Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Hum Genomics ; 18(1): 50, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778374

RESUMEN

Friedreich ataxia (FRDA) is a life-threatening hereditary ataxia; its incidence is 1:50,000 individuals in the Caucasian population. A unique therapeutic drug for FRDA, the antioxidant Omaveloxolone, has been recently approved by the US Food and Drug Administration (FDA). FRDA is a multi-systemic neurodegenerative disease; in addition to a progressive neurodegeneration, FRDA is characterized by hypertrophic cardiomyopathy, diabetes mellitus and musculoskeletal deformities. Cardiomyopathy is the predominant cause of premature death. The onset of FRDA typically occurs between the ages of 5 and 15. Given the complexity and heterogeneity of clinical features and the variability of their onset, the identification of biomarkers capable of assessing disease progression and monitoring the efficacy of treatments is essential to facilitate decision making in clinical practice. We conducted an RNA-seq analysis in peripheral blood mononuclear cells from FRDA patients and healthy donors, identifying a signature of small non-coding RNAs (sncRNAs) capable of distinguishing healthy individuals from the majority of FRDA patients. Among the differentially expressed sncRNAs, microRNAs are a class of small non-coding endogenous RNAs that regulate posttranscriptional silencing of target genes. In FRDA plasma samples, hsa-miR-148a-3p resulted significantly upregulated. The analysis of the Receiver Operating Characteristic (ROC) curve, combining the circulating expression levels of hsa-miR-148a-3p and hsa-miR-223-3p (previously identified by our group), revealed an Area Under the Curve (AUC) of 0.86 (95%, Confidence Interval 0.77-0.95; p-value < 0.0001). An in silico prediction analysis indicated that the IL6ST gene, an interesting marker of neuroinflammation in FRDA, is a common target gene of both miRNAs. Our findings support the evaluation of combined expression levels of different circulating miRNAs as potent epi-biomarkers in FRDA. Moreover, we found hsa-miR-148a-3p significantly over-expressed in Intermediate and Late-Onset Friedreich Ataxia patients' group (IOG and LOG, respectively) compared to healthy individuals, indicating it as a putative prognostic biomarker in this pathology.


Asunto(s)
Biomarcadores , Ataxia de Friedreich , MicroARNs , Humanos , Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Ataxia de Friedreich/sangre , MicroARNs/genética , MicroARNs/sangre , Masculino , Biomarcadores/sangre , Pronóstico , Femenino , Adulto , RNA-Seq , Adolescente , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Niño , Adulto Joven , Persona de Mediana Edad , Preescolar , Curva ROC , Estudios de Casos y Controles
2.
Int J Mol Sci ; 24(16)2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37628866

RESUMEN

Friedreich's ataxia (FRDA) is a rare monogenic disease characterized by multisystem, slowly progressive degeneration. Because of the genetic defect in a non-coding region of FXN gene, FRDA cells exhibit severe deficit of frataxin protein levels. Hence, FRDA pathophysiology is characterized by a plethora of metabolic disruptions related to iron metabolism, mitochondrial homeostasis and oxidative stress. Importantly, an impairment of the antioxidant defences exacerbates the oxidative damage. This appears closely associated with the disablement of key antioxidant proteins, such as the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and the mitochondrial superoxide dismutase (MnSOD). The cytokine interferon gamma (IFN-γ) has been shown to increase frataxin expression in FRDA cells and to improve functional deficits in FRDA mice. Currently, IFN-γ represents a potential therapy under clinical evaluation in FRDA patients. Here, we show that IFN-γ induces a rapid expression of Nrf2 and MnSOD in different cell types, including FRDA patient-derived fibroblasts. Our data indicate that IFN-γ signals two separate pathways to enhance Nrf2 and MnSOD levels in FRDA fibroblasts. MnSOD expression increased through an early transcriptional regulation, whereas the levels of Nrf2 are induced by a post-transcriptional mechanism. We demonstrate that the treatment of FRDA fibroblasts with IFN-γ stimulates a non-canonical Nrf2 activation pathway through p21 and potentiates antioxidant responses under exposure to hydrogen peroxide. Moreover, IFN-γ significantly reduced the sensitivity to hydrogen peroxide-induced cell death in FRDA fibroblasts. Collectively, these results indicate the presence of multiple pathways triggered by IFN-γ with therapeutic relevance to FRDA.


