Open access peer-reviewed chapter

Past, Present and Potential Future Prion Disease Treatment Strategies

Written By

Pamela J. Skinner and Davis M. Seelig

Submitted: 17 May 2016 Reviewed: 07 December 2016 Published: 08 March 2017

DOI: 10.5772/67172

From the Edited Volume

Prion - An Overview

Edited by Yusuf Tutar

Chapter metrics overview

2,040 Chapter Downloads

View Full Metrics

Abstract

The prion diseases are rare and invariably fatal neurodegenerative diseases characterized by a unique, protein‐only pathogenesis. Mechanistically, the prion diseases result from the coerced conversion of a protease‐sensitive form of the cellular prion protein (PrPC) into a protease‐resistant infectious form (PrPres). This chapter reviews the past, present, and potentially future prion disease treatment strategies. This chapter begins with an introduction to prion diseases, the misfolding of prion proteins and what is known about this process, and then proceeds to discuss approaches for treatments. Regarding approaches to treat prion diseases, we discuss (1) small molecule inhibitors, (2) antiprion protein antibodies, (3) prion gene disruption, (4) targeting of the unfolded protein response, and (5) heterologous prion proteins. We elaborate on using heterologous prion proteins to treat prion diseases, as this is an area that we are pursuing. The chapter ends with thoughts on the future direction of prion disease treatment strategies and how these strategies might be applicable to other neurodegenerative diseases involving protein misfolding. The increasing awareness of the role of protein misfolding in many neurodegenerative processes makes the development of an effective treatment strategy for prion diseases a high priority.

Keywords

  • prion
  • treatment
  • CJD
  • GSS
  • PrPres
  • PrPC
  • heterologous prion proteins
  • protein misfolding diseases
  • neurodegenerative disorders

1. Introduction

Prion diseases, or transmissible spongiform encephalopathies (TSEs), are unique, fatal neurodegenerative diseases with infectious, genetic, or sporadic causes. Prion diseases affect humans (e.g. Creutzfeldt‐Jakob disease [CJD], Gerstmann‐Straussler‐Scheinker syndrome [GSS], and fatal familial insomnia [FFI]) and nonhumans (e.g. bovine spongiform encephalopathy [BSE] of cattle, chronic wasting disease [CWD] of cervids, and scrapie of sheep and goats. Irrespective of affected species, prion diseases result in progressive neurocognitive decline following a long incubation period. No effective prion disease treatments exist and most human patients die within 14 months following diagnosis [1]. Notably, many of the fundamental characteristics of prion diseases, including the molecular and biochemical mechanisms underlying the formation, accumulation, and cell‐to‐cell infectivity of misfolded protein and the role of glial‐mediated neuroinflammation aligns prion diseases with more common human neurodegenerative conditions, including Alzheimer's, Parkinson's, and Huntington's diseases.

In the mouse, the prion protein is encoded by the Prnp gene. The nascent 254 amino acid long peptide is then posttranslationally cleaved at its N and C terminus to produce the final 210 amino acid long protein [24]. Structurally, the prion protein is characterized by a disordered aminoterminal tail and a globular C‐terminal domain consisting of three α‐helices and two antiparallel β‐sheets [5, 6]. It is anchored to the outer cell surface membrane via a glycosylphosphatidylinositol (GPI) anchor, which helps tether the protein to the outer cell surface membrane [7].

The hallmark event in the prion disorders is the misfolding of the normal cellular prion protein (denoted PrPC) into a misfolded isoform (commonly denoted as PrPres or PrPSc). In its normal form, PrPC is a monomeric or dimeric protein with abundant alpha helical content, whereas the misfolded variant PrPres is aggregated with a β‐pleated sheet rich conformation [8, 9]. In addition to its structural differences, PrPres is characterized by resistance to protease and chemical disinfection [10]. Although the entirety of the process has not been described, it is widely believed that PrPres replication results from the induced misfolding of PrPC through a nucleation‐dependent polymerization mechanism [11]. This process is included in the model presented in Figure 1.

Figure 1.

A proposed model of heterologous prion protein treatment in the misfolding, nucleation, and formation of amyloid. In the presence of misfolded PrPres, the normal cellular prion protein (PrPc) is induced to misfold. This cycle of misfolding repeats leading to seeds of misfolded oligomers and amyloid deposits. Our studies demonstrate that heterologous prion proteins inhibit this process. We propose that heterologous prion proteins bind directly to PrPC and PrPres to block seed and amyloid formation.

In the pathogenesis of prion diseases, the formation of PrPres is generally believed to be a key event in the disease initiation. Owing to its specificity as a marker of tissue infectivity, PrPres is the most commonly used prion disease diagnostic marker. Although the inciting cause (i.e. PrPres formation) and neuropathologic consequences of prion disease (i.e. gliosis, synaptic dysfunction, spongiosis, and neuronal loss) are well characterized, the mechanism(s) by which the former results in the latter remain unknown. However, it is likely that misfolded PrPC has direct/indirect toxic properties as, as PrPC does not appear to be detrimental [12].

The conversion of PrPC to PrPres is a highly specific process of templated conversion requiring direct interaction between the normal and abnormal forms of the protein [13]. The efficiency of this conversion is predicated upon a number of specific reaction conditions, including the secondary structure of PrPres, homology of the primary and secondary structures between PrPC and PrPres, and the architecture of the PrPC‐PrPres complex [14, 15]. Increased contact between PrPC‐PrPres at residue 129 and the relative rigidity of the β2‐α2 loops in PrPC are two important factors in mediating the efficiency of PrPres formation and TSE susceptibility [1517]. In addition to steric factors, the formation of PrPres is favored by destabilization of PrPC as a number of destabilizing pathogenic mutations in PrP are linked with increasing misfolding rates [1820].

The presence and primary structure of host PrPC are major determinants in conferring susceptibility to prion disease infection. This is most expressly evident by work demonstrating that transgenic mice lacking PrPC are conferred resistance to prion infection [21]. Beyond simple PrPC expression, the degree of sequence homology between infecting prion and host PrPC plays a significant role in determining the efficiency of prion infection and prion replication [22]. Moreover, differences in primary sequence between host PrPC and infectious PrPres have been proposed to underlie the species barrier that mitigates cross species prion infection as well as prion strains [2325]. The importance of prion structure extends beyond simple amino acid homology and is also dependent upon secondary structural variations, including differences in loop/turn structures [23, 26]. In light of complementary in vitro and in vivo work, it appears as though the middle third region of the prion protein is particularly important for the autocatalytic conversion of PrPC to PrPres [27]. The potential clinical relevance of PrPC sequence is demonstrated by work revealing that polymorphisms in this area of the protein can confer prion disease resistance, as mice expressing a variant PrP containing amino acid substitutions in the β2‐α2 loop were resistant to prion infection [28].

