs (miRNAs) as scaffolds for short interfering (siRNAs) may avoid these problems, and miRNA clusters can be engineered to express multiple RNAi effectors, a feature that may prevent RNAi-resistant HCV mutant generation. We exploited the endogenous miRNA-17-92 cluster to generate a polycistronic primary miRNA that is processed into five mature miRNAs that target different regions of the HCV genome. All five anti-HCV miRNAs were active, achieving up to 97% inhibition of Renilla luciferase (RLuc) HCV reporter plasmids. Self-complementary recombinant adeno-associated virus (scAAV) vectors were chosen for therapeutic delivery of the miRNA cluster. Expression of the miRNAs from scAAV inhibited the replication of cell culture-propagated HCV (HCVcc) by 98%, and resulted in up to 93% gene silencing of RLuc-HCV reporter plasmids in mouse liver. No hepatocellular toxicity was observed at scAAV doses as high as 5 × 10(11) vector genomes per mouse, a dose that is approximately five-fold higher than doses of scAAV-shRNA vectors that others have shown previously to be toxic in mouse liver. CONCLUSION: We have demonstrated that exogenous anti-HCV miRNAs induce gene silencing, and when expressed from scAAV vectors inhibit the replication of HCVcc without inducing toxicity. The combination of an AAV vector delivery system and exploitation of the endogenous RNAi pathway is a potentially viable alternative to current HCV treatment regimens.


Assuntos
Hepacivirus/fisiologia , Hepatite C/terapia , MicroRNAs/uso terapêutico , Interferência de RNA , Animais , Linhagem Celular Tumoral , Dependovirus/genética , Vetores Genéticos , Humanos , Masculino , Camundongos , MicroRNAs/genética , Replicação Viral/efeitos dos fármacos
11.
Mol Ther ; 18(7): 1318-29, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20424599

RESUMO

Muscle represents an attractive target tissue for adeno-associated virus (AAV) vector-mediated gene transfer for hemophilia B (HB). Experience with direct intramuscular (i.m.) administration of AAV vectors in humans showed that the approach is safe but fails to achieve therapeutic efficacy. Here, we present a careful evaluation of the safety profile (vector, transgene, and administration procedure) of peripheral transvenular administration of AAV-canine factor IX (cFIX) vectors to the muscle of HB dogs. Vector administration resulted in sustained therapeutic levels of cFIX expression. Although all animals developed a robust antibody response to the AAV capsid, no T-cell responses to the capsid antigen were detected by interferon (IFN)-gamma enzyme-linked immunosorbent spot (ELISpot). Interleukin (IL)-10 ELISpot screening of lymphocytes showed reactivity to cFIX-derived peptides, and restimulation of T cells in vitro in the presence of the identified cFIX epitopes resulted in the expansion of CD4(+)FoxP3(+)IL-10(+) T-cells. Vector administration was not associated with systemic inflammation, and vector spread to nontarget tissues was minimal. At the local level, limited levels of cell infiltrates were detected when the vector was administered intravascularly. In summary, this study in a large animal model of HB demonstrates that therapeutic levels of gene transfer can be safely achieved using a novel route of intravascular gene transfer to muscle.


Assuntos
Dependovirus/genética , Fator IX/genética , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Hemofilia B/terapia , Músculo Esquelético/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Cães , Fator IX/metabolismo , Citometria de Fluxo , Hemofilia B/metabolismo , Humanos , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Músculo Esquelético/patologia
12.
Mol Ther Methods Clin Dev ; 23: 370-389, 2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34761052

RESUMO

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.

13.
J Biol Chem ; 284(34): 22961-9, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19473988

RESUMO

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.


Assuntos
Barreira Hematorretiniana/metabolismo , Barreira Hematorretiniana/fisiopatologia , Fator de Crescimento Insulin-Like I/fisiologia , Retina/metabolismo , Idoso de 80 Anos ou mais , Animais , Barreira Hematorretiniana/patologia , Western Blotting , Bovinos , Feminino , Gliose/metabolismo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Retina/patologia
14.
Hum Gene Ther ; 30(10): 1211-1221, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31482754

