Baseline C-Reactive Protein Levels and Life Prognosis in Parkinson Disease

Hideyuki Sawada, Tomoko Oeda, Atsushi Umemura, Satoshi Tomita, Masayuki Kohsaka, Kwiyoung Park, Kenji Yamamoto, Hiroshi Sugiyama, Hideyuki Sawada, Tomoko Oeda, Atsushi Umemura, Satoshi Tomita, Masayuki Kohsaka, Kwiyoung Park, Kenji Yamamoto, Hiroshi Sugiyama

Abstract

Background: C-reactive protein (CRP) is a biomarker of inflammation, and high levels of CRP correlate with vascular death. Chronic inflammation is considered to be involved in neurodegeneration, although there is no evidence linking it with the process of neurodegenerative diseases.

Objective: To determine the role of baseline CRP levels in the prognosis of patients with Parkinson disease (PD).

Methods: A cohort of 313 patients with a mean age of 69.1 and mean PD duration of 7.9 years was retrospectively followed for a mean observation time of 1,753 days. CRP was measured when patients were not diagnosed with any infections, and levels were repetitively measured to investigate a tendency of "regression to mean." The primary outcome measure was a survival time from study enrollment to death.

Results: During the observation period 56 patients died. Baseline CRP was log-linearly associated with a risk of death in PD. Mean survival time was 3,149 (95% confidence interval; 3,009-3,289) days in patients with CRP ≤ 0.8mg/L (lower two thirds) and 2,620 (2,343-2,897) days in those with CRP > 0.8 mg/L (top third, p < 0.001, log-rank test). The adjusted hazard ratio (HR) per two-fold higher CRP concentration for all deaths was 1.29 (1.10-1.52), and after excluding PD-unrelated deaths, such as cancer or stroke, HR was 1.23 (1.01-1.49) (adjusted for age, sex, PD duration, modified Hohen-Yahr stages, MMSE scores, and serum albumin).

Conclusions: Baseline CRP concentrations were associated with the risk of death and predicted life prognosis of patients with PD. The associations were independent from PD duration, PD severity, cognitive function, ages, and nutritional conditions, suggesting the possibility that subclinical chronic inflammation is associated with a neurodegenerative process in PD.

Conflict of interest statement

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

Figures

Fig 1. Serial changes in CRP levels…
Fig 1. Serial changes in CRP levels in patients with low, mid, and high baseline CRP concentrations.
CRP was measured at study enrollment and during the follow-up period (21–180 days, 181–360 days, 361–720 days, and 721–1080 days after study enrollment). Patients were assigned to three groups: those with low-level ( 0.8 mg/L) CRP at enrollment. Data represent median, and top and bottom error bars represent 75 and 25 percentile values, respectively.
Fig 2. Survival curves of PD patients…
Fig 2. Survival curves of PD patients according to baseline clinical features.
Cumulative survival rates according to CRP levels (A), PD disease duration (B), modified H-Y stages (C), MMSE score (D), age (E), sex (F), NSAID use (G), and serum albumin (H). There was a statistically significant difference in the survival rate between patients with CRP > 0.8 mg/L and ≤ 0.8 mg/L (p = 0.00004) (A), patients with PD disease duration ≤ 8 years and > 8 years (p = 0.00004) (B), those with modified H-Y 1–3 and modified H-Y 4–5 (p = 0.00001) (C), and those with albumin > 4.0mg/dL and albumin ≤ 4.0mg/dL (p = 0.004). However, the survival rate was not influenced by MMSE score, (p = 0.499) (D), age (p = 0.117) (E), sex (p = 0.186) (F), or NSAID use (p = 0.846). Statistical significance was tested using the Log-rank test.
Fig 3. Relative risk of death by…
Fig 3. Relative risk of death by CRP concentration.
Relative risk of death was estimated as hazard ratios (HRs) using Cox proportional hazard models. Unadjusted HRs of all deaths correlated with log CRP concentrations (A). To exclude possible confounders, HRs of all deaths were adjusted for age, sex, PD disease duration, MMSE (≤24 vs. >24), mH-Y (1–3 vs. 4–5), and serum albumin levels, and the analysis showed a log-linear association with CRP concentrations (B). Similarly unadjusted and adjusted HRs of death, excluding PD-unrelated deaths, correlated with CRP concentrations (C, D). Unadjusted and age- and sex-adjusted HRs of deaths from pneumonia correlated with CRP concentrations (E, F). Unadjusted and age- and sex-adjusted HRs of sudden deaths correlated with CRP concentrations, although there was no significance (G, H). Unadjusted and age- and sex-adjusted HRs of deaths from cancer correlated with CRP concentration, although there was no significance (I, J).
Fig 4. Survival curves of PD patients…
Fig 4. Survival curves of PD patients according to CRP levels.
The cumulative survival rate was compared in patients with CRP ≤ 0.8 mg/L and those with CRP > 0.8 mg/L and was stratified by age (A), sex (B), PD duration (C), modified H-Y stage (D), MMSE (E), and serum albumin (F). The cumulative survival rate was significantly higher in patients with CRP ≤ 0.8 mg/L than those with CRP > 0.8 mg/L. Statistical significance was calculated in pooled models.