Given studies that have revealed the pathogenic importance of a precisely formed PrPC‐PrPres complex, it seems reasonable to investigate whether interference with this complex might have therapeutic potential. This approach is best described by Singh and Udgaonkar in their comprehensive review on PrP misfolding, namely to test whether or not “…any ligand, whether small or large, that binds to the native conformation of the [PrPC] protein would stabilize that state and can therefore be expected to decrease the native‐state dynamics that drive misfolding [29].” Support for such an approach has been validated by antibody‐based studies, which have stabilized the α1 region of PrPC and prevented prion disease in animals [30, 31].

In this chapter, we review past, present, and potential future strategies to treat prion diseases.

Advertisement

2. Small molecule inhibitors to treat prion diseases

There are a number of small molecules that have proposed as prion therapeutics, including that either inhibit the misfolding of PrPC or promote the clearance of PrPres. In general, small molecule compounds can be segregated according to their method of action into compounds that either inhibit the misfolding of PrPC (potentially through stabilization) promote the clearance of PrPres. Over the past two decades, many small molecules have been evaluated for their in vitro or in vivo antiprion efficacy.

Of the most commonly examined small molecule candidate therapies, many have not stood up to scrutiny when their in vitro efficacy was tested in vivo. This includes quinacrine, pentosan poylsufate, Congo red, amphotericin B, anthracyclines, and memantine [3239]. Moreover, a subset of these compounds has been shown to extend the lives of prion‐infected animals [4042]. However, as noted by Caughey et al., the “clinical applicability of these compounds is severely limited by a lack of activity when administered after the onset of clinical signs of disease, poor bioavailability to the brain, and/or high toxicity [4246].” Despite the incremental progress in the field, efforts to more efficiently identify and screen test compounds for antiprion activity are ongoing. Early work by Pruisner et al. searched the Available Chemicals Directory for molecules that inhibit prion replication based upon prior studies, identified a number of a family of compounds (pyridine dicarbonitriles) that showed in vitro efficacy in inhibiting prion replication [47, 48]. Follow‐up studies by Reddy et al., who, through the design, synthesis, and screening of a series of related compounds, identified an additional compound that demonstrated efficacy at mitigating PrPres formation [49]. Most recently, Ferriera et al. describe the in silico and in vitro identification and screening of new small organic antiscrapie compounds that decreased PrPres accumulation and inhibit PrP aggregation [46]. Mechanistically, one of the most intriguing families of antiprion compounds is chemical chaperones. Chaperones are cellular constituents that interact with, stabilize, and assisting in the proper folding of nonfolded proteins [50]. When used pharmacologically, chaperones are small compounds that bind to proteins and either induce their refolding or stabilize their structure. Specific chaperones demonstrating in vitro and/or in vivo efficacy in prion disease systems including (along with their mechanism of action): trimethylamine N‐oxide, glycerol, dimethyl sulfoxide (protein stabilization by altering solvent properties), and bile salts [51, 52].

Advertisement

3. Antiprion antibodies to treat prion diseases

Other treatment strategies for prion diseases have been attempted including vaccination and immunotherapy, but these strategies have had limited success [53]. Nonetheless, there have been several promising studies gaining insights into this approach and its potential. To reduce redundancies, we refer the interested reader to the chapter entitled “Immunobiology of Prion Diseases” for more information on this topic.

Advertisement

4. Prion gene disruption to treat prion diseases

Since the cellular prion protein is not essential for life but required for prion disease [54, 55], several groups have worked to develop and test strategies that disrupt normal cellular prion proteins, PrPC. With this in mind, a recent treatment strategy used lentivirus vectors that expressed silencing RNAs directed against the cellular form of the prion protein [56]. These lentiviral vectors were employed to transduce mouse embryonic stem cells and the resultant transduced embryonic stem cells used to create chimeric mice expressing various levels of the silencing RNAs. After infection of these mice with scrapie, mice that were highly chimeric for the transgene and that showed reduced PrPC expression in the brain showed increased survival times. Similarly, Mallucci et al. generated an adult‐onset PrP knockout mouse model with delayed, neuron‐specific deletion of PrPC, which mitigated the clinical and neuropathologic consequences of prion disease [57, 58]. In another study, the same group used RNAi‐driven gene silencing to reduce PrPC expression. Using lenti‐shRNA directed against PrPC, treated mice experienced a significant downregulation of PrPC expression and a delay in prion disease progression [59]. Thus, strategies that reduce or eliminate PrPC using inhibitory RNAs show promise as a treatment for prion diseases.

An alternative approach to reducing PrPC expression is to edit the gene using Zn‐finger nucleases (ZFN), transcription activator‐like effector nucleases (TALEN), or clustered regularly interspaced short palindromic repeats (CRISPR) gene editing systems. Indeed, mice, bovine and goat prion genes have been targeted using these approaches [6062] to produce disease resistant animals, and at least one patent has been filed for gene editing of prion genes in animals [63].

Advertisement

5. Target the unfolded protein response to treat prion diseases

The pivotal event in prion disease pathogenesis is the formation and accumulation of misfolded PrPres in the brain as it initiates a pathologic cascade of glial activation, neuronal hypometabolism, and apoptotic neuronal loss. An increasing body of work indicates that PrPres triggers this pathology, in part, through the activation of the unfolded protein response (UPR) [64]. The UPR is a two‐phase, cytoprotective cascade of the endoplasmic reticulum (ER) that is initiated by misfolded or aggregated protein, and it seeks to resolve cellular and ER stress. In the initial adaptive phase of the UPR, misfolded protein stimulates one (or more) of three sensing proteins: (1) PERK (protein kinase RNA‐like ER kinase), (2) IRE1α (inositol‐requiring protein 1), and/or (3) ATF6 (activating transcription factor‐6). Subsequent homodimerization of two of these proteins (PERK and IRE1α) results in the phosphorylation and activation of intermediate messengers, including eIF2α (eukaryotic initiation factor 2 alpha), ATF4 (activating transcription factor), and XBP1 (X‐box folding protein). The end result of the adaptive phase of the UPR is an attenuation of protein synthesis, an increased synthesis of ER chaperones, and a mitigation of ER protein processing [65, 66]. However, if these initial adaptive efforts fail, the UPR transitions to a second, apoptotic phase involving the activation of caspases 3, 6, 7, and 8.