RESUMO

For most lysosomal storage diseases (LSDs), there is no cure. Gene therapy is an attractive tool for treatment of LSDs caused by deficiencies in secretable lysosomal enzymes, in which neither full restoration of normal enzymatic activity nor transduction of all cells of the affected organ is necessary. However, some LSDs, such as mucopolysaccharidosis type III (MPSIII) diseases or Sanfilippo syndrome, represent a difficult challenge because patients suffer severe neurodegeneration with mild somatic alterations. The disease's main target is the central nervous system (CNS) and enzymes do not efficiently cross the blood-brain barrier (BBB) even if present at very high concentration in circulation. No specific treatment has been approved for MPSIII. In this study, we discuss the adeno-associated virus (AAV) vector-mediated gene transfer strategies currently being developed for MPSIII disease. These strategies rely on local delivery of AAV vectors to the CNS either through direct intraparenchymal injection at several sites or through delivery to the cerebrospinal fluid (CSF), which bathes the whole CNS, or exploit the properties of certain AAV serotypes capable of crossing the BBB upon systemic administration. Although studies in small and large animal models of MPSIII diseases have provided evidence supporting the efficacy and safety of all these strategies, there are considerable differences between the different routes of administration in terms of procedure-associated risks, vector dose requirements, sensitivity to the effect of circulating neutralizing antibodies that block AAV transduction, and potential toxicity. Ongoing clinical studies should shed light on which gene transfer strategy leads to highest clinical benefits while minimizing risks. The development of all these strategies opens a new horizon for treatment of not only MPSIII and other LSDs but also of a wide range of neurological diseases.


Assuntos
Encéfalo/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Mucopolissacaridose III/terapia , Animais , Anticorpos Neutralizantes/biossíntese , Barreira Hematoencefálica/metabolismo , Encéfalo/patologia , Ensaios Clínicos como Assunto , Dependovirus/metabolismo , Modelos Animais de Doenças , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Injeções Intralesionais , Injeções Intravenosas , Lentivirus/genética , Lentivirus/metabolismo , Mucopolissacaridose III/líquido cefalorraquidiano , Mucopolissacaridose III/genética , Mucopolissacaridose III/patologia
15.
EMBO Mol Med ; 10(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29987000

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Fatores de Crescimento de Fibroblastos/genética , Terapia Genética , Resistência à Insulina , Obesidade/terapia , Adipócitos/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal , Diabetes Mellitus Tipo 2/genética , Dieta Hiperlipídica , Metabolismo Energético , Fígado Gorduroso/terapia , Fatores de Crescimento de Fibroblastos/metabolismo , Fibrose/terapia , Técnicas de Transferência de Genes , Hiperplasia/terapia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Músculo Esquelético/metabolismo , Obesidade/genética , Pancreatite/terapia
16.
J Clin Invest ; 113(8): 1149-57, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15085194

RESUMO

IGF-1 has been associated with the pathogenesis of diabetic retinopathy, although its role is not fully understood. Here we show that normoglycemic/normoinsulinemic transgenic mice overexpressing IGF-1 in the retina developed most alterations seen in human diabetic eye disease. A paracrine effect of IGF-1 in the retina initiated vascular alterations that progressed from nonproliferative to proliferative retinopathy and retinal detachment. Eyes from 2-month-old transgenic mice showed loss of pericytes and thickening of basement membrane of retinal capillaries. In mice 6 months and older, venule dilatation, intraretinal microvascular abnormalities, and neovascularization of the retina and vitreous cavity were observed. Neovascularization was consistent with increased IGF-1 induction of VEGF expression in retinal glial cells. In addition, IGF-1 accumulated in aqueous humor, which may have caused rubeosis iridis and subsequently adhesions between the cornea and iris that hampered aqueous humor drainage and led to neovascular glaucoma. Furthermore, all transgenic mice developed cataracts. These findings suggest a role of IGF-1 in the development of ocular complications in long-term diabetes. Thus, these transgenic mice may be used to study the mechanisms that lead to diabetes eye disease and constitute an appropriate model in which to assay new therapies.


Assuntos
Retinopatia Diabética/etiologia , Fator de Crescimento Insulin-Like I/fisiologia , Retina/metabolismo , Animais , Catarata/etiologia , Glaucoma Neovascular/etiologia , Proteína Glial Fibrilar Ácida/análise , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/análise
17.
Mol Metab ; 6(7): 664-680, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28702323

RESUMO

OBJECTIVE: Type 1 diabetes is characterized by autoimmune destruction of ß-cells leading to severe insulin deficiency. Although many improvements have been made in recent years, exogenous insulin therapy is still imperfect; new therapeutic approaches, focusing on preserving/expanding ß-cell mass and/or blocking the autoimmune process that destroys islets, should be developed. The main objective of this work was to test in non-obese diabetic (NOD) mice, which spontaneously develop autoimmune diabetes, the effects of local expression of Insulin-like growth factor 1 (IGF1), a potent mitogenic and pro-survival factor for ß-cells with immunomodulatory properties. METHODS: Transgenic NOD mice overexpressing IGF1 specifically in ß-cells (NOD-IGF1) were generated and phenotyped. In addition, miRT-containing, IGF1-encoding adeno-associated viruses (AAV) of serotype 8 (AAV8-IGF1-dmiRT) were produced and administered to 4- or 11-week-old non-transgenic NOD females through intraductal delivery. Several histological, immunological, and metabolic parameters were measured to monitor disease over a period of 28-30 weeks. RESULTS: In transgenic mice, local IGF1 expression led to long-term suppression of diabetes onset and robust protection of ß-cell mass from the autoimmune insult. AAV-mediated pancreatic-specific overexpression of IGF1 in adult animals also dramatically reduced diabetes incidence, both when vectors were delivered before pathology onset or once insulitis was established. Transgenic NOD-IGF1 and AAV8-IGF1-dmiRT-treated NOD animals had much less islet infiltration than controls, preserved ß-cell mass, and normal insulinemia. Transgenic and AAV-treated islets showed less expression of antigen-presenting molecules, inflammatory cytokines, and chemokines important for tissue-specific homing of effector T cells, suggesting IGF1 modulated islet autoimmunity in NOD mice. CONCLUSIONS: Local expression of Igf1 by AAV-mediated gene transfer counteracts progression to diabetes in NOD mice. This study suggests a therapeutic strategy for autoimmune diabetes in humans.


Assuntos
Diabetes Mellitus Tipo 1/genética , Fator de Crescimento Insulin-Like I/genética , Células Secretoras de Insulina/metabolismo , Animais , Células Cultivadas , Dependovirus/genética , Diabetes Mellitus Tipo 1/terapia , Feminino , Terapia Genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Endogâmicos NOD
18.
Mol Ther Methods Clin Dev ; 6: 1-7, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28626777

RESUMO

Diabetes is a complex metabolic disease that exposes patients to the deleterious effects of hyperglycemia on various organs. Achievement of normoglycemia with exogenous insulin treatment requires the use of high doses of hormone, which increases the risk of life-threatening hypoglycemic episodes. We developed a gene therapy approach to control diabetic hyperglycemia based on co-expression of the insulin and glucokinase genes in skeletal muscle. Previous studies proved the feasibility of gene delivery to large diabetic animals with adeno-associated viral (AAV) vectors. Here, we report the long-term (∼8 years) follow-up after a single administration of therapeutic vectors to diabetic dogs. Successful, multi-year control of glycemia was achieved without the need of supplementation with exogenous insulin. Metabolic correction was demonstrated through normalization of serum levels of fructosamine, triglycerides, and cholesterol and remarkable improvement in the response to an oral glucose challenge. The persistence of vector genomes and therapeutic transgene expression years after vector delivery was documented in multiple samples from treated muscles, which showed normal morphology. Thus, this study demonstrates the long-term efficacy and safety of insulin and glucokinase gene transfer in large animals and especially the ability of the system to respond to the changes in metabolic needs as animals grow older.

19.
JCI Insight ; 1(9): e86696, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27699273

RESUMO

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.


Assuntos
Terapia Genética , Iduronato Sulfatase/genética , Mucopolissacaridose II/terapia , Animais , Dependovirus , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL
20.
Dis Model Mech ; 9(9): 999-1013, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27491071

RESUMO

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.


Assuntos
Progressão da Doença , Mucopolissacaridose III/patologia , Acetiltransferases/deficiência , Acetiltransferases/metabolismo , Animais , Comportamento Animal , Encéfalo/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Glicosaminoglicanos/metabolismo , Homeostase , Humanos , Inflamação/patologia , Longevidade , Lisossomos/metabolismo , Lisossomos/patologia , Lisossomos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Mucopolissacaridose III/enzimologia , Especificidade de Órgãos , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA