1887

Abstract

Prion diseases are a unique group of transmissible, typically sub-acute, neurodegenerative disorders. During central nervous system (CNS) prion disease, the microglia become activated and are thought to provide a protective response by scavenging and clearing prions. The mammalian intestine is host to a large burden of commensal micro-organisms, especially bacteria, termed the microbiota. The commensal microbiota has beneficial effects on host health, including through the metabolism of essential nutrients, regulation of host development and protection against pathogens. The commensal gut microbiota also constitutively regulates the functional maturation of microglia in the CNS, and microglial function is impaired when it is absent in germ-free mice. In the current study, we determined whether the absence of the commensal gut microbiota might also affect prion disease pathogenesis. Our data clearly show that the absence of the commensal microbiota in germ-free mice did not affect prion disease duration or susceptibility after exposure to prions by intraperitoneal or intracerebral injection. Furthermore, the magnitude and distribution of the characteristic neuropathological hallmarks of terminal prion disease in the CNS, including the development of spongiform pathology, accumulation of prion disease-specific protein (PrP), astrogliosis and microglial activation, were similar in conventionally housed and germ-free mice. Thus, although the commensal gut microbiota constitutively promotes the maintenance of the microglia in the CNS under steady-state conditions in naïve mice, our data suggest that dramatic changes to the abundance or complexity of the commensal gut microbiota are unlikely to influence CNS prion disease pathogenesis.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000860
2017-07-01
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/7/1943.html?itemId=/content/journal/jgv/10.1099/jgv.0.000860&mimeType=html&fmt=ahah

References

  1. Prusiner SB, Bolton DC, Groth DF, Bowman KA, Cochran SP et al. Further purification and characterization of scrapie prions. Biochemistry 1982; 21:6942–6950 [View Article][PubMed]
    [Google Scholar]
  2. Legname G, Baskakov IV, Nguyen HO, Riesner D, Cohen FE et al. Synthetic mammalian prions. Science 2004; 305:673–676 [View Article][PubMed]
    [Google Scholar]
  3. Wang F, Wang X, Yuan CG, Ma J. Generating a prion with bacterially expressed recombinant prion protein. Science 2010; 327:1132–1135 [View Article][PubMed]
    [Google Scholar]
  4. Vincenti JE, Murphy L, Grabert K, Mccoll BW, Cancellotti E et al. Defining the microglia response during the time course of chronic neurodegeneration. J Virol 2015; 90:3003–3017 [View Article][PubMed]
    [Google Scholar]
  5. Alibhai J, Blanco RA, Barria MA, Piccardo P, Caughey B et al. Distribution of misfolded prion protein seeding activity alone does not predict regions of neurodegeneration. PLoS Biol 2016; 14:e1002579 [View Article][PubMed]
    [Google Scholar]
  6. Kranich J, Krautler NJ, Falsig J, Ballmer B, Li S et al. Engulfment of cerebral apoptotic bodies controls the course of prion disease in a mouse strain–dependent manner. J Exp Med 2010; 207:2271–2281 [View Article][PubMed]
    [Google Scholar]
  7. Zhan Y, Paolicelli RC, Sforazzini F, Weinhard L, Bolasco G et al. Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat Neurosci 2014; 17:400–406 [View Article][PubMed]
    [Google Scholar]
  8. Prinz M, Priller J. Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat Rev Neurosci 2014; 15:300–312 [View Article][PubMed]
    [Google Scholar]
