#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Prion Replication Occurs in Endogenous Adult Neural Stem Cells and Alters Their Neuronal Fate: Involvement of Endogenous Neural Stem Cells in Prion Diseases


Prion diseases are irreversible progressive neurodegenerative diseases, leading to severe incapacity and death. They are characterized in the brain by prion amyloid deposits, vacuolisation, astrocytosis, neuronal degeneration, and by cognitive, behavioural and physical impairments. There is no treatment for these disorders and stem cell therapy therefore represents an interesting new approach. Gains could not only result from the cell transplantation, but also from the stimulation of endogenous neural stem cells (NSC) or by the combination of both approaches. However, the development of such strategies requires a detailed knowledge of the pathology, particularly concerning the status of the adult neurogenesis and endogenous NSC during the development of the disease. During the past decade, several studies have consistently shown that NSC reside in the adult mammalian central nervous system (CNS) and that adult neurogenesis occurs throughout the adulthood in the subventricular zone of the lateral ventricle or the Dentate Gyrus of the hippocampus. Adult NSC are believed to constitute a reservoir for neuronal replacement during normal cell turnover or after brain injury. However, the activation of this system does not fully compensate the neuronal loss that occurs during neurodegenerative diseases and could even contribute to the disease progression. We investigated here the status of these cells during the development of prion disorders. We were able to show that NSC accumulate and replicate prions. Importantly, this resulted in the alteration of their neuronal fate which then represents a new pathologic event that might underlie the rapid progression of the disease.


Vyšlo v časopise: Prion Replication Occurs in Endogenous Adult Neural Stem Cells and Alters Their Neuronal Fate: Involvement of Endogenous Neural Stem Cells in Prion Diseases. PLoS Pathog 9(8): e32767. doi:10.1371/journal.ppat.1003485
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003485

Souhrn

Prion diseases are irreversible progressive neurodegenerative diseases, leading to severe incapacity and death. They are characterized in the brain by prion amyloid deposits, vacuolisation, astrocytosis, neuronal degeneration, and by cognitive, behavioural and physical impairments. There is no treatment for these disorders and stem cell therapy therefore represents an interesting new approach. Gains could not only result from the cell transplantation, but also from the stimulation of endogenous neural stem cells (NSC) or by the combination of both approaches. However, the development of such strategies requires a detailed knowledge of the pathology, particularly concerning the status of the adult neurogenesis and endogenous NSC during the development of the disease. During the past decade, several studies have consistently shown that NSC reside in the adult mammalian central nervous system (CNS) and that adult neurogenesis occurs throughout the adulthood in the subventricular zone of the lateral ventricle or the Dentate Gyrus of the hippocampus. Adult NSC are believed to constitute a reservoir for neuronal replacement during normal cell turnover or after brain injury. However, the activation of this system does not fully compensate the neuronal loss that occurs during neurodegenerative diseases and could even contribute to the disease progression. We investigated here the status of these cells during the development of prion disorders. We were able to show that NSC accumulate and replicate prions. Importantly, this resulted in the alteration of their neuronal fate which then represents a new pathologic event that might underlie the rapid progression of the disease.


Zdroje

1. PrusinerSB, ScottMR, DeArmondSJ, CohenFE (1998) Prion protein biology. Cell 93: 337–348.

2. PrusinerSB (1991) Molecular biology of prion diseases. Science 252: 1515–1522.

3. GuentchevM, GroschupMH, KordekR, LiberskiPP, BudkaH (1998) Severe, early and selective loss of a subpopulation of GABAergic inhibitory neurons in experimental transmissible spongiform encephalopathies. Brain Pathol 8: 615–623.

4. Bouzamondo-BernsteinE, HopkinsSD, SpilmanP, Uyehara-LockJ, DeeringC, et al. (2004) The neurodegeneration sequence in prion diseases: evidence from functional, morphological and ultrastructural studies of the GABAergic system. J Neuropathol Exp Neurol 63: 882–899.

5. MallucciG, CollingeJ (2005) Rational targeting for prion therapeutics. Nat Rev Neurosci 6: 23–34.

6. Alvarez-BuyllaA, Garcia-VerdugoJM (2002) Neurogenesis in adult subventricular zone. J Neurosci 22: 629–634.

7. GageFH (2000) Mammalian neural stem cells. Science 287: 1433–1438.

8. CameronHA, McKayRD (2001) Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus. J Comp Neurol 435: 406–417.

9. KempermannG, WiskottL, GageFH (2004) Functional significance of adult neurogenesis. Curr Opin Neurobiol 14: 186–191.

10. KimJ, SchaferJ, MingGL (2006) New directions in neurodegeneration. Expert OpinBiolTher 6: 735–738.

11. MingGL, SongH (2011) Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 70: 687–702.

12. JinK, PeelAL, MaoXO, XieL, CottrellBA, et al. (2004) Increased hippocampal neurogenesis in Alzheimer's disease. Proc Natl Acad Sci U S A 101: 343–347.

13. CayreM, CanollP, GoldmanJE (2009) Cell migration in the normal and pathological postnatal mammalian brain. Prog Neurobiol 88: 41–63.

14. JinK, SunY, XieL, PeelA, MaoXO, et al. (2003) Directed migration of neuronal precursors into the ischemic cerebral cortex and striatum. Mol Cell Neurosci 24: 171–189.

15. HaugheyNJ, NathA, ChanSL, BorchardAC, RaoMS, et al. (2002) Disruption of neurogenesis by amyloid beta-peptide, and perturbed neural progenitor cell homeostasis, in models of Alzheimer's disease. J Neurochem 83: 1509–1524.

16. NaYJ, JinJK, LeeYJ, ChoiEK, CarpRI, et al. (2009) Increased neurogenesis in brains of scrapie-infected mice. Neurosci Lett 449: 66–70.

17. MilhavetO, CasanovaD, ChevallierN, McKayRD, LehmannS (2006) Neural stem cell model for prion propagation. Stem Cells 24: 2284–2291.

18. KoyanagiM, TakahashiJ, ArakawaY, DoiD, FukudaH, et al. (2008) Inhibition of the Rho/ROCK pathway reduces apoptosis during transplantation of embryonic stem cell-derived neural precursors. J Neurosci Res 86: 270–280.

19. SteeleAD, EmsleyJG, OzdinlerPH, LindquistS, MacklisJD (2006) Prion protein (PrPc) positively regulates neural precursor proliferation during developmental and adult mammalian neurogenesis. Proc Natl Acad Sci U S A 103: 3416–3421.

20. SantosTG, SilvaIR, Costa-SilvaB, LepiqueAP, MartinsVR, et al. (2011) Enhanced Neural Progenitor/Stem Cells Self-Renewal via the Interaction of Stress-Inducible Protein 1 with the Prion Protein. Stem Cells 29: 1126–1136.

21. Zomosa-SignoretV, ArnaudJD, FontesP, Alvarez-MartinezMT, LiautardJP (2008) Physiological role of the cellular prion protein. Vet Res 39: 9.

22. RamboldAS, MullerV, RonU, Ben-TalN, WinklhoferKF, et al. (2008) Stress-protective signalling of prion protein is corrupted by scrapie prions. Embo J 27: 1974–1984.

23. MilhavetO, McMahonHE, RachidiW, NishidaN, KatamineS, et al. (2000) Prion infection impairs the cellular response to oxidative stress. Proc Natl Acad Sci U S A 97: 13937–13942.

24. CronierS, CarimaloJ, SchaefferB, JaumainE, BeringueV, et al. (2012) Endogenous prion protein conversion is required for prion-induced neuritic alterations and neuronal death. Faseb J 26: 3854–3861.

25. GroteHE, HannanAJ (2007) Regulators of adult neurogenesis in the healthy and diseased brain. Clin Exp Pharmacol Physiol 34: 533–545.

26. Relaño-GinésA, LehmannS, BencsikA, HervaME, TorresJM, et al. (2011) Stem cell therapy extends incubation and survival time in prion infected mice in a time window-dependant manner. Journal of Infectious Diseases 204: 1038–45.

27. CrozetC, LinYL, MettlingC, Mourton-GillesC, CorbeauP, et al. (2004) Inhibition of PrPSc formation by lentiviral gene transfer of PrP containing dominant negative mutations. J Cell Sci 117: 5591–5597.

28. CrozetC, BencsikA, FlamantF, LezmiS, SamarutJ, et al. (2001) Florid plaques in ovine PrP transgenic mice infected with an experimental ovine BSE. EMBO Rep 2: 952–956.

29. CrozetC, FlamantF, BencsikA, AubertD, SamarutJ, et al. (2001) Efficient transmission of two different sheep scrapie isolates in transgenic mice expressing the ovine PrP gene. J Virol 75: 5328–5334.

30. CrozetC, LezmiS, FlamantF, SamarutJ, BaronT, et al. (2007) Peripheral circulation of the prion infectious agent in transgenic mice expressing the ovine prion protein gene in neurons only. J Infect Dis 195: 997–1006.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 8
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#