References

    1. Danesh J, Wheeler JG, Hirschfield GM, Eda S, Eiriksdottir G, Rumley A, et al. C-reactive protein and other circulating markers of inflammation in the prediction of coronary heart disease. N Engl J Med. 2004;350(14):1387–97. Epub 2004/04/09. 10.1056/NEJMoa032804 .
    1. Emerging Risk Factors C, Kaptoge S, Di Angelantonio E, Lowe G, Pepys MB, Thompson SG, et al. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375(9709):132–40. Epub 2009/12/25. 10.1016/S0140-6736(09)61717-7
    1. Heikkila K, Ebrahim S, Lawlor DA. A systematic review of the association between circulating concentrations of C reactive protein and cancer. J Epidemiol Community Health. 2007;61(9):824–33. Epub 2007/08/19. 10.1136/jech.2006.051292
    1. Emberson JR, Whincup PH, Morris RW, Walker M, Lowe GD, Rumley A. Extent of regression dilution for established and novel coronary risk factors: results from the British Regional Heart Study. Eur J Cardiovasc Prev Rehabil. 2004;11(2):125–34. Epub 2004/06/10. .
    1. Perry VH. The influence of systemic inflammation on inflammation in the brain: implications for chronic neurodegenerative disease. Brain Behav Immun. 2004;18(5):407–13. Epub 2004/07/22. 10.1016/j.bbi.2004.01.004 .
    1. Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73(10):768–74. Epub 2009/09/10. 10.1212/WNL.0b013e3181b6bb95
    1. Holmes C, Cunningham C, Zotova E, Culliford D, Perry VH. Proinflammatory cytokines, sickness behavior, and Alzheimer disease. Neurology. 2011;77(3):212–8. Epub 2011/07/15. 10.1212/WNL.0b013e318225ae07
    1. Yasojima K, Schwab C, McGeer EG, McGeer PL. Human neurons generate C-reactive protein and amyloid P: upregulation in Alzheimer's disease. Brain Res. 2000;887(1):80–9. Epub 2001/01/03. .
    1. Hirsch EC, Hunot S. Neuroinflammation in Parkinson's disease: a target for neuroprotection? Lancet Neurol. 2009;8(4):382–97. Epub 2009/03/20. 10.1016/S1474-4422(09)70062-6 .
    1. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology. 1988;38(8):1285–91. Epub 1988/08/01. .
    1. Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, Hashizume Y. Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson's disease brains. Acta Neuropathol. 2003;106(6):518–26. Epub 2003/09/27. 10.1007/s00401-003-0766-2 .
    1. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson's disease. Neurobiol Dis. 2006;21(2):404–12. Epub 2005/09/27. 10.1016/j.nbd.2005.08.002 .
    1. Mortensen RF, Zhong W. Regulation of phagocytic leukocyte activities by C-reactive protein. J Leukoc Biol. 2000;67(4):495–500. Epub 2000/04/19. .
    1. Perry VH, Cunningham C, Holmes C. Systemic infections and inflammation affect chronic neurodegeneration. Nat Rev Immunol. 2007;7(2):161–7. Epub 2007/01/16. 10.1038/nri2015 .
    1. Morgante L, Salemi G, Meneghini F, Di Rosa AE, Epifanio A, Grigoletto F, et al. Parkinson disease survival: a population-based study. Arch Neurol. 2000;57(4):507–12. Epub 2000/04/18. .
    1. Matsumoto H, Sengoku R, Saito Y, Kakuta Y, Murayama S, Imafuku I. Sudden death in Parkinson's disease: a retrospective autopsy study. J Neurol Sci. 2014;343(1–2):149–52. Epub 2014/06/15. 10.1016/j.jns.2014.05.060 .
