1887

Abstract

Neuroinflammation is a prominent component of several neurodegenerative diseases, including multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, tauopathies, amyotrophic lateral sclerosis and prion diseases. In such conditions, the ability to decrease neuroinflammation by drug therapy may influence disease progression. Statins have been used to treat hyperlipidemia as well as reduce neuroinflammation and oxidative stress in various tissues. In previous studies, treatment of scrapie-infected mice with the type 1 statins, simvastatin or pravastatin, showed a small beneficial effect on survival time. In the current study, to increase the effectiveness of statin therapy, we treated infected mice with atorvastatin, a type 2 statin that has improved pharmacokinetics over many type 1 statins. Treatments with either simvastatin or pravastatin were tested for comparison. We evaluated scrapie-infected mice for protease-resistant PrP (PrPres) accumulation, gliosis, neuroinflammation and time until advanced clinical disease requiring euthanasia. All three statin treatments reduced total serum cholesterol ≥40 % in mice. However, gliosis and PrPres deposition were similar in statin-treated and untreated infected mice. Time to euthanasia due to advanced clinical signs was not changed in statin-treated mice relative to untreated mice, a finding at odds with previous reports. Expression of 84 inflammatory genes involved in neuroinflammation was also quantitated. Seven genes were reduced by pravastatin, and one gene was reduced by atorvastatin. In contrast, simvastatin therapy did not reduce any of the tested genes, but did slightly increase the expression of and . Our studies indicate that none of the three statins tested were effective in reducing scrapie-induced neuroinflammation or neuropathogenesis.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000876
2017-08-01
2024-04-20
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/8/2190.html?itemId=/content/journal/jgv/10.1099/jgv.0.000876&mimeType=html&fmt=ahah

References

  1. Budka H, Aguzzi A, Brown P, Brucher JM, Bugiani O et al. Neuropathological diagnostic criteria for Creutzfeldt-Jakob disease (CJD) and other human spongiform encephalopathies (prion diseases). Brain Pathol 1995; 5:459–466 [View Article][PubMed]
    [Google Scholar]
  2. Jeffrey M, González L. Classical sheep transmissible spongiform encephalopathies: pathogenesis, pathological phenotypes and clinical disease. Neuropathol Appl Neurobiol 2007; 33:373–394 [View Article][PubMed]
    [Google Scholar]
  3. Scallet AC, Ye X. Excitotoxic mechanisms of neurodegeneration in transmissible spongiform encephalopathies. Ann N Y Acad Sci 1997; 825:194–205 [View Article][PubMed]
    [Google Scholar]
  4. Black SA, Stys PK, Zamponi GW, Tsutsui S. Cellular prion protein and NMDA receptor modulation: protecting against excitotoxicity. Front Cell Dev Biol 2014; 2:45 [View Article][PubMed]
    [Google Scholar]
  5. Carroll JA, Striebel JF, Race B, Phillips K, Chesebro B. Prion infection of mouse brain reveals multiple new upregulated genes involved in neuroinflammation or signal transduction. J Virol 2015; 89:2388–2404 [View Article][PubMed]
    [Google Scholar]
  6. Carroll JA, Striebel JF, Rangel A, Woods T, Phillips K et al. Prion strain differences in accumulation of PrPSc on neurons and glia are associated with similar expression profiles of neuroinflammatory genes: comparison of three prion strains. PLoS Pathog 2016; 12:e1005551 [View Article][PubMed]
    [Google Scholar]
  7. Tribouillard-Tanvier D, Striebel JF, Peterson KE, Chesebro B. Analysis of protein levels of 24 cytokines in scrapie agent-infected brain and glial cell cultures from mice differing in prion protein expression levels. J Virol 2009; 83:11244–11253 [View Article][PubMed]
    [Google Scholar]
  8. Aiken JM, Williamson JL, Marsh RF. Evidence of mitochondrial involvement in scrapie infection. J Virol 1989; 63:1686–1694[PubMed]
    [Google Scholar]
  9. Choi SI, Ju WK, Choi EK, Kim J, Lea HZ et al. Mitochondrial dysfunction induced by oxidative stress in the brains of hamsters infected with the 263 K scrapie agent. Acta Neuropathol 1998; 96:279–286 [View Article][PubMed]
    [Google Scholar]
  10. Park JH, Kim BH, Park SJ, Jin JK, Jeon YC et al. Association of endothelial nitric oxide synthase and mitochondrial dysfunction in the hippocampus of scrapie-infected mice. Hippocampus 2011; 21:319–333 [View Article][PubMed]
    [Google Scholar]
  11. Sisková Z, Mahad DJ, Pudney C, Campbell G, Cadogan M et al. Morphological and functional abnormalities in mitochondria associated with synaptic degeneration in prion disease. Am J Pathol 2010; 177:1411–1421 [View Article][PubMed]
    [Google Scholar]
  12. Choi HS, Choi YG, Shin HY, Oh JM, Park JH et al. Dysfunction of mitochondrial dynamics in the brains of scrapie-infected mice. Biochem Biophys Res Commun 2014; 448:157–162 [View Article][PubMed]
    [Google Scholar]
  13. Forloni G, Angeretti N, Chiesa R, Monzani E, Salmona M et al. Neurotoxicity of a prion protein fragment. Nature 1993; 362:543–546 [View Article][PubMed]
    [Google Scholar]
  14. Liberski PP, Sikorska B, Bratosiewicz-Wasik J, Gajdusek DC, Brown P. Neuronal cell death in transmissible spongiform encephalopathies (prion diseases) revisited: from apoptosis to autophagy. Int J Biochem Cell Biol 2004; 36:2473–2490 [View Article][PubMed]
    [Google Scholar]
  15. Hope J, Shearman MS, Baxter HC, Chong A, Kelly SM et al. Cytotoxicity of prion protein peptide (PrP106-126) differs in mechanism from the cytotoxic activity of the Alzheimer's disease amyloid peptide, Aβ25-35. Neurodegeneration 1996; 5:1–11 [View Article][PubMed]
    [Google Scholar]
  16. Amor S, Peferoen LA, Vogel DY, Breur M, van der Valk P et al. Inflammation in neurodegenerative diseases– an update. Immunology 2014; 142:151–166 [View Article][PubMed]
    [Google Scholar]
  17. Ransohoff RM. How neuroinflammation contributes to neurodegeneration. Science 2016; 353:777–783 [View Article][PubMed]
    [Google Scholar]
  18. Valera E, Spencer B, Masliah E. Immunotherapeutic approaches targeting Amyloid-β, α-Synuclein, and tau for the treatment of neurodegenerative disorders. Neurotherapeutics 2016; 13:179–189 [View Article][PubMed]
    [Google Scholar]
  19. Zilka N, Korenova M, Novak M. Misfolded tau protein and disease modifying pathways in transgenic rodent models of human tauopathies. Acta Neuropathol 2009; 118:71–86 [View Article][PubMed]
    [Google Scholar]
  20. Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A et al. Who fans the flames of Alzheimer's disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation 2012; 9:47 [View Article][PubMed]
    [Google Scholar]
  21. Wong YC, Krainc D. α-synuclein toxicity in neurodegeneration: mechanism and therapeutic strategies. Nat Med 2017; 23:1–13 [View Article][PubMed]
    [Google Scholar]
  22. Reiss AB, Wirkowski E. Statins in neurological disorders: mechanisms and therapeutic value. Scientific World J 2009; 9:1242–1259 [View Article][PubMed]
    [Google Scholar]
  23. Wang Q, Yan J, Chen X, Li J, Yang Y et al. Statins: multiple neuroprotective mechanisms in neurodegenerative diseases. Exp Neurol 2011; 230:27–34 [View Article][PubMed]
    [Google Scholar]
  24. Bedi O, Dhawan V, Sharma PL, Kumar P. Pleiotropic effects of statins: new therapeutic targets in drug design. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:695–712 [View Article][PubMed]
    [Google Scholar]