Asunto(s)
Ataxia de Friedreich , Interferón gamma , Animales , Ratones , Interferón gamma/farmacología , Factor 2 Relacionado con NF-E2/genética , Antioxidantes/farmacología , Ataxia de Friedreich/genética , Peróxido de Hidrógeno , Superóxido Dismutasa
3.
Int J Mol Sci ; 23(12)2022 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-35742964

RESUMEN

A rare disease is defined by its low prevalence in the general population [...].


Asunto(s)
Enfermedades Raras , Humanos , Enfermedades Raras/genética , Enfermedades Raras/terapia
4.
Front Neurosci ; 16: 814445, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35221903

RESUMEN

Friedreich ataxia is a rare neurodegenerative disorder caused by insufficient levels of the essential mitochondrial protein frataxin. It is a severely debilitating disease that significantly impacts the quality of life of affected patients and reduces their life expectancy, however, an adequate cure is not yet available for patients. Frataxin function, although not thoroughly elucidated, is associated with assembly of iron-sulfur cluster and iron metabolism, therefore insufficient frataxin levels lead to reduced activity of many mitochondrial enzymes involved in the electron transport chain, impaired mitochondrial metabolism, reduced ATP production and inefficient anti-oxidant response. As a consequence, neurons progressively die and patients progressively lose their ability to coordinate movement and perform daily activities. Therapeutic strategies aim at restoring sufficient frataxin levels or at correcting some of the downstream consequences of frataxin deficiency. However, the classical pathways of drug discovery are challenging, require a significant amount of resources and time to reach the final approval, and present a high failure rate. Drug repositioning represents a viable alternative to boost the identification of a therapy, particularly for rare diseases where resources are often limited. In this review we will describe recent efforts aimed at the identification of a therapy for Friedreich ataxia through drug repositioning, and discuss the limitation of such strategies.

5.
Hum Mol Genet ; 31(12): 2010-2022, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35015850

RESUMEN

Frataxin (FXN) deficiency is responsible for Friedreich's ataxia (FRDA) in which, besides the characteristic features of spinocerebellar ataxia, two thirds of patients develop hypertrophic cardiomyopathy that often progresses to heart failure and premature death. Different mechanisms might underlie FRDA pathogenesis. Among them, the role of miRNAs deserves investigations. We carried out an miRNA PCR-array analysis of plasma samples of early-, intermediate- and late-onset FRDA groups, defining a set of 30 differentially expressed miRNAs. Hsa-miR223-3p is the only miRNA shared between the three patient groups and appears upregulated in all of them. The up-regulation of hsa-miR223-3p was further validated in all enrolled patients (n = 37, Fc = +2.3; P < 0.0001). Using a receiver operating characteristic curve analysis, we quantified the predictive value of circulating hsa-miR223-3p for FRDA, obtaining an area under the ROC curve value of 0.835 (P < 0.0001) for all patients. Interestingly, we found a significant positive correlation between hsa-miR223-3p expression and cardiac parameters in typical FRDA patients (onset < 25 years). Moreover, a significant negative correlation between hsa-miR223-3p expression and HAX-1 (HCLS1-associated protein X-1) at mRNA and protein level was observed in all FRDA patients. In silico analyses suggested HAX-1 as a target gene of hsa-miR223-3p. Accordingly, we report that HAX-1 is negatively regulated by hsa-miR223-3p in cardiomyocytes (AC16) and neurons (SH-SY5Y), which are critically affected cell types in FRDA. This study describes for the first time the association between hsa-miR223-3p and HAX-1 expression in FRDA, thus supporting a potential role of this microRNA as non-invasive epigenetic biomarker for FRDA.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Ataxia de Friedreich , MicroARNs , Neuroblastoma , Proteínas Adaptadoras Transductoras de Señales/genética , Ataxia de Friedreich/patología , Humanos , MicroARNs/sangre , Miocitos Cardíacos/metabolismo , Neuroblastoma/metabolismo , ARN Mensajero/genética
6.
Front Neurosci ; 14: 872, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33162876

