eISSN: 1509-572x
ISSN: 1641-4640
Folia Neuropathologica
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2009
vol. 47
 
Share:
Share:

Thoughts on mammalian prion strains

Charles Weissmann

Folia Neuropathol 2009; 47 (2): 104-113
Online publish date: 2009/06/26
Article file
Get citation
 
 

Carlton Gajdusek was one of the most brilliant and entertaining individuals I have encountered. Given a keyword, he would enlarge upon it, drawing on his encyclopaedic knowledge, illustrate it with personal anecdotes that would lead to a new theme, in a monologue that could last for hours and would be continued from a distance with long, handwritten letters and volumes of his memoirs. It is with pleasure that I dedicate this review to the memory of the extraordinary scientist who laid the foundations of prion research (albeit not in these terms).

The nature of the agent causing transmissible spongiform encephalopathies (TSEs), the prion, has been debated since the mid-1960s. The concept of
a “slow” or “unconventional” virus [32] as well as that of a “virino”, an infectious agent proposed to consist of an agent-specific nucleic acid enveloped in a host-specified protein [45], lost support as intense efforts in many laboratories failed to identify a TSE-specific nucleic acid or even a nucleic acid long enough to encode a small protein [82,85]. On the other hand, the biochemical linkage of infectivity and PrPSc, an abnormal form of the normal host protein PrPC, as well as the linkage between the PrP gene and both familial prion diseases [40,65] and susceptibility to prions [78,88], provided support for an updated version of the “protein only” hypothesis [37], namely that the infectious agent is an abnormal conformer of PrP, named PrPSc, that “multiplies” by catalyzing the conversion of PrPC into a likeness of itself [76]. The finding that PrP knockout mice were resistant to scrapie [13] fulfilled a central prediction of the “protein-only” hypothesis and significantly promoted its acceptance, without however proving it.
The discovery that different strains of prions can be propagated in inbred mouse strains expressing but
a single PrP sequence, and the finding that prion strains were selective in regard to the cells in which they can replicate, raised two distinct questions:
(1) How is strain specificity encoded by the prion; and (2) how can cells distinguish between different prion strains, as reflected by their ability to propagate them? Whereas strain-specific properties are associated with distinct conformations of PrP [17,19,76], these conformations may be co-determined by other factors, such as the N-linked glycans [17,19] or by cell-derived components, whose presence or absence within an infected cell might co-determine its competence to replicate a particular strain [101].


Normal and abnormal forms of PrP

Mature murine PrPC consists of 254 amino acids, is encoded within one exon of a single-copy gene and is expressed in almost all tissues, albeit preferentially in the brain [6]. PrPC is highly susceptible to proteinase K (PK) digestion, is attached to the outer surface of the plasma membrane by a glycosylphosphatidylinositol (GPI) anchor and can be readily cleaved off with phosphatidylinositol-specific phospholipase C (PIPLC). PrPC may carry two, one or no asparagine-linked glycans, of which there are 52 or more variants [28,83, 84]. Prion-infected brain or cell cultures contain conformers of PrP that differ from PrPC and are collectively designated as PrPSc. One form of PrPSc, designated rPrPSc (or PrPres), is rich in beta-sheet structure and partially resistant to PK, in contrast to PrPC, which is rich in alpha-helical structure [50,81], and sensitive to PK. Infectivity is invariably associated with multimeric forms of PrP [91] and is irreversibly lost after treatment of infectious preparations with denaturants [77]. Beside rPrPSc there are also disease-related PrP species that are susceptible to standard PK digestion at 37°C, termed sPrPSc (“protease-sensitive PrPSc”) [73,74,85-87,95]. sPrPSc differs from PrPC in that it is precipitable by antibodies reacting with rPrPSc but not with PrPC, such as 15B3 [52,70]; moreover, certain epitopes (90-125 in mouse PrP) that are exposed in PrPC are occluded and only revealed after denaturation, as measured by the conformation-dependent immunoassay (CDI) [87]. It has not been established whether sPrPSc is infectious. The three-dimensional structure of rPrPSc has not been determined but models based on various physical measurements have been proposed [36].

Replication of prions

The protein-only hypothesis proposes that prions consist of a conformational isomer of PrPC, and that their replication comes about by an autocatalytic conversion of PrPC to the pathogenic isoform PrPSc. The “nucleation model” predicates that PrPC is in equilibrium with PrPSc (or a precursor thereof), with the equilibrium strongly favoring PrPC, and that PrPSc is stabilized only when it adds to a crystal-like seed of PrPSc [33,41]. Cleavage of aggregates is postulated to explain the exponential increase of PrPSc during infection [72,92]. The nucleation model is most convincingly supported by the protein misfolding cyclic amplification (PMCA) reaction, which converts PrPC into infectious PrPSc by multiple cycles of sonication and incubation [15,31], but also by studies of “yeast prions” [92,96,103]. PMCA allows extensive replication of infectious prions in a cell-free system containing highly purified (but not completely pure) PrPC and poly(A) or poly(dT) [24]. These experiments should definitively lay to rest the perennial proposal that the infectious agent is a virus [64], even though prions may be secreted from cells in particulate form, as exosomes [1,30,98].


De novo generation of prions

The occurrence of sporadic CJD in humans has been attributed to a rare spontaneous conversion of PrPC to PrPSc, which then becomes self-propagating and transmissible. Similarly, apparently spontaneous rare events in sheep and cattle may give rise to “atypical” forms of scrapie and bovine spongiform encephalopathy, respectively [2,9]. Spontaneous generation of prions occurs in transgenic mice bearing a mutated prion gene [89]. Legname et al. reported that injection of a beta-sheet-rich fibrillar preparation derived from recPrP into mice overexpressing PrP resulted in transmissible neurological disease [54,55]. Finally, it has been reported that prions can be generated in unprimed PMCA experiments [3,24]. These very different lines of evidence argue quite persuasively that prions can be generated de novo.


Prion species

A prion species is defined by the amino acid sequence of the donor’s PrP. Transmission of prions between different animal species frequently results in low attack rates and long incubation times, which are abrogated upon repeated transmission to the recipient species. The so-called “species barrier” is ascribed to differences in the PrP sequences between prion donor and recipient that hinder the accretion of host PrPC to the incoming rPrPSc seed [20]. In the case of trans-species prion transfer to mice, the barrier is often, but not always, overcome by replacing the murine PrP genes by their counterpart from the donor, for example Syrian hamster [78], cattle [51], human [52], or cervids [11]. Replacing the murine PrP gene by its counterpart from another species does not recreate the environment of the donor species; many genes other than PrP play a role in susceptibility to prions [56,57,66,68], and Prusiner and colleagues have proposed a critical role for an as yet hypothetical host-derived “protein X” in prion replication, which could be a chaperone facilitating conversion [43,94, 105]. A “species barrier” may also come about within the same animal species; for example, there are two distinct PrP alleles in different mouse lines, the Prnpa (108L, 189T) and the Prnpb allele (108F, 189V), and transfer of prions between mice with differing PrP alleles is subject to a similar barrier as is transfer between different animal species [67].


Prion strains

Prion populations differing in phenotypic properties, such as pathogenic potential or physicochemical characteristics, but associated with PrPSc having the same amino acid sequence, constitute distinct strains. A large number of different prion strains, originally characterized by the incubation time and the neuropathology they elicit, can be propagated indefinitely in hosts homozygous for the PrP gene (Prnp) [12]. Many of the “classical” prion strains currently propagated in mice and hamsters originated from scrapie-infected sheep or goats [26], but distinct strains occur also in man [21] and cattle [7]. The “protein-only” hypothesis proposes that the strain-specific properties of the prion are enciphered exclusively in the conformation of the cognate PrPSc [76]. Indeed, in many instances different strains are associated with PrPSc species differing in physicochemical properties such as susceptibility to PK digestion [53], electrophoretic mobility following PK treatment (reflecting different cleavage sites in the amino proximal region) [8,21,93], stability towards denaturation agents [74] or the ratio of di, mono and unglycosylated forms [21]. The conformation-dependent immunoassay (CDI) provides a sensitive tool for differentiating between different conformations of PrP associated with distinct prion strains [5,86,87]. The protein-only hypothesis thus postulates that each prion strain is associated with a different isoform of PrP that can convert PrPC to a likeness of itself. The idea that
a protein may assume dozens of different stable conformations appears bizarre if one thinks in terms of functional enzymes, but less so if one considers stable polymers or crystals [29]. The concept of “conformation templating” at the protein level is supported by the cell-free conversion experiments of Caughey and his colleagues [16,58], the seeding experiments with PrPC [42,97] and most convincingly by the PMCA-mediated faithful propagation of prion strains [14].

Experiments with “yeast prion” strains have shown that conformational specificity can be propagated in vitro by pure, unglycosylated proteins [48,92]. Nonetheless, in view of the great multiplicity of mammalian prion strains and the apparent propensity of strains to multiply more efficiently in some cell types than in others, it is conceivable that post-translational modifications of PrP, such as glycosylation, sulfoxidation [22], proline cis-trans isomerism [18] or, more likely, association with some cellular components [101] might favor certain PrP conformations and hence the preferential propagation of particular strains.

When a prion strain is transferred from one host species to another and subsequently returned to and propagated for several passages in the original host species, it may in some instances change, or “mutate”. For example, mouse-derived 139A passaged through hamster and subsequently propagated repeatedly in mouse resulted in a new strain, 139A-H2M; however, Me7 subjected to the same procedure remained apparently unchanged [46,47]. Legname et al. reported that a plethora of distinct prion strains, as characterized by incubation time and stability toward guanidinium chloride, were generated by passaging prions generated in mice inoculated with polymerized recombinant PrP to mice overexpressing wild type PrP [55].


Propagation of prions in cultured cells

In vivo, prions and PrPSc are accumulated mainly in neurons, follicular dendritic cells of the spleen and glia; in cell culture, susceptibility to persistent infection has been described not only for cells of neuronal and glial origin, but also for fibroblastic and epithelial cells, inasmuch as they express the PrP sequence corresponding to the prion donor (84, 85) [4]. Cell lines exposed to prions can become acutely infected, as judged by the production of rPrPSc during
a few days, but persistent propagation of infectivity and rPrPSc occurs only in a subset of these lines [99].
N2a neuroblastoma cell populations are susceptible to persistent infection by mouse prions (Chandler strain), albeit with low efficiency [71], which means that they have to be exposed to high concentrations of prions and that only a small fraction of the population becomes persistently infected [79,80]. It is however possible to select from N2a populations subclones that are susceptible to infection at much lower prion concentrations and that yield a higher proportion of infected cells [10,27,49,71]. Although Nishida et al. selected their clone from transgenic N2a cells that overexpressed PrP, this feature is not required for enhanced susceptibility [27,49,100].


