Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Sep 2.
Published in final edited form as: Expert Rev Neurother. 2021 Sep 2;21(9):983–991. doi: 10.1080/14737175.2021.1965882

Neuroprotective Effect and Potential of Cellular Prion Protein and Its Cleavage Products for Treatment of Neurodegenerative Disorders Part II. Strategies for Therapeutics Development

Emily Dexter 1, Qingzhong Kong 1,2,*
PMCID: PMC8453096  NIHMSID: NIHMS1733905  PMID: 34470554

Abstract

Introduction:

Cellular prion protein (PrPC), some of its derivatives (especially PrP N-terminal N1 peptide and shed PrP), and PrPC-containing exosomes have strong neuroprotective activities, which have been reviewed in the companion article (Part I) and are briefly summarized here.

Areas covered:

We propose that elevating the extracellular levels of a protective PrP form using gene therapy and other approaches is a very promising novel avenue for prophylactic and therapeutic treatments against prion disease, Alzheimer’s disease and several other neurodegenerative diseases. We will dissect the pros and cons of various potential PrP-based treatment options and propose a few strategies that are more likely to succeed. The cited references were obtained from extensive PubMed searches of recent literature, including peer-reviewed original articles and review articles.

Expert opinions:

Concurrent knockdown of cellular PrP expression and elevation of the extracellular levels of a neuroprotective PrP N-terminal peptide via optimized gene therapy vectors is a highly promising broad-spectrum prophylactic and therapeutic strategy against several neurodegenerative diseases, including prion diseases, Alzheimer’s disease and Parkinson’s disease.

Keywords: Aβ and other toxic oligomers, ADAM, alpha-cleavage, Alzheimer’s disease, N1 peptide, neurodegenerative diseases, neuroprotection, prion protein, shedding, therapeutics

1. Overview of protective extracellular PrP forms

The cellular prion protein (PrPC) is a glycosylphosphatidylinositol (GPI)-anchored glycoprotein highly expressed in the nervous system and at various levels in lymphoid, muscle and other tissues16.

The biological and pathological roles of PrP are complicated. On one hand, PrPC is essential for both the replication and pathogenesis of the prion agents in prion diseases (PrD)79, and it plays a key role in the pathogenesis of other neurodegenerative diseases [such as Alzheimer’s (AD) and Parkinson’s (PD)], primarily serving as a receptor for the cytotoxic oligomers of amyloid-β (Aβ), Tau, and α-synuclein (αSyn)1017. In addition, elevated PrPC is associated with several cancers1822. On the other hand, cell surface full-length PrPC seems to play an important role in many biological processes56, such as myelin maintenance23 and neurite outgrowth and neuroprotective signaling2426. Some other PrP forms and PrP processing are also protective or beneficial, which has been reviewed in a companion paper27 and is briefly summarized below.

The PrP N-terminal fragment derived from α-cleavage of PrPC, extracellular full-length PrP from shedding of cell surface PrPC, and exosomes containing PrPC have all shown beneficial and protective effects. The α-cleavage of PrPC occurs at aa110–111 or aa111–112, yielding the membrane-attached C-terminal C1 fragment28 and the N-terminal N1 peptide (88–89 amino acid residues) that is released into the extracellular space. A group of enzymes have been implicated in the α-cleavage of PrPC, including ADAM8, ADAM9, ADAM10, and ADAM17, but all except ADAM8 are still controversial2935. It also appears that more than one enzyme is involved in the α-cleavage of PrPC in the CNS and other tissues31,3334.

The N1 peptide is protective against reactive oxygen species3637. It also protects against toxic Aβ oligomers in AD through inhibiting Aβ oligomerization, accelerating fibril formation (thereby reducing levels of the oligomeric forms), and sequestering Aβ oligomers to prevent toxic signaling and alleviate long-term potentiation (LTP) inhibition3840. The C1 fragment has been reported to inhibit prion replication36 and its negative activities4244 seems to be no significant concerns36,41. Although N1 has not been directly shown to bind to and neutralize Tau and αSyn oligomers, the regions in full-length PrP required for binding to these toxic oligomers are fully contained within N117, so it is reasonable to assume that N1 can neutralize these two oligomers as well.

Recombinant full-length PrP has been reported to induce neuritogenesis4546, and binds to and provides protection against toxic Aβ species38,47. Transgenically expressed anchorless PrP, which is basically the same as the shed PrP, has also been reported to reduce LTP impairment in an AD mouse model48.

Shed PrP is released to the extracellular space through cleavages by a sheddase at or near the C-terminus of PrPC (aa228–231) or within the GPI anchor30,4951. ADAM10 appears to be the primary sheddase for PrPC 30,32,35,52, and the PrP shedding activity of ADAM10 is modulated by ADAM930. While there is no direct evidence demonstrating the neuroprotective activity of shed PrP, shed PrP is expected to show similar protective effect as that of recombinant full-length PrP since it is very similar to recombinant full-length PrP except for its O-linked glycans. In addition, PrP shedding is presumed to be protective against prion diseases and other misfolding diseases since it is supposed to decrease cell surface PrPC levels32,53 thereby reducing the levels of the essential substrate for prion replication and a critical receptor for toxic protein oligomers17,5456.

PrP on exosomes, due to their extracellular location, will not mediate the pathogenesis of toxic oligomers, is a third form of protective extracellular PrP that should be safer for therapeutic purpose5758. PrPC on exosomes has been shown to be protective against Aβ toxicity through accelerating Aβ42 aggregation into its fibrillar form, reducing the uptake of synthetic Aβ, abolishing Aβ42-induced apoptosis5759, and ameliorating Aβ-induced synaptic loss and LTP inhibition in rodent AD models57,59. Since recombinant and cellular PrP can also bind to toxic Tau and αSyn oligomers17, it is reasonable to expect the same protective effect against these toxic oligomers from exosomal PrP.

It is very tempting to harness the protective and beneficial activities of some of the PrP forms and PrP processing described above for therapeutic and preventative treatments against PrD, AD, PD and a few other protein misfolding neurodegenerative diseases involving toxic protein oligomers. Here we present and evaluate various strategies for the development of prophylactic and therapeutic treatments against AD and other neurodegenerative diseases through elevating the extracellular levels of one of the protective PrP forms. Knocking down PrP is also a very attractive strategy to battle a number of neurodegenerative diseases where PrPC plays a critical role in mediating toxicity, but this topic has been reviewed very recently60 and will not be discussed in depth here.

2. Strategies for developing PrP-based therapies

We propose that augmenting the extracellular levels of protective PrP forms is an attractive and broad-spectrum strategy for effective treatment and prevention of many neurodegenerative diseases involving toxic oligomers of PrP, Tau, or α-synuclein, including prion diseases, AD, PD, and other tauopathies (such as progressive supranuclear palsy and frontotemporal lobar degeneration) and synucleinopathies (such as dementia with Lewy bodies, multiple system atrophy, and Lewy body variant of AD). Several PrP forms have been shown to, or are expected to, have protective activities when existing in the extracellular space, and their extracellular levels can be enhanced by a variety of approaches. These include (1) full-length PrP that is shed from cell surface, administered as recombinant PrP, or expressed as anchorless PrP from a gene therapy vector, (2) full-length PrP on exosomes, and (3) N1 peptide released by α-cleavage of PrPC on cell surface, administered as a recombinant peptide, expressed as a secreted peptide from a gene therapy vector, or processed by α-cleavage of a fusion protein expressed from a gene therapy vector. Here we evaluate these strategies to elevate the extracellular levels of each of these protective PrP forms and propose the most feasible options for prophylactic and therapeutic purposes.