  9. de Lucia C, Rinchon A, Olmos-Alonso A, Riecken K, Fehse B et al. Microglia regulate hippocampal neurogenesis during chronic neurodegeneration. Brain Behav Immun 2016; 55:179–190 [View Article][PubMed]
    [Google Scholar]
  10. Williams A, Lucassen PJ, Ritchie D, Bruce M. PrP deposition, microglial activation, and neuronal apoptosis in murine scrapie. Exp Neurol 1997; 144:433–438 [View Article][PubMed]
    [Google Scholar]
  11. Boche D, Cunningham C, Gauldie J, Perry VH. Transforming growth factor-β1-mediated neuroprotection against excitotoxic injury in vivo. J Cereb Blood Flow Metab 2003; 23:1174–1182 [View Article][PubMed]
    [Google Scholar]
  12. Minghetti L, Greco A, Cardone F, Puopolo M, Ladogana A et al. Increased brain synthesis of prostaglandin E2 and F2-isoprostane in human and experimental transmissible spongiform encephalopathies. J Neuropathol Exp Neurol 2000; 59:866–871 [View Article]
    [Google Scholar]
  13. Walsh DT, Perry VH, Minghetti L. Cyclooxygenase-2 is highly expressed in microglial-like cells in a murine model of prion disease. Glia 2000; 29:392–396 [View Article][PubMed]
    [Google Scholar]
  14. Cunningham C, Deacon R, Wells H, Boche D, Waters S et al. Synaptic changes characterize early behavioural signs in the ME7 model of murine prion disease. Eur J Neurosci 2003; 17:2147–2155 [View Article]
    [Google Scholar]
  15. Zhu C, Herrmann US, Falsig J, Abakumova I, Nuvolone M et al. A neuroprotective role for microglia in prion diseases. J Exp Med 2016; 213:1047–1059 [View Article]
    [Google Scholar]
  16. Combrinck MI, Perry VH, Cunningham C. Peripheral infection evokes exaggerated sickness behaviour in pre-clinical murine prion disease. Neuroscience 2002; 112:7–11 [View Article][PubMed]
    [Google Scholar]
  17. Cunningham C, Wilcockson DC, Campion S, Lunnon K, Perry VH. Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. J Neurosci 2005; 25:9275–9284 [View Article][PubMed]
    [Google Scholar]
  18. Sakai K, Hasebe R, Takahashi Y, Song CH, Suzuki A et al. Absence of CD14 delays progression of prion diseases accompanied by increased microglial activation. J Virol 2013; 87:13433–13445 [View Article][PubMed]
    [Google Scholar]
  19. Sommer F, Bäckhed F. The gut microbiotamasters of host development and physiology. Nat Rev Microbiol 2013; 11:227–238 [View Article]
    [Google Scholar]
  20. Russell WR, Hoyles L, Flint HJ, Dumas ME. Colonic bacterial metabolites and human health. Curr Opin Microbiol 2013; 16:246–254 [View Article]
    [Google Scholar]
  21. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science 2012; 336:1268–1273 [View Article]
    [Google Scholar]
  22. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013; 504:446–450 [View Article][PubMed]
    [Google Scholar]
  23. Kamada N, Kim YG, Sham HP, Vallance BA, Puente JL et al. Regulated virulence controls the ability of a pathogen to compete with the gut microbiota. Science 2012; 336:1325–1329 [View Article]
    [Google Scholar]
  24. Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci 2011; 12:453–466 [View Article][PubMed]
    [Google Scholar]
  25. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol 2012; 10:735–742 [View Article]
    [Google Scholar]
  26. Bauer PV, Hamr SC, Duca FA. Regulation of energy balance by a gut–brain axis and involvement of the gut microbiota. Cell Mol Life Sci 2016; 73:737–755 [View Article]
    [Google Scholar]
  27. Erny D, Hrabě de Angelis AL, Prinz M. Communicating systems in the body: how microbiota and microglia cooperate. Immunology 2017; 150:7–15 [View Article]
    [Google Scholar]
