Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670433

ABSTRACT

Friedreich's ataxia is an autosomal recessive neurogenetic disease that is mainly associated with atrophy of the spinal cord and progressive neurodegeneration in the cerebellum. The disease is caused by a GAA-expansion in the first intron of the frataxin gene leading to a decreased level of frataxin protein, which results in mitochondrial dysfunction. Currently, there is no effective treatment to delay neurodegeneration in Friedreich's ataxia. A plausible therapeutic approach is gene therapy. Indeed, Friedreich's ataxia mouse models have been treated with viral vectors en-coding for either FXN or neurotrophins, such as brain-derived neurotrophic factor showing promising results. Thus, gene therapy is increasingly consolidating as one of the most promising therapies. However, several hurdles have to be overcome, including immunotoxicity and pheno-toxicity. We review the state of the art of gene therapy in Friedreich's ataxia, addressing the main challenges and the most feasible solutions for them.


Subject(s)
Friedreich Ataxia , Genetic Therapy , Iron-Binding Proteins , Animals , Disease Models, Animal , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Friedreich Ataxia/therapy , Humans , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/genetics , Mice , Frataxin
2.
J Biol Chem ; 292(31): 12744-12753, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28615439

ABSTRACT

Fe-S cofactors are composed of iron and inorganic sulfur in various stoichiometries. A complex assembly pathway conducts their initial synthesis and subsequent binding to recipient proteins. In this minireview, we discuss how discovery of the role of the mammalian cytosolic aconitase, known as iron regulatory protein 1 (IRP1), led to the characterization of the function of its Fe-S cluster in sensing and regulating cellular iron homeostasis. Moreover, we present an overview of recent studies that have provided insights into the mechanism of Fe-S cluster transfer to recipient Fe-S proteins.


Subject(s)
Homeostasis , Iron Regulatory Protein 1/physiology , Iron/physiology , Models, Molecular , Animals , Apoenzymes/chemistry , Apoenzymes/metabolism , Carbon-Sulfur Lyases/biosynthesis , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/physiology , Electron Transport , Gene Expression Regulation, Enzymologic , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/physiology , Humans , Iron Regulatory Protein 1/biosynthesis , Iron Regulatory Protein 1/chemistry , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/physiology , Iron-Regulatory Proteins/biosynthesis , Iron-Regulatory Proteins/chemistry , Iron-Regulatory Proteins/physiology , Iron-Sulfur Proteins/biosynthesis , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/physiology , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/physiology , Molecular Chaperones/biosynthesis , Molecular Chaperones/chemistry , Molecular Chaperones/physiology , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Response Elements , Succinate Dehydrogenase/biosynthesis , Succinate Dehydrogenase/chemistry , Succinate Dehydrogenase/physiology , Frataxin
3.
Protein Expr Purif ; 149: 7-12, 2018 09.
Article in English | MEDLINE | ID: mdl-29655787

ABSTRACT

The human thyroid peroxidase (hTPO) is an essential enzyme for thyroid hormone biosynthesis and is expressed in thyroid cells. It is an autoantigen against which antibodies are found in the sera of patients with a number of autoimmune thyroid disorders. Overexpression of hTPO has been achieved using the baculovirus expression vector system (BEVS). However, it is produced largely in an aggregated form in the cell lysate fraction, which increases the complexity of protein extraction. In this study, to achieve improved secretory expression of hTPO via BEVS, a truncated recombinant hTPO protein (hTPOt) was engineered by replacing its original signal peptide (SP) in the N-terminal with five heterologous SPs. Our data showed that the SP from the peptidyl-glycine alpha-amidating monooxygenase (PAM), referred to as SPPAM, significantly promoted the secretion of SPPAM-fused hTPOt (p-hTPOt) in High Five cells. Subsequently, we established an optimized scale-up production procedure for p-hTPOt in a 5-L wave-type bioreactor. The secretory p-hTPOt was purified by immobilized metal-chelating affinity chromatography and ion-exchange chromatography, achieving a protein purity of >95%. Finally, the purified p-hTPOt showed high sensitivity and specificity in reactions with positive or negative human serum samples via the double-antigen sandwich method, suggesting potential applications in hTPO-based research and product development.