2.1. PrP N-terminal fragment-based strategies

The PrP N1 peptide protects cells against toxic Aβ oligomers, and it is likely effective against Tau and αSyn oligomers. No significant risks have been identified with extracellular N1, although its reported impact on cell cycle progression36 needs to be assessed. These factors make extracellular N1 and similar extracellular PrP N-terminal peptides highly promising candidates for prophylactic and therapeutic treatments of AD and other neurodegenerative diseases. For simplicity, we will use PrP-N to represent N1 and similar extracellular PrP N-terminal peptides.

There are four main ways to elevate extracellular PrP-N: (1) direct administration of recombinant PrP-N into the ventricular system of the brain, central canal of the spinal cord, or hippocampus and other brain regions, (2) enhancing PrPC α-cleavage activities, (3) CNS expression of a secretive form of PrP-N from a gene therapy vector, and (4) CNS expression of a cell membrane-anchored form of PrP from a gene therapy vector that will be subjected to α-cleavage to release N1 into extracellular space. Some of these strategies have been discussed in an earlier review61.

Enhancing extracellular PrP-N levels through direct administration of a recombinant or synthetic PrP-N peptide is very attractive because it appears to be straightforward and mature technologies exist (Figure 1). There have been reports showing that the cytotoxicity of Aβ aggregates is significantly reduced in vitro or in vivo after preincubation with synthetic PrP N-terminal peptides (PrP2350 or PrP90112)62 or recombinant N1 (PrP23–110)39, although there has been no report of direct infusion with recombinant PrP-N in AD mouse models. In fact, there are many challenges common for the recombinant protein strategy: high costs, biostability, possible immune reactions especially after repeated injections, and detrimental effects on target and non-target cells. For CNS applications, there are also the increased risks of infection and pain associated with repeated invasive intracranial/intrathecal injections. In addition, the recombinant PrP-N strategy will be very costly and likely ineffective if an unmodified N1 peptide is used. The PrP N-terminal domain is unstructured. We and others have found that N1 is highly unstable and easily degraded in tissues6364, suggesting that exogenously administered recombinant N1 may have very short half-life in vivo. It will also require a large amount of the recombinant PrP-N, which is expensive to produce. It is possible to design synthetic modified short PrP-N peptides that have high stability and long half-life in vivo6567, but it will require a lot of experimentation to optimize and validate. Alternatively, fusion with a structured domain may stabilize the N1 peptide as suggested64.

Figure 1. Strategies to develop PrP-based prophylactic and therapeutic treatments against AD and other neurodegenerative diseases.

Figure 1.

Protective extracellular PrPs (full-length, N1 or similar N-terminal fragments [termed PrP-N]) can sequester toxic protein oligomers and neutralize their toxicities in AD and other neurodegenerative diseases. Extracellular PrP levels can be elevated through four approaches. First, direct administration of in vitro prepared PrPC-rich exosomes, recombinant N1, PrP-N or full-length PrP. Second, expression from a gene therapy vector of the secreted N1 or PrP-N, a PrP α-cleavage enzyme, a PrP shedding enzyme, or a protein activating the above α-cleavage or shedding enzymes. Third, enhancing the α-cleavage or shedding of cell surface PrPC through upregulation of the expression of an endogenous gene coding for an α-cleavage or shedding enzyme, or an endogenous gene whose encoded protein activates these endogenous enzymes. Fourth, direct activation of the enzymatic activity of an endogenous PrPC α-cleavage or shedding enzyme. For simplicity, the third and fourth strategies have been simplified as a single concept of increasing α-cleavage or shedding for enhanced production of protective PrP forms in the diagram.

Elevating extracellular PrP-N levels using a gene therapy vector to express PrP-N in the CNS is another appealing strategy (Figure 1). The gene therapy techniques, especially with recombinant adeno-associated virus (rAAV) as the vector, have matured to the point of approved clinical applications for some single-gene diseases in the last few years6870. This strategy has the potential to achieve sustained elevated levels of extracellular N1 or PrP-N for years after a single injection of the gene therapy vector. It will also largely bypass the issue of N1 instability through continuous production and secretion of N1 or PrP-N and will likely achieve a stable elevated N1 or PrP-N concentration in the extracellular space, which may enhance the therapeutic benefits. Mohammadi et al.6364 successfully generated transgenic mice overexpressing N1 without the GPI-anchor signal peptide, but the transgene-expressed N1 was found to be trapped inside the cells due to impaired endoplasmic reticulum translocation, and no protection against Aβ-mediated synaptotoxicity was found in an AD mouse model. Although fusion of a structured domain to the C-terminus of N1 might overcome the intracellular translocation defect of N164, the potential side effects of the fusion tag need to be thoroughly studied. In comparison, adding a structured domain from PrPC itself to the N1 fragment seems to be an attractive alternative since it may not only stabilize the N1 peptide but also solve the secretion defect of N1 without adding a foreign fusion tag. We have demonstrated the feasibility of this approach (unpublished data), but whether the added structured PrP domain will affect the protective activity or leads to undesirable biological activities is still unclear and must be investigated. The gene therapy approach itself also has significant risks, including immunogenicity that will cause inflammation and reduce effectiveness (for viral vectors), chromosomal integrations of the viral DNA (although uncommon for rAAV vectors) that could cause cancers, and high costs and significant difficulty in manufacturing of high-titer viral particles. These concerns can be largely addressed when a non-viral gene therapy vector is used, but the poor transduction efficiency and short duration of gene expression associated with non-viral vectors still need to be significantly improved before clinical use.

Extracellular PrP-N levels can also be augmented through heightened α-cleavage activities, but this strategy is hindered by the uncertainty of the identity of the protease(s) responsible for PrPC α-cleavage. Although several reports implicated the involvement of ADAM8, ADAM9, ADAM10, and ADAM172932,3435, all but ADAM8 remain controversial. The identity of the true α-secretase(s) other than ADAM8 for PrPC must be clarified first.