  25. Kumar A, Sharma N, Gupta A, Kalonia H, Mishra J. Neuroprotective potential of atorvastatin and simvastatin (HMG-CoA reductase inhibitors) against 6-hydroxydopamine (6-OHDA) induced Parkinson-like symptoms. Brain Res 2012; 1471:13–22 [View Article][PubMed]
    [Google Scholar]
  26. Xu YQ, Long L, Yan JQ, Wei L, Pan MQ et al. Simvastatin induces neuroprotection in 6-OHDA-lesioned PC12 via the PI3K/AKT/caspase 3 pathway and anti-inflammatory responses. CNS Neurosci Ther 2013; 19:170–177 [View Article][PubMed]
    [Google Scholar]
  27. Ghosh A, Roy A, Matras J, Brahmachari S, Gendelman HE et al. Simvastatin inhibits the activation of p21ras and prevents the loss of dopaminergic neurons in a mouse model of Parkinson's disease. J Neurosci 2009; 29:13543–13556 [View Article][PubMed]
    [Google Scholar]
  28. Selley ML. Simvastatin prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced striatal dopamine depletion and protein tyrosine nitration in mice. Brain Res 2005; 1037:1–6 [View Article][PubMed]
    [Google Scholar]
  29. Zhao L, Chen T, Wang C, Li G, Zhi W et al. Atorvastatin in improvement of cognitive impairments caused by amyloid β in mice: involvement of inflammatory reaction. BMC Neurol 2016; 16:18 [View Article][PubMed]
    [Google Scholar]
  30. Zhang YY, Fan YC, Wang M, Wang D, Li XH. Atorvastatin attenuates the production of IL-1β, IL-6, and TNF-α in the hippocampus of an amyloid β1-42-induced rat model of Alzheimer's disease. Clin Interv Aging 2013; 8:103–110 [View Article][PubMed]
    [Google Scholar]
  31. Youssef S, Stüve O, Patarroyo JC, Ruiz PJ, Radosevich JL et al. The HMG-CoA reductase inhibitor, atorvastatin, promotes a Th2 bias and reverses paralysis in central nervous system autoimmune disease. Nature 2002; 420:78–84 [View Article][PubMed]
    [Google Scholar]
  32. Stanislaus R, Singh AK, Singh I. Lovastatin treatment decreases mononuclear cell infiltration into the CNS of Lewis rats with experimental allergic encephalomyelitis. J Neurosci Res 2001; 66:155–162 [View Article][PubMed]
    [Google Scholar]
  33. Greenwood J, Walters CE, Pryce G, Kanuga N, Beraud E et al. Lovastatin inhibits brain endothelial cell Rho-mediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitis. Faseb J 2003; 17:905–907 [View Article][PubMed]
    [Google Scholar]
  34. Stanislaus R, Pahan K, Singh AK, Singh I. Amelioration of experimental allergic encephalomyelitis in Lewis rats by lovastatin. Neurosci Lett 1999; 269:71–74 [View Article][PubMed]
    [Google Scholar]
  35. Klotz U. Pharmacological comparison of the statins. Arzneimittelforschung 2003; 53:605–611 [View Article][PubMed]
    [Google Scholar]
  36. Vetrugno V, di Bari MA, Nonno R, Puopolo M, D'Agostino C et al. Oral pravastatin prolongs survival time of scrapie-infected mice. J Gen Virol 2009; 90:1775–1780 [View Article][PubMed]
    [Google Scholar]
  37. Haviv Y, Avrahami D, Ovadia H, Ben-Hur T, Gabizon R et al. Induced neuroprotection independently from PrPSc accumulation in a mouse model for prion disease treated with simvastatin. Arch Neurol 2008; 65:762–775 [View Article][PubMed]
    [Google Scholar]
  38. Kempster S, Bate C, Williams A. Simvastatin treatment prolongs the survival of scrapie-infected mice. Neuroreport 2007; 18:479–482 [View Article][PubMed]
    [Google Scholar]
  39. Mok SW, Thelen KM, Riemer C, Bamme T, Gültner S et al. Simvastatin prolongs survival times in prion infections of the central nervous system. Biochem Biophys Res Commun 2006; 348:697–702 [View Article][PubMed]
    [Google Scholar]
  40. Tribouillard-Tanvier D, Race B, Striebel JF, Carroll JA, Phillips K et al. Early cytokine elevation, PrPres deposition, and gliosis in mouse scrapie: no effect on disease by deletion of cytokine genes IL-12p40 and IL-12p35. J Virol 2012; 86:10377–10383 [View Article][PubMed]
    [Google Scholar]