Prion strain typing by the cell panel assay (CPA)

Classically, the distinction between mouse-adapted prion strains requires determination of incubation times in at least two mouse lines, that extend over 6-10 months. We have developed a cell-based assay, the cell panel assay (CPA), which can distinguish between various murine prion strains within less than 2 weeks [62].

The CPA is based on the standard scrapie cell assay (SSCA), a method for the rapid and sensitive quantification of prions in vitro. In a typical SSCA, 5000 cells are exposed to a serial dilution of a prion preparation for 3 days and after reaching confluence, propagated for
3 splits. After reaching confluence 20 000 cells are filtered onto the membranes of 96-well filter plates. The dried cells are lysed, PK-digested, denatured with guanidinium thiocyanate and PrPSc is visualized by ELISA. rPrPSc-positive cells present as discrete spots and are counted using the Zeiss KS Elispot system. The proportion of rPrPSc-positive cells (“spots/20 000 cells”) is plotted against the logarithm of the brain homogenate dilutions (Fig. 1) [49,63]. The “response index” (RI) of a cell line to a particular prion strain is the reciprocal of the dilution that gives 300 “spots” (or 1.5% of plated cells, a value lying in the linear part of the response curve) under standard assay conditions. RI values can be converted to LD50 units by comparison with the SSCA of a mouse-titered prion preparation run in parallel. The partially automated SSCA is at least as sensitive as the mouse bioassay, allowing quantitation of a 10–7 dilution of RML-infected brain homogenate (about 30 LD50 units/ml), more precise (standard deviation 15-25%) and at least 10 times faster (10 days). The use of semi-automated equipment allows the simultaneous processing of 500 samples or more in quadruplicate by one operator.

The cell panel assay (CPA) is based on the SSCA and is carried out on a set of four cell lines, PK1, R33, CAD5 and LD9, that show different responses to various prion strains [62]. In a typical CPA, the cells of the 4 lines are exposed to serial dilutions of mouse brain homogenates infected with prion strains RML, Me7, 301C or 22L and subjected to the SSCA. Fig. 1A shows that the RML dilutions required to give 300 “spots”) were 5 × 10–7 on CAD5 cells, 8.3 × 10–7 on PK1 cells, and 5 × 10–6 on LD9 cells, from which the cognate RI vales can be calculated (Table I). In contrast, R33 cells failed to yield 300 spots even at 10–3, the lowest dilution of RML used. Figs. 1B-D show the plots for 22L-, Me7- and 301C-infected brain homogenates, respectively. Because distinct prion preparations may contain different levels of prions, the RI must be referred to some other infectivity-related parameter. We arbitrarily chose as reference value the RI obtained on LD9 cells. By the criterion of the RI ratio, CAD5 cells are 9.9, PK1 cells 6.4 and R33 cells < 0.005 times as responsive to RML as LD9 cells (Table I). The CPA as described allows the distinction of RML, 22L, 301C and Me7 prion strains.


Cell tropism of strains

Different prion strains cause different location of lesions and PrPSc deposition in the brain [23] and, as described above, may exhibit different tropism for cell lines (Table I). The uptake of PrPSc by cultured cells appears to be a non-specific process [61] and therefore discrimination between strains probably reflects the capacity of the cell to replicate prions at an appropriate rate. The idea that strain recognition is mediated by the nature of the glycans carried by PrPSc [38] is not supported by the finding that two distinct prion strains could be propagated by PMCA using unglycosylated PrPC [75].

The mechanism underlying specificity for brain areas and for cell lines need not be the same. Persistent infection requires that the rate of PrPSc synthesis be at least equal to the rate of PrPSc depletion [102]. In cell culture, depletion of PrPSc comes about by degradation, secretion and cell division, whereas in brain, where PrPSc accumulates predominantly in neurons, there is no depletion by cell division. Thus, slowing down cell division of cultured cells not only increases the accumulation of PrPSc [34] but may allow cells to become chronically infected by strains to which they are resistant under normal growth conditions (E. Smith and C.W.). The fact that many drugs that “cure” chronically infected cell lines are largely ineffective in abrogating prion disease in vivo [51] reflects at least in part the fact that there is no PrPSc depletion by cell division in the brain. PrPSc degradation has been attributed to the action of cysteine proteases [59,60]; PrPSc of different strains might show different sensitivity to degradation which could contribute to or determine susceptibility to chronic infection. The other side of the equation, the rate of PrPSc synthesis, reflects the activation energy required for the conversion process and thus is a function of both the structure of the PrPSc multimer, which is believed to be strain-dependent, and of the conformational population distribution [104] of the PrPC serving as substrate (Fig. 2). The finding that the rate of PrPSc amplification by PMCA varies considerably for different murine strains (P. Saá and C.I. Lasmezas, personal communication) supports the view that PrPSc structure might be rate-determining also in vivo. The conformational population distribution may depend on post-translational modifications of PrP such as glycosylation or others mentioned above, but perhaps also on association with cellular components [101] which, by favoring certain PrP conformations, could promote preferential propagation of particular strains. The remarkable affinity of PrPC for RNA [35,69, 90] and the requirement for polyanions in the PMCA reaction using purified PrPC as substrate [24,25] support the view that cell components other than PrPC may play an important role in prion propagation [101]. As suggested by Prusiner and his colleagues [43,94,105], the conversion process may be catalyzed by a chaperone; indeed, different strains might require different chaperones and maybe the chaperone content of
a particular cell type may contribute to its capacity for propagating a particular strain.


Outlook

Many basic questions regarding prion strains remain to be answered. Although there is convincing evidence that the PrPSc conformation of distinct strains is different, it is not known to which extent the conformation may depend on features other than the amino acid sequence of the PrP, for example the nature of the glycans or additional, cell-derived components. It would be desirable to obtain large quantities of highly purified PrPSc from a single cell line, infected separately with several different prion strains, determine the glycans carried by each strain-associated PrPSc and search for associated molecules, such as small RNAs or other cell components. The deepest insight will of course be gained once the
3-dimensional structure of PrPSc can be determined at an appropriate resolution, currently still a formidable task. As regards the cell specificity of prions strains, the question to be answered is whether one or more cell components participate in catalyzing the PrPC-to-PrPSc conversion, and if so, whether such components are specific for different strains or sets of strains. The good news is that there are still many opportunities for making interesting discoveries; the sad news, that Carlton is not here to enjoy them.


Acknowledgements

I thank C.A. Baker and S.P. Mahal for critically reading the manuscript.


References

1. Alais S, Simoes S, Baas D, Lehmann S, Raposo G, Darlix JL, Leblanc P Mouse neuroblastoma cells release prion infectivity associated with exosomal vesicles. Biol Cell 2008; 10: 603-615.

2. Baron T, Biacabe AG, Arsac JN, Benestad S, Groschup MH. Atypical transmissible spongiform encephalopathies (TSEs) in ruminants. Vaccine 2007; 25: 5625-5630.

3. Barria MA, Mukherjee A, Gonzalez-Romero D, Morales R, Soto C. De novo generation of infectious prions in vitro produces a new disease phenotype. PLoS Pathog 2009; 5:e1000421.

4. Bedecs K. Cell culture models to unravel prion protein function and aberrancies in prion diseases. Methods Mol Biol 2008; 459: 1-20.

5. Bellon A, Seyfert-Brandt W, Lang W, Baron H, Groner A, Vey M. Improved conformation-dependent immunoassay: suitability for human prion detection with enhanced sensitivity. J Gen Virol 2003; 84: 1921-1925.

6. Bendheim PE, Brown HR, Rudelli RD, Scala LJ, Goller NL, Wen GY, Kascsak RJ, Cashman NR, Bolton DC. Nearly ubiquitous tissue distribution of the scrapie agent precursor protein. Neurology 1992; 42: 149-156.

7. Beringue V, Bencsik A, Le Dur A, Reine F, Lai TL, Chenais N, Tilly G, Biacabe AG, Baron T, Vilotte JL, Laude H. Isolation from cattle of a prion strain distinct from that causing bovine spongiform encephalopathy. PLoS Pathog 2006; 2(10): e112.

8. Bessen RA, Marsh RF. Biochemical and physical properties of the prion protein from two strains of the transmissible mink encephalopathy agent. J Virol 1992; 66: 2096-2101.

9. Biacabe AG, Morignat E, Vulin J, Calavas D, Baron TG. Atypical bovine spongiform encephalopathies, France, 2001-2007. Emerg Infect Dis 2008; 14: 298-300.

10. Bosque PJ, Prusiner SB. Cultured cell sublines highly susceptible to prion infection. J Virol 2000; 74: 4377-4386.

11. Browning SR, Mason GL, Seward T, Green M, Eliason GA, Mathiason C, Miller MW, Williams ES, Hoover E, Telling GC. Transmission of prions from mule deer and elk with chronic wasting disease to transgenic mice expressing cervid PrP. J Virol 2004; 78: 13345-13350.

12. Bruce ME, Fraser H, McBride PA, Scott JR, Dickinson AG. The basis of strain variation in scrapie. In: Prusiner SB, Collinge J, Powell J, Anderton B (eds.). Prion Diseases of Humans and Animals. Ellis Horwood: New York, London 1992: pp. 497-508.

13. Büeler H, Aguzzi A, Sailer A, Greiner RA, Autenried P, Aguet M, Weissmann C. Mice devoid of PrP are resistant to scrapie. Cell 1993; 73: 1339-1347.

14. Castilla J, Morales R, Saa P, Barria M, Gambetti P, Soto C. Cell-free propagation of prion strains. EMBO J 2008; 27: 2557-2566.

15. Castilla J, Saa P, Morales R, Abid K, Maundrell K, Soto C. Protein misfolding cyclic amplification for diagnosis and prion propagation studies. Methods Enzymol 2006; 412: 3-21.

