Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Hum Mol Genet ; 29(3): 471-482, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31943004

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cardiomyopathy, Hypertrophic/pathology , Friedreich Ataxia/complications , Gene Expression Regulation , Heart Failure/pathology , Iron-Binding Proteins/metabolism , Myocytes, Cardiac/pathology , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/metabolism , Female , Heart Failure/etiology , Heart Failure/metabolism , Humans , Iron-Binding Proteins/genetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Myocytes, Cardiac/metabolism , Young Adult , Frataxin
2.
Mov Disord ; 34(3): 323-334, 2019 03.
Article in English | MEDLINE | ID: mdl-30624801

ABSTRACT

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.


Subject(s)
Friedreich Ataxia/drug therapy , Iron-Binding Proteins/metabolism , Pyridazines/therapeutic use , Cell Line , Drug Evaluation, Preclinical , Drug Repositioning , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Humans , Iron-Binding Proteins/genetics , Nitriles , Pyrimidines , Frataxin
3.
Cell Rep ; 18(8): 2007-2017, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228265

ABSTRACT

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.


Subject(s)
Friedreich Ataxia/metabolism , Iron-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Catalysis , Cell Line , HEK293 Cells , Humans , Mitochondrial Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Frataxin
4.
Hum Mol Genet ; 24(15): 4296-305, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25948553

ABSTRACT

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.


Subject(s)
Friedreich Ataxia/genetics , Iron-Binding Proteins/biosynthesis , src-Family Kinases/genetics , Adenosine Triphosphate/deficiency , Adenosine Triphosphate/genetics , Enzyme Inhibitors/pharmacology , Friedreich Ataxia/drug therapy , Friedreich Ataxia/pathology , Gene Expression Regulation/drug effects , Humans , Iron-Binding Proteins/genetics , Oxidation-Reduction , Ubiquitination/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism , Frataxin
5.
Neurobiol Dis ; 75: 91-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25549872

ABSTRACT

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.


Subject(s)
Aconitate Hydratase/metabolism , Friedreich Ataxia/drug therapy , Friedreich Ataxia/metabolism , Iron-Binding Proteins/metabolism , Neuroprotective Agents/pharmacology , Binding Sites , Cell Line , Drug Design , Fluorescence , HEK293 Cells , Humans , Immunoblotting , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/genetics , Molecular Docking Simulation , Mutation , Neuroprotective Agents/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitination/drug effects , Frataxin
6.
Hum Mol Genet ; 20(7): 1253-61, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21216878

ABSTRACT

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.


Subject(s)
Friedreich Ataxia/metabolism , Iron-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitination , Aconitate Hydratase/genetics , Aconitate Hydratase/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Friedreich Ataxia/drug therapy , Friedreich Ataxia/genetics , HEK293 Cells , Humans , Iron-Binding Proteins/genetics , Mutation, Missense , Proteasome Endopeptidase Complex/genetics , Ubiquitin/genetics , Frataxin
7.
Hum Mol Genet ; 19(7): 1221-9, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20053667

ABSTRACT

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.


Subject(s)
Aconitate Hydratase/metabolism , Cytosol/metabolism , Iron Regulatory Protein 1/metabolism , Iron-Binding Proteins/pharmacology , Aconitate Hydratase/genetics , Cells, Cultured , Friedreich Ataxia/genetics , Gene Expression Regulation , Humans , Iron Regulatory Protein 1/genetics , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL
...