The selection of an optimal α-secretase ADAM for augmentation will be both a challenging task and a critical factor for outcomes. Of the known ADAMs implicated in PrP processing, ADAM10 was initially reported to contribute to the α-cleavage of PrPC 29,31,35,71, and it seems to be an attractive candidate to be enhanced for a number of reasons described below. But the PrPC α-cleavage activity of ADAM10 is strongly disputed and appears to be ruled out by later experiments with neuron-specific conditional ADAM10-knockout or ADAM10-overexpressing mice or cells30,32,54. Nevertheless, ADAM10 remains an interesting protein for therapeutic purposes even if it is not involved in PrPC α-cleavage. One of the reasons is that ADAM10 is the primary PrP sheddase56 that produces an extracellular PrP form (termed shed PrP) with protective activities38,4548, and it plays a key role in the non-amyloidogenic α-cleavage of APP7273, making it a notable target for AD therapy development7274. Transgenic mice overexpressing ADAM10 in neurons showed increased neurotrophic N-terminal APP domain (APPsα), reduced soluble Aβ peptides, diminished Aβ plaques, and partially rescued LTP74. Additionally, the increased levels of ADAM10 did not seem to cause significant detrimental phenotypic alterations29,54,7475, suggesting that enhancing APP α-cleavage activities through augmenting ADAM10 is a viable strategy. It is worth noting that high-level neuronal overexpression of ADAM10 was reported to cause more seizures and stronger neuronal damage and inflammation after kainate-induced epileptic seizures76. In addition, ADAM10 is known to promote cancer77 and it is a target for cancer and autoimmunity therapies78, suggesting that enhancement of ADAM10 activity needs to be done carefully and kept at moderate levels. Moreover, transgenic neuronal overexpression of ADAM10 in mice led to a decrease in total PrP levels rather than an increase of specific PrPC cleavage products as well as enhanced survival of the mice when challenged with prions54. If the same is true when the ADAM10 activity is elevated through administration of recombinant ADAM10 or expression from a gene therapy vector, augmenting ADAM10 should still protect against AD, PD, and other neurodegenerative diseases through decreasing cell surface PrPC levels. Whether ADAM8 and/or the yet-to-be identified other PrPC α-cleavage enzyme(s) can be targeted to fight AD and other neurodegenerative diseases awaits further studies.

The PrP α-cleavage activities can be enhanced by different approaches (Figure 1): (1) infusion with a bioactive recombinant α-secretase, (2) intracranial expression of a α-secretase from a gene therapy vector, (3) stimulation of the expression of an endogenous α-secretase gene, and (4) enhancement of the activity of an endogenous α-secretase protein.

Infusion with a recombinant α-secretase appears to be a simple solution, but it could be complicated in reality. First, as discussed earlier, we need to select an α-secretase that will have a strong impact on the PrPC α-cleavage activities in the CNS yet with minimal side effects, and it is critical to factor in the substrate spectrum of the α-secretase. All ADAMs implicated in PrPC α-cleavage have many substrates79 and enhanced shedding of some of the unintended substrates due to elevated activity of one PrPC α-cleavage enzyme may have serious deleterious consequences, such as promoting inflammation or tumor metastasis. Second, we must find the best route for administration of the chosen recombinant α-secretase. The expression of α-secretase is usually region and cell type specific in the brain; conceptually, a broad increase of α-secretase activity in the CNS could lead to detrimental cleavages of some substrates in undesirable brain regions and/or cell types. This concern can be partially addressed via localized administration of the recombinant α-secretase to a proper brain region. Third, ideally a modified or variant form of the recombinant α-secretase that does not promote cancer progression can be designed for therapeutic purposes, since all ADAM enzymes implicated in PrPC α-cleavage have been reported to play a role in promoting cancers7780. Administration of a recombinant precursor (inactive) form of the chosen α-secretase will likely be safer since natural cellular regulations may limit the activation of the recombinant precursor to brain areas where this α-secretase is normally expressed. Fourth, the common limitations of recombinant protein therapy described earlier also need to be addressed.

Increased PrP α-cleavage activities can also be achieved through expression of a α-cleavage enzyme from a gene therapy vector (Figure 1), which will come with all the benefits of the gene therapy approach, including sustained effect after a single treatment. Nevertheless, there are many issues to consider with this approach. First, same as for the recombinant protein approach, the choice of the α-secretase enzyme is critical and expression of its precursor form may be safer. Second, the selection/design of an optimal gene therapy vector is paramount that will enable efficient transduction of the CNS cells and effective and authentic cell-type specific expression of the α-secretase gene. The vector backbone and the gene expression cassette for the selected α-secretase need to be evaluated for efficiency of cell entry, specificity of cell targeting, and vector-carried α-secretase gene transcription activity in CNS cells. Furthermore, a choice must be made between integrating type of vectors (retroviral vectors) and non-integrating vectors (rAAV and non-viral plasmid DNA). Viral vectors are often chosen over non-viral vectors because of their high efficiency of cell entry and long duration of expression. Third, the route of gene therapy vector administration is also very important since it will affect the organ distribution as well as brain regional distribution of the gene therapy vector. Systemic administration (e.g. intravenous) will likely lead to broad expression of the α-secretase in peripheral organs (such as liver and lungs) that may have unpredictable consequences and would also require the vector to penetrate efficiently the blood-brain-barrier in order to reach the CNS. In contrast, direct injection of a gene therapy vector to the CNS, intrathecally or intracerebroventricularly, may address the disadvantages of systematic treatments, but it is much more invasive. Fourth, a proper dosage range needs to be determined to achieve adequate prophylactic or therapeutic protections with the least side effects and costs. Fifth, the previously discussed general risks associated with gene therapy also need to be addressed.

Modulation of endogenous α-secretase gene activity can also yield increased levels of extracellular N1 (Figure 1). This approach may be better than the other two approaches described above, because it will likely yield a more authentic expression profile thereby carrying less side effects. There have been reports on regulation of various putative α-secretase gene activities29,8182. Expression of dimeric PrP from a gene therapy vector may also be feasible since transgenic expression of a dimeric PrP has been shown to enhance PrP α-cleavage and appears to be safe8385. In addition, identifying small molecules that specifically activate a proper PrP α-cleavage enzyme through established conventional methods would also be worth trying8687.

Enhancing α-cleavage activities carries significant risks as detailed in the companion paper (Part I) and briefly summarized below. First, all ADAMs implicated in PrPC a-cleavage (ADAM8, ADAM9, ADAM10, and ADAM17) have been reported to promote cancer78,80. Second, enhancing the α-cleavage will also lead to increase levels of the C1 fragment, which has been reported to make cells more sensitive to staurosporine42 and implicated in primary myopathy involving p53 in an inducible transgenic mouse model overexpressing PrP in the skeletal muscles4344. However, the negative effects of C1 may be countered by N1 through down-regulation of p53 expression36. In addition, transgenic mice overexpressing GPI-anchored C1 did not appear to exhibit gross neurological deficits or histological lesions41, suggesting that C1 may be pro-apoptotic only under apoptotic stimuli but not under normal conditions33.

2.2. Exosomal PrP

Enhancing extracellular PrP levels through augmenting PrP-containing exosomes is another attractive alternative for PrP-based therapies since they have been reported to be protective against the toxicity of Aβ, Tau, and αSyn oligomers5759,88. The exosome approach has significant advantages over the recombinant PrP approach. Exosomes are capable of crossing the blood-brain barrier89,90. Exosomes can be engineered for various purposes9192 including as a safe and effective delivery vehicle for anti-cancer agents93. Exosomes have been successfully modified to carry a siRNA cargo and target brain cells, achieving a 62% knockdown of BACE1 (a therapeutic target in Alzheimer’s disease) in wild-type mice when administered intravenously94. Several studies used exosomes to target brain cancers93 and traumatic brain injuries95.