    1. Driver JA, Logroscino G, Lu L, Gaziano JM, Kurth T. Use of non-steroidal anti-inflammatory drugs and risk of Parkinson's disease: nested case-control study. BMJ. 2011;342:d198 Epub 2011/01/22. 10.1136/bmj.d198
    1. Gao X, Chen H, Schwarzschild MA, Ascherio A. Use of ibuprofen and risk of Parkinson disease. Neurology. 2011;76(10):863–9. Epub 2011/03/04. 10.1212/WNL.0b013e31820f2d79
    1. Ben-Shlomo Y, Marmot MG. Survival and cause of death in a cohort of patients with parkinsonism: possible clues to aetiology? J Neurol Neurosurg Psychiatry. 1995;58(3):293–9. Epub 1995/03/01.
    1. Sawada H, Oeda T, Umemura A, Tomita S, Hayashi R, Kohsaka M, et al. Subclinical elevation of plasma C-reactive protein and illusions/hallucinations in subjects with Parkinson's disease: case-control study. PLoS One. 2014;9(1):e85886 Epub 2014/02/06. 10.1371/journal.pone.0085886
    1. Arima H, Kubo M, Yonemoto K, Doi Y, Ninomiya T, Tanizaki Y, et al. High-sensitivity C-reactive protein and coronary heart disease in a general population of Japanese: the Hisayama study. Arteriosclerosis, thrombosis, and vascular biology. 2008;28(7):1385–91. Epub 2008/04/12. 10.1161/ATVBAHA.107.157164 .
    1. Nakanishi S, Yamane K, Kamei N, Okubo M, Kohno N. Elevated C-reactive protein is a risk factor for the development of type 2 diabetes in Japanese Americans. Diabetes care. 2003;26(10):2754–7. Epub 2003/09/30. .
    1. Levy G, Tang MX, Louis ED, Cote LJ, Alfaro B, Mejia H, et al. The association of incident dementia with mortality in PD. Neurology. 2002;59(11):1708–13. Epub 2002/12/11. .
    1. de Lau LM, Breteler MM. Epidemiology of Parkinson's disease. Lancet Neurol. 2006;5(6):525–35. Epub 2006/05/23. 10.1016/S1474-4422(06)70471-9 .
    1. Shimizu K, Ogura H, Wasa M, Hirose T, Shimazu T, Nagasaka H, et al. Refractory hypoglycemia and subsequent cardiogenic shock in starvation and refeeding: Report of three cases. Nutrition. 2014;30(9):1090–2. Epub 2014/06/15. 10.1016/j.nut.2014.01.007 .
    1. Brochard V, Combadiere B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119(1):182–92. Epub 2008/12/24. 10.1172/JCI36470
    1. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci U S A. 2009;106(31):13010–5. Epub 2009/08/05. 10.1073/pnas.0903691106
    1. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci. 2010;30(20):6838–51. Epub 2010/05/21. 10.1523/JNEUROSCI.5699-09.2010
    1. Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci. 2008;28(30):7687–98. Epub 2008/07/25. 10.1523/JNEUROSCI.0143-07.2008
    1. Reynolds AD, Glanzer JG, Kadiu I, Ricardo-Dukelow M, Chaudhuri A, Ciborowski P, et al. Nitrated alpha-synuclein-activated microglial profiling for Parkinson's disease. J Neurochem. 2008;104(6):1504–25. Epub 2007/11/27. 10.1111/j.1471-4159.2007.05087.x .
    1. Cao S, Standaert DG, Harms AS. The gamma chain subunit of Fc receptors is required for alpha-synuclein-induced pro-inflammatory signaling in microglia. J Neuroinflammation. 2012;9:259 Epub 2012/11/29. 10.1186/1742-2094-9-259