Subject(s)
Autoantigens/biosynthesis , Bioreactors , Iodide Peroxidase/biosynthesis , Iron-Binding Proteins/biosynthesis , Animals , Autoantigens/genetics , Baculoviridae/metabolism , Chromatography, Affinity , Chromatography, Ion Exchange , Escherichia coli , Gene Expression , Humans , Iodide Peroxidase/genetics , Iron-Binding Proteins/genetics , Mixed Function Oxygenases/chemistry , Multienzyme Complexes/chemistry , Protein Sorting Signals , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sf9 Cells/metabolism , Signal Transduction
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.
J Biol Chem ; 290(30): 18584-95, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26063801

ABSTRACT

Cadmium is a well known mutagenic metal that can enter cells via nonspecific metal transporters, causing several cellular damages and eventually leading to death. In the yeast Saccharomyces cerevisiae, the transcription factor Yap1 plays a key role in the regulation of several genes involved in metal stress response. We have previously shown that Yap1 represses the expression of FET4, a gene encoding a low affinity iron transporter able to transport metals other than iron. Here, we have studied the relevance of this repression in cell tolerance to cadmium. Our results indicate that genomic deletion of Yap1 increases FET4 transcript and protein levels. In addition, the cadmium toxicity exhibited by this strain is completely reversed by co-deletion of FET4 gene. These data correlate well with the increased intracellular levels of cadmium observed in the mutant yap1. Rox1, a well known aerobic repressor of hypoxic genes, conveys the Yap1-mediated repression of FET4. We further show that, in a scenario where the activity of Yap1 or Rox1 is compromised, cells activate post-transcriptional mechanisms, involving the exoribonuclease Xrn1, to compensate the derepression of FET4. Our data thus reveal a novel protection mechanism against cadmium toxicity mediated by Yap1 that relies on the aerobic repression of FET4 and results in the impairment of cadmium uptake.


Subject(s)
Cadmium/metabolism , Cation Transport Proteins/biosynthesis , Iron-Binding Proteins/biosynthesis , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Stress, Physiological/genetics , Transcription Factors/metabolism , Biological Transport/genetics , Cadmium/toxicity , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper Transport Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exoribonucleases/metabolism , Gene Expression Regulation, Fungal , Iron/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mutation , Repressor Proteins/genetics , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
6.
Hum Mol Genet ; 23(7): 1829-41, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24242291

ABSTRACT

Friedreich ataxia (FRDA) is a neurodegenerative disease characterized by a decreased expression of the mitochondrial protein frataxin. Major neurological symptoms of the disease are due to degeneration of dorsal root ganglion (DRG) sensory neurons. In this study we have explored the neurodegenerative events occurring by frataxin depletion on primary cultures of neurons obtained from rat DRGs. Reduction of 80% of frataxin levels in these cells was achieved by transduction with lentivirus containing shRNA silencing sequences. Frataxin depletion caused mitochondrial membrane potential decrease, neurite degeneration and apoptotic cell death. A marked increase of free intracellular Ca(2+) levels and alteration in Ca(2+)-mediated signaling pathways was also observed, thus suggesting that altered calcium homeostasis can play a pivotal role in neurodegeneration caused by frataxin deficiency. These deleterious effects were reverted by the addition of a cell-penetrant TAT peptide coupled to the BH4, the anti-apoptotic domain of Bcl-x(L). Treatment of cultured frataxin-depleted neurons with TAT-BH4 was able to restore the free intracellular Ca(2+) levels and protect the neurons from degeneration. These observations open the possibility of new therapies of FRDA based on modulating the Ca(2+) signaling and prevent apoptotic process to protect DRG neurons from neurodegeneration.


Subject(s)
Apoptosis/genetics , Ganglia, Spinal/cytology , Iron-Binding Proteins/genetics , Sensory Receptor Cells/cytology , bcl-X Protein/genetics , Animals , Calcium/metabolism , Calcium Signaling/genetics , Cells, Cultured , Friedreich Ataxia/genetics , Gene Products, tat/genetics , Homeostasis , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/metabolism , Membrane Potential, Mitochondrial/genetics , Mitochondrial Proteins/metabolism , Neurites/pathology , Neurodegenerative Diseases , Protein Structure, Tertiary/genetics , RNA Interference , RNA, Small Interfering , Rats , Frataxin
7.
Exp Cell Res ; 335(1): 51-61, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25929520