RESUMEN

The identification of efficient markers of disease progression and response to possibly effective treatments is a key priority for slowly progressive, rare and neurodegenerative diseases, such as Friedreich's ataxia. Various imaging modalities have documented specific abnormalities in Friedreich's ataxia that could be tracked to provide useful indicators of efficacy in clinical trials. Advanced MRI imaging (diffusion tensor imaging, DTI; functional MRI, fMRI; and resting-state fMRI, rs-fMRI) and retinal imaging (optical coherence tomography, OCT) were tested longitudinally in a small group of Friedreich's ataxia patients participating in an open-label clinical trial testing the safety and the efficacy of 6-month treatment with interferon gamma. While the DTI indices documented the slow progression of fractional anisotropy loss, fMRI and rs-fMRI were significantly modified during and after treatment. The fMRI changes significantly correlated with the Scale for the Assessment and Rating of Ataxia, which is used to monitor clinical response. OCT documented the known thickness reduction of the retinal nerve fiber layer thickness, but there was no change over time. This pilot study provides indications for the potential utility of fMRI and rs-fMRI as ancillary measures in clinical trials for Friedreich's ataxia.

7.
Hum Mol Genet ; 29(3): 471-482, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31943004

RESUMEN

Frataxin deficiency, responsible for Friedreich's ataxia (FRDA), is crucial for cell survival since it critically affects viability of neurons, pancreatic beta cells and cardiomyocytes. In FRDA, the heart is frequently affected with typical manifestation of hypertrophic cardiomyopathy, which can progress to heart failure and cause premature death. A microarray analysis performed on FRDA patient's lymphoblastoid cells stably reconstituted with frataxin, indicated HS-1-associated protein X-1 (HAX-1) as the most significantly upregulated transcript (FC = +2, P < 0.0006). quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR) and western blot analysis performed on (I) HEK293 stably transfected with empty vector compared to wild-type frataxin and (II) lymphoblasts from FRDA patients show that low frataxin mRNA and protein expression correspond to reduced levels of HAX-1. Frataxin overexpression and silencing were also performed in the AC16 human cardiomyocyte cell line. HAX-1 protein levels are indeed regulated through frataxin modulation. Moreover, correlation between frataxin and HAX-1 was further evaluated in peripheral blood mononuclear cells (PBMCs) from FRDA patients and from non-related healthy controls. A regression model for frataxin which included HAX-1, group membership and group* HAX-1 interaction revealed that frataxin and HAX-1 are associated both at mRNA and protein levels. Additionally, a linked expression of FXN, HAX-1 and antioxidant defence proteins MnSOD and Nrf2 was observed both in PBMCs and AC16 cardiomyocytes. Our results suggest that HAX-1 could be considered as a potential biomarker of cardiac disease in FRDA and the evaluation of its expression might provide insights into its pathogenesis as well as improving risk stratification strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cardiomiopatía Hipertrófica/patología , Ataxia de Friedreich/complicaciones , Regulación de la Expresión Génica , Insuficiencia Cardíaca/patología , Proteínas de Unión a Hierro/metabolismo , Miocitos Cardíacos/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Anciano , Cardiomiopatía Hipertrófica/etiología , Cardiomiopatía Hipertrófica/metabolismo , Femenino , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Humanos , Proteínas de Unión a Hierro/genética , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Adulto Joven , Frataxina
9.
Nucleic Acids Res ; 47(20): 10728-10743, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31584077

