Skip to main content

Advertisement

Log in

Evaluation of Long Non-coding RNA Expression Profiles in Peripheral Blood Mononuclear Cells of Patients with Parkinson’s Disease

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

As in many biological processes, the long non-coding RNAs (lncRNA) are currently known to have important roles in Parkinson’s disease (PD). The aim of the study is to evaluate differentiated expressions of lncRNAs and their target mRNAs in the peripheral blood cells of individuals with Parkinson’s disease. The peripheral blood samples were taken from 10 Parkinson’s diagnosed people aging 50 years and more and from 10 healthy people as for the control group. Total RNA was isolated from peripheral blood mononuclear cells (PBMC), and a total of 5 samples were selected and evaluated by microarray analysis. lncRNAs with high fold change (fc < 1.5/fc > 1.5) were determined as a result of the analysis. Following this, the expression changes of some lncRNAs and their target mRNAs were examined by quantitative simultaneous polymerase chain reaction (qRT-PCR) in all individuals in the patient and control groups. Also, in order to determine the molecular level basic activities of lncRNAs determined by microarray analysis and which biological process and biochemical pathway they were in, Gene Ontology (GO) analysis (http://geneontology.org/) database was used. Thirteen upregulated and 31 downregulated lncRNAs whose expression changes were determined by microarray analysis and confirmed by qRT-PCR method were found in Parkinson’s patients. As they were evaluated by GO analysis, lncRNAs were expressed differently in patient and control groups and they are found to be related with the processes such as macromolecule metabolic processes, immune system, gene expression, cell activation, ATPase activity, DNA packaging complex, signal receptor activity, immune receptor activity, and protein binding were found to be significant.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

We’re sorry, something doesn't seem to be working properly.

Please try refreshing the page. If that doesn't work, please contact support so we can address the problem.

Data Availability

All the data generated and analyzed during this study are included in the published article.

References

  1. Wirdefeldt K, Adami HO, Cole P, Trichopoulos D, Mandel J (2011) Epidemiology and etiology of Parkinson’s disease: a review of the evidence. Eur J Epidemiol 26(1):1–58

    Article  Google Scholar 

  2. Schapira AHV, Chaudhuri KR, Jenner P (2017) Non-motor features of Parkinson disease. Nat Rev Neurosci 18(7):435–450

    Article  CAS  PubMed  Google Scholar 

  3. Chaudhuri KR, Prieto-Jurcynska C, Naidu Y, Mitra T, Frades-Payo B, Tluk S, Ruessmann A, Odin P et al (2010) The nondeclaration of nonmotor symptoms of Parkinson’s disease to health care professionals: an international study using the nonmotor symptoms questionnaire. Mov Disord 25(6):704–709

    Article  PubMed  Google Scholar 

  4. Miller DB, O’Callaghan JP (2015) Biomarkers of Parkinson’s disease: present and future. Metabolism 64(3):40–46

    Article  Google Scholar 

  5. Niu M, Xu R, Wang J, Hou B, Xie A (2016) MiR-133b ameliorates axon degeneration induced by MPP(+) via targeting RhoA. Neuroscience 325:39–49

    Article  CAS  PubMed  Google Scholar 

  6. Kim J, Fiesel FC, Belmonte KC, Hudec R, Wang WX, Kim C, Nelson PT, Springer W et al (2016) miR-27a and miR-27b regulate autophagic clearance of damaged mitochondria by targeting PTEN-induced putative kinase 1 (PINK1). Mol Neurodegener 11(55):1–15

    Google Scholar 

  7. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J et al (2001) Initial sequencing and analysis of the human genome. Nature 409(6822):860–921

    Article  CAS  PubMed  Google Scholar 

  8. Zhang P, Wu W, Chen Q, Chen M (2019) Non-coding RNAs and their ıntegrated networks. J Integr Bioinform 16(3):1–12

    Article  Google Scholar 

  9. Fatica A, Bozzoni I (2013) Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet 15(1):7–21

    Article  PubMed  Google Scholar 

  10. Mercer TR, Dinger ME, Mattick JS (2009) Long noncoding RNAs: insights into functions. Nat Rev Genet 10(3):155–159

    Article  CAS  PubMed  Google Scholar 

  11. Klattenhoff CA, Scheuermann JC, Surface LE, Bradley RK, Fields PA, Steinhauser ML, Ding H, Butty VL et al (2013) Braveheart, a long noncoding RNA required for cardiovascular lineage commitment. Cell 152(3):570–583

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ng SY, Lin L, Soh BS, Stanton LW (2013) Long noncoding RNAs in development and disease of the central nervous system. Trends Genet 29(8):461–468

    Article  CAS  PubMed  Google Scholar 

  13. Coupland KG, Kim WS, Halliday GM, Hallupp M, Dobson-Stone C, Kwok JB (2016) Role of the long non-coding RNA MAPTAS1 in regulation of microtubule associated protein tau (MAPT) expression in Parkinson’s disease. PLoS One 11(6):e0157924

    Article  PubMed  PubMed Central  Google Scholar 

  14. Carrieri C, Forrest AR, Santoro C, Persichetti F, Carninci P, Zucchelli S, Gustincich S (2015) Expression analysis of the long non-coding RNA antisense to Uchl1 (AS Uchl1) during dopaminergic cells’ differentiation in vitro and in neurochemical models of Parkinson’s disease. Front Cell Neurosci 9:114

    Article  PubMed  PubMed Central  Google Scholar 

  15. Jiao F, Wang Q, Zhang P, Bu L, Yan J, Tian B (2017) Expression signatures of long non-coding RNA in the substantia nigra of pre-symptomatic mouse model of Parkinson’s disease. Behav Brain Res 331:123–130

    Article  CAS  PubMed  Google Scholar 

  16. Lin D, Liang Y, Jing X, Chen Y, Lei M, Zeng Z, Zhou T, Wu X et al (2018) Microarray analysis of an synthetic α-synuclein induced cellular model reveals the expression profile of long non-coding RNA in Parkinson’s disease. Brain Res 1678:384–396

    Article  CAS  PubMed  Google Scholar 

  17. Kraus TFJ, Haider M, Spanner J, Steinmaurer M, Dietinger V, Kretzschmar HA (2017) Altered long noncoding RNA expression precedes the course of Parkinson’s disease-a preliminary report. Mol Neurobiol 54(4):2869–2877

    Article  CAS  PubMed  Google Scholar 

  18. Ni Y, Huang H, Chen Y, Cao M, Zhou H, Zhang Y (2017) Investigation of long non-coding RNA expression profiles in the substantia nigra of Parkinson’s disease. Cell Mol Neurobiol 37(2):329–338

    Article  CAS  PubMed  Google Scholar 

  19. Boros FA, Maszlag-Torok R, Vecsei L, Klivenyi P (2020) Increased level of NEAT1 long non-coding RNA is detectable in peripheral blood cells of patients with Parkinson’s disease. Brain Res 1730:146672

    Article  CAS  PubMed  Google Scholar 

  20. Wang D, Gao H, Li Y, Jiang S, Yong Y, Yang X (2020) Genome-scale expression pattern of long non-coding RNAs in Chinese Uyghur patients with Parkinson’s disease. Med Sci Monit 26:e925888

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Soreq L, Guffanti A, Salomonis N, Simchovitz A, Israel Z, Bergman H, Soreq H (2014) Long non-coding RNA and alternative splicing modulations in Parkinson’s leukocytes identified by RNA sequencing. PLoS Comput Biol 10(3):e1003517

    Article  PubMed  PubMed Central  Google Scholar 

  22. Chi LM, Wang LP, Jiao D (2019) Identification of differentially expressed genes and long noncoding RNAs associated with Parkinson’s disease. Parkinson’s Dis 6078251:1–7

    Google Scholar 

  23. Zhou Y, Gu C, Li J, Zhu L, Huang G, Dai J, Huang H (2018) Aberrantly expressed long noncoding RNAs and genes in Parkinson’s disease. Neuropsychiatr Dis Treat 14:3219–3229

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hu J-Z, Rong Z-J, Li M, Li P, Jiang LY, Luo ZX, Duan CY, Cao Y et al (2019) Silencing of lncRNA PKIA-AS1 attenuates spinal nerve ligation-induced neuropathic pain through epigenetic downregulation of CDK6 expression. Front Cell Neurosci 13:50

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Amer AA, Baltimore D (1996) An essential role for NF-kB in preventing TNF-alpha-induced cell death. Science 274(5288):782–784

    Article  Google Scholar 

  26. Shih RH, Wang CY, Yang CM (2015) NF-kappaB signaling pathways in neurological ınflammation: a mini review. Front Mol Neurosci 8:77

    Article  PubMed  PubMed Central  Google Scholar 

  27. Torres-Odio S, Key J, Hoepken HH, Canet-Pons J, Valek L, Roller B, Walter M, Morales-Gordo B et al (2017) Progression of pathology in PINK1-deficient mouse brain from splicing via ubiquitination, ER stress, and mitophagy changes to neuroinflammation. J Neuroinflammation 14:154

    Article  PubMed  PubMed Central  Google Scholar 

  28. Shen M, Mattox W (2012) Activation and repression functions of an SR splicing regulator depend on exonic versus intronic-binding position. Nucleic Acids Res 40:428–437

    Article  CAS  PubMed  Google Scholar 

  29. Jeong S (2017) SR proteins: binders, regulators, and connectors of RNA. Mol Cells 40(1):1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tesseur I, Nguyen A, Chang B, Li L, Woodling NS, Wyss-Coray T, Luo J (2017) Deficiency in neuronal TGF-β signaling leads to nigrostriatal degeneration and activation of TGF-β signaling protects against MPTP neurotoxicity in mice. J Neurosci 37(17):4584–4592

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lagna G, Hata A, Hemmati-Brivanlou A, Massagué J (1996) Partnership between DPC4 and SMAD proteins in TGF-β signalling pathways. Nature 383:832–836

    Article  CAS  PubMed  Google Scholar 

  32. Massague J (1998) TGF-beta signal transduction. Annu Rev Biochem 67:753–791

    Article  CAS  PubMed  Google Scholar 

  33. Varelas X, Samavarchi-Tehrani P, Narimatsu M, Weiss A, Cockburn K, Larsen BG, Rossant J, Wrana JL (2010) The Crumbs complex couples cell density sensing to Hippo-dependent control of the TGF-β–SMAD pathway. Dev Cell 19:831–844

    Article  CAS  PubMed  Google Scholar 

  34. Li PP, Li RG, Huang YQ, Lu JP, Zhang WJ, Wang ZY (2021) lncRNA OTUD6B-AS1 promotes paclitaxel resistance in triple negative breast cancer by regulation of miR-26a-5p/MTDH pathway-mediated autophagy and genomic instability. Aging (Albany NY) 13(21):24171–24191

    Article  CAS  PubMed  Google Scholar 

  35. Cui SY, Zhang W, Cui ZM, Yi H, Xu DW, Liu W, Zhu XH (2021) Knockdown of long non-coding RNA LEF1-AS1 attenuates apoptosis and inflammatory injury of microglia cells following spinal cord injury. J Orthop Surg Res 16(1):1–11

    Article  Google Scholar 

  36. Takata M, Pachera E, Frank-Bertoncelj M, Kozlova A, Jüngel A, Whitfield ML, Assassi S, Calcagni M et al (2019) OTUD6B-AS1 might be a novel regulator of apoptosis in systemic sclerosis. Front Immunol 10:100

    Article  Google Scholar 

  37. Bhattacharya S, Ghosh MK (2014) Cell death and deubiquitinases: perspectives in cancer. Biomed Res Int 2014:435197

    Article  PubMed  PubMed Central  Google Scholar 

  38. Messemaker TC, Chadli L, Cai G, Goelela VS, Boonstra M, Annemarie L, Dorjée AL, Andersen SN et al (2018) Antisense long non-coding RNAs are deregulated in skin tissue of patients with systemic sclerosis. J Invest Dermatol 138(4):826–835

    Article  CAS  PubMed  Google Scholar 

  39. Mullen RD, Colvin SC, Hunter CS, Savage JJ, Walvoord EC, Bhangoo AP, Ten S, Weigel J et al (2007) Roles of the LHX3 and LHX4 LIM-homeodomain factors in pituitary development. Mol Cell Endocrinol 400(1–2):44–51

    Google Scholar 

  40. Bretzner F, Brownstone RM (2013) Lhx3-Chx10 reticulospinal neurons in locomotor circuits. J Neurosci 33(37):14681–14692

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Raveney BJE, Oki S, Hohjoh H, Nakamura M, Sato W, Murata M, Yamamuraa T (2015) Eomesodermin-expressing T-helper cells are essential for chronic neuroinflammation. Nat Commun 6:8437

    Article  CAS  PubMed  Google Scholar 

  42. Oki S (2019) Eomes-expressing T-helper cells as potential target of therapy in chronic neuroinflammation. Neurochem Int 130:104348

    Article  CAS  PubMed  Google Scholar 

  43. Xin C, Liu J (2021) Long non-coding RNAs in Parkinson’s disease. Neurochem Res 46:1031–1042

    Article  CAS  PubMed  Google Scholar 

  44. Kuo MC, Liu SC, Hsu YF, Wu RM (2021) The role of noncoding RNAs in Parkinson’s disease: biomarkers and associations with pathogenic pathways. J Biomed Sci 28(1):78

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhang H, Yao L, Zheng Z, Koc S, Lu G (2022) The role of non-coding RNAs in the pathogenesis of Parkinson’s disease: recent advancement. Pharmaceuticals (Basel, Switzerland) 15(7):811

    Article  CAS  PubMed  Google Scholar 

  46. Huang T, Zhao JY, Pan RR, Jiang T, Fu XX, Huang Q, Wang XX, Gong PY et al (2023) Dysregulation of circulatory levels of lncRNAs in Parkinson’s disease. Mol Neurobiol 60(1):317–328

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Dr. Elif Avcı and Dr. Ahmet Mağrur Karagülmez in the Department of Neurology, Pamukkale University, for their diligent assistance with the data collection for this study.

Funding

This study is supported by Pamukkale University Scientific Research Projects Coordinatorship with the project number 2020SABE024.

Author information

Authors and Affiliations

Authors

Contributions

F. Gizem Sarıekiz, A. Gaye Tomatir, and P. Elvan Tokgun designed the study. L. Sinan Bir selected the patients included in the study. F. Gizem sarıekiz and p. Elvan Tokgun performed the wet lab of the study. F. Gizem Sarıekiz, A. Gaye Tomatir, and P. Elvan Tokgun analyzed the data and wrote the manuscript. F. Gizem Sarıekiz, A. Gaye Tomatir, P. Elvan Tokgun, and L. Sinan Bir read and confirmed the study.

Corresponding author

Correspondence to Fatma Gizem Sarıekiz.

Ethics declarations

Ethics Approval and Consent to Participate

All protocols used in this study were approved by the Ethics Commitee of Pamukkale University Hospital, Turkey. All procedures performed in this study involving human participants were in accordance with the ethical standards of the institutional commitee. Informed concent was obtained from all individual participants included in the study.

Consent for Publication

The informed consent was obtained from study participants.

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sarıekiz, F.G., Tomatır, A.G., Tokgün, P.E. et al. Evaluation of Long Non-coding RNA Expression Profiles in Peripheral Blood Mononuclear Cells of Patients with Parkinson’s Disease. Mol Neurobiol 60, 6201–6211 (2023). https://doi.org/10.1007/s12035-023-03470-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-023-03470-0

Keywords

Navigation