  41. Chen C, Lin J, Smolarek T, Tremaine L. P-glycoprotein has differential effects on the disposition of statin acid and lactone forms in mdr1a/b knockout and wild-type mice. Drug Metab Dispos 2007; 35:1725–1729 [View Article][PubMed]
    [Google Scholar]
  42. Johnson-Anuna LN, Eckert GP, Keller JH, Igbavboa U, Franke C et al. Chronic administration of statins alters multiple gene expression patterns in mouse cerebral cortex. J Pharmacol Exp Ther 2005; 312:786–793 [View Article][PubMed]
    [Google Scholar]
  43. Wong K, Bumpstead S, van der Weyden L, Reinholdt LG, Wilming LG et al. Sequencing and characterization of the FVB/NJ mouse genome. Genome Biol 2012; 13:R72 [View Article][PubMed]
    [Google Scholar]
  44. Sellers RS. Translating mouse models: immune variation and efficacy testing. Toxicol Pathol 2017; 45:134–145 [CrossRef]
    [Google Scholar]
  45. Wilson NO, Solomon W, Anderson L, Patrickson J, Pitts S et al. Pharmacologic inhibition of CXCL10 in combination with anti-malarial therapy eliminates mortality associated with murine model of cerebral malaria. PLoS One 2013; 8:e60898 [View Article][PubMed]
    [Google Scholar]
  46. Mcmanus RM, Mills KH, Lynch MA. T cells-protective or pathogenic in Alzheimer's disease?. J Neuroimmune Pharmacol 2015; 10:547–560 [View Article][PubMed]
    [Google Scholar]
  47. Belnoue E, Kayibanda M, Vigario AM, Deschemin JC, van Rooijen N et al. On the pathogenic role of brain-sequestered αβ CD8+ T cells in experimental cerebral malaria. J Immunol 2002; 169:6369–6375 [View Article][PubMed]
    [Google Scholar]
  48. Jäger A, Dardalhon V, Sobel RA, Bettelli E, Kuchroo VK. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J Immunol 2009; 183:7169–7177 [View Article][PubMed]
    [Google Scholar]
  49. Ben-Nun A, Wekerle H, Cohen IR. The rapid isolation of clonable antigen-specific T lymphocyte lines capable of mediating autoimmune encephalomyelitis. Eur J Immunol 1981; 11:195–199 [View Article][PubMed]
    [Google Scholar]
  50. Williams AE, Lawson LJ, Perry VH, Fraser H. Characterization of the microglial response in murine scrapie. Neuropathol Appl Neurobiol 1994; 20:47–55 [View Article][PubMed]
    [Google Scholar]
  51. Betmouni S, Perry VH, Gordon JL. Evidence for an early inflammatory response in the central nervous system of mice with scrapie. Neuroscience 1996; 74:1–5 [View Article][PubMed]
    [Google Scholar]
  52. Lewicki H, Tishon A, Homann D, Mazarguil H, Laval F et al. T cells infiltrate the brain in murine and human transmissible spongiform encephalopathies. J Virol 2003; 77:3799–3808 [View Article][PubMed]
    [Google Scholar]
  53. Neuhaus O, Strasser-Fuchs S, Fazekas F, Kieseier BC, Niederwieser G et al. Statins as immunomodulators: comparison with interferon-β1b in MS. Neurology 2002; 59:990–997 [View Article][PubMed]
    [Google Scholar]
  54. Undela K, Gudala K, Malla S, Bansal D. Statin use and risk of Parkinson's disease: a meta-analysis of observational studies. J Neurol 2013; 260:158–165 [View Article][PubMed]
    [Google Scholar]
  55. Friedman B, Lahad A, Dresner Y, Vinker S. Long-term statin use and the risk of Parkinson's disease. Am J Manag Care 2013; 19:626–632[PubMed]
    [Google Scholar]
  56. Gao X, Simon KC, Schwarzschild MA, Ascherio A. Prospective study of statin use and risk of Parkinson disease. Arch Neurol 2012; 69:380–384 [View Article][PubMed]
    [Google Scholar]
  57. Huang X, Alonso A, Guo X, Umbach DM, Lichtenstein ML et al. Statins, plasma cholesterol, and risk of Parkinson's disease: a prospective study. Mov Disord 2015; 30:552–559 [View Article][PubMed]
    [Google Scholar]
  58. Tison F, Nègre-Pagès L, Meissner WG, Dupouy S, Li Q et al. Simvastatin decreases levodopa-induced dyskinesia in monkeys, but not in a randomized, placebo-controlled, multiple cross-over ("n-of-1") exploratory trial of simvastatin against levodopa-induced dyskinesia in Parkinson's disease patients. Parkinsonism Relat Disord 2013; 19:416–421 [View Article][PubMed]