16. Caughey B, Raymond GJ, Priola SA, Kocisko DA, Race RE, Bessen RA, Lansbury PT, Jr., Chesebro B. Methods for studying prion protein (PrP) metabolism and the formation of protease-resistant PrP in cell culture and cell-free systems. An update. Mol Biotechnol 1999; 3: 45-55.

17. Chien P, Weissman JS, DePace AH. Emerging principles of conformation-based prion inheritance. Annu Rev Biochem 2004; 73: 617-656.

18. Cohen E, Taraboulos A. Scrapie-like prion protein accumulates in aggresomes of cyclosporin A-treated cells. EMBO J 2003; 22: 404-417.

19. Collinge J. Prion diseases of humans and animals: their causes and molecular basis. Annu Rev Neurosci 2001; 24: 519-550.

20. Collinge J, Clarke AR. A general model of prion strains and their pathogenicity. Science 2007; 318: 930-936.

21. Collinge J, Sidle KC, Meads J, Ironside J, Hill AF. Molecular analysis of prion strain variation and the aetiology of ‘new variant’ CJD [see comments]. Nature 1996; 383: 685-690.

22. Colombo G, Meli M, Morra G, Gabizon R, Gasset M. Methionine sulfoxides on prion protein Helix-3 switch on the alpha-fold destabilization required for conversion. PLoS ONE 2009; 4: e4296.

23. DeArmond SJ, Yang SL, Lee A, Bowler R, Taraboulos A, Groth D, Prusiner SB. Three scrapie prion isolates exhibit different accumulation patterns of the prion protein scrapie isoform. Proc Natl Acad Sci U S A 1993; 90: 6449-6453.

24. Deleault NR, Harris BT, Rees JR, Supattapone S. Formation of native prions from minimal components in vitro. Proc Natl Acad Sci U S A 2007; 104: 9741-9746.

25. Deleault NR, Lucassen RW, Supattapone S. RNA molecules stimulate prion protein conversion. Nature 2003; 425: 717-720.

26. Dickinson AG. Scrapie in sheep and goats. In: Kimberlin RH (ed.). Slow Virus Diseases of Animals and Man. Chapter 10, Elsevier/North Holland: Amsterdam, 1976: pp. 209-41.

27. Enari M, Flechsig E, Weissmann C. Scrapie prion protein accumulation by scrapie-infected neuroblastoma cells abrogated by exposure to a prion protein antibody. Proc Natl Acad Sci U S A 2001; 98: 9295-9299.

28. Endo T, Groth D, Prusiner SB, Kobata A. Diversity of oligosaccharide structures linked to asparagines of the scrapie prion protein. Biochemistry 1989; 28: 8380-8388.

29. Faber HR, Matthews BW. A mutant T4 lysozyme displays five different crystal conformations. Nature 1990; 348: 263-266.

30. Fevrier B, Vilette D, Archer F, Loew D, Faigle W, Vidal M, Laude H, Raposo G. Cells release prions in association with exosomes. Proc Natl Acad Sci U S A 2004; 101: 9683-9688.

31. Fujihara A, Atarashi R, Fuse T, Ubagai K, Nakagaki T, Yamaguchi N, Ishibashi D, Katamine S, Nishida N. Hyperefficient PrP(Sc) amplification of mouse-adapted BSE and scrapie strain by protein misfolding cyclic amplification technique. FEBS J 2009; 276: 2841-2848.

32. Gajdusek DC. Unconventional viruses and the origin and disappearance of Kuru. Science 1977; 197: 943-960.

33. Gajdusek DC. Transmissible and non-transmissible amyloidoses: Autocatalytic post-translational conversion of host precursor proteins to beta-pleated configurations. J Neuroimmunology 1988; 20: 95-110.

34. Ghaemmaghami S, Phuan PW, Perkins B, Ullman J, May BC, Cohen FE, Prusiner SB. Cell division modulates prion accumulation in cultured cells. Proc Natl Acad Sci U S A 2007; 104: 17971-17976.

35. Gomes MP, Cordeiro Y, Silva JL. The peculiar interaction between mammalian prion protein and RNA. Prion 2008; 2: 64-66.

36. Govaerts C, Wille H, Prusiner SB, Cohen FE. Evidence for assembly of prions with left-handed beta-helices into trimers. Proc Natl Acad Sci U S A 2004; 101: 8342-8347.

37. Griffith JS. Self-replication and scrapie. Nature 1967; 215: 1043-1044.

38. Hecker R, Taraboulos A, Scott M, Pan KM, Yang SL, Torchia M, Jendroska K, DeArmond SJ, Prusiner SB. Replication of distinct scrapie prion isolates is region specific in brains of transgenic mice and hamsters. Genes Dev 1992; 6: 1213-1228.

39. Hill AF, Joiner S, Beck JA, Campbell TA, Dickinson A, Poulter M, Wadsworth JD, Collinge J. Distinct glycoform ratios of protease resistant prion protein associated with PRNP point mutations. Brain 2006; 129: 676-685.

40. Hsiao K, Baker HF, Crow TJ, Poulter M, Owen F, Terwilliger JD, Westaway D, Ott J, Prusiner SB. Linkage of a prion protein missense variant to Gerstmann-Sträussler syndrome. Nature 1989; 338: 342-345.

41. Jarrett JT, Lansbury PJ. Seeding “one-dimensional crystallization” of amyloid: a pathogenic mechanism in Alzheimer’s disease and scrapie? Cell 1993; 73: 1055-1058.

42. Jones EM, Surewicz WK. Fibril conformation as the basis of species- and strain-dependent seeding specificity of mammalian prion amyloids. Cell 2005; 121: 63-72.

43. Kaneko K, Zulianello L, Scott M, Cooper CM, Wallace AC, James TL, Cohen FE, Prusiner SB. Evidence for protein X binding to
a discontinuous epitope on the cellular prion protein during scrapie prion propagation. Proc Natl Acad Sci U S A 1997; 94: 10069-10074.

44. Khalili-Shirazi A, Summers L, Linehan J, Mallinson G, Anstee D, Hawke S, Jackson GS, Collinge J. PrP glycoforms are associated in a strain-specific ratio in native PrPSc. J Gen Virol 2005; 86: 2635-2644.

45. Kimberlin RH. Scrapie agent: Prions or virinos? Nature 1982; 297: 107-108.

46. Kimberlin RH, Cole S, Walker CA. Temporary and permanent modifications to a single strain of mouse scrapie on transmission to rats and hamsters. J Gen Virol 1987; 68: 1875-1881.

47. Kimberlin RH, Walker CA, Fraser H. The genomic identity of different strains of mouse scrapie is expressed in hamsters and preserved on reisolation in mice. J Gen Virol 1989; 70: 2017-2025.

48. King CY, Diaz-Avalos R. Protein-only transmission of three yeast prion strains. Nature 2004; 428: 319-323.

49. Klohn PC, Stoltze L, Flechsig E, Enari M, Weissmann C. A quantitative, highly sensitive cell-based infectivity assay for mouse scrapie prions. Proc Natl Acad Sci U S A 2003; 100: 11666-11671.

50. Knaus KJ, Morillas M, Swietnicki W, Malone M, Surewicz WK, Yee VC. Crystal structure of the human prion protein reveals
a mechanism for oligomerization. Nat Struct Biol 2001; 8: 770-774.

51. Kocisko DA, Morrey JD, Race RE, Chen J, Caughey B. Evaluation of new cell culture inhibitors of protease-resistant prion protein against scrapie infection in mice. J Gen Virol 2004; 85: 2479-2483.

52. Korth C, Stierli B, Streit P, Moser M, Schaller O, Fischer R, Schulz-Schaeffer W, Kretzschmar H, Raeber A, Braun U, Ehrensperger F, Hornemann S, Glockshuber R, Riek R, Billeter M, Wuthrich K, Oesch B. Prion (PrPSc)-specific epitope defined by a monoclonal antibody. Nature 1997; 390: 74-77.

53. Kuczius T, Groschup MH. Differences in proteinase K resistance and neuronal deposition of abnormal prion proteins characterize bovine spongiform encephalopathy (BSE) and scrapie strains. Mol Med 1999; 5: 406-418.

54. Legname G, Baskakov IV, Nguyen HO, Riesner D, Cohen FE, DeArmond SJ, Prusiner SB. Synthetic mammalian prions. Science 2004; 305: 673-676.

55. Legname G, Nguyen HO, Peretz D, Cohen FE, DeArmond SJ, Prusiner SB. Continuum of prion protein structures enciphers
a multitude of prion isolate-specified phenotypes. Proc Natl Acad Sci U S A 2006; 103: 19105-19110.

56. Lloyd SE, Maytham EG, Pota H, Grizenkova J, Molou E, Uphill J, Hummerich H, Whitfield J, Alpers MP, Mead S, Collinge J. HECTD2 is associated with susceptibility to mouse and human prion disease. PLoS Genet 2009; 5:e1000383.

57. Lloyd SE, Uphill JB, Targonski PV, Fisher EM, Collinge J. Identification of genetic loci affecting mouse-adapted bovine spongiform encephalopathy incubation time in mice. Neurogenetics 2002; 4: 77-81.

58. Lucassen R, Nishina K, Supattapone S. In vitro amplification of protease-resistant prion protein requires free sulfhydryl groups. Biochemistry 2003; 42: 4127-4135.

59. Luhr KM, Nordstrom EK, Low P, Kristensson K. Cathepsin B and L are involved in degradation of prions in GT1-1 neuronal cells. Neuroreport 2004; 15: 1663-1667.

60. Luhr KM, Nordstrom EK, Low P, Ljunggren HG, Taraboulos A, Kristensson K. Scrapie protein degradation by cysteine proteases in CD11c+ dendritic cells and GT1-1 neuronal cells. J Virol 2004; 78: 4776-4782.

61. Magalhaes AC, Baron GS, Lee KS, Steele-Mortimer O, Dorward D, Prado MA, Caughey B. Uptake and neuritic transport of scrapie prion protein coincident with infection of neuronal cells. J Neurosci 2005; 25: 5207-5216.

62. Mahal SP, Baker CA, Demczyk CA, Smith EW, Julius C, Weissmann C. Prion strain discrimination in cell culture: the cell panel assay. Proc Natl Acad Sci U S A 2007; 104: 20908-20913.

63. Mahal SP, Demczyk CA, Smith EW, Klohn PC, Weissmann C. Assaying Prions in Cell Culture: The Standard Scrapie Cell Assay (SSCA) and the Scrapie Cell Assay in End Point Format (SCEPA). In: Hill AF (ed.). Methods in Molecular Biology: Prions. Humana Press: Totowa, NJ, 2008.

64. Manuelidis L, Yu ZX, Barquero N, Mullins B. Cells infected with scrapie and Creutzfeldt-Jakob disease agents produce intracellular 25-nm virus-like particles. Proc Natl Acad Sci U S A 2007; 104: 1965-1970.

65. Mead S. Prion disease genetics. Eur J Hum Genet 2006; 4: 273-281.

66. Mead S, Poulter M, Uphill J, Beck J, Whitfield J, Webb TE, Campbell T, Adamson G, Deriziotis P, Tabrizi SJ, Hummerich H, Verzilli C, Alpers MP, Whittaker JC, Collinge J. Genetic risk factors for variant Creutzfeldt-Jakob disease: a genome-wide association study. Lancet Neurol 2009; 8: 57-66.

67. Moore RC, Hope J, McBride PA, McConnell I, Selfridge J, Melton DW, Manson JC. Mice with gene targetted prion protein alterations show that Prnp, Sinc and Prni are congruent. Nat Genet 1998; 8: 118-125.

68. Moreno CR, Lantier F, Lantier I, Sarradin P, Elsen JM. Detection of new quantitative trait Loci for susceptibility to transmissible spongiform encephalopathies in mice. Genetics 2003; 165: 2085-2091.

69. Nandi PK, Sizaret PY. Murine recombinant prion protein induces ordered aggregation of linear nucleic acids to condensed globular structures. Arch Virol 2001; 46: 327-345.

70. Nazor KE, Kuhn F, Seward T, Green M, Zwald D, Purro M, Schmid J, Biffiger K, Power AM, Oesch B, Raeber AJ, Telling GC. Immunodetection of disease-associated mutant PrP, which accelerates disease in GSS transgenic mice. EMBO J 2005; 24: 2472-2480.

71. Nishida N, Harris DA, Vilette D, Laude H, Frobert Y, Grassi J, Casanova D, Milhavet O, Lehmann S. Successful transmission of three mouse-adapted scrapie strains to murine neuroblastoma cell lines overexpressing wild-type mouse prion protein. J Virol 2000; 74: 320-325.

72. Orgel LE. Prion replication and secondary nucleation. Chem Biol 1996; 3: 413-414.

73. Pastrana MA, Sajnani G, Onisko B, Castilla J, Morales R, Soto C, Requena JR. Isolation and Characterization of a Proteinase
K-Sensitive PrP(Sc) Fraction. Biochemistry 2006; 45: 15710-15717.

74. Peretz D, Scott MR, Groth D, Williamson RA, Burton DR, Cohen FE, Prusiner SB. Strain-specified relative conformational stability of the scrapie prion protein. Protein Sci 2001; 10: 854-863.

75. Piro JR, Harris BT, Nishina K, Soto C, Morales R, Rees JR, Supattapone S. Prion Protein Glycosylation is Not Required for Strain-Specific Neurotropism. J Virol 2009; 18: 18.

76. Prusiner SB. Molecular biology of prion diseases. Science 1991; 252: 1515-1522.

77. Prusiner SB, Groth D, Serban A, Stahl N, Gabizon R. Attempts to restore scrapie prion infectivity after exposure to protein denaturants. Proc Natl Acad Sci U S A 1993; 90: 2793-2797.

78. Prusiner SB, Scott M, Foster D, Pan KM, Groth D, Mirenda C, Torchia M, Yang SL, Serban D, Carlson GA, Hoppe PC, Westaway D, DeArmond SJ. Transgenetic studies implicate interactions between homologous PrP isoforms in scrapie prion replication. Cell 1990; 63: 673-686.

79. Race RE, Caughey B, Graham K, Ernst D, Chesebro B. Analyses of frequency of infection, specific infectivity, and prion protein biosynthesis in scrapie-infected neuroblastoma cell clones. J Virol 1988; 62: 2845-2849.

80. Race RE, Fadness LH, Chesebro B. Characterization of scrapie infection in mouse neuroblastoma cells. J Gen Virol 1987; 68: 1391-1399.

81. Riek R, Wider G, Billeter M, Hornemann S, Glockshuber R, Wuthrich K. Prion protein NMR structure and familial human spongiform encephalopathies. Proc Natl Acad Sci U S A 1998; 95: 11667-11672.

82. Riesner D, Kellings K, Wiese U, Wulfert M, Mirenda C, Prusiner SB. Prions and nucleic acids: search for “residual” nucleic acids and screening for mutations in the PrP-gene. Dev Biol Stand 1993; 80: 173-181.

83. Rudd PM, Endo T, Colominas C, Groth D, Wheeler SF, Harvey DJ, Wormald MR, Serban H, Prusiner SB, Kobata A, Dwek RA. Glycosylation differences between the normal and pathogenic prion protein isoforms. Proc Natl Acad Sci U S A 1999; 96: 13044-13049.

84. Rudd PM, Wormald MR, Wing DR, Prusiner SB, Dwek RA. Prion glycoprotein: structure, dynamics, and roles for the sugars. Biochemistry 2001; 40: 3759-3766.

85. Safar JG, Kellings K, Serban A, Groth D, Cleaver JE, Prusiner SB, Riesner D Search for a prion-specific nucleic acid. J Virol 2005; 79: 10796-10806.

86. Safar JG, Scott M, Monaghan J, Deering C, Didorenko S, Vergara J, Ball H, Legname G, Leclerc E, Solforosi L, Serban H, Groth D, Burton DR, Prusiner SB, Williamson RA. Measuring prions causing bovine spongiform encephalopathy or chronic wasting disease by immunoassays and transgenic mice. Nat Biotechnol 2002; 20: 1147-1150.

87. Safar JG, Wille H, Itri V, Groth D, Serban H, Torchia M, Cohen FE, Prusiner SB. Eight prion strains have PrP(Sc) molecules with different conformations. Nat Med 1998; 4: 1157-1165.

88. Scott M, Foster D, Mirenda C, Serban D, Coufal F, Walchli M, Torchia M, Groth D, Carlson G, DeArmond SJ, Westaway D, Prusiner SB. Transgenic mice expressing hamster prion protein produce species- specific scrapie infectivity and amyloid plaques. Cell 1989; 59: 847-857.

89. Sigurdson CJ, Nilsson KP, Hornemann S, Heikenwalder M, Manco G, Schwarz P, Ott D, Rulicke T, Liberski PP, Julius C, Falsig J, Stitz L, Wuthrich K, Aguzzi A. De novo generation of a transmissible spongiform encephalopathy by mouse transgenesis. Proc Natl Acad Sci U S A 2009; 106: 304-309.

90. Silva JL, Lima LM, Foguel D, Cordeiro Y. Intriguing nucleic-acid-binding features of mammalian prion protein. Trends Biochem Sci 2008; 33: 132-140.

91. Silveira JR, Raymond GJ, Hughson AG, Race RE, Sim VL, Hayes SF, Caughey B. The most infectious prion protein particles. Nature 2005; 437: 257-261.

92. Tanaka M, Collins SR, Toyama BH, Weissman JS. The physical basis of how prion conformations determine strain phenotypes. Nature 2006; 442: 585-589.

93. Telling GC, Parchi P, DeArmond SJ, Cortelli P, Montagna P, Gabizon R, Mastrianni J, Lugaresi E, Gambetti P, Prusiner SB. Evidence for the conformation of the pathologic isoform of the prion protein enciphering and propagating prion diversity. Science 1996; 274: 2079-2082.

94. Telling GC, Scott M, Mastrianni J, Gabizon R, Torchia M, Cohen FE, DeArmond SJ, Prusiner SB. Prion propagation in mice expressing human and chimeric PrP transgenes implicates the interaction of cellular PrP with another protein. Cell 1995; 83: 79-90.

95. Tremblay P, Ball HL, Kaneko K, Groth D, Hegde RS, Cohen FE, DeArmond SJ, Prusiner SB, Safar JG. Mutant PrPSc conformers induced by a synthetic peptide and several prion strains. J Virol 2004; 78: 2088-2099.

96. Uptain SM, Lindquist S. Prions as protein-based genetic elements. Annu Rev Microbiol 2002; 56: 703-741.

97. Vanik DL, Surewicz KA, Surewicz WK. Molecular basis of barriers for interspecies transmissibility of mammalian prions. Mol Cell 2004; 14: 139-145.

98. Vella LJ, Sharples RA, Nisbet RM, Cappai R, Hill AF. The role of exosomes in the processing of proteins associated with neurodegenerative diseases. Eur Biophys J 2008; 37: 323-332.

99. Vorberg I, Raines A, Priola SA. Acute formation of protease-resistant prion protein does not always lead to persistent scrapie infection in vitro. J Biol Chem 2004; 279: 29218-29225.

100. Vorberg I, Raines A, Story B, Priola SA. Susceptibility of common fibroblast cell lines to transmissible spongiform encephalopathy agents. J Infect Dis 2004; 189: 431-439.

101. Weissmann C. A “unified theory” of prion propagation. Nature 1991; 352: 679-683.

102. Weissmann C. The state of the prion. Nat Rev Microbiol 2004; 2: 861-871.

103. Wickner RB, Taylor KL, Edskes HK, Maddelein ML, Moriyama H, Roberts BT. Yeast prions act as genes composed of self-propagating protein amyloids. Adv Protein Chem 2004; 57: 313-334.

104. Zhuravlev PI, Materese CK, Papoian GA. Deconstructing the Native State: Energy Landscapes, Function, and Dynamics of Globular Proteins. J Phys Chem B 2009; 19: 19.

105. Zulianello L, Kaneko K, Scott M, Erpel S, Han D, Cohen FE, Prusiner SB Dominant-negative inhibition of prion formation diminished by deletion mutagenesis of the prion protein. J Virol 2000; 74: 4351-4360.
Copyright: © 2009 Mossakowski Medical Research Centre Polish Academy of Sciences and the Polish Association of Neuropathologists. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.