RESUMEN

Friedreich's ataxia (FRDA) is an untreatable disorder with neuro- and cardio-degenerative progression. This monogenic disease is caused by the hyper-expansion of naturally occurring GAA repeats in the first intron of the FXN gene, encoding for frataxin, a protein implicated in the biogenesis of iron-sulfur clusters. As the genetic defect interferes with FXN transcription, FRDA patients express a normal frataxin protein but at insufficient levels. Thus, current therapeutic strategies are mostly aimed to restore physiological FXN expression. We have previously described SINEUPs, natural and synthetic antisense long non-coding RNAs, which promote translation of partially overlapping mRNAs through the activity of an embedded SINEB2 domain. Here, by in vitro screening, we have identified a number of SINEUPs targeting human FXN mRNA and capable to up-regulate frataxin protein to physiological amounts acting at the post-transcriptional level. Furthermore, FXN-specific SINEUPs promote the recovery of disease-associated mitochondrial aconitase defects in FRDA-derived cells. In summary, we provide evidence that SINEUPs may be the first gene-specific therapeutic approach to activate FXN translation in FRDA and, more broadly, a novel scalable platform to develop new RNA-based therapies for haploinsufficient diseases.


Asunto(s)
Ataxia de Friedreich/genética , Regulación de la Expresión Génica , Proteínas de Unión a Hierro/genética , Modelos Biológicos , ARN no Traducido/metabolismo , Aconitato Hidratasa/metabolismo , Línea Celular , Fibroblastos/metabolismo , Humanos , Linfocitos/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , Frataxina
10.
Mov Disord ; 34(3): 323-334, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30624801

RESUMEN

BACKGROUND: Friedreich's ataxia is an autosomal-recessive cerebellar ataxia caused by mutation of the frataxin gene, resulting in decreased frataxin expression, mitochondrial dysfunction, and oxidative stress. Currently, no treatment is available for Friedreich's ataxia patients. Given that levels of residual frataxin critically affect disease severity, the main goal of a specific therapy for Friedreich's ataxia is to increase frataxin levels. OBJECTIVES: With the aim to accelerate the development of a new therapy for Friedreich's ataxia, we took a drug repositioning approach to identify market-available drugs able to increase frataxin levels. METHODS: Using a cell-based reporter assay to monitor variation in frataxin amount, we performed a high-throughput screening of a library containing 853 U.S. Food and Drug Administration-approved drugs. RESULTS: Among the potentially interesting candidates isolated from the screening, we focused our attention on etravirine, an antiviral drug currently in use as an anti-human immunodeficiency virus therapy. Here, we show that etravirine can promote a significant increase in frataxin levels in cells derived from Friedreich's ataxia patients, by enhancing frataxin messenger RNA translation. Importantly, frataxin accumulation in treated patient cell lines is comparable to frataxin levels in unaffected carrier cells, suggesting that etravirine could be therapeutically relevant. Indeed, etravirine treatment restores the activity of the iron-sulphur cluster containing enzyme aconitase and confers resistance to oxidative stress in cells derived from Friedreich's ataxia patients. CONCLUSIONS: Considering its excellent safety profile along with its ability to increase frataxin levels and correct some of the disease-related defects, etravirine represents a promising candidate as a therapeutic for Friedreich's ataxia. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Ataxia de Friedreich/tratamiento farmacológico , Proteínas de Unión a Hierro/metabolismo , Piridazinas/uso terapéutico , Línea Celular , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Humanos , Proteínas de Unión a Hierro/genética , Nitrilos , Pirimidinas , Frataxina
11.
Cell Rep ; 18(8): 2007-2017, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228265

RESUMEN

Friedreich ataxia (FRDA) is a severe genetic neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin. To date, there is no therapy to treat this condition. The amount of residual frataxin critically affects the severity of the disease; thus, attempts to restore physiological frataxin levels are considered therapeutically relevant. Frataxin levels are controlled by the ubiquitin-proteasome system; therefore, inhibition of the frataxin E3 ligase may represent a strategy to achieve an increase in frataxin levels. Here, we report the identification of the RING E3 ligase RNF126 as the enzyme that specifically mediates frataxin ubiquitination and targets it for degradation. RNF126 interacts with frataxin and promotes its ubiquitination in a catalytic activity-dependent manner, both in vivo and in vitro. Most importantly, RNF126 depletion results in frataxin accumulation in cells derived from FRDA patients, highlighting the relevance of RNF126 as a new therapeutic target for Friedreich ataxia.


Asunto(s)
Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Catálisis , Línea Celular , Células HEK293 , Humanos , Proteínas Mitocondriales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Frataxina
12.
Hum Mol Genet ; 24(15): 4296-305, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25948553

RESUMEN

Defective expression of frataxin is responsible for the inherited, progressive degenerative disease Friedreich's Ataxia (FRDA). There is currently no effective approved treatment for FRDA and patients die prematurely. Defective frataxin expression causes critical metabolic changes, including redox imbalance and ATP deficiency. As these alterations are known to regulate the tyrosine kinase Src, we investigated whether Src might in turn affect frataxin expression. We found that frataxin can be phosphorylated by Src. Phosphorylation occurs primarily on Y118 and promotes frataxin ubiquitination, a signal for degradation. Accordingly, Src inhibitors induce accumulation of frataxin but are ineffective on a non-phosphorylatable frataxin-Y118F mutant. Importantly, all the Src inhibitors tested, some of them already in the clinic, increase frataxin expression and rescue the aconitase defect in frataxin-deficient cells derived from FRDA patients. Thus, Src inhibitors emerge as a new class of drugs able to promote frataxin accumulation, suggesting their possible use as therapeutics in FRDA.


Asunto(s)
Ataxia de Friedreich/genética , Proteínas de Unión a Hierro/biosíntesis , Familia-src Quinasas/genética , Adenosina Trifosfato/deficiencia , Adenosina Trifosfato/genética , Inhibidores Enzimáticos/farmacología , Ataxia de Friedreich/tratamiento farmacológico , Ataxia de Friedreich/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Unión a Hierro/genética , Oxidación-Reducción , Ubiquitinación/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Frataxina
13.
Neurobiol Dis ; 75: 91-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25549872

RESUMEN

Friedreich ataxia is an inherited neurodegenerative disease that leads to progressive disability. There is currently no effective treatment and patients die prematurely. The underlying genetic defect leads to reduced expression of the mitochondrial protein frataxin. Frataxin insufficiency causes mitochondrial dysfunction and ultimately cell death, particularly in peripheral sensory ganglia. There is an inverse correlation between the amount of residual frataxin and the severity of disease progression; therefore, therapeutic approaches aiming at increasing frataxin levels are expected to improve patients' conditions. We previously discovered that a significant amount of frataxin precursor is degraded by the ubiquitin/proteasome system before its functional mitochondrial maturation. We also provided evidence for the therapeutic potential of small molecules that increase frataxin levels by docking on the frataxin ubiquitination site, thus preventing frataxin ubiquitination and degradation. We called these compounds ubiquitin-competing molecules (UCM). By extending our search for effective UCM, we identified a set of new and more potent compounds that more efficiently promote frataxin accumulation. Here we show that these compounds directly interact with frataxin and prevent its ubiquitination. Interestingly, these UCM are not effective on the ubiquitin-resistant frataxin mutant, indicating their specific action on preventing frataxin ubiquitination. Most importantly, these compounds are able to promote frataxin accumulation and aconitase rescue in cells derived from patients, strongly supporting their therapeutic potential.


Asunto(s)
Aconitato Hidratasa/metabolismo , Ataxia de Friedreich/tratamiento farmacológico , Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/metabolismo , Fármacos Neuroprotectores/farmacología , Sitios de Unión , Línea Celular , Diseño de Fármacos , Fluorescencia , Células HEK293 , Humanos , Immunoblotting , Proteínas de Unión a Hierro/química , Proteínas de Unión a Hierro/genética , Simulación del Acoplamiento Molecular , Mutación , Fármacos Neuroprotectores/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ubiquitinación/efectos de los fármacos , Frataxina
14.
Exp Gerontol ; 48(2): 191-201, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23247094

RESUMEN

Severe mitochondria deficiency leads to a number of devastating degenerative disorders, yet, mild mitochondrial dysfunction in different species, including the nematode Caenorhabditis elegans, can have pro-longevity effects. This apparent paradox indicates that cellular adaptation to partial mitochondrial stress can induce beneficial responses, but how this is achieved is largely unknown. Complete absence of frataxin, the mitochondrial protein defective in patients with Friedreich's ataxia, is lethal in C. elegans, while its partial deficiency extends animal lifespan in a p53 dependent manner. In this paper we provide further insight into frataxin control of C. elegans longevity by showing that a substantial reduction of frataxin protein expression is required to extend lifespan, affect sensory neurons functionality, remodel lipid metabolism and trigger autophagy. We find that Beclin and p53 genes are required to induce autophagy and concurrently reduce lipid storages and extend animal lifespan in response to frataxin suppression. Reciprocally, frataxin expression modulates autophagy in the absence of p53. Human Friedreich ataxia-derived lymphoblasts also display increased autophagy, indicating an evolutionarily conserved response to reduced frataxin expression. In sum, we demonstrate a causal connection between induction of autophagy and lifespan extension following reduced frataxin expression, thus providing the rationale for investigating autophagy in the pathogenesis and treatment of Friedreich's ataxia and possibly other human mitochondria-associated disorders.


Asunto(s)
Autofagia , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Ataxia de Friedreich/metabolismo , Silenciador del Gen , Proteínas de Unión a Hierro/metabolismo , Metabolismo de los Lípidos , Longevidad , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Humanos , Proteínas de Unión a Hierro/genética , Mitocondrias/metabolismo , Interferencia de ARN , Células Receptoras Sensoriales/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Frataxina
15.
Hum Mol Genet ; 21(13): 2855-61, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22447512

RESUMEN

Friedreich's ataxia (FRDA) is the most common hereditary ataxia, affecting ∼3 in 100 000 individuals in Caucasian populations. It is caused by intronic GAA repeat expansions that hinder the expression of the FXN gene, resulting in defective levels of the mitochondrial protein frataxin. Sensory neurons in dorsal root ganglia (DRG) are particularly damaged by frataxin deficiency. There is no specific therapy for FRDA. Here, we show that frataxin levels can be upregulated by interferon gamma (IFNγ) in a variety of cell types, including primary cells derived from FRDA patients. IFNγ appears to act largely through a transcriptional mechanism on the FXN gene. Importantly, in vivo treatment with IFNγ increases frataxin expression in DRG neurons, prevents their pathological changes and ameliorates the sensorimotor performance in FRDA mice. These results disclose new roles for IFNγ in cellular metabolism and have direct implications for the treatment of FRDA.


Asunto(s)
Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Interferón gamma/farmacología , Interferón gamma/fisiología , Proteínas de Unión a Hierro/biosíntesis , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ataxia de Friedreich/tratamiento farmacológico , Ataxia de Friedreich/patología , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Células HeLa , Humanos , Interferón gamma/uso terapéutico , Proteínas de Unión a Hierro/genética , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Transcripción Genética , Activación Transcripcional , Frataxina
16.
Hum Mol Genet ; 20(7): 1253-61, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21216878

RESUMEN

Friedreich's ataxia (FRDA) is a devastating orphan disease, with no specific treatment. The disease is caused by reduced expression of the protein frataxin, which results in mitochondrial defects and oxidative damage. Levels of residual frataxin critically affect onset and progression of the disease. Understanding the molecular mechanisms that regulate frataxin stability and degradation may, therefore, be exploited for the design of effective therapeutics. Here we show that frataxin is degraded by the ubiquitin-proteasome system and that K(147) is the critical residue responsible for frataxin ubiquitination and degradation. Accordingly, a K(147)R substitution generates a more stable frataxin. We then disclose a set of lead compounds, computationally selected to target the molecular cleft harboring K(147), that can prevent frataxin ubiquitination and degradation, and increase frataxin levels in cells derived from FRDA patients. Moreover, treatment with these compounds induces substantial recovery of aconitase activity and adenosine-5'-triphosphate levels in FRDA cells. Thus, we provide evidence for the therapeutic potential of directly interfering with the frataxin degradation pathway.