ABSTRACT

It is generally accepted that Friedreich's ataxia (FRDA) is caused by a deficiency in frataxin expression, a mitochondrial protein involved in iron homeostasis, which mainly affects the brain, dorsal root ganglia of the spinal cord, heart and in certain cases the pancreas. However, there is little knowledge as to other possible genes that may be affected in this disorder, and which can contribute to its complexity. In the current study we compared human periodontal ligament cells gene expression of healthy individuals and FRDA patients. The expression of active-caspase 3, as well as other apoptosis-related genes, was increased in the FRDA cells. Furthermore, iron-sulphur cluster genes, as well as oxidative stress-related genes were overexpressed in FRDA. Moreover, brain-derived neurotrophic factor, neuregulin 1 and miR-132 were all upregulated. These three genes are capable of regulating the expression of each other. Interestingly, when the cells from FRDA patients were co-cultured in the presence of idebenone and deferiprone, caspase expression decreased while antioxidant gene expression, as well as frataxin expression, increased. Regarding epigenetic mechanisms, the frataxin gene was hypermethylated, compared to the healthy counterparts, in the upstream GAA repetitive region. Of the three DNA methyltransferases, DNMT1 but not DNMT3׳s gene expression was higher in FRDA cells. In conclusion, our data show that FRDA cells present altered expression of genes related to cell cycle, oxidative stress and iron homeostasis which may be implicated in the increased apoptotic levels. Also, the altered expression is in a certain degree normalized in the presence of idebenone and deferiprone.


Subject(s)
Caspase 3/biosynthesis , Friedreich Ataxia/genetics , Iron-Binding Proteins/biosynthesis , MicroRNAs/biosynthesis , Oxidative Stress/genetics , Antioxidants/pharmacology , Apoptosis/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Brain-Derived Neurotrophic Factor/biosynthesis , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation , Decitabine , Deferiprone , Epigenesis, Genetic , Gene Expression Profiling , Humans , Iron Chelating Agents/pharmacology , Neuregulin-1/biosynthesis , Periodontal Ligament/cytology , Pyridones/pharmacology , Superoxide Dismutase/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Frataxin
8.
Biochim Biophys Acta ; 1840(10): 3022-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24997422

ABSTRACT

BACKGROUND: The neurodegenerative disease Friedreich's ataxia is the result of frataxin deficiency. Frataxin is a mitochondrial protein involved in iron-sulfur cluster (Fe-S) cofactor biogenesis, but its functional role in this pathway is debated. This is due to the interconnectivity of iron metabolic and oxidative stress response pathways that make distinguishing primary effects of frataxin deficiency challenging. Since Fe-S cluster assembly is conserved, frataxin overexpression phenotypes in a simple eukaryotic organism will provide additional insight into frataxin function. METHODS: The Schizosaccharomyces pombe frataxin homologue (fxn1) was overexpressed from a plasmid under a thiamine repressible promoter. The S. pombe transformants were characterized at several expression strengths for cellular growth, mitochondrial organization, iron levels, oxidative stress, and activities of Fe-S cluster containing enzymes. RESULTS: Observed phenotypes were dependent on the amount of Fxn1 overexpression. High Fxn1 overexpression severely inhibited S. pombe growth, impaired mitochondrial membrane integrity and cellular respiration, and led to Fxn1 aggregation. Cellular iron accumulation was observed at moderate Fxn1 overexpression but was most pronounced at high levels of Fxn1. All levels of Fxn1 overexpression up-regulated oxidative stress defense and mitochondrial Fe-S cluster containing enzyme activities. CONCLUSIONS: Despite the presence of oxidative stress and accumulated iron, activation of Fe-S cluster enzymes was common to all levels of Fxn1 overexpression; therefore, Fxn1 may regulate the efficiency of Fe-S cluster biogenesis in S. pombe. GENERAL SIGNIFICANCE: We provide evidence that suggests that dysregulated Fe-S cluster biogenesis is a primary effect of both frataxin overexpression and deficiency as in Friedreich's ataxia.


Subject(s)
Friedreich Ataxia/metabolism , Fungal Proteins/metabolism , Iron-Binding Proteins/biosynthesis , Iron/metabolism , Mitochondrial Membranes/metabolism , Schizosaccharomyces/metabolism , Friedreich Ataxia/genetics , Fungal Proteins/genetics , Iron-Binding Proteins/genetics , Oxidative Stress/genetics , Schizosaccharomyces/genetics , Sequence Homology, Amino Acid , Up-Regulation , Frataxin
9.
Lancet ; 384(9942): 504-13, 2014 Aug 09.
Article in English | MEDLINE | ID: mdl-24794816

ABSTRACT

BACKGROUND: Friedreich's ataxia is a progressive degenerative disorder caused by deficiency of the frataxin protein. Expanded GAA repeats within intron 1 of the frataxin (FXN) gene lead to its heterochromatinisation and transcriptional silencing. Preclinical studies have shown that the histone deacetylase inhibitor nicotinamide (vitamin B3) can remodel the pathological heterochromatin and upregulate expression of FXN. We aimed to assess the epigenetic and neurological effects and safety of high-dose nicotinamide in patients with Friedreich's ataxia. METHODS: In this exploratory, open-label, dose-escalation study in the UK, male and female patients (aged 18 years or older) with Friedreich's ataxia were given single doses (phase 1) and repeated daily doses of 2-8 g oral nicotinamide for 5 days (phase 2) and 8 weeks (phase 3). Doses were gradually escalated during phases 1 and 2, with individual maximum tolerated doses used in phase 3. The primary outcome was the upregulation of frataxin expression. We also assessed the safety and tolerability of nicotinamide, used chromatin immunoprecipitation to investigate changes in chromatin structure at the FXN gene locus, and assessed the effect of nicotinamide treatment on clinical scales for ataxia. This study is registered with ClinicalTrials.gov, number NCT01589809. FINDINGS: Nicotinamide was generally well tolerated; the main adverse event was nausea, which in most cases was mild, dose-related, and resolved spontaneously or after dose reduction, use of antinausea drugs, or both. Phase 1 showed a dose-response relation for proportional change in frataxin protein concentration from baseline to 8 h post-dose, which increased with increasing dose (p=0·0004). Bayesian analysis predicted that 3·8 g would result in a 1·5-times increase and 7·5 g in a doubling of frataxin protein concentration. Phases 2 and 3 showed that daily dosing at 3·5-6 g resulted in a sustained and significant (p<0·0001) upregulation of frataxin expression, which was accompanied by a reduction in heterochromatin modifications at the FXN locus. Clinical measures showed no significant changes. INTERPRETATION: Nicotinamide was associated with a sustained improvement in frataxin concentrations towards those seen in asymptomatic carriers during 8 weeks of daily dosing. Further investigation of the long-term clinical benefits of nicotinamide and its ability to ameliorate frataxin deficiency in Friedreich's ataxia is warranted. FUNDING: Ataxia UK, Ataxia Ireland, Association Suisse de l'Ataxie de Friedreich, Associazione Italiana per le Sindromi Atassiche, UK National Institute for Health Research, European Friedreich's Ataxia Consortium for Translational Studies, and Imperial Biomedical Research Centre.


Subject(s)
Friedreich Ataxia/drug therapy , Iron-Binding Proteins/drug effects , Niacinamide/administration & dosage , Vitamin B Complex/administration & dosage , Adult , Chromatin/drug effects , Chromatin/genetics , Dose-Response Relationship, Drug , Epigenesis, Genetic , Female , Friedreich Ataxia/genetics , Humans , Iron-Binding Proteins/biosynthesis , Male , Middle Aged , Treatment Outcome , United Kingdom , Young Adult , Frataxin
10.
Growth Factors ; 32(1): 1-10, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24438103

ABSTRACT

SOCS3 (suppressor of cytokine signaling 3) inhibits the intracellular signaling cascade initiated by exposure of cells to cytokines. SOCS3 regulates signaling via two distinct mechanisms: directly inhibiting the catalytic activity of Janus kinases (JAKs) that initiate the intracellular signaling cascade and catalysing the ubiquitination of signaling components by recruiting components of an E3 ubiquitin ligase complex. Here we investigate the latter mode-of-action biochemically by reconstructing a SOCS3-based E3 ubiquitin ligase complex in vitro using fully purified, recombinant components and examining its ability to promote the ubiquitination of molecules involved in the cytokine signaling cascade. We show that SOCS3 is an active substrate recruitment module for a Cullin5-based E3 ligase and have defined the core protein components required for ubiquitination. SOCS3-induced polyubiquitination was rapid and could proceed through a number of different ubiquitin lysines. SOCS3 catalyzed the ubiquitination of both the IL-6 receptor common chain (gp130) and JAK2.


Subject(s)
Cytokine Receptor gp130/genetics , Janus Kinase 2/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Cell Line , Cloning, Molecular , Cullin Proteins/biosynthesis , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cytokine Receptor gp130/metabolism , Elongin , Humans , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/genetics , Janus Kinase 2/metabolism , Mice , NEDD8 Protein , Phosphorylation , Protein Binding , Signal Transduction/genetics , Spodoptera/cytology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Ubiquitin-Conjugating Enzymes/biosynthesis , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitination/genetics , Ubiquitins/chemistry
11.
Hum Mol Genet ; 21(13): 2855-61, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22447512

ABSTRACT

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.


Subject(s)
Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Interferon-gamma/pharmacology , Interferon-gamma/physiology , Iron-Binding Proteins/biosynthesis , Animals , Cells, Cultured , Disease Models, Animal , Friedreich Ataxia/drug therapy , Friedreich Ataxia/pathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HeLa Cells , Humans , Interferon-gamma/therapeutic use , Iron-Binding Proteins/genetics , Mice , Mice, Transgenic , Mitochondria/metabolism , Transcription, Genetic , Transcriptional Activation , Frataxin
12.
J Neurochem ; 126 Suppl 1: 147-54, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23859350

ABSTRACT

The genetic defect in Friedreich's ataxia (FRDA) is the expansion of a GAA·TCC triplet in the first intron of the FXN gene, which encodes the mitochondrial protein frataxin. Previous studies have established that the repeats reduce transcription of this essential gene, with a concomitant decrease in frataxin protein in affected individuals. As the repeats do not alter the FXN protein coding sequence, one therapeutic approach would be to increase transcription of pathogenic FXN genes. Histone posttranslational modifications near the expanded repeats are consistent with heterochromatin formation and FXN gene silencing. In an effort to find small molecules that would reactivate this silent gene, histone deacetylase inhibitors were screened for their ability to up-regulate FXN gene expression in patient cells and members of the pimelic 2-aminobenzamide family of class I histone deacetylase inhibitors were identified as potent inducers of FXN gene expression and frataxin protein. Importantly, these molecules up-regulate FXN expression in human neuronal cells derived from patient-induced pluripotent stem cells and in two mouse models for the disease. Preclinical studies of safety and toxicity have been completed for one such compound and a phase I clinical trial in FRDA patients has been initiated. Furthermore, medicinal chemistry efforts have identified improved compounds with superior pharmacological properties.


Subject(s)
Gene Expression/physiology , Histone Deacetylase Inhibitors/therapeutic use , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/genetics , Animals , Clinical Trials as Topic , Gene Silencing , Heterochromatin/metabolism , Histone Deacetylase Inhibitors/chemistry , Humans , Iron-Binding Proteins/drug effects , Mice , Structure-Activity Relationship , Frataxin
13.
Hum Mol Genet ; 20(14): 2807-22, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21531789

ABSTRACT

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.


Subject(s)
Apoptosis , Gene Silencing , Iron-Binding Proteins/biosynthesis , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Friedreich Ataxia/therapy , Humans , Iron-Binding Proteins/genetics , Models, Biological , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Frataxin
14.
Clin Exp Nephrol ; 17(3): 424-30, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23180044

ABSTRACT

BACKGROUND: The mitochondrial protein frataxin regulates iron metabolism for heme and iron sulfur cluster synthesis in the mitochondria and could be associated with the regulation of oxidative stress. To clarify the expression of frataxin and its association with uremia, we evaluated the mRNA and protein levels of frataxin in the polymorphonuclear leukocytes (PMNLs) of patients on hemodialysis (HD). METHODS: Uremic patients on HD (n = 18) and healthy control subjects (n = 18) were investigated. PMNLs were isolated by differential centrifugation. The mRNA levels of frataxin in isolated leukocytes were quantified by TaqMan real-time polymerase chain reaction. Frataxin protein expression in the cell lysate was evaluated using SDS-polyacrylamide gel electrophoresis and Western blotting. RESULTS: The frataxin/glyceraldehyde-3-phosphate dehydrogenase mRNA ratio in PMNLs from uremic patients was significantly lower than that in control subjects. Frataxin protein expression in uremic patients was also significantly lower than that in controls. Multiple regression analysis showed that frataxin mRNA levels were independently associated with the serum levels of both the oxidative stress marker malondialdehyde and the proinflammatory cytokine tumor necrosis factor-α. CONCLUSION: The downregulation of frataxin seems to be linked with uremic status, which is usually associated with chronic inflammation and the acceleration of oxidative stress. Mitochondrial iron regulation may play a role in several comorbidities and in the poor prognosis in uremic patients. Further investigation is needed to elucidate whether reduced frataxin levels are linked to the pathological status of uremic patients and whether uremic substances affect frataxin expression.


Subject(s)
Iron-Binding Proteins/biosynthesis , Renal Dialysis , Uremia/metabolism , Aged , Down-Regulation , Female , Humans , Kidney Failure, Chronic/metabolism , Male , Middle Aged , Mitochondrial Proteins/metabolism , Neutrophils/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/blood , Frataxin
15.
J Nutr Biochem ; 114: 109258, 2023 04.
Article in English | MEDLINE | ID: mdl-36587874

ABSTRACT

Emerging evidence supports the beneficial effect of quercetin on liver mitochondrial disorders. However, the molecular mechanism by which quercetin protects mitochondria is limited, especially in alcoholic liver disease. In this study, C57BL/6N mice were fed with Lieber De Carli liquid diet (28% ethanol-derived calories) for 12 weeks plus a single binge ethanol and intervened with quercetin (100 mg/kg.bw). Moreover, HepG2CYP2E1+/+ were stimulated with ethanol (100 mM) and quercetin (50 µM) to investigate the effects of mitochondrial protein frataxin. The results indicated that quercetin alleviated alcohol-induced histopathological changes and mitochondrial functional disorders in mice livers. Consistent with increased PINK1, Parkin, Bnip3 and LC3II as well as decreased p62, TOM20 and VDAC1 expression, the inhibition of mitophagy by ethanol was blocked by quercetin. Additionally, quercetin improved the imbalance of iron metabolism-related proteins expression in alcohol-fed mice livers. Compared with ethanol-treated Lv-empty HepG2CYP2E1+/+ cells, frataxin deficiency further exacerbated the inhibition of mitochondrial function. Conversely, restoration of frataxin expression ameliorated the effect of ethanol. Furthermore, frataxin deficiency reduced the protective effects of quercetin on mitochondria disordered by ethanol. Attentively, ferric ammonium citrate (FAC) and deferiprone decreased or increased frataxin expression in HepG2CYP2E1+/+, respectively. Notably, we further found FAC reversed the increasing effect of quercetin on frataxin expression. Ultimately, silencing NCOA4 attenuated the inhibition of quercetin on LDH release and mitochondrial membrane potential increase, and similar results were observed by adding FAC. Collectively, these findings demonstrated quercetin increased frataxin expression through regulating iron level, thereby mitigating ethanol-induced mitochondrial dysfunction.


Subject(s)
Iron , Liver Diseases, Alcoholic , Liver , Mitochondria, Liver , Quercetin , Animals , Mice , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Ethanol/toxicity , Iron/metabolism , Liver/metabolism , Mice, Inbred C57BL , Quercetin/pharmacology , Quercetin/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Liver Diseases, Alcoholic/metabolism , Iron-Binding Proteins/biosynthesis , Iron-Binding Proteins/metabolism , Frataxin
16.
J Biol Chem ; 286(6): 4209-15, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21127046

ABSTRACT

Expansion of a GAA · TTC repeat in the first intron of the frataxin (FXN) gene causes an mRNA deficit that results in Friedreich ataxia (FRDA). The region flanking the repeat on FRDA alleles is associated with more extensive DNA methylation than is seen on normal alleles and histone modifications typical of repressed genes. However, whether these changes are responsible for the mRNA deficit is controversial. Using chromatin immunoprecipitation and cell lines from affected and unaffected individuals, we show that certain marks of active chromatin are also reduced in the promoter region of the FXN gene in patient cells. Thus, the promoter chromatin may be less permissive for transcription initiation than it is on normal alleles. Furthermore, we show that the initiating form of RNA polymerase II and histone H3 trimethylated on lysine 4, a chromatin mark tightly linked to transcription initiation, are both present at lower levels on FRDA alleles. In addition, a mark of transcription elongation, trimethylated H3K36, shows a reduced rate of accumulation downstream of the repeat. Our data thus suggest that repeat expansion reduces both transcription initiation and elongation in FRDA cells. Our findings may have implications for understanding the mechanism responsible for FRDA as well as for therapeutic approaches to reverse the transcription deficit.


Subject(s)
DNA Repeat Expansion , Friedreich Ataxia/metabolism , Iron-Binding Proteins/biosynthesis , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Transcription, Genetic , Alleles , Cells, Cultured , Chromatin Immunoprecipitation/methods , DNA Methylation/genetics , Female , Friedreich Ataxia/genetics , Histones/genetics , Histones/metabolism , Humans , Introns , Iron-Binding Proteins/genetics , Male , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/genetics , Frataxin
17.
Am J Physiol Lung Cell Mol Physiol ; 303(6): L519-27, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22842216

ABSTRACT

Cigarette smoke (CS) is a well-established risk factor in the development of chronic obstructive pulmonary disease (COPD). In contrast, the extent to which CS exposure contributes to the development of the systemic manifestations of COPD, such as skeletal muscle dysfunction and wasting, remains largely unknown. Decreased skeletal muscle capillarization has been previously reported in early stages of COPD and might play an important role in the development of COPD-associated skeletal muscle abnormalities. To investigate the effects of chronic CS exposure on skeletal muscle capillarization and exercise tolerance, a mouse model of CS exposure was used. The 129/SvJ mice were exposed to CS for 6 mo, and the expression of putative elements of the hypoxia-angiogenic signaling cascade as well as muscle capillarization were studied. Additionally, functional tests assessing exercise tolerance/endurance were performed in mice. Compared with controls, skeletal muscles from CS-exposed mice exhibited significantly enhanced expression of von Hippel-Lindau tumor suppressor (VHL), ubiquitin-conjugating enzyme E2D1 (UBE2D1), and prolyl hydroxylase-2 (PHD2). In contrast, hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) expression was reduced. Furthermore, reduced muscle fiber cross-sectional area, decreased skeletal muscle capillarization, and reduced exercise tolerance were also observed in CS-exposed animals. Taken together, the current results provide evidence linking chronic CS exposure and induction of VHL expression in skeletal muscles leading toward impaired hypoxia-angiogenesis signal transduction, reduced muscle fiber cross-sectional area, and decreased exercise tolerance.


Subject(s)
Muscle, Skeletal/blood supply , Smoking/physiopathology , Animals , Capillaries/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor-Proline Dioxygenases , Iron-Binding Proteins/biosynthesis , Mice , Muscle, Skeletal/cytology , Procollagen-Proline Dioxygenase/biosynthesis , Ubiquitin-Conjugating Enzymes/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Von Hippel-Lindau Tumor Suppressor Protein/biosynthesis
18.
J Biol Chem ; 285(49): 38486-501, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20889968

ABSTRACT

Friedreich ataxia (FRDA) is an autosomal recessive degenerative disease caused by insufficient expression of frataxin (FXN), a mitochondrial iron-binding protein required for Fe-S cluster assembly. The development of treatments to increase FXN levels in FRDA requires elucidation of the steps involved in the biogenesis of functional FXN. The FXN mRNA is translated to a precursor polypeptide that is transported to the mitochondrial matrix and processed to at least two forms, FXN(42-210) and FXN(81-210). Previous reports suggested that FXN(42-210) is a transient processing intermediate, whereas FXN(81-210) represents the mature protein. However, we find that both FXN(42-210) and FXN(81-210) are present in control cell lines and tissues at steady-state, and that FXN(42-210) is consistently more depleted than FXN(81-210) in samples from FRDA patients. Moreover, FXN(42-210) and FXN(81-210) have strikingly different biochemical properties. A shorter N terminus correlates with monomeric configuration, labile iron binding, and dynamic contacts with components of the Fe-S cluster biosynthetic machinery, i.e. the sulfur donor complex NFS1·ISD11 and the scaffold ISCU. Conversely, a longer N terminus correlates with the ability to oligomerize, store iron, and form stable contacts with NFS1·ISD11 and ISCU. Monomeric FXN(81-210) donates Fe(2+) for Fe-S cluster assembly on ISCU, whereas oligomeric FXN(42-210) donates either Fe(2+) or Fe(3+). These functionally distinct FXN isoforms seem capable to ensure incremental rates of Fe-S cluster synthesis from different mitochondrial iron pools. We suggest that the levels of both isoforms are relevant to FRDA pathophysiology and that the FXN(81-210)/FXN(42-210) molar ratio should provide a useful parameter to optimize FXN augmentation and replacement therapies.


Subject(s)
Friedreich Ataxia/metabolism , Gene Expression Regulation , Iron-Binding Proteins/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Protein Precursors/biosynthesis , Adolescent , Adult , Carbon-Sulfur Lyases/genetics , Carbon-Sulfur Lyases/metabolism , Cell Line, Transformed , Child , Female , Friedreich Ataxia/genetics , Humans , Iron/metabolism , Iron-Binding Proteins/genetics , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Male , Mitochondria/genetics , Mitochondrial Proteins/genetics , Protein Biosynthesis/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Precursors/genetics , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Frataxin
19.
Hum Mol Genet ; 17(15): 2265-73, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18424449

ABSTRACT

Friedreich ataxia (FA) is a progressive neurodegenerative disease caused by expansion of a trinucleotide repeat within the first intron of the gene that encodes frataxin. In our study, we investigated the regulation of frataxin expression by iron and demonstrated that frataxin mRNA levels decrease significantly in multiple human cell lines treated with the iron chelator, desferal (DFO). In addition, frataxin mRNA and protein levels decrease in fibroblast and lymphoblast cells derived from both normal controls and from patients with FA when treated with DFO. Lymphoblasts and fibroblasts of FA patients have evidence of cytosolic iron depletion, as indicated by increased levels of iron regulatory protein 2 (IRP2) and/or increased IRE-binding activity of IRP1. We postulate that this inferred cytosolic iron depletion occurs as frataxin-deficient cells overload their mitochondria with iron, a downstream regulatory effect that has been observed previously when mitochondrial iron-sulfur cluster assembly is disrupted. The mitochondrial iron overload and presumed cytosolic iron depletion potentially further compromise function in frataxin-deficient cells by decreasing frataxin expression. Thus, our results imply that therapeutic efforts should focus on an approach that combines iron removal from mitochondria with a treatment that increases cytosolic iron levels to maximize residual frataxin expression in FA patients.


Subject(s)
Friedreich Ataxia/metabolism , Iron Overload/metabolism , Iron-Binding Proteins/biosynthesis , Iron/metabolism , Mitochondria/metabolism , Cell Line , Cytosol/metabolism , Deferoxamine/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Friedreich Ataxia/genetics , Gene Expression Regulation , Humans , Iron Chelating Agents/pharmacology , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Iron-Binding Proteins/genetics , Lymphocytes/drug effects , Lymphocytes/metabolism , Mitochondria/drug effects , RNA, Messenger/metabolism , Frataxin
20.
Mol Cell Biol ; 40(23)2020 11 06.
Article in English | MEDLINE | ID: mdl-32989015

ABSTRACT

Fanconi anemia (FA) is a unique DNA damage repair pathway. To date, 22 genes have been identified that are associated with the FA pathway. A defect in any of those genes causes genomic instability, and the patients bearing the mutation become susceptible to cancer. In our earlier work, we identified that Fanconi anemia protein G (FANCG) protects the mitochondria from oxidative stress. In this report, we have identified eight patients having a mutation (C.65G>C), which converts arginine at position 22 to proline (p.Arg22Pro) in the N terminus of FANCG. The mutant protein, hFANCGR22P, is able to repair the DNA and able to retain the monoubiquitination of FANCD2 in the FANCGR22P/FGR22P cell. However, it lost mitochondrial localization and failed to protect mitochondria from oxidative stress. Mitochondrial instability in the FANCGR22P cell causes the transcriptional downregulation of mitochondrial iron-sulfur cluster biogenesis protein frataxin (FXN) and the resulting iron deficiency of FA protein FANCJ, an iron-sulfur-containing helicase involved in DNA repair.


Subject(s)
Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Genomic Instability/genetics , Iron-Binding Proteins/biosynthesis , Mitochondria/pathology , RNA Helicases/genetics , Amino Acid Sequence/genetics , Cell Line, Tumor , DNA Damage/genetics , DNA Repair/genetics , Down-Regulation/genetics , Fanconi Anemia/genetics , Fanconi Anemia/pathology , HEK293 Cells , HeLa Cells , Humans , Iron-Binding Proteins/genetics , Iron-Sulfur Proteins/genetics , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL