Altered gene expression and DNA damage in peripheral blood cells from Friedreich's ataxia patients: cellular model of pathology

Astrid C Haugen, Nicholas A Di Prospero, Joel S Parker, Rick D Fannin, Jeff Chou, Joel N Meyer, Christopher Halweg, Jennifer B Collins, Alexandra Durr, Kenneth Fischbeck, Bennett Van Houten, Astrid C Haugen, Nicholas A Di Prospero, Joel S Parker, Rick D Fannin, Jeff Chou, Joel N Meyer, Christopher Halweg, Jennifer B Collins, Alexandra Durr, Kenneth Fischbeck, Bennett Van Houten

Abstract

The neurodegenerative disease Friedreich's ataxia (FRDA) is the most common autosomal-recessively inherited ataxia and is caused by a GAA triplet repeat expansion in the first intron of the frataxin gene. In this disease, transcription of frataxin, a mitochondrial protein involved in iron homeostasis, is impaired, resulting in a significant reduction in mRNA and protein levels. Global gene expression analysis was performed in peripheral blood samples from FRDA patients as compared to controls, which suggested altered expression patterns pertaining to genotoxic stress. We then confirmed the presence of genotoxic DNA damage by using a gene-specific quantitative PCR assay and discovered an increase in both mitochondrial and nuclear DNA damage in the blood of these patients (p<0.0001, respectively). Additionally, frataxin mRNA levels correlated with age of onset of disease and displayed unique sets of gene alterations involved in immune response, oxidative phosphorylation, and protein synthesis. Many of the key pathways observed by transcription profiling were downregulated, and we believe these data suggest that patients with prolonged frataxin deficiency undergo a systemic survival response to chronic genotoxic stress and consequent DNA damage detectable in blood. In conclusion, our results yield insight into the nature and progression of FRDA, as well as possible therapeutic approaches. Furthermore, the identification of potential biomarkers, including the DNA damage found in peripheral blood, may have predictive value in future clinical trials.

Trial registration: ClinicalTrials.gov NCT00229632.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Selected gene classifications according to…
Figure 1. Selected gene classifications according to biological processes.
Significantly regulated genes (SAM FDRp≤0.05 in updown, over, or under [expression] output lists). The percent of total (displayed with a gray ball) is based on the number of significantly changed genes out of the total number of genes assigned to each gene ontology term. The change in expression for each GO term is depicted in yellow for upregulation and blue for downregulation.
Figure 2. Analogous gene expression responses in…
Figure 2. Analogous gene expression responses in blood from children and adults with FRDA.
(A) A common gene signature is representative of a genotoxic stress response found by Gene Set Analysis. Twenty-three genotoxic stress response gene sets were searched for common genes. This heat map, generated by unsupervised clustering, displays the genes present in at least 6 of the 23 gene sets and compares transcript levels between FRDA patients, the adult and children cohorts, and controls. Yellow = upregulated; Blue = downregulated. C = Control; P = Patient. Note that while three patients and one control did not segregate with their respective groups, all adult patients and controls clustered in two separate groups in this unsupervised clustering. (B) Heat-map generated by unsupervised clustering of FRDA and control samples, which displays the overlap of significantly differentially expressed genes (SAM FDRp≤0.007). C = control; P = patient. (C) A selected list of significant GO groups representing the overlap gene list described in (B). All controls used for comparison to the FRDA children are young adults (see Table S1). The percent of total (displayed with a gray ball) is based on the number of significantly changed genes out of the total number of genes assigned to each gene ontology term. The gene number for each GO group is shown with a blue bar, the intensity of which is indicative of the p-value.
Figure 3. Nuclear and mitochondrial DNA damage…
Figure 3. Nuclear and mitochondrial DNA damage are identified by QPCR analysis of blood DNA from 47 patients with FRDA and 15 controls.
These data represent the number of excess lesions found per 10 kb of DNA from both mtDNA and nDNA genomes in FRDA patients as compared to the controls. A significant number of nuclear (0.53 lesions/10 kb) and mitochondrial DNA lesions (0.81 lesions/10 kb) were observed (p<0.0001, respectively, by Mann-Whitney U test). There is a significantly higher number of mitochondrial lesions than nuclear lesions (p<0.002 by Mann-Whitney U test), and both types of lesions are highly correlated (p<0.0001 by Spearman's Rank Correlation). Error bars represent the standard error of the mean.
Figure 4. Patients with lower levels of…
Figure 4. Patients with lower levels of frataxin correlate with age of onset of disease and have more compromised mitochondrial and protein biosynthetic function.
(A) Real-time PCR of frataxin levels in all patients with available RNA (27) were compared to controls (10). Levels of frataxin are relative to the average ΔCT of the controls (dotted line). The error bars represent standard deviations. Brackets encompass the patients stratified by their expression distribution into those expressing higher levels of frataxin (n = 6) and those expressing lower levels of frataxin (n = 21) (see Figure S1). (B) Global gene expression changes were analyzed in the patients with low levels of frataxin vs. patients with high levels of frataxin. Significantly differentially expressed genes (SAM FDR≤8%; p≤0.05; n = 973) were further examined for gene ontology groups categorized by biological process. The same list of genes was analyzed with IPA (Ingenuity Systems), which identified significant biological functions and canonical pathways. All gene groups contain mostly downregulated genes, indicating compromised mitochondrial and biosynthetic function in patients with the lowest expression of frataxin. (C) Age of onset plotted against the Real-time PCR frataxin levels yields a correlation of r2 = 0.305 (p = 0.00277).
Figure 5. Model of Friedreich's ataxia pathology…
Figure 5. Model of Friedreich's ataxia pathology based on this study.
Data presented in this study are consistent with a dysregulation of mitochondrial function, decreased oxidative phosphorylation, increased ROS production, and subsequent mitochondrial and nuclear DNA damage. These factors contribute to decreased signaling and altered DNA transactions, which are likely to result in subsequent loss of protein synthesis and decreased protein degradation, as suggested in the transcription profiling. These alterations may cause tissue damage, altered immune response, and the clinical pathology associated with FRDA.

References

    1. Campuzano V, Montermini L, Lutz Y, Cova L, Hindelang C, et al. Frataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranes. Hum Mol Genet. 1997;6:1771–1780.
    1. Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, et al. Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science. 1996;271:1423–1427.
    1. Ohshima K, Montermini L, Wells RD, Pandolfo M. Inhibitory effects of expanded GAA.TTC triplet repeats from intron I of the Friedreich ataxia gene on transcription and replication in vivo. J Biol Chem. 1998;273:14588–14595.
    1. Bidichandani SI, Ashizawa T, Patel PI. The GAA triplet-repeat expansion in Friedreich ataxia interferes with transcription and may be associated with an unusual DNA structure. Am J Hum Genet. 1998;62:111–121.
    1. Muhlenhoff U, Richhardt N, Ristow M, Kispal G, Lill R. The yeast frataxin homolog Yfh1p plays a specific role in the maturation of cellular Fe/S proteins. Hum Mol Genet. 2002;11:2025–2036.
    1. Stehling O, Elsasser HP, Bruckel B, Muhlenhoff U, Lill R. Iron-sulfur protein maturation in human cells: evidence for a function of frataxin. Hum Mol Genet. 2004;13:3007–3015.
    1. Schulz JB, Dehmer T, Schols L, Mende H, Hardt C, et al. Oxidative stress in patients with Friedreich ataxia. Neurology. 2000;55:1719–1721.
    1. Wong A, Yang J, Cavadini P, Gellera C, Lonnerdal B, et al. The Friedreich's ataxia mutation confers cellular sensitivity to oxidant stress which is rescued by chelators of iron and calcium and inhibitors of apoptosis. Hum Mol Genet. 1999;8:425–430.
    1. Chantrel-Groussard K, Geromel V, Puccio H, Koenig M, Munnich A, et al. Disabled early recruitment of antioxidant defenses in Friedreich's ataxia. Hum Mol Genet. 2001;10:2061–2067.
    1. Shoichet SA, Baumer AT, Stamenkovic D, Sauer H, Pfeiffer AF, et al. Frataxin promotes antioxidant defense in a thiol-dependent manner resulting in diminished malignant transformation in vitro. Hum Mol Genet. 2002;11:815–821.
    1. Gakh O, Park S, Liu G, Macomber L, Imlay JA, et al. Mitochondrial iron detoxification is a primary function of frataxin that limits oxidative damage and preserves cell longevity. Hum Mol Genet. 2006;15:467–479.
    1. Napoli E, Taroni F, Cortopassi GA. Frataxin, iron-sulfur clusters, heme, ROS, and aging. Antioxid Redox Signal. 2006;8:506–516.
    1. Puccio H, Simon D, Cossee M, Criqui-Filipe P, Tiziano F, et al. Mouse models for Friedreich ataxia exhibit cardiomyopathy, sensory nerve defect and Fe-S enzyme deficiency followed by intramitochondrial iron deposits. Nat Genet. 2001;27:181–186.
    1. Calabrese V, Lodi R, Tonon C, D'Agata V, Sapienza M, et al. Oxidative stress, mitochondrial dysfunction and cellular stress response in Friedreich's ataxia. J Neurol Sci. 2005;233:145–162.
    1. Foury F, Cazzalini O. Deletion of the yeast homologue of the human gene associated with Friedreich's ataxia elicits iron accumulation in mitochondria. FEBS Lett. 1997;411:373–377.
    1. Karthikeyan G, Santos JH, Graziewicz MA, Copeland WC, Isaya G, et al. Reduction in frataxin causes progressive accumulation of mitochondrial damage. Hum Mol Genet. 2003;12:3331–3342.
    1. Lamarche JB, Cote M, Lemieux B. The cardiomyopathy of Friedreich's ataxia morphological observations in 3 cases. Can J Neurol Sci. 1980;7:389–396.
    1. Waldvogel D, van Gelderen P, Hallett M. Increased iron in the dentate nucleus of patients with Friedrich's ataxia. Ann Neurol. 1999;46:123–125.
    1. Emond M, Lepage G, Vanasse M, Pandolfo M. Increased levels of plasma malondialdehyde in Friedreich ataxia. Neurology. 2000;55:1752–1753.
    1. Bradley JL, Homayoun S, Hart PE, Schapira AH, Cooper JM. Role of oxidative damage in Friedreich's ataxia. Neurochem Res. 2004;29:561–567.
    1. Seznec H, Simon D, Bouton C, Reutenauer L, Hertzog A, et al. Friedreich ataxia: the oxidative stress paradox. Hum Mol Genet. 2005;14:463–474.
    1. Di Prospero NA, Baker A, Jeffries N, Fischbeck KH. Neurological effects of high-dose idebenone in patients with Friedreich's ataxia: a randomised, placebo-controlled trial. Lancet Neurol. 2007;6:878–886.
    1. Myers LM, Lynch DR, Farmer JM, Friedman LS, Lawson JA, et al. Urinary isoprostanes in Friedreich ataxia: lack of correlation with disease features. Mov Disord. 2008;23:1920–1922.
    1. Michael S, Petrocine SV, Qian J, Lamarche JB, Knutson MD, et al. Iron and iron-responsive proteins in the cardiomyopathy of Friedreich's ataxia. Cerebellum. 2006;5:257–267.
    1. Tan G, Napoli E, Taroni F, Cortopassi G. Decreased expression of genes involved in sulfur amino acid metabolism in frataxin-deficient cells. Hum Mol Genet. 2003;12:1699–1711.
    1. Coppola G, Choi SH, Santos MM, Miranda CJ, Tentler D, et al. Gene expression profiling in frataxin deficient mice: microarray evidence for significant expression changes without detectable neurodegeneration. Neurobiol Dis. 2006;22:302–311.
    1. Schoenfeld RA, Napoli E, Wong A, Zhan S, Reutenauer L, et al. Frataxin deficiency alters heme pathway transcripts and decreases mitochondrial heme metabolites in mammalian cells. Hum Mol Genet. 2005;14:3787–3799.
    1. Harrill AH, Watkins PB, Su S, Ross PK, Harbourt DE, et al. Mouse population-guided resequencing reveals that variants in CD44 contribute to acetaminophen-induced liver injury in humans. Genome Res 2009
    1. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98:5116–5121.
    1. Efron B, Tibshirani R. On testing the significance of sets of genes. Tech report. 2006:1–32.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550.
    1. Hu T, Gibson DP, Carr GJ, Torontali SM, Tiesman JP, et al. Identification of a gene expression profile that discriminates indirect-acting genotoxins from direct-acting genotoxins. Mutat Res. 2004;549:5–27.
    1. Santos JH, Meyer JN, Mandavilli BS, Van Houten B. Quantitative PCR-based measurement of nuclear and mitochondrial DNA damage and repair in mammalian cells. Methods Mol Biol. 2006;314:183–199.
    1. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci U S A. 1997;94:514–519.
    1. Santos JH, Hunakova L, Chen Y, Bortner C, Van Houten B. Cell sorting experiments link persistent mitochondrial DNA damage with loss of mitochondrial membrane potential and apoptotic cell death. J Biol Chem. 2003;278:1728–1734.
    1. Mandavilli BS, Boldogh I, Van Houten B. 3-nitropropionic acid-induced hydrogen peroxide, mitochondrial DNA damage, and cell death are attenuated by Bcl-2 overexpression in PC12 cells. Brain Res Mol Brain Res. 2005;133:215–223.
    1. Mandavilli BS, Ali SF, Van Houten B. DNA damage in brain mitochondria caused by aging and MPTP treatment. Brain Res. 2000;885:45–52.
    1. Salazar JJ, Van Houten B. Preferential mitochondrial DNA injury caused by glucose oxidase as a steady generator of hydrogen peroxide in human fibroblasts. Mutat Res. 1997;385:139–149.
    1. Van Houten B, Cheng S, Chen Y. Measuring gene-specific nucleotide excision repair in human cells using quantitative amplification of long targets from nanogram quantities of DNA. Mutat Res. 2000;460:81–94.
    1. Filla A, De Michele G, Cavalcanti F, Pianese L, Monticelli A, et al. The relationship between trinucleotide (GAA) repeat length and clinical features in Friedreich ataxia. Am J Hum Genet. 1996;59:554–560.
    1. Pianese L, Turano M, Lo Casale MS, De Biase I, Giacchetti M, et al. Real time PCR quantification of frataxin mRNA in the peripheral blood leucocytes of Friedreich ataxia patients and carriers. J Neurol Neurosurg Psychiatry. 2004;75:1061–1063.
    1. Chou JW, Zhou T, Kaufmann WK, Paules RS, Bushel PR. Extracting gene expression patterns and identifying co-expressed genes from microarray data reveals biologically responsive processes. BMC Bioinformatics. 2007;8:427.
    1. Trouillas P, Takayanagi T, Hallett M, Currier RD, Subramony SH, et al. International Cooperative Ataxia Rating Scale for pharmacological assessment of the cerebellar syndrome. The Ataxia Neuropharmacology Committee of the World Federation of Neurology. J Neurol Sci. 1997;145:205–211.
    1. Onuki J, Chen Y, Teixeira PC, Schumacher RI, Medeiros MH, et al. Mitochondrial and nuclear DNA damage induced by 5-aminolevulinic acid. Arch Biochem Biophys. 2004;432:178–187.
    1. Burnett R, Melander C, Puckett JW, Son LS, Wells RD, et al. DNA sequence-specific polyamides alleviate transcription inhibition associated with long GAA.TTC repeats in Friedreich's ataxia. Proc Natl Acad Sci U S A. 2006;103:11497–11502.
    1. Lodi R, Cooper JM, Bradley JL, Manners D, Styles P, et al. Deficit of in vivo mitochondrial ATP production in patients with Friedreich ataxia. Proc Natl Acad Sci U S A. 1999;96:11492–11495.
    1. Vorgerd M, Schols L, Hardt C, Ristow M, Epplen JT, et al. Mitochondrial impairment of human muscle in Friedreich ataxia in vivo. Neuromuscul Disord. 2000;10:430–435.
    1. Thierbach R, Schulz TJ, Isken F, Voigt A, Mietzner B, et al. Targeted disruption of hepatic frataxin expression causes impaired mitochondrial function, decreased life span and tumor growth in mice. Hum Mol Genet. 2005;14:3857–3864.
    1. Lewis PD, Corr JB, Arlett CF, Harcourt SA. Increased sensitivity to gamma irradiation of skin fibroblasts in Friedreich's ataxia. Lancet. 1979;2:474–475.
    1. Evans HJ, Vijayalaxmi, Pentland B, Newton MS. Mutagen hypersensitivity in Friedreich's ataxia. Ann Hum Genet. 1983;47:193–204.
    1. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000;408:239–247.
    1. Schulz TJ, Thierbach R, Voigt A, Drewes G, Mietzner B, et al. Induction of oxidative metabolism by mitochondrial frataxin inhibits cancer growth: Otto Warburg revisited. J Biol Chem. 2006;281:977–981.
    1. De Pas T, Martinelli G, De Braud F, Peccatori F, Catania C, et al. Friedreich's ataxia and intrathecal chemotherapy in a patient with lymphoblastic lymphoma. Ann Oncol. 1999;10:1393.
    1. Kidd A, Coleman R, Whiteford M, Barron LH, Simpson SA, et al. Breast cancer in two sisters with Friedreich's ataxia. Eur J Surg Oncol. 2001;27:512–514.
    1. Ackroyd R, Shorthouse AJ, Stephenson TJ. Gastric carcinoma in siblings with Friedreich's ataxia. Eur J Surg Oncol. 1996;22:301–303.
    1. Barr H, Page R, Taylor W. Primary small bowel ganglioneuroblastoma and Friedreich's ataxia. J R Soc Med. 1986;79:612–613.
    1. Lill R, Muhlenhoff U. Maturation of iron-sulfur proteins in eukaryotes: mechanisms, connected processes, and diseases. Annu Rev Biochem. 2008;77:669–700.
    1. Klinge S, Hirst J, Maman JD, Krude T, Pellegrini L. An iron-sulfur domain of the eukaryotic primase is essential for RNA primer synthesis. Nat Struct Mol Biol. 2007;14:875–877.
    1. Lukianova OA, David SS. A role for iron-sulfur clusters in DNA repair. Curr Opin Chem Biol. 2005;9:145–151.
    1. Rudolf J, Makrantoni V, Ingledew WJ, Stark MJ, White MF. The DNA repair helicases XPD and FancJ have essential iron-sulfur domains. Mol Cell. 2006;23:801–808.
    1. Sturm B, Bistrich U, Schranzhofer M, Sarsero JP, Rauen U, et al. Friedreich's ataxia, no changes in mitochondrial labile iron in human lymphoblasts and fibroblasts: a decrease in antioxidative capacity? J Biol Chem. 2005;280:6701–6708.
    1. Paupe V, Dassa EP, Goncalves S, Auchere F, Lonn M, et al. Impaired nuclear Nrf2 translocation undermines the oxidative stress response in friedreich ataxia. PLoS ONE. 2009;4:e4253.
    1. Niedernhofer LJ, Garinis GA, Raams A, Lalai AS, Robinson AR, et al. A new progeroid syndrome reveals that genotoxic stress suppresses the somatotroph axis. Nature. 2006;444:1038–1043.
    1. Paschen W, Proud CG, Mies G. Shut-down of translation, a global neuronal stress response: mechanisms and pathological relevance. Curr Pharm Des. 2007;13:1887–1902.
    1. Mukhopadhyay D, Riezman H. Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science. 2007;315:201–205.
    1. Herrmann J, Lerman LO, Lerman A. Ubiquitin and ubiquitin-like proteins in protein regulation. Circ Res. 2007;100:1276–1291.
    1. Zeeberg BR, Feng W, Wang G, Wang MD, Fojo AT, et al. GoMiner: a resource for biological interpretation of genomic and proteomic data. Genome Biol. 2003;4:R28.
    1. Dennis G, Jr, Sherman BT, Hosack DA, Yang J, Gao W, et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003;4:P3.
    1. Eisen MB, Spellman PT, Brown PO, Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci U S A. 1998;95:14863–14868.
    1. Heid CA, Stevens J, Livak KJ, Williams PM. Real time quantitative PCR. Genome Res. 1996;6:986–994.

Source: PubMed

3
Abonner