Gene Expression Differences in Peripheral Blood of Parkinson's Disease Patients with Distinct Progression Profiles

Raquel Pinho, Leonor C Guedes, Lilach Soreq, Patrícia P Lobo, Tiago Mestre, Miguel Coelho, Mário M Rosa, Nilza Gonçalves, Pauline Wales, Tiago Mendes, Ellen Gerhardt, Christiane Fahlbusch, Vincenzo Bonifati, Michael Bonin, Gabriel Miltenberger-Miltényi, Fran Borovecki, Hermona Soreq, Joaquim J Ferreira, Tiago F Outeiro, Raquel Pinho, Leonor C Guedes, Lilach Soreq, Patrícia P Lobo, Tiago Mestre, Miguel Coelho, Mário M Rosa, Nilza Gonçalves, Pauline Wales, Tiago Mendes, Ellen Gerhardt, Christiane Fahlbusch, Vincenzo Bonifati, Michael Bonin, Gabriel Miltenberger-Miltényi, Fran Borovecki, Hermona Soreq, Joaquim J Ferreira, Tiago F Outeiro

Abstract

The prognosis of neurodegenerative disorders is clinically challenging due to the inexistence of established biomarkers for predicting disease progression. Here, we performed an exploratory cross-sectional, case-control study aimed at determining whether gene expression differences in peripheral blood may be used as a signature of Parkinson's disease (PD) progression, thereby shedding light into potential molecular mechanisms underlying disease development. We compared transcriptional profiles in the blood from 34 PD patients who developed postural instability within ten years with those of 33 patients who did not develop postural instability within this time frame. Our study identified >200 differentially expressed genes between the two groups. The expression of several of the genes identified was previously found deregulated in animal models of PD and in PD patients. Relevant genes were selected for validation by real-time PCR in a subset of patients. The genes validated were linked to nucleic acid metabolism, mitochondria, immune response and intracellular-transport. Interestingly, we also found deregulation of these genes in a dopaminergic cell model of PD, a simple paradigm that can now be used to further dissect the role of these molecular players on dopaminergic cell loss. Altogether, our study provides preliminary evidence that expression changes in specific groups of genes and pathways, detected in peripheral blood samples, may be correlated with differential PD progression. Our exploratory study suggests that peripheral gene expression profiling may prove valuable for assisting in prediction of PD prognosis, and identifies novel culprits possibly involved in dopaminergic cell death. Given the exploratory nature of our study, further investigations using independent, well-characterized cohorts will be essential in order to validate our candidates as predictors of PD prognosis and to definitively confirm the value of gene expression analysis in aiding patient stratification and therapeutic intervention.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Hierarchical clustering of genes detected…
Fig 1. Hierarchical clustering of genes detected as differentially expressed in rapid versus slow progression PD patients.
Classification based on expression signals of the bootstrapping t-test-detected genes resulted in only five mis-classified slow and 11 mis-classified rapid progression patients. The color bar denotes z-score adjusted expression values, green used for lower expression and purple for higher expression levels. Eucledian distance and average linkage methods were used.
Fig 2. Functional network of the differentially…
Fig 2. Functional network of the differentially expressed genes altered in PD patients having slow versus rapid disease progression.
GO network analysis on the GO biological process terms that were found as highly enriched in blood cells from slow versus rapid progression patients. The node circle size represents the number of altered genes in the category, and the categories colours correspond to the functional group.
Fig 3. Selected genes investigated by qPCR…
Fig 3. Selected genes investigated by qPCR in PD patients-derived samples.
-ΔCts plotted for 10 genes chosen for qPCR validation in cDNA obtained from 16 patients with either slow or rapid progression of the disease. Data is expressed as means ± SD of triplicates. T-test was used for statistical analysis with significance level of p

Fig 4. Selected genes investigated in a…

Fig 4. Selected genes investigated in a LUHMES/MPP + cell model of PD.

(A) LUHMES cells…
Fig 4. Selected genes investigated in a LUHMES/MPP+ cell model of PD.
(A) LUHMES cells treated with solvent (DMSO) or 2.5μM MPP+ stained for Tyrosine Hydroxylase (TH), TUJ1 and nucleus or (B) observed in bright field, showed robust loss of neurite integrity upon MPP+ treatment. (C) Cell viability analysis assessed by ToxiLigh assay revealed a significant increase of adenylate kinase content in the supernatants of MPP+ treated cells. (D) -Δcts plotted for seven genes chosen for validation using D8 differentiated LUHMES cells exposed to 2.5μm MPP+. (E) Correlation between fold-change expression values obtained in LUHMES/MPP+ model and PD patients. Data is expressed as means ± SD of triplicate samples. T-test was used for statistical analysis with significance level of p<0.05. *p<0.05; **p<0.01. Scale bar 100 μm.
Fig 4. Selected genes investigated in a…
Fig 4. Selected genes investigated in a LUHMES/MPP+ cell model of PD.
(A) LUHMES cells treated with solvent (DMSO) or 2.5μM MPP+ stained for Tyrosine Hydroxylase (TH), TUJ1 and nucleus or (B) observed in bright field, showed robust loss of neurite integrity upon MPP+ treatment. (C) Cell viability analysis assessed by ToxiLigh assay revealed a significant increase of adenylate kinase content in the supernatants of MPP+ treated cells. (D) -Δcts plotted for seven genes chosen for validation using D8 differentiated LUHMES cells exposed to 2.5μm MPP+. (E) Correlation between fold-change expression values obtained in LUHMES/MPP+ model and PD patients. Data is expressed as means ± SD of triplicate samples. T-test was used for statistical analysis with significance level of p<0.05. *p<0.05; **p<0.01. Scale bar 100 μm.

References

    1. de Lau LM, Breteler MM. Epidemiology of Parkinson's disease. Lancet Neurol. 2006;5(6):525–35. Epub 2006/05/23. doi: S1474-4422(06)70471-9 [pii] 10.1016/S1474-4422(06)70471-9 .
    1. Newman EJ, Breen K, Patterson J, Hadley DM, Grosset KA, Grosset DG. Accuracy of Parkinson's disease diagnosis in 610 general practice patients in the West of Scotland. Mov Disord. 2009;24(16):2379–85. Epub 2009/11/06. 10.1002/mds.22829 .
    1. Schrag A, Ben-Shlomo Y, Quinn N. How valid is the clinical diagnosis of Parkinson's disease in the community? J Neurol Neurosurg Psychiatry. 2002;73(5):529–34. Epub 2002/10/25. .
    1. Tolosa E, Wenning G, Poewe W. The diagnosis of Parkinson's disease. The Lancet Neurology. 2006;5(1):75–86. 10.1016/S1474-4422(05)70285-4 .
    1. Evans JR, Mason SL, Williams-Gray CH, Foltynie T, Brayne C, Robbins TW, et al. The natural history of treated Parkinson's disease in an incident, community based cohort. Journal of neurology, neurosurgery, and psychiatry. 2011;82(10):1112–8. 10.1136/jnnp.2011.240366 .
    1. Shtilbans A, Henchcliffe C. Biomarkers in Parkinson's disease: an update. Curr Opin Neurol. 2012;25(4):460–5. Epub 2012/07/10. 10.1097/WCO.0b013e3283550c0d 00019052-201208000-00012 [pii]. .
    1. Gotovac K, Hajnsek S, Pasic MB, Pivac N, Borovecki F. Personalized medicine in neurodegenerative diseases: how far away? Mol Diagn Ther. 2014;18(1):17–24. Epub 2013/10/15. 10.1007/s40291-013-0058-z .
    1. Borovecki F, Lovrecic L, Zhou J, Jeong H, Then F, Rosas HD, et al. Genome-wide expression profiling of human blood reveals biomarkers for Huntington's disease. Proc Natl Acad Sci U S A. 2005;102(31):11023–8. Epub 2005/07/27. 0504921102 [pii] 10.1073/pnas.0504921102 .
    1. Soreq L, Bergman H, Goll Y, Greenberg DS, Israel Z, Soreq H. Deep brain stimulation induces rapidly reversible transcript changes in Parkinson's leucocytes. J Cell Mol Med. 2012;16(7):1496–507. 10.1111/j.1582-4934.2011.01444.x .
    1. Soreq L, Guffanti A, Salomonis N, Simchovitz A, Israel Z, Bergman H, et al. Long non-coding RNA and alternative splicing modulations in Parkinson's leukocytes identified by RNA sequencing. PLoS computational biology. 2014;10(3):e1003517 10.1371/journal.pcbi.1003517
    1. Scherzer CR, Eklund AC, Morse LJ, Liao Z, Locascio JJ, Fefer D, et al. Molecular markers of early Parkinson's disease based on gene expression in blood. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(3):955–60. 10.1073/pnas.0610204104
    1. Soreq L, Israel Z, Bergman H, Soreq H. Advanced microarray analysis highlights modified neuro-immune signaling in nucleated blood cells from Parkinson's disease patients. Journal of neuroimmunology. 2008;201–202:227–36. 10.1016/j.jneuroim.2008.06.019 .
    1. Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson's disease: a clinico-pathological study of 100 cases. Journal of neurology, neurosurgery, and psychiatry. 1992;55(3):181–4.
    1. Hely MA, Morris JG, Traficante R, Reid WG, O'Sullivan DJ, Williamson PM. The sydney multicentre study of Parkinson's disease: progression and mortality at 10 years. Journal of neurology, neurosurgery, and psychiatry. 1999;67(3):300–7.
    1. Williams-Gray CH, Mason SL, Evans JR, Foltynie T, Brayne C, Robbins TW, et al. The CamPaIGN study of Parkinson's disease: 10-year outlook in an incident population-based cohort. Journal of neurology, neurosurgery, and psychiatry. 2013;84(11):1258–64. 10.1136/jnnp-2013-305277 .
    1. Kempster PA, Williams DR, Selikhova M, Holton J, Revesz T, Lees AJ. Patterns of levodopa response in Parkinson's disease: a clinico-pathological study. Brain. 2007;130(Pt 8):2123–8. 10.1093/brain/awm142 .
    1. Goetz CG, Tilley BC, Shaftman SR, Stebbins GT, Fahn S, Martinez-Martin P, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord. 2008;23(15):2129–70. 10.1002/mds.22340 .
    1. Smyth GK. Limma: Linear Models for Microarray Data In: Gentleman Robert, Carey Vincent J., Huber Wolfgang, Irizarry Rafael A., Dudoit S, editors. Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Statistics for Biology and Health: Springer; New York; 2005. p. 397–420.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8(1):118–27. Epub 2006/04/25. 10.1093/biostatistics/kxj037 .
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic acids research. 2009;37(1):1–13. 10.1093/nar/gkn923
    1. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome research. 2003;13(11):2498–504. 10.1101/gr.1239303
    1. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 2009;25(8):1091–3. 10.1093/bioinformatics/btp101
    1. Scholz D, Poltl D, Genewsky A, Weng M, Waldmann T, Schildknecht S, et al. Rapid, complete and large-scale generation of post-mitotic neurons from the human LUHMES cell line. J Neurochem. 2011;119(5):957–71. Epub 2011/03/26. 10.1111/j.1471-4159.2011.07255.x .
    1. Rozen S, Skaletsky H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 2000;132:365–86. Epub 1999/11/05. .
    1. Dheda K, Huggett JF, Bustin SA, Johnson MA, Rook G, Zumla A. Validation of housekeeping genes for normalizing RNA expression in real-time PCR. Biotechniques. 2004;37(1):112–4, 6, 8–9. Epub 2004/07/31. .
    1. Tomlinson CL, Stowe R, Patel S, Rick C, Gray R, Clarke CE. Systematic review of levodopa dose equivalency reporting in Parkinson's disease. Mov Disord. 2010;25(15):2649–53. 10.1002/mds.23429 .
    1. Klein C, Westenberger A. Genetics of Parkinson's disease. Cold Spring Harbor perspectives in medicine. 2012;2(1):a008888 10.1101/cshperspect.a008888
    1. Abbas N, Lucking CB, Ricard S, Durr A, Bonifati V, De Michele G, et al. A wide variety of mutations in the parkin gene are responsible for autosomal recessive parkinsonism in Europe. French Parkinson's Disease Genetics Study Group and the European Consortium on Genetic Susceptibility in Parkinson's Disease. Hum Mol Genet. 1999;8(4):567–74. .
    1. Soreq L, Bergman H, Israel Z, Soreq H. Exon arrays reveal alternative splicing aberrations in Parkinson's disease leukocytes. Neuro-degenerative diseases. 2012;10(1–4):203–6. 10.1159/000332598 .
    1. Soreq L, Ben-Shaul Y, Israel Z, Bergman H, Soreq H. Meta-analysis of genetic and environmental Parkinson's disease models reveals a common role of mitochondrial protection pathways. Neurobiol Dis. 2012;45(3):1018–30. Epub 2011/12/27. 10.1016/j.nbd.2011.12.021 S0969-9961(11)00396-2 [pii]. .
    1. Dumitriu A, Latourelle JC, Hadzi TC, Pankratz N, Garza D, Miller JP, et al. Gene expression profiles in Parkinson disease prefrontal cortex implicate FOXO1 and genes under its transcriptional regulation. PLoS genetics. 2012;8(6):e1002794 10.1371/journal.pgen.1002794
    1. Hauser MA, Li YJ, Xu H, Noureddine MA, Shao YS, Gullans SR, et al. Expression profiling of substantia nigra in Parkinson disease, progressive supranuclear palsy, and frontotemporal dementia with parkinsonism. Arch Neurol. 2005;62(6):917–21. 10.1001/archneur.62.6.917 .
    1. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, et al. PGC-1alpha, a potential therapeutic target for early intervention in Parkinson's disease. Science translational medicine. 2010;2(52):52ra73 10.1126/scitranslmed.3001059
    1. Conn KJ, Ullman MD, Larned MJ, Eisenhauer PB, Fine RE, Wells JM. cDNA microarray analysis of changes in gene expression associated with MPP+ toxicity in SH-SY5Y cells. Neurochemical research. 2003;28(12):1873–81. .
    1. Locascio JJ, Eberly S, Liao Z, Liu G, Hoesing AN, Duong K, et al. Association between alpha-synuclein blood transcripts and early, neuroimaging-supported Parkinson's disease. Brain. 2015;138(Pt 9):2659–71. 10.1093/brain/awv202
    1. Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219(4587):979–80. .
    1. Jenner P. The MPTP-treated primate as a model of motor complications in PD: primate model of motor complications. Neurology. 2003;61(6 Suppl 3):S4–11. .
    1. Harvey SH, Sheedy DM, Cuddihy AR, O'Connell MJ. Coordination of DNA damage responses via the Smc5/Smc6 complex. Mol Cell Biol. 2004;24(2):662–74. Epub 2004/01/01. .
    1. Hirano T. The ABCs of SMC proteins: two-armed ATPases for chromosome condensation, cohesion, and repair. Genes Dev. 2002;16(4):399–414. Epub 2002/02/19. 10.1101/gad.955102 .
    1. Parenti R, Paratore S, Torrisi A, Cavallaro S. A natural antisense transcript against Rad18, specifically expressed in neurons and upregulated during beta-amyloid-induced apoptosis. Eur J Neurosci. 2007;26(9):2444–57. Epub 2007/11/01. EJN5864 [pii] 10.1111/j.1460-9568.2007.05864.x .
    1. Francisconi S, Codenotti M, Ferrari-Toninelli G, Uberti D, Memo M. Preservation of DNA integrity and neuronal degeneration. Brain Res Brain Res Rev. 2005;48(2):347–51. Epub 2005/04/27. S0165-0173(04)00201-2 [pii] 10.1016/j.brainresrev.2004.12.023 .
    1. Uberti D, Ferrari Toninelli G, Memo M. Involvement of DNA damage and repair systems in neurodegenerative process. Toxicol Lett. 2003;139(2–3):99–105. Epub 2003/03/12. S037842740200423X [pii]. .
    1. Rolig RL, McKinnon PJ. Linking DNA damage and neurodegeneration. Trends Neurosci. 2000;23(9):417–24. Epub 2000/08/15. S0166-2236(00)01625-8 [pii]. .
    1. Jeppesen DK, Bohr VA, Stevnsner T. DNA repair deficiency in neurodegeneration. Prog Neurobiol. 2011;94(2):166–200. Epub 2011/05/10. 10.1016/j.pneurobio.2011.04.013 S0301-0082(11)00064-5 [pii]. .
    1. Hu H, Wang B, Borde M, Nardone J, Maika S, Allred L, et al. Foxp1 is an essential transcriptional regulator of B cell development. Nat Immunol. 2006;7(8):819–26. Epub 2006/07/05. ni1358 [pii] 10.1038/ni1358 .
    1. Feng X, Ippolito GC, Tian L, Wiehagen K, Oh S, Sambandam A, et al. Foxp1 is an essential transcriptional regulator for the generation of quiescent naive T cells during thymocyte development. Blood. 2010;115(3):510–8. Epub 2009/12/08. 10.1182/blood-2009-07-232694 [pii]. .
    1. Jackson BC, Carpenter C, Nebert DW, Vasiliou V. Update of human and mouse forkhead box (FOX) gene families. Hum Genomics. 2010;4(5):345–52. Epub 2010/07/24. 2611113G3H18HNN3 [pii]. .
    1. Lee JY, Parks JS. ATP-binding cassette transporter AI and its role in HDL formation. Curr Opin Lipidol. 2005;16(1):19–25. Epub 2005/01/15. 00041433-200502000-00005 [pii]. .
    1. Krimbou L, Marcil M, Genest J. New insights into the biogenesis of human high-density lipoproteins. Curr Opin Lipidol. 2006;17(3):258–67. Epub 2006/05/09. 10.1097/01.mol.0000226117.43178.7c 00041433-200606000-00009 [pii]. .
    1. Mena NP, Bulteau AL, Salazar J, Hirsch EC, Nunez MT. Effect of mitochondrial complex I inhibition on Fe-S cluster protein activity. Biochem Biophys Res Commun. 2011;409(2):241–6. 10.1016/j.bbrc.2011.04.137 .
    1. Tanji K, Tanaka T, Mori F, Kito K, Takahashi H, Wakabayashi K, et al. NUB1 suppresses the formation of Lewy body-like inclusions by proteasomal degradation of synphilin-1. Am J Pathol. 2006;169(2):553–65. Epub 2006/08/01. S0002-9440(10)62737-X [pii] 10.2353/ajpath.2006.051067 .
    1. Richet E, Pooler AM, Rodriguez T, Novoselov SS, Schmidtke G, Groettrup M, et al. NUB1 modulation of GSK3beta reduces tau aggregation. Hum Mol Genet. 2012;21(24):5254–67. Epub 2012/09/12. 10.1093/hmg/dds376 [pii]. .
    1. Tanji K, Mori F, Kito K, Kakita A, Mimura J, Itoh K, et al. Synphilin-1-binding protein NUB1 is colocalized with nonfibrillar, proteinase K-resistant alpha-synuclein in presynapses in Lewy body disease. J Neuropathol Exp Neurol. 2011;70(10):879–89. Epub 2011/09/23. 10.1097/NEN.0b013e3182303745 00005072-201110000-00005 [pii]. .
    1. Bendor J, Lizardi-Ortiz JE, Westphalen RI, Brandstetter M, Hemmings HC Jr, Sulzer D, et al. AGAP1/AP-3-dependent endocytic recycling of M5 muscarinic receptors promotes dopamine release. EMBO J. 2010;29(16):2813–26. Epub 2010/07/29. 10.1038/emboj.2010.154 [pii]. .
    1. Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome biology. 2003;4(5):P3 Epub 2003/05/08.
    1. Hosack DA, Dennis G Jr, Sherman BT, Lane HC, Lempicki RA. Identifying biological themes within lists of genes with EASE. Genome biology. 2003;4(10):R70 Epub 2003/10/02. 10.1186/gb-2003-4-10-r70

Source: PubMed

3
Abonnieren