Asunto(s)
Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Aconitato Hidratasa/genética , Aconitato Hidratasa/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Sustitución de Aminoácidos , Ataxia de Friedreich/tratamiento farmacológico , Ataxia de Friedreich/genética , Células HEK293 , Humanos , Proteínas de Unión a Hierro/genética , Mutación Missense , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética , Frataxina
17.
Atherosclerosis ; 210(2): 400-6, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20122687

RESUMEN

OBJECTIVE: The development of atherosclerotic lesions associates with the proliferation of vascular smooth muscle cells (VSMC), and their migration from arterial tunica media to the intima. Fibroblast growth factor (FGF)-2 can trigger either phenomena, which are accompanied by the functional impairment of the p53 transcription factor. However, FGF-2 impact on p53 function in VSMC is largely unknown. METHODS AND RESULTS: RT-PCR and Western blot analyses assayed FGF-2 effect on human primary VSMC expression of p53-induced molecules with a role in atherogenesis. Confocal microscopy evaluated whether FGF-2 could affect p53 distribution inside VSMC. Results indicate that VSMC exposure to FGF-2 at amounts stimulating the proliferation and migration of these cells promotes p53 phosphorylation and transient accumulation in VSMC nuclei. This is followed by an increase in the expression of the p53-induced thrombospondin (TSP)-1, a VSMC growth and motility factor, and human double minute 2 (HDM2), an antagonist of p53 transcriptional and growth suppressive activity. At later time points, in agreement with the increase of HDM2 and with the capability of this protein to export nuclear p53 to the cytoplasm, the content of p53 in VSMC nuclei is reduced, and the expression of the p53-targeted TSP-l and HDM2 is diminished. CONCLUSIONS: Since FGF-2, p53, TSP-1, and HDM2 are expressed in human atherosclerotic lesions, the in vitro effects of FGF-2 described herein may be operative in vivo, providing a molecular mechanism for FGF-2 pro-atherogenic activity.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Músculo Liso Vascular/citología , Proteína p53 Supresora de Tumor/metabolismo , Aterosclerosis , Western Blotting , Movimiento Celular , Células Cultivadas , Citoplasma/metabolismo , Humanos , Modelos Biológicos , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombospondina 1/metabolismo
18.
Hum Mol Genet ; 19(7): 1221-9, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20053667

RESUMEN

The inability to produce normal levels of the mitochondrial protein frataxin causes the hereditary degenerative disorder Friedreich's Ataxia (FRDA), a syndrome characterized by progressive gait instability, cardiomyopathy and high incidence of diabetes. Frataxin is an iron-binding protein involved in the biogenesis of iron-sulfur clusters (ISC), prosthetic groups allowing essential cellular functions such as oxidative phosphorylation, enzyme catalysis and gene regulation. Although several evidence suggest that frataxin acts as an iron-chaperone within the mitochondrial compartment, we have recently demonstrated the existence of a functional extramitochondrial pool of mature frataxin in various human cell types. Here, we show that a similar proteolytic process generates both mature mitochondrial and extramitochondrial frataxin. To address the physiological function of human extramitochondrial frataxin, we searched for ISC-dependent interaction partners. We demonstrate that the extramitochondrial form of frataxin directly interacts with cytosolic aconitase/iron regulatory protein-1 (IRP1), a bifunctional protein alternating between an enzymatic and a RNA-binding function through the 'iron-sulfur switch' mechanism. Importantly, we found that the cytosolic aconitase defect and consequent IRP1 activation occurring in FRDA cells are reversed by the action of extramitochondrial frataxin. These results provide new insight into the control of cytosolic aconitase/IRP1 switch and expand current knowledge about the molecular pathogenesis of FRDA.