  28. Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 2015; 18:965–977 [View Article]
    [Google Scholar]
  29. Donaldson DS, Mabbott NA. The influence of the commensal and pathogenic gut microbiota on prion disease pathogenesis. J Gen Virol 2016; 97:1725–1738 [View Article]
    [Google Scholar]
  30. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci 2017; 20:145–155 [View Article]
    [Google Scholar]
  31. Lev M, Raine CS, Levenson SM. Enhanced survival of germfree mice after infection with irradiated scrapie brain. Experientia 1971; 27:1358–1359 [View Article]
    [Google Scholar]
  32. Wade WF, Dees C, German TL, Marsh RF. Effect of bacterial Flora and mouse genotype (euthymic or athymic) on scrapie pathogenesis. J Leukoc Biol 1986; 40:525–532[PubMed]
    [Google Scholar]
  33. Mabbott NA, Mackay F, Minns F, Bruce ME. Temporary inactivation of follicular dendritic cells delays neuroinvasion of scrapie. Nat Med 2000; 6:719–720 [View Article][PubMed]
    [Google Scholar]
  34. Montrasio F, Frigg R, Glatzel M, Klein MA, Mackay F et al. Impaired prion replication in spleens of mice lacking functional follicular dendritic cells. Science 2000; 288:1257–1259 [View Article][PubMed]
    [Google Scholar]
  35. Mcculloch L, Brown KL, Bradford BM, Hopkins J, Bailey M et al. Follicular dendritic cell-specific prion protein (PrPC) expression alone is sufficient to sustain prion infection in the spleen. PLoS Pathog 2011; 7:e1002402 [View Article][PubMed]
    [Google Scholar]
  36. Obata T, Goto Y, Kunisawa J, Sato S, Sakamoto M et al. Indigenous opportunistic bacteria inhabit mammalian gut-associated lymphoid tissues and share a mucosal antibody-mediated symbiosis. Proc Natl Acad Sci USA 2010; 107:7419–7424 [View Article][PubMed]
    [Google Scholar]
  37. Lécuyer E, Rakotobe S, Lengliné-Garnier H, Lebreton C, Picard M et al. Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses. Immunity 2014; 40:608–620 [View Article]
    [Google Scholar]
  38. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med 2009; 1:6ra14 [View Article]
    [Google Scholar]
  39. Magnusson KR, Hauck L, Jeffrey BM, Elias V, Humphrey A et al. Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience 2015; 300:128–140 [View Article][PubMed]
    [Google Scholar]
  40. Zhu C, Schwarz P, Abakumova I, Aguzzi A. Unaltered prion pathogenesis in a mouse model of high-fat diet-induced insulin resistance. PLoS One 2015; 10:e0144983 [View Article]
    [Google Scholar]
  41. Prinz M, Huber G, Macpherson AJS, Heppner FL, Glatzel M et al. Oral prion infection requires normal numbers of Peyer's patches but not of enteric lymphocytes. Am J Pathol 2003; 162:1103–1111 [View Article]
    [Google Scholar]
  42. Horiuchi M, Furuoka H, Kitamura N, Shinagaw M. Alymphoplasia mice are resistant to prion infection via oral route. Jpn J Vet Res 2006; 53:149–157[PubMed]
    [Google Scholar]
  43. Glaysher BR, Mabbott NA. Role of the GALT in scrapie agent neuroinvasion from the intestine. J Immunol 2007; 178:3757–3766 [View Article][PubMed]
    [Google Scholar]
  44. Donaldson DS, Else KJ, Mabbott NA. The gut-associated lymphoid tissues in the small intestine, not the large intestine, play a major role in oral prion disease pathogenesis. J Virol 2015; 89:9532–9547 [View Article][PubMed]
    [Google Scholar]
  45. Bradford BM, Reizis B, Mabbott NA. Oral prion disease pathogenesis is impeded in the specific absence of CXCR5-expressing dendritic cells. J Virol 2017; 91:e00124-17 [View Article]
    [Google Scholar]
  46. Donaldson DS, Bradford BM, Artis D, Mabbott NA. Reciprocal regulation of lymphoid tissue development in the large intestine by IL-25 and IL-23. Mucosal Immunol 2015; 8:582–595 [View Article]