    1. Li YN, Qin XJ, Kuang F, Wu R, Duan XL, Ju G, et al. Alterations of Fc gamma receptor I and Toll-like receptor 4 mediate the antiinflammatory actions of microglia and astrocytes after adrenaline-induced blood-brain barrier opening in rats. J Neurosci Res. 2008;86(16):3556–65. Epub 2008/08/30. 10.1002/jnr.21810 .
    1. Closhen D, Bender B, Luhmann HJ, Kuhlmann CR. CRP-induced levels of oxidative stress are higher in brain than aortic endothelial cells. Cytokine. 2010;50(2):117–20. Epub 2010/03/09. 10.1016/j.cyto.2010.02.011 .
    1. Ibi M, Sawada H, Kume T, Katsuki H, Kaneko S, Shimohama S, et al. Depletion of intracellular glutathione increases susceptibility to nitric oxide in mesencephalic dopaminergic neurons. J Neuroch. 1999;73(4):1696–703. Epub 1999/09/29. .
    1. Sawada H, Kawamura T, Shimohama S, Akaike A, Kimura J. Different mechanisms of glutamate-induced neuronal death between dopaminergic and non-dopaminergic neurons in rat mesencephalic culture. J Neurosci Res. 1996;43(4):503–10. Epub 1996/02/15. .
    1. Ebadi M, Sharma SK. Peroxynitrite and mitochondrial dysfunction in the pathogenesis of Parkinson's disease. Antioxidants & redox signaling. 2003;5(3):319–35. Epub 2003/07/26. 10.1089/152308603322110896 .
    1. Sawada H, Shimohama S, Tamura Y, Kawamura T, Akaike A, Kimura J. Methylphenylpyridium ion (MPP+) enhances glutamate-induced cytotoxicity against dopaminergic neurons in cultured rat mesencephalon. J Neurosci Res. 1996;43(1):55–62. Epub 1996/01/01. 10.1002/jnr.490430107 .
    1. Sharma SK, Ebadi M. Metallothionein attenuates 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in dopaminergic neurons. Antioxidants & redox signaling. 2003;5(3):251–64. Epub 2003/07/26. 10.1089/152308603322110832 .
    1. Kuhlmann CR, Librizzi L, Closhen D, Pflanzner T, Lessmann V, Pietrzik CU, et al. Mechanisms of C-reactive protein-induced blood-brain barrier disruption. Stroke. 2009;40(4):1458–66. Epub 2009/02/28. 10.1161/STROKEAHA.108.535930 .
    1. Lunnon K, Teeling JL, Tutt AL, Cragg MS, Glennie MJ, Perry VH. Systemic inflammation modulates Fc receptor expression on microglia during chronic neurodegeneration. J Immunol. 2011;186(12):7215–24. Epub 2011/05/17. 10.4049/jimmunol.0903833 .
    1. Cerejeira J, Firmino H, Vaz-Serra A, Mukaetova-Ladinska EB. The neuroinflammatory hypothesis of delirium. Acta Neuropathol. 2010;119(6):737–54. Epub 2010/03/24. 10.1007/s00401-010-0674-1 .
    1. Perry VH. Contribution of systemic inflammation to chronic neurodegeneration. Acta Neuropathol. 2010;120(3):277–86. Epub 2010/07/21. 10.1007/s00401-010-0722-x .
    1. Umemura A, Oeda T, Tomita S, Hayashi R, Kohsaka M, Park K, et al. Delirium and high Fever are associated with subacute motor deterioration in Parkinson disease: a nested case-control study. PLoS One. 2014;9(6):e94944 Epub 2014/06/03. 10.1371/journal.pone.0094944
    1. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–8. Epub 2012/12/21. 10.1038/nature11729
    1. Codolo G, Plotegher N, Pozzobon T, Brucale M, Tessari I, Bubacco L, et al. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS One. 2013;8(1):e55375 Epub 2013/02/06. 10.1371/journal.pone.0055375

Source: PubMed

3
Iratkozz fel