    [Google Scholar]
  59. Sparks DL, Sabbagh M, Connor D, Soares H, Lopez J et al. Statin therapy in Alzheimer's disease. Acta Neurol Scand Suppl 2006; 185:78–86 [View Article][PubMed]
    [Google Scholar]
  60. Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer's disease. Ther Adv Neurol Disord 2013; 6:19–33 [View Article][PubMed]
    [Google Scholar]
  61. Feldman HH, Doody RS, Kivipelto M, Sparks DL, Waters DD et al. Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease: LEADe. Neurology 2010; 74:956–964 [View Article][PubMed]
    [Google Scholar]
  62. Simons M, Schwärzler F, Lütjohann D, von Bergmann K, Beyreuther K et al. Treatment with simvastatin in normocholesterolemic patients with Alzheimer's disease: A 26-week randomized, placebo-controlled, double-blind trial. Ann Neurol 2002; 52:346–350 [View Article][PubMed]
    [Google Scholar]
  63. Trompet S, van Vliet P, de Craen AJ, Jolles J, Buckley BM et al. Pravastatin and cognitive function in the elderly. Results of the PROSPER study. J Neurol 2010; 257:85–90 [View Article][PubMed]
    [Google Scholar]
  64. Pihl-Jensen G, Tsakiri A, Frederiksen JL. Statin treatment in multiple sclerosis: a systematic review and meta-analysis. CNS Drugs 2015; 29:277–291 [View Article][PubMed]
    [Google Scholar]
  65. Birnbaum G, Cree B, Altafullah I, Zinser M, Reder AT. Combining beta interferon and atorvastatin may increase disease activity in multiple sclerosis. Neurology 2008; 71:1390–1395 [View Article][PubMed]
    [Google Scholar]
  66. Lanzillo R, Orefice G, Quarantelli M, Rinaldi C, Prinster A et al. Atorvastatin combined to interferon to verify the efficacy (ACTIVE) in relapsing-remitting active multiple sclerosis patients: a longitudinal controlled trial of combination therapy. Mult Scler 2010; 16:450–454 [View Article][PubMed]
    [Google Scholar]
  67. Togha M, Karvigh SA, Nabavi M, Moghadam NB, Harirchian MH et al. Simvastatin treatment in patients with relapsing-remitting multiple sclerosis receiving interferon beta 1a: a double-blind randomized controlled trial. Mult Scler 2010; 16:848–854 [View Article][PubMed]
    [Google Scholar]
  68. Sorensen PS, Lycke J, Erälinna JP, Edland A, Wu X et al. Simvastatin as add-on therapy to interferon β-1a for relapsing-remitting multiple sclerosis (SIMCOMBIN study): a placebo-controlled randomised phase 4 trial. Lancet Neurol 2011; 10:691–701 [View Article][PubMed]
    [Google Scholar]
  69. Demaimay R, Adjou KT, Beringue V, Demart S, Lasmézas CI et al. Late treatment with polyene antibiotics can prolong the survival time of scrapie-infected animals. J Virol 1997; 71:9685–9689[PubMed]
    [Google Scholar]
  70. Pocchiari M, Schmittinger S, Masullo C. Amphotericin B delays the incubation period of scrapie in intracerebrally inoculated hamsters. J Gen Virol 1987; 68:219–223 [View Article][PubMed]
    [Google Scholar]
  71. Priola SA, Raines A, Caughey WS. Porphyrin and phthalocyanine antiscrapie compounds. Science 2000; 287:1503–1506 [View Article][PubMed]
    [Google Scholar]
  72. Striebel JF, Race B, Carroll JA, Phillips K, Chesebro B. Knockout of fractalkine receptor Cx3cr1 does not alter disease or microglial activation in prion-infected mice. J Gen Virol 2016; 97:1481–1487 [View Article][PubMed]
    [Google Scholar]
  73. Meade-White K, Race B, Trifilo M, Bossers A, Favara C et al. Resistance to chronic wasting disease in transgenic mice expressing a naturally occurring allelic variant of deer prion protein. J Virol 2007; 81:4533–4539 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.000876
Loading
/content/journal/jgv/10.1099/jgv.0.000876
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error