    [Google Scholar]
  47. Kashyap PC, Marcobal A, Ursell LK, Larauche M, Duboc H et al. Complex interactions among diet, gastrointestinal transit, and gut microbiota in humanized mice. Gastroenterology 2013; 144:967–977 [View Article]
    [Google Scholar]
  48. Reikvam DH, Erofeev A, Sandvik A, Grcic V, Jahnsen FL et al. Depletion of murine intestinal microbiota: effects on gut mucosa and epithelial gene expression. PLoS One 2011; 6:e17996 [View Article]
    [Google Scholar]
  49. Lunnon K, Teeling JL, Tutt AL, Cragg MS, Glennie MJ et al. Systemic inflammation modulates Fc receptor expression on microglia during chronic neurodegeneration. J Immunol 2011; 186:7215–7224 [View Article]
    [Google Scholar]
  50. Cunningham C, Campion S, Lunnon K, Murray CL, Woods JFC et al. Systemic inflammation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biol Psychiatry 2009; 65:304–312 [View Article]
    [Google Scholar]
  51. Sigurdson CJ, Heikenwalder M, Manco G, Barthel M, Schwarz P et al. Bacterial colitis increases susceptibility to oral prion disease. J Infect Dis 2009; 199:243–252 [View Article][PubMed]
    [Google Scholar]
  52. Donaldson DS, Sehgal A, Rios D, Williams IR, Mabbott NA. Increased abundance of M cells in the gut epithelium dramatically enhances oral prion disease susceptibility. PLoS Pathog 2016; 12:e1006075 [View Article]
    [Google Scholar]
  53. Valleron A-J, Boelle P-Y, Will R, Cesbron J-Y. Estimation of epidemic size and incubation time based on age characteristics of vCJD in the United Kingdom. Science 2001; 294:1726–1728 [View Article]
    [Google Scholar]
  54. Wilesmith JW. BSE: Epidemiological approaches, trials and tribulations. Prev Vet Med 1993; 18:33–42 [View Article]
    [Google Scholar]
  55. Diack AB, Head MW, Mccutcheon S, Boyle A, Knight R et al. Variant CJD. 18 years of research and surveillance. Prion 2014; 8:286–295 [CrossRef]
    [Google Scholar]
  56. Gill ON, Spencer Y, Richard-Loendt A, Kelly C, Dabaghian R et al. Prevalent abnormal prion protein in human appendixes after bovine spongiform encephalopathy epizootic: large scale survey. BMJ 2013; 347:f5675 [View Article][PubMed]
    [Google Scholar]
  57. Lorenz RG, Chaplin DD, Mcdonald KG, Mcdonough JS, Newberry RD. Isolated lymphoid follicle formation is inducible and dependent upon lymphotoxin-sufficient B lymphocytes, lymphotoxin β receptor, and TNF receptor I function. J Immunol 2003; 170:5475–5482 [View Article]
    [Google Scholar]
  58. Pabst O, Herbrand H, Friedrichsen M, Velaga S, Dorsch M et al. Adaptation of solitary intestinal lymphoid tissue in response to microbiota and chemokine receptor CCR7 signaling. J Immunol 2006; 177:6824–6832 [View Article]
    [Google Scholar]
  59. Fraser H, Dickinson AG. The sequential development of the brain lesions of scrapie in three strains of mice. J Comp Pathol 1968; 78:301–311 [View Article][PubMed]
    [Google Scholar]
  60. Mccutcheon S, Langeveld JPM, Tan BC, Gill AC, de Wolf C et al. Prion protein-specific antibodies that detect multiple TSE agents with high sensitivity. PLoS One 2014; 9:e91143 [View Article]
    [Google Scholar]
  61. Inman CF, Rees LEN, Barker E, Haverson K, Stokes CR et al. Validation of computer-assisted, pixel-based analysis of multiple-colour immunofluorescence histology. J Immunol Methods 2005; 302:156–167 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.000860
Loading
/content/journal/jgv/10.1099/jgv.0.000860
Loading

Data & Media loading...

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