Previous work has demonstrated involvement of both phases of the UPR in human and rodent prion disease [67, 68]. In addition to triggering apoptosis, it is increasing clear that the UPR is able to induce the deleterious, glial‐mediated inflammatory response that is characteristic of both prion and other neurodegenerative diseases [69]. Specifically, Moreno et al. have shown that prion replications results in unchecked eIF2α activation that contributes to synaptic failure, neuronal loss, and clinical deficits in prion‐infected mice [70]. However, the role of the UPR in human prion disease is less clear. Although Hetz et al. demonstrated increased levels of ER stress associated with misfolded protein in the brains of human patients with sporadic or variant CJD [71], subsequent immunohistochemical studies examining the brains of human patients with CJD for activated forms of PERK and eIF2α have failed confirm consistent involvement of the UPR [72].

Despite the inconclusive mechanistic data linking the UPR with prion disease pathogenesis, a small number of groups have examined the efficacy of therapeutic strategies directed at mitigating its activation. The therapeutic potential of targeting the UPR pathway is best demonstrated by work performed by Mallucci and Moreno. In their initial studies, they report that genetic mitigation of eIF2α activation decreases synaptic loss and neuronal loss in prion‐infected mice [70]. Moreover, in follow‐up work they demonstrate that upstream blockade of UPR activation through pharmacological inhibition of the activation of PERK reverses cognitive deficits and prevents clinical disease in prion‐infected mice [67]. A smaller body of work has revealed that pharmacologic inhibition of the UPR using the neuroprotective, antiapoptotic bile acids tauroursodeoxycholic acid (TUDCA) and ursodeoxycholic acid (UDCA), results in decreased levels of activated eIP2α in organotypic cerebellar slices as well as decreased neuroinflammation and prolonged survival in mice. The reported benefits of bile acids result, in part, from their ability to inhibit the UPR activation across all three sensing pathways as reflected by lower levels of phosphorylated eIF2α, ATF4, PERK, ATF6, and IRE1α [7376].

Advertisement

6. Heterologous prion proteins to treat prion diseases

The concept of heterologous prion proteins (HetPrP) as potential therapeutics is based on a body of research, including studies performed in cell free, cell culture, and animal models, in which prion proteins from different species were allowed to interact. Horiuchi et al. demonstrate that inclusion of a heterologous species PrPc in a cell‐free conversion system was capable of interfering with the formation of PrPres between two homologous species [24]. When they divide the process of PrPres formation into two steps, namely initial binding between PrPC and PrPres followed by acquisition of protease resistance, the interfering effect of HetPrP appears to occur during the latter [24]. Further, the expression of hamster prion protein (HaPrP) in scrapie‐infected mouse cells in vitro lead to near complete elimination of PrPres [77] supporting a role for heterologous HaPrP in either inhibiting PrPres production or enhancing its clearance. Moreover, the induced expression rabbit prion proteins in scrapie‐infected mouse cells led to substantially less PrPres as compared to mouse cells that do not express rabbit prion proteins, supporting a role for rabbit prion proteins interfering with mouse PrPres formation [23].

In our work, we extended these in vitro observations into the mouse using the rocky mountain laboratories (RML)-Chandler strain of scrapie and HetPrP therapy using bacterially expressed and purified recombinant HaPrP amino acids 23‐231 [78]. For this study, mice were intracerebrally inoculated with an RML‐Chandler strain brain homogenate combined with either recombinant HaPrP or vehicle control. The following day, mice were treated with HaPrP orally. We assessed the effect of HaPrP dosage using two treatment groups, including a high dose of recombinant protein (0.7 mg/ml, high dose) and a low dose (0.35 mg/ml). Lastly, two control groups were included, those being a mock treatment group comprised of mice that were infected and treated with vehicle only, and mice that were not infected and not treated. We assessed the impact of treatment on clinical disease by evaluating mice daily following infection, weekly during the first months and then daily in later months for signs of scrapie‐related symptoms including decreased motility, flattened stature, ataxic gait, hind limb paresis, dull eyes, weight loss, and kyphosis.

Treatment with the high dose HaPrP effectively and significantly delayed the onset of clinical symptoms, and prolonged survival compared to the vehicle‐treated animals [78]. Moreover, when the study was terminated at 452 days postinfection, half of the high‐dose‐treated animals were still free of scrapie symptoms. Figure 2 shows the survival times.

Figure 2.

Treatment with heterologous recombinant HaPrP prolonged survival. Kaplan‐Meier plots showing the survival times of mock‐treated (orange, n = 5), low‐dose‐treated (blue, n = 5), high‐dose‐treated mice (purple, n = 6) and uninfected (red, n = 10). We tested for differences between groups using a modified version of the Gehan‐Wilcoxon test and found a statistically significant difference between the mock infected group and the high‐dose group (p = 0.0348).

In addition to abrogating the clinical signs of prion disease, mice receiving the high‐dose of HaPrP, compared to mice treated with a low dose of HaPrP or with vehicle only, accumulated significantly less PrPres in both brain and spleen. Furthermore, HaPrP partially mitigated the neuropathologic consequences of prion infection as high‐dose‐treated animals showed a trend towards fewer activated astrocytes as revealed by immunohistochemistry for glial fibrillary acidic protein and less severe neuropil spongiosis in total brain and highly significant reductions in the thalamus.

Although we demonstrated that treatment with HetPrPC inhibits both the formation of PrPres and the clinical consequences of prion infection, the mechanism underlying this phenomenon is not known. We think that HetPrP binds to both PrPres and PrPC and blocks the production and elongation of PrPres chains and amyloid formation. This is modeled in Figure 1.

The work of Horiuchi et al. offers two possible mechanistic models for this interference, based upon number and type of binding sites for PrPC on PrPres [24]. They posit in a “one binding system,” that the binding of HetPrP to a growing PrPres oligomer creates an aggregate that is incapable of generating the steric interactions necessary for the continued production of PrPres. Alternately, they propose in a “two binding system” that the growing PrPres oligomer contains two binding sites, namely a conversion‐inducing site and a nonconverting site. In this two‐site system, HetPrP interferes with the formation of PrPres by binding and blockading conversion site without blocking the nonconverting site.

In addition to biochemical mechanisms described, it is possible that the protective effect of HetPrP in our study resulted from an evoked immune response that impacted PrPres formations. However, our data do not support this hypothesis. By western blot analysis of serum from study mice, we did not detect the presence of antihamster PrP antibodies in treated compared to control animals. Lastly, it is important to note that because mice were simultaneously intracerebrally inoculated with both scrapie prions and HaPrP, it is quite likely that the HaPrP in the inoculum served to inactivate the scrapie prion by binding to PrPres and forming an inactive complex due to sequence incongruence.

It is increasingly apparent that HetPrP treatment safely inhibits the PrPC to PrPres conversion process. In vitro and in vivo studies render feasible the prospect of treating human prion diseases with HetPrP. While demonstrating efficacy, in our study the treatment regime used (intracerebral instillation of HetPrP at the time of infection followed by oral ingestion of heterologous PrPC) which is not ideal for treating patients with existing prion disease. Delivery via intracerebral injection is certainly not anticipated to allow HetPrP to make contact with and inactivate all PrPres in the system. As such, future studies are needed to develop more practical HetPrP delivery modalities as well as to evaluate potentially more effective HetPrP sequences.

While a wide range of mammal species are susceptible to prion infection, the efficiency of interspecies transmission is varied and governed by a “species barrier,” the integrity of which is inversely proportional to the strength of the interaction between host PrPC and incoming PrPres. Interestingly, rabbits have been shown to be unusually resistant to prion disease inoculation, as attempts to transmit CJD, Kuru, sheep scrapie, TME, and mouse‐adapted scrapie to rabbits failed [79, 80]. While subsequent groups have confirmed that the rabbit is not absolutely resistant or prion infection, there is general agreement that they are only minimally susceptible [81, 82]. The degree of primary sequence homology is important in determining the robustness of the species barrier. The rabbit prion protein shows relatively low sequence homology to other species prion proteins. Based upon this work, we propose that a rabbit PrP‐based HetPrP treatment strategy may be more effective than HaPrP at inhibiting prion disease.

While we used IC injection of HetPrP in our study, the clinical evolution of this approach necessitates a more effective and simpler means of delivery. One such approach could be delivery via the bloodstream and use blood vessels to efficiently deliver HetPrP to all areas of the brain. In addition, it may be possible to use peptides derived from HetPrP rather than whole proteins. Indeed Chabry et al. showed in vitro inhibition of PrP conversation with synthetic peptides derived from mouse and hamster PrP [83, 84]. Another such possibility for HetPrP treatment is the adoption of a gene therapy‐based approach using lentiviral vectors. Thus, further studies are warranted to optimize both the form of HetPrP as well as its mode of delivery.

In related studies, other groups have found promising therapeutic results as well. Meier et al. engineered PrPC fused to immunoglobulin Fcgamma, termed PrP‐Fc(2) [85]. Wild‐type mice expressing PrP‐Fc(2) and subsequently infected with scrapie prions showed delayed PrPres accumulation and onset of disease [85]. In follow‐up studies, they further showed that expression of PrP‐Fc(2) transduced by a lentiviral vector at 170 days postinfection was able to reduce prion infectivity by 3–4 logs [86]. Toupet et al. created a recombinant lentiviral vector that transduces expression of a dominant negative mouse prion protein that recapitulates sheep PrPQ171R and human PrPE219K polymorphisms associated with prion disease resistance [87]. They showed that chronic injection of this vector directly into the brains of prion disease infected mice led to reduced astrocytic gliosis and extended survival [87]. Moreover, Soto et al. designed beta sheet breaker peptides corresponding to the conserved region of PrP 115‐122 that is thought to play a central role in conversion of PrPC to PrPres [8891]. These beta sheet peptides partly reversed PrPres to PrPc in vitro, and when mixed with scrapie prions and injected into mice, decreased infectivity by 90–95% [88]. Thus, multiple strategies have been developed and tested in mice that use prion proteins or related peptides to target and reduce prion infectivity and have demonstrated efficacy.

Advertisement

7. Potential future strategies to treat prion diseases

Understanding pathogenesis is key to developing new therapies for prion diseases. For example, we [92, 93] and others have gained insights into prion disease pathogenesis by studying in changes in gene expression that occur during the disease process. These expression alterations provide insights to underlying pathological processes, and key mediators of these processes might be targeted in future prion treatment strategies. Another example comes from Hetz et al., who determined in a scrapie infected cell culture system that PrPsc toxicity and apoptosis induction was associated in an increase in an endoplasmic reticulum resident enzyme caspase‐12, and a corresponding increase in caspase‐12 was also seen in humans affected by CJD [94]. With this knowledge of a key process in pathogenesis, they were able to inhibit apoptosis by overexpression of a catalytic mutant of caspase‐12 [94]. In another set of studies, the 37 kDa/67 kDa laminin receptor LPR/LR was targeted based on knowledge that LPR/LR is a cell surface receptor for PrPc [95] and required for PrPres propagation in scrapie‐infected cells [96]. Zuber et al. created and infused single‐chain Fv antibodies directed against LPR/LR into mice just prior to inoculation with scrapie prions and weekly afterwards, and found an ~40% reduction in PrPres in spleen [97]. In similar experiments, Pflanz et al. injected lentiviral vectors that transduce small interfering RNAs directed against LPR/LR precursor mRNA into the brains of mice, then infected them with scrapie, and found a 41% reduction in PrPres and prolongation of the preclinical phase [98]. Thus, gaining understanding of the molecular event underlying prion disease pathogenesis can identify potential targets for future prion disease therapeutics.

Moving forward to a viable treatment and cure for prion diseases in humans will likely involve a combination of therapies. For example, this might involve a combination of approaches such as gene editing to create disease resistant prion gene alleles, a drug that inhibits apoptosis, a small molecule that stabilizes PrPc and regular injections of heterologous prion proteins that bind and clear nascent PrPres.

Importantly, strategies that work for treating prion diseases may also be effective when applied to other neurodegenerative diseases that involve protein misfolding, such as Alzheimer's disease and Huntington's chorea. There is increasing evidence of underlying similarities in the pathogenesis of protein misfolding neurodegenerative diseases. Hence, similar cure strategies may be feasible.

Advertisement

8. Conclusion

In conclusion, an ever‐expanding understanding of basic prion pathogenesis, combined with the rapidly ever‐expanding development of new biotechnologies, combined with existing strategies to treat prion diseases, will likely to lead to a feasible and effective treatment for prion diseases in the near future. Already, innovations such as genome editing, inhibitory RNAs, and improved gene therapy vectors are being applied to and advancing treatment strategies to create improved treatments. In addition, strategies that show efficacy that target separate components of disease pathogenesis can be combined. Thus, in the coming years, the outlook is very promising for the development of an effective treatment and potential cure for individuals with prion diseases. Furthermore, strategies used to treat prion diseases might be broadly applicable and effective when applied to other protein misfolding diseases. The increasing awareness of the role of protein misfolding in many neurodegenerative processes makes the development of an effective treatment strategy for prion diseases a high priority.

Advertisement

Abbreviations and acronyms

TSEsTransmissible spongiform encephalopathies
CJDCreutzfeldt‐Jakob disease
GSSGerstmann‐Straussler‐Scheinker syndrome
FFIFatal familial insomnia
BSEBovine spongiform encephalopathy
CWDChronic wasting disease
Denoted PrPCCellular prion protein
Commonly denoted as PrPres or PrPScMisfolded isoform
HetPrPHeterologous prion proteins
HaPrPHamster prion protein

References

  1. 1. Valleron AJ, Boelle PY, Will R, Cesbron JY. Estimation of epidemic size and incubation time based on age characteristics of vCJD in the United Kingdom. Science. 2001;294(5547):1726–1728. doi: 10.1126/science.1066838.
  2. 2. Basler K, Oesch B, Scott M, et al. Scrapie and cellular PrP isoforms are encoded by the same chromosomal gene. Cell. 1986;46(386272089):417–428.
  3. 3. Mange A, Beranger F, Peoc’h K, Onodera T, Frobert Y, Lehmann S. Alpha‐ and beta‐ cleavages of the amino‐terminus of the cellular prion protein. Biol Cell. 2004;96(2):125–132. doi: 10.1016/j.biolcel.2003.11.007.
  4. 4. Westaway D, Prusiner SB. Conservation of the cellular gene encoding the scrapie prion protein. Nucleic Acids Res. 1986;14(586176712):2035–2044.
  5. 5. Riek R, Hornemann S, Wider G, Glockshuber R, Wuthrich K. NMR characterization of the full‐length recombinant murine prion protein, mPrP(23‐231). FEBS Lett. 1997;413(297424376):282–288.
  6. 6. Riek R, Hornemann S, Wider G, Billeter M, Glockshuber R, Wuthrich K. NMR structure of the mouse prion protein domain PrP(121‐321). Nature. 1996;382(658796317593):180–182.
  7. 7. Stahl N, Borchelt DR, Hsiao K, Prusiner SB. Scrapie prion protein contains a phosphatidylinositol glycolipid. Cell. 1987;51(288027007):229–240.
  8. 8. Pan KM, Baldwin M, Nguyen J, et al. Conversion of alpha‐helices into beta‐sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci U S A. 1993;90(2394068524):10962–10966.
  9. 9. Gasset M, Baldwin MA, Fletterick RJ, Prusiner SB. Perturbation of the secondary structure of the scrapie prion protein under conditions that alter infectivity. Proc Natl Acad Sci U S A. 1993;90(193126320):1–5.
  10. 10. Taylor DM. Inactivation of BSE agent. Dev Biol Stand. 1991;7592175414:97–102.
  11. 11. Jarrett JT, Lansbury PT, Jr. Seeding “one‐dimensional crystallization” of amyloid: a pathogenic mechanism in Alzheimer’s disease and scrapie? Cell. 1993;73(693292067):1055–1058.
  12. 12. Hetz C, Maundrell K, Soto C. Is loss of function of the prion protein the cause of prion disorders? Trends Mol Med. 2003;9(6):237–243. doi: S1471491403000698 [pii].
  13. 13. Caughey B, Chesebro B. Transmissible spongiform encephalopathies and prion protein interconversions. Adv Virus Res. 2001;56:277–311.
  14. 14. Prusiner SB, Scott M, Foster D, et al. Transgenetic studies implicate interactions between homologous PrP isoforms in scrapie prion replication. Cell. 1990;63(491029499):673–686.
  15. 15. Mallik S, Yang W, Norstrom EM, Mastrianni JA. Live cell fluorescence resonance energy transfer predicts an altered molecular association of heterologous PrPSc with PrPC. J Biol Chem. 2010;285(12):8967–8975. doi: 10.1074/jbc.M109.058107 [doi].
  16. 16. Bett C, Fernandez‐Borges N, Kurt TD, et al. Structure of the beta2‐alpha2 loop and interspecies prion transmission. FASEB J. 2012;26(7):2868–2876. doi: 10.1096/fj.11‐200923.
  17. 17. Giachin G, Biljan I, Ilc G, Plavec J, Legname G. Probing early misfolding events in prion protein mutants by NMR spectroscopy. Molecules. 2013;18(8):9451–9476. doi: 10.3390/molecules18089451.
  18. 18. Singh J, Udgaonkar JB. Structural effects of multiple pathogenic mutations suggest a model for the initiation of misfolding of the prion protein. Angew Chem Int Ed Engl. 2015;54(26):7529–7533. doi: 10.1002/anie.201501011.
  19. 19. Liemann S, Glockshuber R. Influence of amino acid substitutions related to inherited human prion diseases on the thermodynamic stability of the cellular prion protein. Biochemistry (N Y). 1999;38(1199178830):3258–3267.
  20. 20. Apetri AC, Surewicz K, Surewicz WK. The effect of disease‐associated mutations on the folding pathway of human prion protein. J Biol Chem. 2004;279(17):18008–18014. doi: 10.1074/jbc.M313581200.
  21. 21. Bueler H, Aguzzi A, Sailer A, et al. Mice devoid of PrP are resistant to scrapie. Cell. 1993;73(793313963):1339–1347.
  22. 22. Rigter A, Bossers A. Sheep scrapie susceptibility‐linked polymorphisms do not modulate the initial binding of cellular to disease‐associated prion protein prior to conversion. J Gen Virol. 2005;86(Pt 9):2627–2634. doi: 86/9/2627.
  23. 23. Vorberg I, Groschup MH, Pfaff E, Priola SA. Multiple amino acid residues within the rabbit prion protein inhibit formation of its abnormal isoform. J Virol. 2003;77(3):2003–2009.
  24. 24. Horiuchi M, Priola SA, Chabry J, Caughey B. Interactions between heterologous forms of prion protein: Binding, inhibition of conversion, and species barriers. Proc Natl Acad Sci U S A. 2000;97(1120283912):5836–5841.
  25. 25. Priola SA, Vorberg I. Molecular aspects of disease pathogenesis in the transmissible spongiform encephalopathies. Mol Biotechnol. 2006;33(1):71–88.
  26. 26. Moore RA, Taubner LM, Priola SA. Prion protein misfolding and disease. Curr Opin Struct Biol. 2009;19(1):14–22. doi: 10.1016/j.sbi.2008.12.007.
  27. 27. Priola SA. Prion protein and species barriers in the transmissible spongiform encephalopathies. Biomed Pharmacother. 1999;53(1):27–33. doi: S0753332299800572.
  28. 28. Kurt TD, Jiang L, Bett C, Eisenberg D, Sigurdson CJ. A proposed mechanism for the promotion of prion conversion involving a strictly conserved tyrosine residue in the beta2‐alpha2 loop of PrPC. J Biol Chem. 2014;289(15):10660–10667. doi: 10.1074/jbc.M114.549030.
  29. 29. Singh J, Udgaonkar JB. Molecular mechanism of the misfolding and oligomerization of the prion protein: Current understanding and its implications. Biochemistry. 2015;54(29):4431–4442. doi: 10.1021/acs.biochem.5b00605.
  30. 30. Heppner FL, Musahl C, Arrighi I, et al. Prevention of scrapie pathogenesis by transgenic expression of anti‐prion protein antibodies. Science. 2001;294(5540):178–182.
  31. 31. White AR, Enever P, Tayebi M, et al. Monoclonal antibodies inhibit prion replication and delay the development of prion disease. Nature. 2003;422(6927):80–83. doi: 10.1038/nature01457.
  32. 32. Haik S, Brandel JP, Salomon D, et al. Compassionate use of quinacrine in Creutzfeldt‐Jakob disease fails to show significant effects. Neurology. 2004;63(12):2413–2415. doi: 63/12/2413.
  33. 33. Whittle IR, Knight RS, Will RG. Unsuccessful intraventricular pentosan polysulphate treatment of variant creutzfeldt‐jakob disease. Acta Neurochir (Wien). 2006;148(6):677–9; discussion 679. doi: 10.1007/s00701‐006‐0772‐y.
  34. 34. Farquhar C, Dickinson A, Bruce M. Prophylactic potential of pentosan polysulphate in transmissible spongiform encephalopathies. Lancet. 1999;353(9147):117. doi: S0140‐6736(98)05395‐1.
  35. 35. Caughey B, Ernst D, Race RE. Congo red inhibition of scrapie agent replication. J Virol. 1993;67(1093381832):6270–6272.
  36. 36. Demaimay R, Adjou KT, Beringue V, et al. Late treatment with polyene antibiotics can prolong the survival time of scrapie‐infected animals. J Virol. 1997;71(1298037685):9685–9689.
  37. 37. Tagliavini F, McArthur RA, Canciani B, et al. Effectiveness of anthracycline against experimental prion disease in syrian hamsters. Science. 1997;276(531597293259):1119–1122.
  38. 38. Muller WE, Ushijima H, Schroder HC, et al. Cytoprotective effect of NMDA receptor antagonists on prion protein (PrionSc)‐induced toxicity in rat cortical cell cultures. Eur J Pharmacol. 1993;246(394039507):261–267.
  39. 39. Muller WE, Laplanche JL, Ushijima H, Schroder HC. Novel approaches in diagnosis and therapy of Creutzfeldt‐Jakob disease. Mech Ageing Dev. 2000;116(2‐3):193–218. doi: S0047637400001123.
  40. 40. Cashman NR, Caughey B. Prion diseases—close to effective therapy? Nat Rev Drug Discov. 2004;3(10):874–884.
  41. 41. Trevitt CR, Collinge J. A systematic review of prion therapeutics in experimental models. Brain. 2006;129(Pt 9):2241–2265. doi: awl150.
  42. 42. Sim VL. Prion disease: Chemotherapeutic strategies. Infect Disord Drug Targets. 2012;12(2):144–160. doi: IDDT‐EPUP‐20120314‐001.
  43. 43. Collins SJ, Lewis V, Brazier M, Hill AF, Fletcher A, Masters CL. Quinacrine does not prolong survival in a murine Creutzfeldt‐Jakob disease model. Ann Neurol. 2002;52(4):503–506. doi: 10.1002/ana.10336.
  44. 44. Caughey B, Caughey WS, Kocisko DA, Lee KS, Silveira JR, Morrey JD. Prions and transmissible spongiform encephalopathy (TSE) chemotherapeutics: a common mechanism for anti‐TSE compounds? Acc Chem Res. 2006;39(9):646–653. doi: 10.1021/ar050068p.
  45. 45. Kocisko DA, Caughey B. Mefloquine, an antimalaria drug with antiprion activity in vitro, lacks activity in vivo. J Virol. 2006;80(2):1044–1046. doi: 80/2/1044.
  46. 46. Ferreira NC, Marques IA, Conceicao WA, et al. Anti‐prion activity of a panel of aromatic chemical compounds: In vitro and in silico approaches. PLoS One. 2014;9(1):e84531. doi: 10.1371/journal.pone.0084531.
  47. 47. Perrier V, Wallace AC, Kaneko K, Safar J, Prusiner SB, Cohen FE. Mimicking dominant negative inhibition of prion replication through structure‐based drug design. Proc Natl Acad Sci U S A. 2000;97(1120283952):6073–6078.
  48. 48. Kaneko K, Wille H, Mehlhorn I, et al. Molecular properties of complexes formed between the prion protein and synthetic peptides. J Mol Biol. 1997;270(4):574–586. doi: S0022–2836(97)91135‐9.
  49. 49. Reddy TR, Mutter R, Heal W, et al. Library design, synthesis, and screening: pyridine dicarbonitriles as potential prion disease therapeutics. J Med Chem. 2006;49(2):607–615. doi: 10.1021/jm050610f.
  50. 50. Hartl FU, Hayer‐Hartl M. Molecular chaperones in the cytosol: from nascent chain to folded protein. Science. 2002;295(5561):1852–1858. doi: 10.1126/science.1068408.
  51. 51. Tatzelt J, Prusiner SB, Welch WJ. Chemical chaperones interfere with the formation of scrapie prion protein. EMBO J. 1996;15(2397133266):6363–6373.
  52. 52. Cortez LM, Campeau J, Norman G, et al. Bile acids reduce prion conversion, reduce neuronal loss, and prolong male survival in models of prion disease. J Virol. 2015;89(15):7660–7672. doi: JVI.01165‐15.
  53. 53. Li L, Napper S, Cashman NR. Immunotherapy for prion diseases: Opportunities and obstacles. Immunotherapy. 2010;2(2):269–282. doi: 10.2217/imt.10.3.
  54. 54. Brandner S, Isenmann S, Raeber A, et al. Normal host prion protein necessary for scrapie‐induced neurotoxicity. Nature. 1996;379(656396149246):339–343.
  55. 55. Brandner S, Raeber A, Sailer A, et al. Normal host prion protein (PrPC) is required for scrapie spread within the central nervous system. Proc Natl Acad Sci U S A. 1996;93(2397075133):13148–13151.
  56. 56. Pfeifer A, Eigenbrod S, Al‐Khadra S, et al. Lentivector‐mediated RNAi efficiently suppresses prion protein and prolongs survival of scrapie‐infected mice. J Clin Invest. 2006;116(12):3204–3210. doi: 10.1172/JCI29236.
  57. 57. Mallucci GR, Ratte S, Asante EA, et al. Post‐natal knockout of prion protein alters hippocampal CA1 properties, but does not result in neurodegeneration. EMBO J. 2002;21(3):202–210. doi: 10.1093/emboj/21.3.202.
  58. 58. Mallucci G, Dickinson A, Linehan J, Klohn PC, Brandner S, Collinge J. Depleting neuronal PrP in prion infection prevents disease and reverses spongiosis. Science. 2003;302(5646):871–874. doi: 10.1126/science.1090187.
  59. 59. White MD, Farmer M, Mirabile I, Brandner S, Collinge J, Mallucci GR. Single treatment with RNAi against prion protein rescues early neuronal dysfunction and prolongs survival in mice with prion disease. Proc Natl Acad Sci U S A. 2008;105(29):10238–10243. doi: 10.1073/pnas.0802759105.
  60. 60. Bevacqua RJ, Fernandez‐Martin R, Savy V, et al. Efficient edition of the bovine PRNP prion gene in somatic cells and IVF embryos using the CRISPR/Cas9 system. Theriogenology. 2016;86(8):1886–1896.e1. doi: 10.1016/j.theriogenology.2016.06.010.
  61. 61. Kaczmarczyk L, Mende Y, Zevnik B, Jackson WS. Manipulating the prion protein gene sequence and expression levels with CRISPR/Cas9. PLoS One. 2016;11(4):e0154604. doi: 10.1371/journal.pone.0154604.
  62. 62. Ni W, Qiao J, Hu S, et al. Efficient gene knockout in goats using CRISPR/Cas9 system. PLoS One. 2014;9(9):e106718. doi: 10.1371/journal.pone.0106718.
  63. 63. Weinstein E, Simmons P, Cui X, inventors. Genomic editing of prion disorder‐related genes in animals. Patent US20110023147 A1. 2011.
  64. 64. Ferreiro E, Costa R, Marques S, Cardoso SM, Oliveira CR, Pereira CM. Involvement of mitochondria in endoplasmic reticulum stress‐induced apoptotic cell death pathway triggered by the prion peptide PrP(106–126). J Neurochem. 2008;104(3):766–776. doi: JNC5048.
  65. 65. Maly DJ, Papa FR. Druggable sensors of the unfolded protein response. Nat Chem Biol. 2014;10(11):892–901. doi: 10.1038/nchembio.1664.
  66. 66. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519–529. doi: nrm2199.
  67. 67. Moreno JA, Halliday M, Molloy C, et al. Oral treatment targeting the unfolded protein response prevents neurodegeneration and clinical disease in prion‐infected mice. Sci Transl Med. 2013;5(206):206ra138. doi: 10.1126/scitranslmed.3006767; 10.1126/scitranslmed.3006767.
  68. 68. Hetz C, Maundrell K, Soto C. Is loss of function of the prion protein the cause of prion disorders? Trends Mol Med. 2003;9(6):237–243. doi: S1471491403000698.
  69. 69. Salminen A, Kauppinen A, Suuronen T, Kaarniranta K, Ojala J. ER stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology. J Neuroinflammation. 2009;6:41‐2094‐6‐41. doi: 10.1186/1742‐2094‐6‐41.
  70. 70. Moreno JA, Radford H, Peretti D, et al. Sustained translational repression by eIF2alpha‐P mediates prion neurodegeneration. Nature. 2012;485(7399):507–511. doi: 10.1038/nature11058.
  71. 71. Hetz CA, Soto C. Stressing out the ER: a role of the unfolded protein response in prion‐related disorders. Curr Mol Med. 2006;6(1):37–43.
  72. 72. Unterberger U, Hoftberger R, Gelpi E, Flicker H, Budka H, Voigtlander T. Endoplasmic reticulum stress features are prominent in Alzheimer disease but not in prion diseases in vivo. J Neuropathol Exp Neurol. 2006;65(4):348–357. doi: 10.1097/01.jnen.0000218445.30535.6f.
  73. 73. Lo AC, Callaerts‐Vegh Z, Nunes AF, Rodrigues CM, D’Hooge R. Tauroursodeoxycholic acid (TUDCA) supplementation prevents cognitive impairment and amyloid deposition in APP/PS1 mice. Neurobiol Dis. 2013;50:21–29. doi: 10.1016/j.nbd.2012.09.003.
  74. 74. Castro‐Caldas M, Carvalho AN, Rodrigues E, et al. Tauroursodeoxycholic acid prevents MPTP‐induced dopaminergic cell death in a mouse model of Parkinson’s disease. Mol Neurobiol. 2012;46(2):475–486. doi: 10.1007/s12035‐012‐8295‐4.
  75. 75. Keene CD, Rodrigues CM, Eich T, Chhabra MS, Steer CJ, Low WC. Tauroursodeoxycholic acid, a bile acid, is neuroprotective in a transgenic animal model of Huntington’s disease. Proc Natl Acad Sci U S A. 2002;99(16):10671–10676. doi: 10.1073/pnas.162362299.
  76. 76. Dromparis P, Paulin R, Stenson TH, Haromy A, Sutendra G, Michelakis ED. Attenuating endoplasmic reticulum stress as a novel therapeutic strategy in pulmonary hypertension. Circulation. 2013;127(1):115–125. doi: 10.1161/CIRCULATIONAHA.112.133413.
  77. 77. Priola SA, Caughey B, Race RE, Chesebro B. Heterologous PrP molecules interfere with accumulation of protease‐resistant PrP in scrapie‐infected murine neuroblastoma cells. J Virol. 1994;68(8):4873–4878.
  78. 78. Skinner PJ, Kim HO, Bryant D, et al. Treatment of prion disease with heterologous prion proteins. PLoS One. 2015;10(7):e0131993. doi: 10.1371/journal.pone.0131993.
  79. 79. Gibbs CJ,Jr., Gajdusek DC. Experimental subacute spongiform virus encephalopathies in primates and other laboratory animals. Science. 1973;182(10773250694):67–68.
  80. 80. Barlow RM, Rennie JC. The fate of ME7 scrapie infection in rats, guinea‐pigs and rabbits. Res Vet Sci. 1976;21(176245654):110–111.
  81. 81. Chianini F, Fernandez‐Borges N, Vidal E, et al. Rabbits are not resistant to prion infection. Proc Natl Acad Sci U S A. 2012;109(13):5080–5085. doi: 10.1073/pnas.1120076109.
  82. 82. Vidal E, Fernandez‐Borges N, Pintado B, et al. Bovine spongiform encephalopathy induces misfolding of alleged prion‐resistant species cellular prion protein without altering its pathobiological features. J Neurosci. 2013;33(18):7778–7786. doi: 10.1523/JNEUROSCI.0244‐13.2013.
  83. 83. Begley DJ. Transport of prion proteins across the blood‐brain barrier. Exp Neurol. 2009;220(2):217–218. doi: 10.1016/j.expneurol.2009.08.006.
  84. 84. Chabry J, Caughey B, Chesebro B. Specific inhibition of in vitro formation of protease‐resistant prion protein by synthetic peptides. J Biol Chem. 1998;273(2198250777):13203–13207.
  85. 85. Meier P, Genoud N, Prinz M, et al. Soluble dimeric prion protein binds PrP(sc) in vivo and antagonizes prion disease. Cell. 2003;113(1):49–60.
  86. 86. Genoud N, Ott D, Braun N, et al. Antiprion prophylaxis by gene transfer of a soluble prion antagonist. Am J Pathol. 2008;172(5):1287–1296. doi: 10.2353/ajpath.2008.070836.
  87. 87. Toupet K, Compan V, Crozet C, et al. Effective gene therapy in a mouse model of prion diseases. PLoS One. 2008;3(7):e2773. doi: 10.1371/journal.pone.0002773.
  88. 88. Soto C, Kascsak RJ, Saborio GP, et al. Reversion of prion protein conformational changes by synthetic beta‐ sheet breaker peptides. Lancet. 2000;355(919920137584):192–197.
  89. 89. De Gioia L, Selvaggini C, Ghibaudi E, et al. Conformational polymorphism of the amyloidogenic and neurotoxic peptide homologous to residues 106‐126 of the prion protein. J Biol Chem. 1994;269(1194179145):7859–7862.
  90. 90. Zhang H, Kaneko K, Nguyen JT, et al. Conformational transitions in peptides containing two putative alpha‐ helices of the prion protein. J Mol Biol. 1995;250(495341684):514–526.
  91. 91. Chabry J, Caughey B, Chesebro B. Specific inhibition of in vitro formation of protease‐resistant prion protein by synthetic peptides. J Biol Chem. 1998;273(21):13203–13207.
  92. 92. Kim HO, Snyder GP, Blazey TM, Race RE, Chesebro B, Skinner PJ. Prion disease induced alterations in gene expression in spleen and brain prior to clinical symptoms. Adv Appl Bioinform Chem. 2008;1:29–50.
  93. 93. Skinner PJ, Abbassi H, Chesebro B, Race RE, Reilly C, Haase AT. Gene expression alterations in brains of mice infected with three strains of scrapie. BMC Genomics. 2006;7:114.
  94. 94. Hetz C, Russelakis‐Carneiro M, Maundrell K, Castilla J, Soto C. Caspase‐12 and endoplasmic reticulum stress mediate neurotoxicity of pathological prion protein. EMBO J. 2003;22(20):5435–5445. doi: 10.1093/emboj/cdg537.
  95. 95. Gauczynski S, Peyrin JM, Haik S, et al. The 37‐kDa/67‐kDa laminin receptor acts as the cell‐surface receptor for the cellular prion protein. EMBO J. 2001;20(21):5863–5875. doi: 10.1093/emboj/20.21.5863.
  96. 96. Leucht C, Simoneau S, Rey C, et al. The 37 kDa/67 kDa laminin receptor is required for PrP(sc) propagation in scrapie‐infected neuronal cells. EMBO Rep. 2003;4(3):290–295. doi: 10.1038/sj.embor.embor768.
  97. 97. Zuber C, Knackmuss S, Rey C, et al. Single chain fv antibodies directed against the 37 kDa/67 kDa laminin receptor as therapeutic tools in prion diseases. Mol Immunol. 2008;45(1):144–151. doi: S0161‐5890(07)00197‐6.
  98. 98. Pflanz H, Vana K, Mitteregger G, et al. Microinjection of lentiviral vectors expressing small interfering RNAs directed against laminin receptor precursor mRNA prolongs the pre‐clinical phase in scrapie‐infected mice. J Gen Virol. 2009;90(Pt 1):269–274. doi: 10.1099/vir.0.004168‐0.

Written By

Pamela J. Skinner and Davis M. Seelig

Submitted: 17 May 2016 Reviewed: 07 December 2016 Published: 08 March 2017