Asunto(s)
Aconitato Hidratasa/metabolismo , Citosol/metabolismo , Proteína 1 Reguladora de Hierro/metabolismo , Proteínas de Unión a Hierro/farmacología , Aconitato Hidratasa/genética , Células Cultivadas , Ataxia de Friedreich/genética , Regulación de la Expresión Génica , Humanos , Proteína 1 Reguladora de Hierro/genética , Frataxina
19.
Blood ; 111(2): 829-37, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17932249

RESUMEN

Ataxia telangiectasia (A-T) is a rare cancer-predisposing genetic disease, caused by the lack of functional ATM kinase, a major actor of the double strand brakes (DSB) DNA-damage response. A-T patients show a broad and diverse phenotype, which includes an increased rate of lymphoma and leukemia development. Fas-induced apoptosis plays a fundamental role in the homeostasis of the immune system and its defects have been associated with autoimmunity and lymphoma development. We therefore investigated the role of ATM kinase in Fas-induced apoptosis. Using A-T lymphoid cells, we could show that ATM deficiency causes resistance to Fas-induced apoptosis. A-T cells up-regulate FLIP protein levels, a well-known inhibitor of Fas-induced apoptosis. Reconstitution of ATM kinase activity was sufficient to decrease FLIP levels and to restore Fas sensitivity. Conversely, genetic and pharmacologic ATM kinase inactivation resulted in FLIP protein up-regulation and Fas resistance. Both ATM and FLIP are aberrantly regulated in Hodgkin lymphoma. Importantly, we found that reconstitution of ATM kinase activity decreases FLIP protein levels and restores Fas sensitivity in Hodgkin lymphoma-derived cells. Overall, these data identify a novel molecular mechanism through which ATM kinase may regulate the immune system homeostasis and impair lymphoma development.


Asunto(s)
Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Enfermedad de Hodgkin/metabolismo , Linfocitos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Receptor fas/metabolismo , Animales , Apoptosis/genética , Ataxia Telangiectasia , Proteínas de la Ataxia Telangiectasia Mutada , Autoinmunidad/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Homeostasis/genética , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Linfocitos/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Receptor fas/genética
20.
Hum Mol Genet ; 16(13): 1534-40, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17468497

RESUMEN

The defective expression of frataxin causes the hereditary neurodegenerative disorder Friedreich's ataxia (FRDA). Human frataxin is synthesized as a 210 amino acid precursor protein, which needs proteolytic processing into mitochondria to be converted into the functional mature form. In vitro processing of human frataxin was previously described to yield a 155 amino acid mature form, corresponding to residues 56-210 (frataxin(56-210)). Here, we studied the maturation of frataxin by in vivo overexpression in human cells. Our data show that the main form of mature frataxin is generated by a proteolytic cleavage between Lys80 and Ser81, yielding a 130 amino acid protein (frataxin(81-210)). This maturation product corresponds to the endogenous frataxin detected in human heart, peripheral blood lymphocytes or dermal fibroblasts. Moreover, we demonstrate that frataxin(81-210) is biologically functional, as it rescues aconitase defects in frataxin-deficient cells derived from FRDA patients. Importantly, our data indicate that frataxin(56-210) can be produced in vivo when the primary 80-81 maturation site is unavailable, suggesting the existence of proteolytic mechanisms that can actively control the size of the mature product, with possible functional implications.


Asunto(s)
Proteínas de Unión a Hierro/metabolismo , Aconitato Hidratasa/metabolismo , Secuencia de Aminoácidos , Línea Celular , Células Cultivadas , Fibroblastos/metabolismo , Ataxia de Friedreich/genética , Eliminación de Gen , Regulación de la Expresión Génica , Homocigoto , Humanos , Proteínas de Unión a Hierro/química , Linfocitos/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Frataxina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...