There are two ways to achieve higher levels of PrP-carrying exosomes in the CNS: (1) CNS injection with PrP-containing exosomes generated from cultured cells and (2) enhancement of endogenous exosome generation, but only the former seems feasible at this point (Figure 1). Many methods have been developed for efficient isolation of exosomes9697, including from brain cell types59,88,9899. Exosomes carrying high levels of PrPC may be obtained from cultured cells naturally or engineered to express PrPC at high levels. It should be possible to design an exosomal PrP that is more protective and carries less side effects. The exosomes can also be further engineered to target brain cells for less invasive peripheral administration.

Elevating extracellular PrP levels through administration of PrP-carrying exosomes share some of the risks with the recombinant full-length PrP strategy, such as unwanted functions of the PrP protein, the need for repeated treatments, and likely high costs. In addition, exosomes carry many other proteins and lipids on its surface100 as well as proteins, nucleic acids and other molecules internally, which could have detrimental effects101103. Several of the caveats associated with exosomes have been reviewed recently91. For example, exosomes from prion-infected cells have been reported to carry prion infectivity104 and facilitate the replication and spread of prions105106. Careful selection of cells for exosome production and targeted engineering of the exosomes will reduce the risks.

2.3. Shed PrP or recombinant full-length PrP

Elevating extracellular PrP levels for therapeutic purposes can also be achieved through enhancing shedding of endogenous PrPC or infusion with recombinant full-length PrP. Recombinant full-length PrP or transgenic expression of anchorless PrP that is secreted into extracellular space have shown protective effects38,4548. It is reasonable to expect similar protective activities from full-length PrP shed from PrPC on cell surfaces. Enhancing PrP shedding was indeed found to be protective against prions in most reports30,5354,56.

Direct CNS infusion with recombinant full-length PrP seems simple (Figure 1), but there have been no published reports with this approach. Full-length PrP carries unique risks, including promotion of replication and spread of prions107109 and induction of CNS inflammation110111. In addition, the common caveats related to the use of recombinant proteins also apply.

Enhancing shedding of cell surface PrPC is an alternative to direct infusion of full-length recombinant PrP. This strategy aims to augment PrP shedding from cell surface through enhancing the activity of ADAM10, the primary PrP sheddase, which is supposed to lead to increased shedding of PrPC and α-cleavage of PrPC and APP, all of which are protective7274. It can be achieved through four strategies that are very similar to those of the of α-secretase activity enhancement strategy (Figure 1): (1) infusion with a bioactive recombinant sheddase (ADAM10), (2) CNS expression of ADAM10 from a gene therapy vector, (3) stimulation of the expression of the endogenous ADAM10 gene, and (4) enhancing the activity of the endogenous ADAM10 protein. The last two approaches are of particular interest since they will likely preserve the authentic spatial expression profile of the endogenous ADAM10 gene, thereby minimizing potential side effects. Again, activating ADAM9 appears to be an attractive strategy since ADAM9 has been reported to promote shedding of active ADAM1081 and upregulate the α-cleavage of PrPC 29,81 and APP72. The same pros and cons described earlier for the α-secretase activity enhancement approach also apply here. Moreover, there are multiple potential risks associated with the ADAM10 enhancement approach, including promoting cancer and autoimmunity7778 as well as facilitating prion replication and inducing CNS inflammation due to increases level of shed PrP108,109. Despite these concerns, it may still be possible to elevate ADAM10 activity safely since transgenic mice overexpressing ADAM10 appear largely normal29,54,7475.

3. Expert opinion

Extracellular PrP forms, including shed PrP, exosomal PrP and PrP N-terminal fragments (PrP-N), have strong and broad spectrum therapeutic potentials for several neurodegenerative diseases due to their protective activities against PrD, AD, PD and other protein misfolding neurodegenerative diseases.

We have summarized a variety of strategies to harness the power of protective PrP forms, including direct infusion with recombinant PrP or its N-terminal fragments, infusion of recombinant enzymes that release PrP or its N-terminal fragments from cell surfaces, infusion with exosomes carrying PrP, expression of a secreted form of PrP or its N-terminal fragments from a gene therapy vector, or enhancing the expression/activities of an endogenous a-secretase or sheddase (Figure 1). All of these strategies have their own strengths and weaknesses. Because N1 has shown multiple protective activities and the N1 peptide has no known side effect aside from its impact on cell cycle progression, the most promising approach seems to be expression of a well-designed PrP-N from an optimal gene therapy vector that can achieve sustained high-level expression and secretion of N1 or PrP-N in the CNS after a single injection. The second attractive approach is elevating ADAM9 activities that will activate ADAM10 to simultaneously enhance the α-cleavage of APP and the shedding of PrPC. However, the cancer promoting effect of ADAM9112 needs to be evaluated thoroughly to validate the safety of this approach and a safe and effective range of ADAM9 elevation must be established. The third appealing strategy is peripheral administration of engineered exosomes that target brain cells and carry an optimized PrP with strong protective activities and minimal hazardous activities. Both the exosomes and the PrP molecules they carry can be engineered to maximize the delivery efficiency and protective activities while minimizing unwanted side effects. These approaches, especially those delivered as gene therapies, also have the potential to be used safely as prophylactics for disease prevention when applied before disease onset or at preclinical stages. AD and other neurodegenerative diseases result from progressive damages to dendrites, axons and synapses, as well as loss of neurons. Neuronal loss in most CNS regions is irreversible but minimal during preclinical and early phases of the diseases. Therefore, early and accurate diagnosis of the neurodegenerative diseases, including the subtypes or strains, combined with early and prompt interventions (including with our proposed strategies), will be critical for favorable clinical outcomes. There will be significant challenges with these strategies, ranging from direct side effects of the elevated PrP form, the negative impact stemming from the molecules adopted to elevate the extracellular PrP forms (such as the problems associated with enhancing an ADAM enzyme activity), and hurdles and complications from the technologies employed (such as the difficulty in generating the required amount of clinical grade recombinant proteins or gene therapy vectors, immune response to the recombinant protein or vector, and potential carcinogenicity of the gene therapy vector).

In addition, knocking down endogenous PrP gene expression with antisense oligonucleotides or siRNA has shown great potential in delaying onset and slowing down progression in AD and PrD mouse models60,113120. It will likely be much more effective to use two gene therapy vectors to simultaneously overexpress a secreted PrP-N and knock down the endogenous PrP expression. This combined strategy should achieve strong neuroprotection (through PrP-N) and significant reduction of toxic signaling mediated by the cell surface PrPC (through the knockdown of the endogenous PrP gene). We are in the process of testing this combined strategy for treatment and prevention of human prion diseases in humanized transgenic mouse models.

Article highlights:

  • The extracellular prion protein (PrP) forms, such as shed PrP, exosomal PrP and PrP N-terminal fragments (PrP-N) can inhibit and neutralize key toxic molecules and prevent toxic signaling in prion diseases (PrD), Alzheimer’s disease (AD), Parkinson’s Disease (PD) and a few other related protein misfolding neurodegenerative diseases.

  • Several strategies to harness the protective powers of the extracellular PrP forms are explored, including direct infusion with a recombinant PrP form, infusion of recombinant enzymes that release the PrP form from the cell surface (ADAM10 or ADAM8 or ADAM17), peripheral infusion with PrP-rich exosomes engineered to target the brain, or increased expression of a secreted form of PrP from a gene therapy vector, or enhancing the expression/activities of an endogenous secretase or sheddase.

  • Expected challenges for each strategy are discussed, including unwanted increased biological activity of the PrP form, indirect side effects of increased PrP cleavage enzyme activity, difficulty in generating the sufficient recombinant proteins or gene therapy vectors for treatment, risk of immune responses to recombinant proteins or gene therapy vector, and potential carcinogenicity of the gene therapy vector.

  • The optimal strategy to increase PrP-N levels consists of a gene therapy vector that can achieve sustained high-level expression and secretion of PrP-N in the CNS after a single injection.

  • Another appealing strategy is to elevate the ADAM9 activity that will activate ADAM10 to enhance both the α-cleavage of APP and the shedding of PrPC.

  • Simultaneous knockdown of cellular PrP expression and elevation of the extracellular levels of a PrP-N via optimized gene therapy vectors should be a highly promising broad -spectrum prophylactic and therapeutic strategy against several neurodegenerative diseases, including PrD, AD and PD.

Acknowledgements

The figure was created with BioRender.com.

Funding

This work was partially supported by NIH R01 NS109532 and a research grant from CJD Foundation.

Footnotes

Declaration of interests

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or conflict with the subject matter or materials discussed in this manuscript apart from those disclosed.

References

Papers of special note have been highlighted as either of interest (*) or of considerable interest (**) to readers.

  • 1.Riek R, Hornemann S, Wider G, et al. NMR structure of the mouse prion protein domain PrP(121–231). Nature. 1996;382:180–2 [DOI] [PubMed] [Google Scholar]
  • 2.Zahn R, Liu A, Lührs T, et al. NMR solution structure of the human prion protein. Proc Natl Acad Sci U S A. 2000;97:145–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Adle-Biassette H, Verney C, Peoc’h K, et al. Immunohistochemical Expression of Prion Protein (PrPC) in the Human Forebrain During Development, Journal of Neuropathology & Experimental Neurology, 2006;65:698–706 [DOI] [PubMed] [Google Scholar]
  • 4.Peralta OA, Eyestone WH. Quantitative and qualitative analysis of cellular prion protein (PrP(C)) expression in bovine somatic tissues. Prion. 2009;3:161–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Castle AR, Gill AC. Physiological Functions of the Cellular Prion Protein. Front Mol Biosci. 2017;6;4:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Linden R The Biological Function of the Prion Protein: A Cell Surface Scaffold of Signaling Modules. Front Mol Neurosci. 2017;10:77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Aguzzi A, Heikenwalder M. Pathogenesis of prion diseases: current status and future outlook. Nat Rev Microbiol. 2006;4:765–75 [DOI] [PubMed] [Google Scholar]
  • 8.Colby DW and Prusiner SB. Prions. Cold Spring Harb Perspect Biol. 2011;3:a006833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sigurdson CJ, Bartz JC, Glatzel M. Cellular and Molecular Mechanisms of Prion Disease. Annu Rev Pathol. 2019January24;14:497–516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Laurén J, Gimbel DA, Nygaard HB, et al. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457:1128–32 [DOI] [PMC free article] [PubMed] [Google Scholar]; **First report on cellular PrP as a mediator of the toxicity of Aβ oligomers
  • 11.Um JW, Nygaard HB, Heiss JK, et al. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci. 2012;15:1227–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Haas LT, Salazar SV, Kostylev MA, et al. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in Alzheimer’s disease. Brain. 2016;139:526–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Aulić S, Masperone L, Narkiewicz J, et al. α-Synuclein amyloids hijack prion protein to gain cell entry, facilitate cell-to-cell spreading and block prion replication. Sci Rep. 2017;7:10050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Salazar SV, Strittmatter SM. Cellular prion protein as a receptor for amyloid-β oligomers in Alzheimer’s disease. Biochem Biophys Res Commun. 2017;483:1143–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shafiei SS, Guerrero-Muñoz MJ, Castillo-Carranza DL. Tau Oligomers: Cytotoxicity, Propagation, and Mitochondrial Damage. Front Aging Neurosci. 2017;9:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Urrea L, Segura-Feliu M, Masuda-Suzukake M, et al. Involvement of Cellular Prion Protein in α-Synuclein Transport in Neurons Mol Neurobiol. 2018;55:1847–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Corbett GT, Wang Z, Hong W, et al. Young-Pearse TL, Billinton A, Walsh DM. PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins. Acta Neuropathol. 2020;139:503–26 [DOI] [PMC free article] [PubMed] [Google Scholar]; **Cellular PrP is a key receptor mediating the toxicity of soluble aggregates (oligomers) of Aβ, Tau, and αSyn
  • 18.Pan Y, Zhao L, Liang J, et al. Cellular prion protein promotes invasion and metastasis of gastric cancer. FASEB J. 2006;20:1886–8 [DOI] [PubMed] [Google Scholar]
  • 19.Han H, Bearss DJ, Browne LW, et al. Identification of differentially expressed genes in pancreatic cancer cells using cDNA microarray. Cancer Res. 2002;62:2890–6 [PubMed] [Google Scholar]
  • 20.Li C, Yu S, Nakamura F, et al. Binding of pro-prion to filamin A disrupts cytoskeleton and correlates with poor prognosis in pancreatic cancer. J Clin Invest. 2009;119:2725–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sy MS, Li C, Yu S, et al. The fatal attraction between pro-prion and filamin A: prion as a marker in human cancers. Biomark Med. 2010;4:453–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Antony H, Wiegmans AP, Wei MQ, et al. Potential roles for prions and protein-only inheritance in cancer. Cancer Metastasis Rev. 2012;31:1–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bremer J, Baumann F, Tiberi C, et al. Axonal prion protein is required for peripheral myelin maintenance. Nat Neurosci. 2010;13:310–8 [DOI] [PubMed] [Google Scholar]
  • 24.Chiarini LB, Freitas AR, Zanata SM, et al. Cellular prion protein transduces neuroprotective signals. EMBO J. 2002;21:3317–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chen S, Mangé A, Dong L, et al. Prion protein as trans-interacting partner for neurons is involved in neurite outgrowth and neuronal survival. Mol. Cell. Neurosci, 2003;22:227–33 [DOI] [PubMed] [Google Scholar]
  • 26.Lopes MH, Hajj GN, Muras AG, et al. Interaction of cellular prion and stress-inducible protein 1 promotes neuritogenesis and neuroprotection by distinct signaling pathways. J Neurosci. 2005;25:11330–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dexter E, Kong Q. Neuroprotective Effect and Potential of Cellular Prion Protein and Its Cleavage Products for Treatment of Neurodegenerative Disorders. Part II. Strategies for Therapeutics Development. Expert Review of Neurotherapeutics. 2021; [DOI] [PMC free article] [PubMed] [Google Scholar]; **The companion article reviewing the relevant literature
  • 28.Chen SG, Teplow DB, Parchi P, et al. Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem. 1995;270:19173–80 [DOI] [PubMed] [Google Scholar]
  • 29.Cissé MA, Sunyach C, Lefranc-Jullien S, et al. The disintegrin ADAM9 indirectly contributes to the physiological processing of cellular prion by modulating ADAM10 activity. J. Biol. Chem 2005;280:40624–31 [DOI] [PubMed] [Google Scholar]
  • 30.Taylor DR, Parkin ET, Cocklin SL, et al. Role of ADAMs in the ectodomain shedding and conformational conversion of the prion protein. J Biol Chem. 2009;284:22590–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Vincent B, Paitel E, Saftig P, et al. The disintegrins ADAM10 and TACE contribute to the constitutive and phorbol ester-regulated normal cleavage of the cellular prion protein. J Biol Chem. 2001;276:37743–6 [DOI] [PubMed] [Google Scholar]
  • 32.Altmeppen HC, Prox J, Puig B, et al. Lack of a-disintegrin-and-metalloproteinase ADAM10 leads to intracellular accumulation and loss of shedding of the cellular prion protein in vivo. Mol Neurodegener. 2011;6:36. [DOI] [PMC free article] [PubMed] [Google Scholar]; **ADAM10 is the primary PrP sheddase and appears not involved in PrP α-cleavage
  • 33.Liang J, Kong Q. α-Cleavage of cellular prion protein. Prion, 2012a;6:453–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liang J, Wang W, Sorensen D, et al. Cellular prion protein regulates its own α-cleavage through ADAM8 in skeletal muscle. J Biol Chem. 2012b;287:16510–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.McDonald AJ, Dibble JP, Evans EG, et al. A new paradigm for enzymatic control of α-cleavage and β-cleavage of the prion protein. J Biol Chem. 2014b;289:803–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Guillot-Sestier MV, Sunyach C, Druon C, et al. The alpha-secretase-derived N-terminal product of cellular prion, N1, displays neuroprotective function in vitro and in vivo. J Biol Chem. 2009; 284:35973–86 [DOI] [PMC free article] [PubMed] [Google Scholar]; **First report on the neuroprotective effect of N1 in vitro and in vivo
  • 37.Guillot-Sestier MV, Sunyach C, Ferreira ST, et al. α-Secretase-derived fragment of cellular prion, N1, protects against monomeric and oligomeric amyloid β (Aβ)-associated cell death. J Biol Chem. 2011;287:5021–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fluharty BR, Biasini E, Stravalaci M, et al. An N-terminal fragment of the prion protein binds to amyloid-β oligomers and inhibits their neurotoxicity in vivo. J Biol Chem. 2013;288:7857–66 [DOI] [PMC free article] [PubMed] [Google Scholar]; *First report on the neuroprotective effect of N1 against the toxicity of Aβ oligomers in vivo
  • 39.Béland M, Bédard M, Tremblay G, et al. Aβ induces its own prion protein N-terminal fragment (PrPN1)-mediated neutralization in amorphous aggregates. Neurobiol Aging. 2014a;35:1537–48 [DOI] [PubMed] [Google Scholar]
  • 40.Scott-McKean JJ, Surewicz K, Choi JK, et al. Soluble prion protein and its N-terminal fragment prevent impairment of synaptic plasticity by Aβ oligomers: Implications for novel therapeutic strategy in Alzheimer’s disease. Neurobiol Dis. 2016;91:124–31 [DOI] [PMC free article] [PubMed] [Google Scholar]; *Both recombinant PrP and recombinant N1 neutralize the toxicity of Aβ oligomers
  • 41.Westergard L, Turnbaugh JA, Harris DA. A naturally occurring C-terminal fragment of the prion protein (PrP) delays disease and acts as a dominant-negative inhibitor of PrPSc formation. J Biol Chem. 2011;286:44234–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sunyach C, Cisse MA, da Costa CA, et al. The C-terminal products of cellular prion protein processing, C1 and C2, exert distinct influence on p53-dependent sTaurosporine-induced caspase-3 activation. J Biol Chem. 2007;282:1956–63 [DOI] [PubMed] [Google Scholar]
  • 43.Huang S, Liang J, Zheng M, et al. Regulated over-expression of PrP in the skeletal muscles leads to myopathy in transgenic mice. Proc Natl Acad Sci USA. 2007;104:6800–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Liang J, Parchaliuk D, Medina S, et al. Activation of p53-regulated pro-apoptotic signaling pathways in PrP-mediated myopathy. BMC Genomics. 2009;10:201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kanaani J, Prusiner SB, Diacovo J, et al. Recombinant prion protein induces rapid polarization and development of synapses in embryonic rat hippocampal neurons in vitro. Journal of Neurochemistry, 2005;95: 1373–86 [DOI] [PubMed] [Google Scholar]
  • 46.Amin L, Xuan TA Nguyen Rolle IG, et al. Characterization of prion protein function by focal neurite stimulation Cell Sci. 2016;129: 3878–91 [DOI] [PubMed] [Google Scholar]
  • 47.Nieznanski K, Choi JK, Chen S, et al. Soluble prion protein inhibits amyloid-β (Aβ) fibrillization and toxicity. J Biol Chem. 2012;287:33104–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Calella AM, Farinelli M, Nuvolone M, et al. Prion protein and Abeta-related synaptic toxicity impairment. EMBO Mol Med. 2010;2:306–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tagliavini F, Prelli F, Porro M, et al. A soluble form of prion protein in human cerebrospinal fluid: implications for prion-related encephalopathies. Biochem Biophys Res Commun 1992;184:1398–404 [DOI] [PubMed] [Google Scholar]
  • 50.Borchelt DR, Rogers M, Stahl N, et al. Release of the cellular prion protein from cultured cells after loss of its glycoinositol phospholipid anchor. Glycobiology. 1993;3:319–29 [DOI] [PubMed] [Google Scholar]
  • 51.Parizek P, Roeckl C, Weber J, et al. Similar turnover and shedding of the cellular prion protein in primary lymphoid and neuronal cells. J Biol. Chem 2001;276:44627–32 [DOI] [PubMed] [Google Scholar]
  • 52.McDonald AJ, Millhauser GL. PrP overdrive: does inhibition of α-cleavage contribute to PrP(C) toxicity and prion disease? Prion. 2014a;8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Heiseke A, Schöbel S, Lichtenthaler SF, et al. The Novel Sorting Nexin SNX33 Interferes with Cellular PrPSc Formation by Modulation of PrPc Shedding. Traffic, 2008;9:1116–29 [DOI] [PubMed] [Google Scholar]
  • 54.Endres K, Mitteregger G, Kojro E, et al. Influence of ADAM10 on prion protein processing and scrapie infectiosity in vivo. Neurobiol Dis 2009; 36:233–41 [DOI] [PubMed] [Google Scholar]
  • 55.Watts JC, Giles K, Patel S, et al. Evidence that bank vole PrP is a universal acceptor for prions. PLoS Pathog 2014;10: e1003990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Altmeppen HC, Prox J, Krasemann S, et al. The sheddase ADAM10 is a potent modulator of prion disease. eLife, 2015;4, e04260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.An K, Klyubin I, Kim Y,J et al. Exosomes neutralize synaptic-plasticity-disrupting activity of Aβ assemblies in vivo. Mol. Brain 2013;6,47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Falker C, Hartmann A, Guett I, et al. Exosomal cellular prion protein drives fibrillization of amyloid beta and counteracts amyloid beta-mediated neurotoxicity. J Neurochem. 2016;137:88–100 [DOI] [PubMed] [Google Scholar]; **PrP on exosomes neutralizes Aβ toxicity
  • 59.Yuyama K, Sun H, Sakai S, et al. Decreased Amyloid-beta Pathologies by Intracerebral Loading of Glycosphingolipid-enriched Exosomes in Alzheimer Model Mice. Journal of Biochemistry. 2014;289:24488–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Legname G, Scialò C. On the role of the cellular prion protein in the uptake and signaling of pathological aggregates in neurodegenerative diseases. Prion. 2020;14:257–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Béland M, Roucou X. Taking advantage of physiological proteolytic processing of the prion protein for a therapeutic perspective in prion and Alzheimer diseases. Prion. 2014b;8:106–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Nieznanska H, Bandyszewska M, Surewicz K, et al. Identification of prion protein-derived peptides of potential use in Alzheimer’s disease therapy. Biochim Biophys Acta. 2018;1864;2143–53 [DOI] [PubMed] [Google Scholar]
  • 63.Mohammadi B, Linsenmeier L, Shafiq M, et al. Transgenic Overexpression of the Disordered Prion Protein N1 Fragment in Mice Does Not Protect Against Neurodegenerative Diseases Due to Impaired ER Translocation. Mol Neurobiol. 2020;57:2812–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mohammadi B, Glatzel M, Altmeppen HC. Disordered structure and flexible roles: using the prion protein N1 fragment for neuroprotective and regenerative therapy. Neural Regen Res. 2021;16:1431–2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Müller MM. Post-Translational Modifications of Protein Backbones: Unique Functions, Mechanisms, and Challenges. Biochemistry. 2018;57:177–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fisher E, Pavlenko K, Vlasov A, et al. Peptide-Based Therapeutics for Oncology. Pharmaceut Med. 2019;33:9–20 [DOI] [PubMed] [Google Scholar]
  • 67.Ding Y, Ting JP, Liu J, et al. Impact of non-proteinogenic amino acids in the discovery and development of peptide therapeutics. Amino Acids. 2020;52:1207–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.San Sebastian W, Samaranch L, Kells AP, et al. Gene therapy for misfolding protein diseases of the central nervous system. Neurotherapeutics. 2013;10:498–510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Naidoo J, Stanek LM, Ohno K, et al. Extensive Transduction and Enhanced Spread of a Modified AAV2 Capsid in the Non-human Primate CNS. Mol Ther. 2018;26:2418–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358–78 [DOI] [PMC free article] [PubMed] [Google Scholar]; *Excellent review on AAV vectors for gene therapy
  • 71.Vincent B, Paitel E, Frobert Y, et al. Phorbol ester-regulated cleavage of normal prion protein in HEK293 human cells and murine neurons. J Biol Chem. 2000;275:35612–6 [DOI] [PubMed] [Google Scholar]
  • 72.Lammich S, Kojro E, Postina R, et al. Constitutive and regulated alpha-secretase cleavage of Alzheimer’s amyloid precursor protein by a disintegrin metalloprotease. Proc Natl Acad Sci USA. 1999;96:3922–7 [DOI] [PMC free article] [PubMed] [Google Scholar]; **ADAM10 is an α-secretase for APP
  • 73.Kuhn PH, Wang H, Dislich B, Colombo A, Zeitschel U, Ellwart JW, Kremmer E, Rossner S, Lichtenthaler SF. ADAM10 is the physiologically relevant, constitutive alpha-secretase of the amyloid precursor protein in primary neurons. EMBO J. 2010;29:3020–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Postina R, Schroeder A, Dewachter I, et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J. Clin. Invest 2004;113,1456–64 [DOI] [PMC free article] [PubMed] [Google Scholar]; **Moderate neuronal ADAM10 overexpression is safe and effective in AD mice
  • 75.Prinzen C, Trümbach D, Wurst W, et al. Differential gene expression in ADAM10 and mutant ADAM10 transgenic mice. BMC Genomics. 2009;10:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Clement AB, Hanstein R, Schröder A, et al. Effects of neuron-specific ADAM10 modulation in an in vivo model of acute excitotoxic stress. Neuroscience. 2008;152:459–68 [DOI] [PubMed] [Google Scholar]
  • 77.Mullooly M, McGowan PM, Crown J, et al. The ADAMs family of proteases as targets for the treatment of cancer. Cancer Biol Ther. 2016;17:870–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Smith TM Jr, Tharakan A, Martin RK. Targeting ADAM10 in Cancer and Autoimmunity. Front Immunol. 2020;11:499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Dreymueller D, Pruessmeyer J, Groth E, Ludwig A. The role of ADAM-mediated shedding in vascular biology. Eur J Cell Biol. 2012;91:472–85 [DOI] [PubMed] [Google Scholar]
  • 80.Conrad C, Benzel J, Dorzweiler K, et al. ADAM8 in invasive cancers: links to tumor progression, metastasis, and chemoresistance. Clin Sci (Lond). 2019;133:83–99 [DOI] [PubMed] [Google Scholar]
  • 81.Tousseyn T, Thathiah A, Jorissen E, et al. ADAM10, the rate-limiting protease of regulated intramembrane proteolysis of Notch and other proteins, is processed by ADAMS-9, ADAMS-15, and the gamma-secretase. J Biol Chem. 2009;284:11738–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Matthews AL, Noy PJ, Reyat JS, et al. Regulation of A disintegrin and metalloproteinase (ADAM) family sheddases ADAM10 and ADAM17: The emerging role of tetraspanins and rhomboids. Platelets. 2017;28:333–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Béland M, Motard J, Barbarin A, et al. PrP(C) homodimerization stimulates the production of PrPC cleaved fragments PrPN1 and PrPC1. J Neurosci. 2012;32:13255–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Meier P, Genoud N, Prinz M, et al. Soluble dimeric prion protein binds PrP(Sc) in vivo and antagonizes prion disease. Cell 2003;113:49–60 [DOI] [PubMed] [Google Scholar]
  • 85.Engelke AD, Gonsberg A, Thapa S, et al. Dimerization of the cellular prion protein inhibits propagation of scrapie prions. J Biol Chem. 2018;293:8020–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dai J, Liu ZQ, Wang XQ, et al. Discovery of Small Molecules for Up-Regulating the Translation of Antiamyloidogenic Secretase, a Disintegrin and Metalloproteinase 10 (ADAM10), by Binding to the G-Quadruplex-Forming Sequence in the 5’ Untranslated Region (UTR) of Its mRNA. J Med Chem. 2015;58:3875–91 [DOI] [PubMed] [Google Scholar]
  • 87.Wetzel S, Seipold L, Saftig P. The metalloproteinase ADAM10: A useful therapeutic target? Biochim Biophys Acta Mol Cell Res. 2017;1864:2071–81 [DOI] [PubMed] [Google Scholar]
  • 88.Yuyama K, Sun H, Usuki S, et al. A potential function for neuronal exosomes: Sequestering intracerebral amyloid-beta peptide. Febs Letters. 2015;589:84–8 [DOI] [PubMed] [Google Scholar]; *Brain infusion of neuronal exosomes reduces Aβ and amyloid depositions in AD mice
  • 89.Yang T, Martin P, Fogarty B, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in danio rerio. Pharm. Res 2015;32, 2003–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Chen C, Liu L, Ma F et al. Elucidation of Exosome Migration Across the Blood–Brain Barrier Model In Vitro. Cel. Mol. Bioeng 2016;9,509–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Li X, Corbett AL, Taatizadeh E, et al. Challenges and opportunities in exosome research-Perspectives from biology, engineering, and cancer therapy. APL bioengineering, 2019;3, 011503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Jafari D, Shajari S, Jafari R, et al. Designer Exosomes: A New Platform for Biotechnology Therapeutics. BioDrugs. 2020;34:567–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Song H, Liu B, Dong B, et al. Exosome-Based Delivery of Natural Products in Cancer Therapy. Front Cell Dev Biol. 2021;9:650426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol 2011;29, 341–5 [DOI] [PubMed] [Google Scholar]
  • 95.Xiong Y, Mahmood A, Chopp M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen Res. 2017;12:19–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Stam J, Bartel S, Bischoff R, et al. Isolation of extracellular vesicles with combined enrichment methods. J Chromatogr B Analyt Technol Biomed Life Sci. 2021;1169:122604. [DOI] [PubMed] [Google Scholar]
  • 97.Kurian TK, Banik S, Gopal D, et al. Elucidating Methods for Isolation and Quantification of Exosomes: A Review. Mol Biotechnol. 2021;63:249–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gao B, Zhou S, Sun C, et al. Brain Endothelial Cell-Derived Exosomes Induce Neuroplasticity in Rats with Ischemia/Reperfusion Injury. ACS Chem Neurosci. 2020;11:2201–13 [DOI] [PubMed] [Google Scholar]
  • 99.Tu YK, Hsueh YH. Extracellular vesicles isolated from human olfactory ensheathing cells enhance the viability of neural progenitor cells. Neurol Res. 2020;42:959–67 [DOI] [PubMed] [Google Scholar]
  • 100.Maia J, Caja S, Strano Moraes MC, et al. Exosome-Based Cell-Cell Communication in the Tumor Microenvironment. Front Cell Dev Biol. 2018;6:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Emmanouilidou E, Melachroinou K, Roumeliotis T, et al. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci. 2010;30:6838–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hu G, Yao H, Chaudhuri AD, et al. Exosome-mediated shuttling of microRNA-29 regulates HIV Tat and morphine-mediated neuronal dysfunction. Cell Death Dis. 2012;3:e381–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Saman S, Kim W, Raya M, et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J Biol Chem. 2012;287: 3842–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fevrier B, Vilette D, Archer F, et al. Cells release prions in association with exosomes. Proc Natl Acad Sci U S A. 2004;101:9683–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Vella LJ, Sharples RA, Lawson VA, et al. Packaging of prions into exosomes is associated with a novel pathway of PrP processing. J Pathol. 2007;211:582–90 [DOI] [PubMed] [Google Scholar]
  • 106.Cervenakova L, Saa P, Yakovleva O, et al. Are prions transported by plasma exosomes? Transfus Apher. Sci 2016;55, 70–83 [DOI] [PubMed] [Google Scholar]
  • 107.Chesebro B, Trifilo M, Race R, et al. Anchorless prion protein results in infectious amyloid disease without clinical scrapie. Science 2005;308:1435–9 [DOI] [PubMed] [Google Scholar]
  • 108.Chesebro B, Race B, Meade-White K, et al. Fatal transmissible amyloid encephalopathy: a new type of prion disease associated with lack of prion protein membrane anchoring. PLoS Pathog. 2010;6:e1000800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Stohr J, Watts JC, Legname G, et al. Spontaneous generation of anchorless prions in transgenic mice. Proc Natl Acad Sci USA 2011;108:21223–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Roberts TK, Eugenin EA, Morgello S, et al. PrPC, the cellular isoform of the human prion protein, is a novel biomarker of HIV-associated neurocognitive impairment and mediates neuroinflammation. Am J Pathol. 2010;177:1848–1860 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Megra BW, Eugenin EA, Berman JW. The Role of Shed PrPc in the Neuropathogenesis of HIV Infection. J Immunol. 2017;199:224–232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chou CW, Huang YK, Kuo TT, et al. An Overview of ADAM9: Structure, Activation, and Regulation in Human Diseases. Int J Mol Sci. 2020;21:7790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Daude N Specific inhibition of pathological prion protein accumulation by small interfering RNAs. J Cell Sci. 2003;116: 2775–9 [DOI] [PubMed] [Google Scholar]
  • 114.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;116:3204–10 [DOI] [PMC free article] [PubMed] [Google Scholar]; **First to demonstrate the effectiveness and safety of anti-PrP RNAi for prion disease treatment in mice
  • 115.Kong Q RNAi: a novel strategy for the treatment of prion diseases. J Clin Invest. 2006; 116:3101–3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.White MD, Farmer M, Mirabile I, et al. Single treatment with RNAi against prion protein rescues early neuronal dysfunction and prolongs survival in mice with prion disease. Proc Natl Acad Sci. 2008; 105: 10238–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Pulford B, Reim N, Bell A, et al. Liposome-siRNA-peptide complexes cross the blood-brain barrier and significantly decrease PrPC on neuronal cells and PrPRES in infected cell cultures. PLoS One. 2010;5: 1–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Onodera T, Sakudo A, Tsubone H et al. Review of studies that have used knockout mice to assess normal function of prion protein under immunological or pathophysiological stress. Microbiol Immunol, 2014;58: 361–74 [DOI] [PubMed] [Google Scholar]
  • 119.Bender H, Noyes N, Annis JL, PrPC knockdown by liposome-siRNA-peptide complexes (LSPCs) prolongs survival and normal behavior of prion-infected mice immunotolerant to treatment. PLoS One. 2019;14:e0219995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Raymond GJ, Zhao HT, Race B, et al. Antisense oligonucleotides extend survival of prion-infected mice. JCI Insight. 2019;5:131175. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES