Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Sci Rep ; 14(1): 8391, 2024 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-38600238

RESUMO

Friedreich's ataxia is a degenerative and progressive multisystem disorder caused by mutations in the highly conserved frataxin (FXN) gene that results in FXN protein deficiency and mitochondrial dysfunction. While gene therapy approaches are promising, consistent induction of therapeutic FXN protein expression that is sub-toxic has proven challenging, and numerous therapeutic approaches are being tested in animal models. FXN (hFXN in humans, mFXN in mice) is proteolytically modified in mitochondria to produce mature FXN. However, unlike endogenous hFXN, endogenous mFXN is further processed into N-terminally truncated, extra-mitochondrial mFXN forms of unknown function. This study assessed mature exogenous hFXN expression levels in the heart and liver of C57Bl/6 mice 7-10 months after intravenous administration of a recombinant adeno-associated virus encoding hFXN (AAVrh.10hFXN) and examined the potential for hFXN truncation in mice. AAVrh.10hFXN induced dose-dependent expression of hFXN in the heart and liver. Interestingly, hFXN was processed into truncated forms, but found at lower levels than mature hFXN. However, the truncations were at different positions than mFXN. AAVrh.10hFXN induced mature hFXN expression in mouse heart and liver at levels that approximated endogenous mFXN levels. These results suggest that AAVrh.10hFXN can likely induce expression of therapeutic levels of mature hFXN in mice.


Assuntos
Frataxina , Ataxia de Friedreich , Humanos , Animais , Camundongos , Coração , Processamento de Proteína Pós-Traducional , Fígado/metabolismo , Terapia Genética , Proteínas de Ligação ao Ferro/metabolismo , Ataxia de Friedreich/terapia , Ataxia de Friedreich/tratamento farmacológico
2.
Commun Biol ; 6(1): 1093, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37891254

RESUMO

Deficiency in human mature frataxin (hFXN-M) protein is responsible for the devastating neurodegenerative and cardiodegenerative disease of Friedreich's ataxia (FRDA). It results primarily through epigenetic silencing of the FXN gene by GAA triplet repeats on intron 1 of both alleles. GAA repeat lengths are most commonly between 600 and 1200 but can reach 1700. A subset of approximately 3% of FRDA patients have GAA repeats on one allele and a mutation on the other. FRDA patients die most commonly in their 30s from heart disease. Therefore, increasing expression of heart hFXN-M using gene therapy offers a way to prevent early mortality in FRDA. We used rhesus macaque monkeys to test the pharmacology of an adeno-associated virus (AAV)hu68.CB7.hFXN therapy. The advantage of using non-human primates for hFXN-M gene therapy studies is that hFXN-M and monkey FXN-M (mFXN-M) are 98.5% identical, which limits potential immunologic side-effects. However, this presented a formidable bioanalytical challenge in quantification of proteins with almost identical sequences. This could be overcome by the development of a species-specific quantitative mass spectrometry-based method, which has revealed for the first time, robust transgene-specific human protein expression in monkey heart tissue. The dose response is non-linear resulting in a ten-fold increase in monkey heart hFXN-M protein expression with only a three-fold increase in dose of the vector.


Assuntos
Ataxia de Friedreich , Proteínas de Ligação ao Ferro , Animais , Humanos , Macaca mulatta , Proteínas de Ligação ao Ferro/genética , Coração , Ataxia de Friedreich/genética , Ataxia de Friedreich/terapia , Ataxia de Friedreich/metabolismo , Terapia Genética , Frataxina
3.
Hum Gene Ther ; 34(13-14): 605-615, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37166361

RESUMO

Friedreich's ataxia (FA) is a life-threatening autosomal recessive disorder characterized by neurological and cardiac dysfunction. Arrhythmias and heart failure are the main cause of premature death. From prior studies in murine models of FA, adeno-associated virus encoding the normal human frataxin gene (AAVrh.10hFXN) effectively treated the cardiac manifestations of the disease. However, the therapeutic dose window is limited by high level of human frataxin (hFXN) gene expression associated with toxicity. As a therapeutic goal, since FA heterozygotes have no clinical manifestations of FA, we estimated the level of frataxin (FXN) necessary to convert the heart of a homozygote to that of a heterozygote. In noncardiac cells, FA heterozygotes have 30-80% of normal FXN levels (17.7-47.2 ng/mg, average 32.5 ng/mg) and FA homozygotes 2-30% normal levels (1.2-17.7 ng/mg, average 9.4 ng/mg). Therefore, an AAV vector would need to augment endogenous in an FA homozygote by >8.3 ng/mg. To determine the required dose of AAVrh.10hFXN, we administered 1.8 × 1011, 5.7 × 1011, or 1.8 × 1012 gc/kg of AAVrh.10hFXN intravenously (IV) to muscle creatine kinase (mck)-Cre conditional knockout Fxn mice, a cardiac and skeletal FXN knockout model. The minimally effective dose was 5.7 × 1011 gc/kg, resulting in cardiac hFXN levels of 6.1 ± 4.2 ng/mg and a mild (p < 0.01 compared with phosphate-buffered saline controls) improvement in mortality. A dose of 1.8 × 1012 gc/kg resulted in cardiac hFXN levels of 33.7 ± 6.4 ng/mg, a significant improvement in ejection fraction and fractional shortening (p < 0.05, both comparisons) and a 21.5% improvement in mortality (p < 0.001). To determine if the significantly effective dose of 1.8 × 1012 gc/kg could achieve human FA heterozygote levels in a large animal, this dose was administered IV to nonhuman primates. After 12 weeks, the vector-expressed FXN in the heart was 17.8 ± 4.9 ng/mg, comparable to the target human levels. These data identify both minimally and significantly effective therapeutic doses that are clinically relevant for the treatment of the cardiac manifestations of FA.


Assuntos
Ataxia de Friedreich , Insuficiência Cardíaca , Humanos , Camundongos , Animais , Ataxia de Friedreich/genética , Ataxia de Friedreich/terapia , Coração , Proteínas de Ligação ao Ferro/genética , Camundongos Knockout
4.
J Neurol ; 270(1): 208-222, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36152050

RESUMO

This narrative review aims at providing an update on the management of inherited cerebellar ataxias (ICAs), describing main clinical entities, genetic analysis strategies and recent therapeutic developments. Initial approach facing a patient with cerebellar ataxia requires family medical history, physical examination, exclusions of acquired causes and genetic analysis, including Next-Generation Sequencing (NGS). To guide diagnosis, several algorithms and a new genetic nomenclature for recessive cerebellar ataxias have been proposed. The challenge of NGS analysis is the identification of causative variant, trio analysis being usually the most appropriate option. Public genomic databases as well as pathogenicity prediction software facilitate the interpretation of NGS results. We also report on key clinical points for the diagnosis of the main ICAs, including Friedreich ataxia, CANVAS, polyglutamine spinocerebellar ataxias, Fragile X-associated tremor/ataxia syndrome. Rarer forms should not be neglected because of diagnostic biomarkers availability, disease-modifying treatments, or associated susceptibility to malignancy. Diagnostic difficulties arise from allelic and phenotypic heterogeneity as well as from the possibility for one gene to be associated with both dominant and recessive inheritance. To complicate the phenotype, cerebellar cognitive affective syndrome can be associated with some subtypes of cerebellar ataxia. Lastly, we describe new therapeutic leads: antisense oligonucleotides approach in polyglutamine SCAs and viral gene therapy in Friedreich ataxia. This review provides support for diagnosis, genetic counseling and therapeutic management of ICAs in clinical practice.


Assuntos
Ataxia Cerebelar , Ataxia de Friedreich , Ataxias Espinocerebelares , Humanos , Ataxia Cerebelar/diagnóstico , Ataxia Cerebelar/genética , Ataxia Cerebelar/terapia , Ataxia de Friedreich/diagnóstico , Ataxia de Friedreich/genética , Ataxia de Friedreich/terapia , Mutação , Ataxia/genética , Ataxias Espinocerebelares/genética
5.
Stem Cells Transl Med ; 10(8): 1157-1169, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33734599

RESUMO

Friedreich ataxia (FRDA) is an autosomal recessive disease characterized by degeneration of dorsal root ganglia (DRG) sensory neurons, which is due to low levels of the mitochondrial protein Frataxin. To explore cell replacement therapies as a possible approach to treat FRDA, we examined transplantation of sensory neural progenitors derived from human embryonic stem cells (hESC) and FRDA induced pluripotent stem cells (iPSC) into adult rodent DRG regions. Our data showed survival and differentiation of hESC and FRDA iPSC-derived progenitors in the DRG 2 and 8 weeks post-transplantation, respectively. Donor cells expressed neuronal markers, including sensory and glial markers, demonstrating differentiation to these lineages. These results are novel and a highly significant first step in showing the possibility of using stem cells as a cell replacement therapy to treat DRG neurodegeneration in FRDA as well as other peripheral neuropathies.


Assuntos
Ataxia de Friedreich , Células-Tronco Pluripotentes Induzidas , Doenças do Sistema Nervoso Periférico , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/terapia , Gânglios Espinais , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Receptoras Sensoriais
6.
Hum Mol Genet ; 28(8): 1274-1285, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30544254

RESUMO

Friedreich ataxia (FA) is currently an incurable inherited mitochondrial neurodegenerative disease caused by reduced levels of frataxin. Cardiac failure constitutes the main cause of premature death in FA. While adeno-associated virus-mediated cardiac gene therapy was shown to fully reverse the cardiac and mitochondrial phenotype in mouse models, this was achieved at high dose of vector resulting in the transduction of almost all cardiomyocytes, a dose and biodistribution that is unlikely to be replicated in clinic. The purpose of this study was to define the minimum vector biodistribution corresponding to the therapeutic threshold, at different stages of the disease progression. Correlative analysis of vector cardiac biodistribution, survival, cardiac function and biochemical hallmarks of the disease revealed that full rescue of the cardiac function was achieved when only half of the cardiomyocytes were transduced. In addition, meaningful therapeutic effect was achieved with as little as 30% transduction coverage. This therapeutic effect was mediated through cell-autonomous mechanisms for mitochondria homeostasis, although a significant increase in survival of uncorrected neighboring cells was observed. Overall, this study identifies the biodistribution thresholds and the underlying mechanisms conditioning the success of cardiac gene therapy in Friedreich ataxia and provides guidelines for the development of the clinical administration paradigm.


Assuntos
Cardiomiopatias/metabolismo , Ataxia de Friedreich/fisiopatologia , Miócitos Cardíacos/fisiologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Ataxia de Friedreich/terapia , Terapia Genética/métodos , Humanos , Proteínas de Ligação ao Ferro/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Distribuição Tecidual , Frataxina
7.
Mol Ther ; 26(8): 1940-1952, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29853274

RESUMO

Friedreich ataxia (FA) is a rare mitochondrial disease characterized by sensory and spinocerebellar ataxia, hypertrophic cardiomyopathy, and diabetes, for which there is no treatment. FA is caused by reduced levels of frataxin (FXN), an essential mitochondrial protein involved in the biosynthesis of iron-sulfur (Fe-S) clusters. Despite significant progress in recent years, to date, there are no good models to explore and test therapeutic approaches to stop or reverse the ganglionopathy and the sensory neuropathy associated to frataxin deficiency. Here, we report a new conditional mouse model with complete frataxin deletion in parvalbumin-positive cells that recapitulate the sensory ataxia and neuropathy associated to FA, albeit with a more rapid and severe course. Interestingly, although fully dysfunctional, proprioceptive neurons can survive for many weeks without frataxin. Furthermore, we demonstrate that post-symptomatic delivery of frataxin-expressing AAV allows for rapid and complete rescue of the sensory neuropathy associated with frataxin deficiency, thus establishing the pre-clinical proof of concept for the potential of gene therapy in treating FA neuropathy.


Assuntos
Ataxia de Friedreich/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Proteínas de Ligação ao Ferro/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Ataxia de Friedreich/genética , Humanos , Camundongos , Camundongos Knockout , Resultado do Tratamento , Frataxina
8.
Sci Transl Med ; 9(413)2017 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-29070698

RESUMO

Friedreich's ataxia (FRDA) is an incurable autosomal recessive neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin due to an intronic GAA-repeat expansion in the FXN gene. We report the therapeutic efficacy of transplanting wild-type mouse hematopoietic stem and progenitor cells (HSPCs) into the YG8R mouse model of FRDA. In the HSPC-transplanted YG8R mice, development of muscle weakness and locomotor deficits was abrogated as was degeneration of large sensory neurons in the dorsal root ganglia (DRGs) and mitochondrial capacity was improved in brain, skeletal muscle, and heart. Transplanted HSPCs engrafted and then differentiated into microglia in the brain and spinal cord and into macrophages in the DRGs, heart, and muscle of YG8R FRDA mice. We observed the transfer of wild-type frataxin and Cox8 mitochondrial proteins from HSPC-derived microglia/macrophages to FRDA mouse neurons and muscle myocytes in vivo. Our results show the HSPC-mediated phenotypic rescue of FRDA in YG8R mice and suggest that this approach should be investigated further as a strategy for treating FRDA.


Assuntos
Ataxia de Friedreich/terapia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Animais , Comportamento Animal , Diferenciação Celular , Modelos Animais de Doenças , Fibroblastos/metabolismo , Ataxia de Friedreich/patologia , Ataxia de Friedreich/fisiopatologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Locomoção , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Sistema Nervoso/patologia , Fagocitose , Células Receptoras Sensoriais/patologia , Frataxina
9.
J Gene Med ; 19(11): 376-386, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29044877

RESUMO

BACKGROUND: Friedreich's ataxia (FA) is an autosomal recessive neurodegenerative disease caused by mutations in the frataxin gene (FXN), which lead to reduced levels of the essential mitochondrial protein frataxin. Currently, there is no effective cure. METHODS: With the aim of developing a gene therapy for FA neuropathology, we describe the construction and preliminary characterization of a high-capacity nonreplicative genomic herpes simplex virus type 1 vector (H24B-FXNlac vector) carrying a reduced version of the human FXN genomic locus, comprising the 5-kb promoter and the FXN cDNA with the inclusion of intron 1. RESULTS: We show that the transgene cassette contains the elements necessary to preserve physiological neuronal regulation of human FXN expression. Transduction of cultured fetal rat dorsal root ganglia neurons with the H24B-FXNlac vector results in sustained expression of human FXN transcripts and frataxin protein. Rat footpad inoculation with the H24B-FXNlac vector results in human FXN transgene delivery to the dorsal root ganglia, with expression persisting for at least 1 month. CONCLUSIONS: The results of the present study support the feasibility of using this vector for sustained neuronal expression of human frataxin for FA gene therapy.


Assuntos
Gânglios Espinais/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Proteínas de Ligação ao Ferro/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/terapia , Gânglios Espinais/citologia , Terapia Genética/métodos , Células HEK293 , Humanos , Proteínas de Ligação ao Ferro/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo , Transgenes/genética , Células Vero , Frataxina
10.
Gene Ther ; 23(7): 606-14, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27082765

RESUMO

Artificially designed transcription activator-like effector (TALE) proteins fused to a transcription activation domain (TAD), such as VP64, are able to activate specific eukaryotic promoters. They thus provide a good tool for targeted gene regulation as a therapy. However, the efficacy of such an agent in vivo remains to be demonstrated as the majority of studies have been carried out in cell culture. We produced an adeno-associated virus 9 (AAV9) coding for a TALEfrat#8 containing 13 repeat variable diresidues able to bind to the proximal promoter of human frataxin (FXN) gene. This TALEfrat#8 was fused with a 3XFLAG at its N terminal and a VP64 TAD at its C terminal, and driven by a CAG promoter. This AAV9_3XFLAG-TALEfrat#8-VP64 was injected intraperitoneally to 9-day-old and 4-month-old YG8R mice. After 1 month, the heart, muscle and liver were removed and their FXN mRNA and FXN protein were analyzed. The results show that the AAV9_3XFLAG-TALEfrat#8-VP64 increased the FXN mRNA and FXN protein in the three organs studied. These results corroborate our previous in vitro studies in the FRDA human fibroblasts. Our study indicates that an AAV coding for a TALE protein coupled with a TAD may be used to increase gene expression in vivo as a possible treatment not only for FRDA but also for other haploinsufficiency diseases.


Assuntos
Dependovirus/genética , Ataxia de Friedreich/terapia , Terapia Genética/métodos , Proteínas de Ligação ao Ferro/genética , Animais , Células Cultivadas , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Humanos , Injeções Intraperitoneais , Proteínas de Ligação ao Ferro/metabolismo , Camundongos , Especificidade de Órgãos , Regiões Promotoras Genéticas , Frataxina
12.
Int J Cardiol ; 203: 964-71, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26625322

RESUMO

BACKGROUND: Friedreich's ataxia (FRDA), a recessive neurodegenerative disorder commonly associated with hypertrophic cardiomyopathy, is caused by silencing of the frataxin (FXN) gene encoding the mitochondrial protein involved in iron-sulfur cluster biosynthesis. METHODS: Application of our previously established FRDA human induced pluripotent stem cell (hiPSC) derived cardiomyocytes model as a platform to assess the efficacy of treatment with either the antioxidant coenzyme Q10 analog, idebenone (IDE) or the iron chelator, deferiprone (DFP), which are both under clinical trial. RESULTS: DFP was able to more significantly suppress synthesis of reactive oxygen species (ROS) than IDE at the dosages of 25 µM and 10nM respectively which agreed with the reduced rate of intracellular accumulation of iron by DFP treatment from 25 to 50 µM. With regard to cardiac electrical-contraction (EC) coupling function, decay velocity of calcium handling kinetics in FRDA-hiPSC-cardiomyocytes was significantly improved by DFP treatment but not by IDE. Further mechanistic studies revealed that DFP also modulated iron induced mitochondrial stress as reflected by mitochondria network disorganization and decline level of respiratory chain protein, succinate dehydrogenase (CxII) and cytochrome c oxidase (COXIV). In addition, iron-response protein (IRP-1) regulatory loop was overridden by DFP as reflected by resumed level of ferritin (FTH) back to basal level and the attenuated transferrin receptor (TSFR) mRNA level suppression thereby reducing further iron uptake. CONCLUSIONS: DFP modulated iron homeostasis in FRDA-hiPSC-cardiomyocytes and effectively relieved stress-stimulation related to cardiomyopathy. The resuming of redox condition led to the significantly improved cardiac prime events, cardiac electrical-coupling during contraction.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Ataxia de Friedreich/terapia , Células-Tronco Pluripotentes Induzidas , Ferro/metabolismo , Miócitos Cardíacos/metabolismo , Piridonas/farmacologia , Ubiquinona/análogos & derivados , Antioxidantes/farmacologia , Deferiprona , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Regulação da Expressão Gênica , Homeostase , Humanos , Quelantes de Ferro/farmacologia , Proteínas de Ligação ao Ferro/biossíntese , Proteínas de Ligação ao Ferro/genética , Miócitos Cardíacos/patologia , Estresse Oxidativo , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquinona/farmacologia , Frataxina
13.
Mol Ther ; 23(1): 130-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25070719

RESUMO

The main objective of this work is to demonstrate the feasibility of using bone marrow-derived stem cells in treating a neurodegenerative disorder such as Friedreich's ataxia. In this disease, the dorsal root ganglia of the spinal cord are the first to degenerate. Two groups of mice were injected intrathecally with mesenchymal stem cells isolated from either wild-type or Fxntm1Mkn/Tg(FXN)YG8Pook (YG8) mice. As a result, both groups presented improved motor skills compared to nontreated mice. Also, frataxin expression was increased in the dorsal root ganglia of the treated groups, along with lower expression of the apoptotic markers analyzed. Furthermore, the injected stem cells expressed the trophic factors NT3, NT4, and BDNF, which bind to sensory neurons of the dorsal root ganglia and increase their survival. The expression of antioxidant enzymes indicated that the stem cell-treated mice presented higher levels of catalase and GPX-1, which are downregulated in the YG8 mice. There were no significant differences in the use of stem cells isolated from wild-type and YG8 mice. In conclusion, bone marrow mesenchymal stem cell transplantation, both autologous and allogeneic, is a feasible therapeutic option to consider in delaying the neurodegeneration observed in the dorsal root ganglia of Friedreich's ataxia patients.


Assuntos
Ataxia de Friedreich/terapia , Gânglios Espinais/patologia , Proteínas de Ligação ao Ferro/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Catalase/genética , Catalase/metabolismo , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/patologia , Gânglios Espinais/metabolismo , Expressão Gênica , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Injeções Espinhais , Proteínas de Ligação ao Ferro/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Atividade Motora , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neurotrofina 3/genética , Neurotrofina 3/metabolismo , Transplante Autólogo , Transplante Homólogo , Glutationa Peroxidase GPX1 , Frataxina
14.
Med Hypotheses ; 83(3): 296-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24962209

RESUMO

We advance the overarching hypothesis that stem cell therapy is a potent treatment for Friedreich's ataxia (FRDA). Here, we discuss the feasibility of autologous transplantation in FRDA, highlighting the need for the successful isolation of the FRDA patient's bone marrow-derived mesenchymal stem cells, followed by characterization that these cells maintain the GAA repeat expansion and the reduced FXN mRNA expression, both hallmark features of FRDA. Next, we discuss the need for assessment of the proliferative capability and pluripotency of FRDA patient's bone marrow-derived mesenchymal stem cells. In particular, we view the need for characterizing the in vitro differentiation of bone marrow-derived mesenchymal stem cells into the two cell types primarily affected in FRDA, peripheral neurons and cardiomyocytes. Finally, we discuss the need to test the application of bone marrow-derived mesenchymal stem cells as potent autologous donor cells for FRDA. The demonstration of the functional correction of the mutated gene in these cells will be a critical endpoint of determining the potential of stem cell therapy in FRDA. We envision a gene-based cell transplant strategy as a likely therapeutic approach for FRDA, involving stable insertion of functional human bacterial artificial chromosomes or BACs containing the intact FXN gene into stem cells, thereafter leading to the expression of frataxin protein in differentiated neurons/cardiomyocytes.


Assuntos
Ataxia de Friedreich/fisiopatologia , Ataxia de Friedreich/terapia , Terapia Genética/métodos , Transplante de Células-Tronco , Transplante Autólogo , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Proteínas de Ligação ao Ferro/metabolismo , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/citologia , Neurônios/metabolismo , Expansão das Repetições de Trinucleotídeos , Frataxina
15.
Hum Gene Ther ; 25(8): 684-93, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24749505

RESUMO

Neurodegenerative disorders such as Friedreich ataxia (FRDA) present significant challenges in developing effective therapeutic intervention. Current treatments aim to manage symptoms and thus improve quality of life, but none can cure, nor are proven to slow, the neurodegeneration inherent to this disease. The primary clinical features of FRDA include progressive ataxia and shortened life span, with complications of cardiomyopathy being the major cause of death. FRDA is most commonly caused by an expanded GAA trinucleotide repeat in the first intron of FXN that leads to reduced levels of frataxin, a mitochondrial protein important for iron metabolism. The GAA expansion in FRDA does not alter the coding sequence of FXN. It results in reduced production of structurally normal frataxin, and hence any increase in protein level is expected to be therapeutically beneficial. Recently, there has been increased interest in developing novel therapeutic applications like cell and/or gene therapies, and these cutting-edge applications could provide effective treatment options for FRDA. Importantly, since individuals with FRDA produce frataxin at low levels, increased expression should not elicit an immune response. Here we review the advances to date and highlight the future potential for cell and gene therapy to treat this debilitating disease.


Assuntos
Ataxia de Friedreich/terapia , Terapia Genética , Transplante de Células-Tronco , Animais , Células da Medula Óssea/fisiologia , Vetores Genéticos , Humanos , Transdução Genética
16.
Nat Med ; 20(5): 542-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24705334

RESUMO

Cardiac failure is the most common cause of mortality in Friedreich's ataxia (FRDA), a mitochondrial disease characterized by neurodegeneration, hypertrophic cardiomyopathy and diabetes. FRDA is caused by reduced levels of frataxin (FXN), an essential mitochondrial protein involved in the biosynthesis of iron-sulfur (Fe-S) clusters. Impaired mitochondrial oxidative phosphorylation, bioenergetics imbalance, deficit of Fe-S cluster enzymes and mitochondrial iron overload occur in the myocardium of individuals with FRDA. No treatment exists as yet for FRDA cardiomyopathy. A conditional mouse model with complete frataxin deletion in cardiac and skeletal muscle (Mck-Cre-Fxn(L3/L-) mice) recapitulates most features of FRDA cardiomyopathy, albeit with a more rapid and severe course. Here we show that adeno-associated virus rh10 vector expressing human FXN injected intravenously in these mice fully prevented the onset of cardiac disease. Moreover, later administration of the frataxin-expressing vector, after the onset of heart failure, was able to completely reverse the cardiomyopathy of these mice at the functional, cellular and molecular levels within a few days. Our results demonstrate that cardiomyocytes with severe energy failure and ultrastructure disorganization can be rapidly rescued and remodeled by gene therapy and establish the preclinical proof of concept for the potential of gene therapy in treating FRDA cardiomyopathy.


Assuntos
Cardiomiopatias/terapia , Ataxia de Friedreich/genética , Ataxia de Friedreich/terapia , Proteínas de Ligação ao Ferro/genética , Animais , Cardiomiopatias/genética , Cardiomiopatias/patologia , Modelos Animais de Doenças , Ataxia de Friedreich/patologia , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Proteínas de Ligação ao Ferro/administração & dosagem , Proteínas Ferro-Enxofre/biossíntese , Camundongos , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Miócitos Cardíacos/patologia , Frataxina
17.
J Bone Joint Surg Br ; 94(5): 684-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22529092

RESUMO

We reviewed 31 consecutive patients with Friedreich's ataxia and scoliosis. There were 24 males and seven females with a mean age at presentation of 15.5 years (8.6 to 30.8) and a mean curve of 51° (13° to 140°). A total of 12 patients had thoracic curvatures, 11 had thoracolumbar and eight had double thoracic/lumbar. Two patients had long thoracolumbar collapsing scoliosis with pelvic obliquity and four had hyperkyphosis. Left-sided thoracic curves in nine patients (45%) and increased thoracic kyphosis differentiated these deformities from adolescent idiopathic scoliosis. There were 17 patients who underwent a posterior instrumented spinal fusion at mean age of 13.35 years, which achieved and maintained good correction of the deformity. Post-operative complications included one death due to cardiorespiratory failure, one revision to address nonunion and four patients with proximal junctional kyphosis who did not need extension of the fusion. There were no neurological complications and no wound infections. The rate of progression of the scoliosis in children kept under simple observation and those treated with bracing was less for lumbar curves during bracing and similar for thoracic curves. The scoliosis progressed in seven of nine children initially treated with a brace who later required surgery. Two patients presented after skeletal maturity with balanced curves not requiring correction. Three patients with severe deformities who would benefit from corrective surgery had significant cardiac co-morbidities.


Assuntos
Ataxia de Friedreich/complicações , Escoliose/etiologia , Adolescente , Adulto , Braquetes , Criança , Contraindicações , Progressão da Doença , Feminino , Seguimentos , Ataxia de Friedreich/diagnóstico por imagem , Ataxia de Friedreich/terapia , Humanos , Vértebras Lombares/cirurgia , Masculino , Radiografia , Escoliose/diagnóstico por imagem , Escoliose/terapia , Fusão Vertebral/efeitos adversos , Fusão Vertebral/métodos , Vértebras Torácicas/cirurgia , Resultado do Tratamento , Adulto Jovem
18.
Hum Mol Genet ; 20(14): 2807-22, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21531789

RESUMO

Friedreich's ataxia (FRDA) is an autosomal recessive disease caused by mutations that produce a deficiency in frataxin. Despite the importance of neurodegeneration in FRDA, little is known about the consequences of frataxin deficiency in neuronal cells. Here we describe a neuronal cell model for FRDA based on the use of lentiviral vectors that carry minigenes encoding frataxin-specific shRNAs that silence the expression of this gene. These lentivectors can knockdown frataxin expression in human neuroblastoma SH-SY5Y cells, which results in large-scale cell death in differentiated neuron-like cells but not in undifferentiated neuroblastoma cells. Frataxin-deficient neuron-like cells appear to die through apoptosis that is accompanied by up-regulation of p53, PUMA and Bax and activation of caspase-3. No significant autophagy is observed in frataxin-deficient neuron-like cells and the pharmacological activation of autophagy does not significantly increase neuronal cell death in response to the frataxin deficiency. Cell death triggered by frataxin knockdown can be impaired by interference with p53, caspase inhibitors and gene transfer of FXN. These results suggest that frataxin gene silencing in human neuron-like cells may constitute a useful cell model to characterize the molecular changes triggered by frataxin deficiency in neurons, as well as to search for therapies that may protect against neurodegeneration.


Assuntos
Apoptose , Inativação Gênica , Proteínas de Ligação ao Ferro/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/terapia , Humanos , Proteínas de Ligação ao Ferro/genética , Modelos Biológicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , Frataxina
19.
Gene Ther ; 18(10): 1015-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21490681

RESUMO

Novel gene-based therapies for disease will depend in many cases on long-term persistent transgene expression. To develop gene therapy strategies for Friedreich's ataxia (FRDA), we have examined the persistence of transgene expression in the brain in vivo provided by the entire 135 kb FXN genomic DNA locus delivered as an infectious bacterial artificial chromosome (iBAC) herpes simplex virus type 1 (HSV-1)-based vector injected in the adult mouse cerebellum. We constructed genomic DNA-reporter fusion vectors carrying a complete 135 kb FXN genomic locus with an insertion of the Escherichia coli lacZ gene at the ATG start codon (iBAC-FXN-lacZ). SHSY5Y human neuroblastoma cells transduced by iBAC-FXN-lacZ showed high efficiency of vector delivery and LacZ expression. Direct intracranial injection of iBAC-FXN-lacZ into the adult mouse cerebellum resulted in a large number of easily detectable transduced cells, with LacZ expression driven by the FXN genomic locus, which persisted for at least 75 days. Green fluorescent protein expression driven from the same vector but by the strong HSV-1 IE4/5 promoter was transient. Our data demonstrate for the first time sustained transgene expression in vivo by infectious delivery of a genomic DNA locus >100 kb in size. Such an approach may be suitable for gene rescue strategies in neurological disease, such as FRDA.


Assuntos
Cerebelo , Ataxia de Friedreich/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Proteínas de Ligação ao Ferro/administração & dosagem , Animais , Células Cultivadas , Cromossomos Artificiais Bacterianos/genética , Primers do DNA/genética , Escherichia coli , Ataxia de Friedreich/genética , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Herpesvirus Humano 1 , Imuno-Histoquímica , Proteínas de Ligação ao Ferro/genética , Óperon Lac/genética , Camundongos , Microscopia de Fluorescência , Transgenes/genética , Frataxina
20.
Artigo em Inglês | MEDLINE | ID: mdl-20862363

RESUMO

Artificial chromosomes and minichromosome-like episomes are large DNA molecules capable of containing whole genomic loci, and be maintained as nonintegrating, replicating molecules in proliferating human somatic cells. Authentic human artificial chromosomes are very difficult to engineer because of the difficulties associated with centromere structure, so they are not widely used for gene-therapy applications. However, OriP/EBNA1-based episomes, which they lack true centromeres, can be maintained stably in dividing cells as they bind to mitotic chromosomes and segregate into daughter cells. These episomes are more easily engineered than true human artificial chromosomes and can carry entire genes along with all their regulatory sequences. Thus, these constructs may facilitate the long-term persistence and physiological regulation of the expression of therapeutic genes, which is crucial for some gene therapy applications. In particular, they are promising vectors for gene therapy in inherited diseases that are caused by recessive mutations, for example haemophilia A and Friedreich's ataxia. Interestingly, the episome carrying the frataxin gene (deficient in Friedreich's ataxia) has been demonstrated to rescue the susceptibility to oxidative stress which is typical of fibroblasts from Friedreich's ataxia patients. This provides evidence of their potential to treat genetic diseases linked to recessive mutations through gene therapy.


Assuntos
Cromossomos Artificiais Humanos/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Expressão Gênica , Terapia Genética/métodos , Plasmídeos/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ataxia de Friedreich/terapia , Hemofilia A/terapia , Herpesvirus Humano 4/genética , Humanos , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/uso terapêutico , Frataxina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA