Ukr.Biochem.J. 2022; Volume 94, Issue 1, Jan-Feb, pp. 33-43

doi: https://doi.org/10.15407/ubj94.01.033

Systemic inflammation biomarkers in 6-OHDA- and LPS-induced Parkinson’s disease in rats

Zh. Oliynyk*, M. Rudyk, V. Svyatetska, T. Dovbynchuk, G. Tolstanova, L. Skivka

ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Ukraine;
*e-mail: ojankin@yahoo.com

Received: 14 December 2021; Accepted: 21 January 2022

Hematological and immunological markers of systemic inflammation were studied in 6-hydroxydopamine (6-OHDA)- and lipopolysaccharide (LPS)-induced models of Parkinson’s disease (PD). Experiments were carried out on adult male Wistar rats: 1 – intact animals; 2 – sham-operated animals and 3 – 6-OHDA- and LPS-lesioned animals. PD development was confirmed by the results of behavioral testing (apomorphine test, open field test) and immunohistochemical detection of the loss of dopaminergic neurons. Hematological indices (complete blood count and differential leukocyte count (DLC)) were examined using hematological analyser. Immunological indices included phenotypic (CD206 and CD80/86) and metabolic (oxidative metabolism and phagocytic activity) characteristics of circulating monocytes (Mo) and granulocytes (Gr), which were determined by flow cytometry, as well as plasma levels of C-reactive protein, which were determined by ELISA. LPS-induced PD was associated with neutrophilia, 1.9 times increased neutrophil-to-lymphocyte ratio, 3 times increased platelet-to-lymphocyte ratio, and 3 times increased systemic immune inflammation index as compared to intact animals. Functional profile of circulating phagocytes from LPS-lesioned animals was characterized by the pro-inflammtory metabolic shift, as was indicated by 5 times increased oxidative metabolism indices and up-regulated CD80/86 expression along with decreased phagocytic activity and CD206 expression. 6-OHDA-lesioned rats demonstrated decreased DLC indices as compared to intact and sham-operated rats. Functional profile of circulating phagocytes in this model was characterized by anti-inflammatory shift. The results obtained from this study demonstrated that stereotaxic LPS-induced PD is appropriate rodent model for the study of systemic inflammation which is inherent for the disease pathophysiology.

Keywords: , , , , , , , ,


References:

  1. MacMahon Copas AN, McComish SF, Fletcher JM, Caldwell MA. The Pathogenesis of Parkinson’s Disease: A Complex Interplay Between Astrocytes, Microglia, and T Lymphocytes? Front Neurol. 2021;12:666737. PubMed, PubMedCentral, CrossRef
  2. Béjot Y, Yaffe K. Ageing Population: A Neurological Challenge. Neuroepidemiology. 2019;52(1-2):76-77. PubMed, CrossRef
  3. Mishra A, Bandopadhyay R, Singh PK, Mishra PS, Sharma N, Khurana N. Neuroinflammation in neurological disorders: pharmacotherapeutic targets from bench to bedside. Metab Brain Dis. 2021;36(7):1591-1626. PubMed, CrossRef
  4. Süβ P, Lana AJ, Schlachetzki JCM. Chronic peripheral inflammation: a possible contributor to neurodegenerative diseases. Neural Regen Res. 2021;16(9):1711-1714. PubMed, PubMedCentral, CrossRef
  5. Sadek HL, Almohari SF, Renno WM. The Inflammatory Cytokines in the Pathogenesis of Parkinson’s Disease. J Alzheimers Dis Parkinsonism. 2014; 4:148. CrossRef
  6. Wu Y, Hu Y, Wang B, Li S, Ma C, Liu X, Moynagh PN, Zhou J, Yang S. Dopamine Uses the DRD5-ARRB2-PP2A Signaling Axis to Block the TRAF6-Mediated NF-κB Pathway and Suppress Systemic Inflammation. Mol Cell. 2020;78(1):42-56.e6. PubMed, CrossRef
  7. Beers DR, Zhao W, Neal DW, Thonhoff JR, Thome AD, Faridar A, Wen S, Wang J, Appel SH. Elevated acute phase proteins reflect peripheral inflammation and disease severity in patients with amyotrophic lateral sclerosis. Sci Rep. 2020;10(1):15295. PubMed, PubMedCentral, CrossRef
  8. Ferrari CC, Tarelli R. Parkinson’s disease and systemic inflammation. Parkinsons Dis. 2011;2011:436813. PubMed, PubMedCentral, CrossRef
  9. Pajares M , Rojo AI, Manda G, Boscá L, Cuadrado A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic Implications. Cells. 2020;9(7):1687. PubMed, PubMedCentral, CrossRef
  10. Ortiz GG, González-Usigli H, Pacheco-Moisés FP, Mireles-Ramírez MA. Physiology and Pathology of Neuroimmunology: Role of Inflammation in Parkinson’s Disease. In book: Physiology and Pathology of Immunology. Edition 1. In Tech Open. 2017; 173-197.
  11. Potashkin JA, Blume SR, Runkle NK. Limitations of animal models of Parkinson’s disease. Parkinsons Dis. 2010;2011:658083. PubMed, PubMedCentral, CrossRef
  12. Stefani A, Cerroni R, Pierantozzi M, D’Angelo V, Grandi L, Spanetta M, Galati S. Deep brain stimulation in Parkinson’s disease patients and routine 6-OHDA rodent models: Synergies and pitfalls. Eur J Neurosci. 2021;53(7):2322-2343. PubMed, CrossRef
  13. Deng I, Corrigan F, Zhai G, Zhou XF, Bobrovskaya L. Lipopolysaccharide animal models of Parkinson’s disease: Recent progress and relevance to clinical disease. Brain Behav Immun Health. 2020;4:100060. PubMed, PubMedCentral, CrossRef
  14. Pott Godoy MC, Tarelli R, Ferrari CC, Sarchi MI, Pitossi FJ. Central and systemic IL-1 exacerbates neurodegeneration and motor symptoms in a model of Parkinson’s disease. Brain. 2008;131(Pt 7):1880-1894. PubMed, PubMedCentral, CrossRef
  15. Talanov SA, Oleshko NN, Tkachenko MN, Sagach VF. Pharmacoprotective influences on different links of the mechanism underlying 6-hydroxydopamine-induced degeneration of nigro-striatal dopaminergic neurons. Neurophysiology. 2006;38(2):128-133. CrossRef
  16. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates. 6th Edition, San Diego: Academic Press, 2007; 456 р.
  17. Hoban DB, Connaughton E, Connaughton C, Hogan G, Thornton C, Mulcahy P, Moloney TC, Dowd E. Further characterisation of the LPS model of Parkinson’s disease: a comparison of intra-nigral and intra-striatal lipopolysaccharide administration on motor function, microgliosis and nigrostriatal neurodegeneration in the rat. Brain Behav Immun. 2013;27(1):91-100. PubMed, CrossRef
  18.  Leite-Almeida H, Almeida-Torres L, Mesquita AR, Pertovaara A, Sousa N, Cerqueira JJ, Almeida A. The impact of age on emotional and cognitive behaviours triggered by experimental neuropathy in rats. Pain. 2009;144(1-2):57-65. PubMed, CrossRef
  19. Iancu R, Mohapel P, Brundin P, Paul G. Behavioral characterization of a unilateral 6-OHDA-lesion model of Parkinson’s disease in mice. Behav Brain Res. 2005;162(1):1-10. PubMed, CrossRef
  20. Walsh S, Finn DP, Dowd E. Time-course of nigrostriatal neurodegeneration and neuroinflammation in the 6-hydroxydopamine-induced axonal and terminal lesion models of Parkinson’s disease in the rat. Neuroscience. 2011;175:251-261. PubMed, CrossRef
  21. Pauletti G, Dandekar S, Rong H, Ramos L, Peng H, Seshadri R, Slamon DJ. Assessment of methods for tissue-based detection of the HER-2/neu alteration in human breast cancer: a direct comparison of fluorescence in situ hybridization and immunohistochemistry. J Clin Oncol. 2000;18(21):3651-3664. PubMed, CrossRef
  22. Erdogan T. Role of systemic immune-inflammation index in asthma and NSAID-exacerbated respiratory disease. Clin Respir J. 2021;15(4):400-405. PubMed, CrossRef
  23. Rudyk MP, Pozur VV, Voieikova DO, Hurmach YV, Khranovska NM, Skachkova OV, Svyatetska VM, Fedorchuk OG, Skivka LM, Berehova TV, Ostapchenko LI. Sex-based differences in phagocyte metabolic profile in rats with monosodium glutamate-induced obesity. Sci Rep. 2018;8(1):5419. PubMed, PubMedCentral, CrossRef
  24. Bezard E, Yue Z, Kirik D, Spillantini MG. Animal models of Parkinson’s disease: limits and relevance to neuroprotection studies. Mov Disord. 2013;28(1):61-70. PubMed, PubMedCentral, CrossRef
  25. Liu M, Bing G. Lipopolysaccharide animal models for Parkinson’s disease. Parkinsons Dis. 2011;2011:327089. PubMed, PubMedCentral, CrossRef
  26. Eidson LN, Kannarkat GT, Barnum CJ, Chang J, Chung J, Caspell-Garcia C, Taylor P, Mollenhauer B, Schlossmacher MG, Ereshefsky L,Yen M, Kopil C, Frasier M, Marek K, Hertzberg VS, Tansey MG. Candidate inflammatory biomarkers display unique relationships with alpha-synuclein and correlate with measures of disease severity in subjects with Parkinson’s disease. J Neuroinflammation. 2017;14(1):164. PubMed, PubMedCentral, CrossRef
  27. Minalyan A, Gabrielyan L, Pietra C, Taché Y, Wang L. Multiple Beneficial Effects of Ghrelin Agonist, HM01 on Homeostasis Alterations in 6-Hydroxydopamine Model of Parkinson’s Disease in Male Rats. Front Integr Neurosci. 2019;13:13. PubMed, PubMedCentral, CrossRef
  28. Luo J, Chen C, Li Q. White blood cell counting at point-of-care testing: A review. Electrophoresis. 2020;41(16-17):1450-1468. PubMed, CrossRef
  29. Lin JX, Lin JP, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu R, Zheng CH, Huang CM, Li P. Complete blood count-based inflammatory score (CBCS) is a novel prognostic marker for gastric cancer patients after curative resection. BMC Cancer. 2020;20(1):11. PubMed, PubMedCentral, CrossRef
  30. Jensen MP, Jacobs BM, Dobson R, Bandres-Ciga S, Blauwendraat C, Schrag A, Noyce AJ, IPDGC. Lower Lymphocyte Count is Associated With Increased Risk of Parkinson’s Disease. Ann Neurol. 2021;89(4):803-812. PubMed, CrossRef
  31. Jiang S, Wang Y, Gao H, Luo Q, Wang D, Li Y, Yong Y, Yang X. Cell Ratio Differences in Peripheral Blood between Early- and Late-Onset Parkinson’s Disease: A Case-Control Study. Biomed Res Int. 2019;2019:2072635. PubMed, PubMedCentral, CrossRef
  32. Ingersoll MA, Platt AM, Potteaux S, Randolph GJ. Monocyte trafficking in acute and chronic inflammation. Trends Immunol. 2011;32(10):470-477. PubMed, PubMedCentral, CrossRef
  33. Kara SP, Altunan B, Unal A. Investigation of the peripheral inflammation (neutrophil-lymphocyte ratio) in two neurodegenerative diseases of the central nervous system. Neurol Sci. 2021;1-9. PubMed, PubMedCentral, CrossRef
  34. Sanjari Moghaddam H, Ghazi Sherbaf F, Mojtahed Zadeh M, Ashraf-Ganjouei A, Aarabi MH. Association Between Peripheral Inflammation and DATSCAN Data of the Striatal Nuclei in Different Motor Subtypes of Parkinson Disease. Front Neurol. 2018;9:234. PubMed, PubMedCentral, CrossRef
  35. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122(3):787-795. PubMed, PubMedCentral, CrossRef
  36. Tan HY, Wang N, Li S, Hong M, Wang X, Feng Y. The Reactive Oxygen Species in Macrophage Polarization: Reflecting Its Dual Role in Progression and Treatment of Human Diseases. Oxid Med Cell Longev. 2016;2016:2795090. PubMed, PubMedCentral, CrossRef
  37. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front Immunol. 2019;10:1084. PubMed, PubMedCentral, CrossRef
  38. Dilek N, Vuillefroy de Silly R, Blancho G, Vanhove B. Myeloid-derived suppressor cells: mechanisms of action and recent advances in their role in transplant tolerance. Front Immunol. 2012;3:208. PubMed, PubMedCentral, CrossRef

Creative CommonsThis work is licensed under a Creative Commons Attribution 4.0 International License.