Next Article in Journal
Contrasting Effects of an Atherogenic Diet and High-Protein/Unsaturated Fatty Acids Diet on the Accelerated Aging Mouse Model SAMP8 Phenotype
Previous Article in Journal
Outcome Prediction by Diffusion Tensor Imaging (DTI) in Patients with Traumatic Injuries of the Median Nerve
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Systematic Review of Sporadic Creutzfeldt-Jakob Disease: Pathogenesis, Diagnosis, and Therapeutic Attempts

by
Maria Carolina Jurcau
1,
Anamaria Jurcau
2,*,
Razvan Gabriel Diaconu
1,
Vlad Octavian Hogea
1 and
Vharoon Sharma Nunkoo
3
1
Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
2
Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
3
Neurorehabilitation Ward, Clinical Emergency County Hospital Bihor, 410169 Oradea, Romania
*
Author to whom correspondence should be addressed.
Neurol. Int. 2024, 16(5), 1039-1065; https://doi.org/10.3390/neurolint16050079
Submission received: 3 August 2024 / Revised: 9 September 2024 / Accepted: 14 September 2024 / Published: 20 September 2024

Abstract

:
Creutzfeldt-Jakob disease is a rare neurodegenerative and invariably fatal disease with a fulminant course once the first clinical symptoms emerge. Its incidence appears to be rising, although the increasing figures may be related to the improved diagnostic tools. Due to the highly variable clinical picture at onset, many specialty physicians should be aware of this disease and refer the patient to a neurologist for complete evaluation. The diagnostic criteria have been changed based on the considerable progress made in research on the pathogenesis and on the identification of reliable biomarkers. Moreover, accumulated knowledge on pathogenesis led to the identification of a series of possible therapeutic targets, although, given the low incidence and very rapid course, the evaluation of safety and efficacy of these therapeutic strategies is challenging.

1. Introduction

Creutzfeldt-Jakob disease (CJD), first described in 1920 by German neurologist Hans Gerhard Creutzfeldt and shortly afterward by Alfons Maria Jakob [1], is a rare, rapidly progressive, and invariably fatal neurodegenerative disease [2]. It entered the spotlight in the 1990s when the outbreak of the bovine spongiform encephalopathy and the variant CJD transmitted to humans led to the initiation of international surveillance programs for CJD worldwide [3,4,5].
The annual incidence is around 1–2/1 million inhabitants but increases with age [2]. However, national surveillance studies drew attention to an increase in the incidence of prion diseases, although it is not clear whether this reflects an actual increase in the incidence of the diseases or increased awareness of physicians and improved availability of diagnostic tools [6,7]. Moreover, recent case reports linked the onset of CJD to infections with SARS-CoV-2 or vaccination against COVID-19 [8,9].
The disease belongs to a group of neurodegenerative conditions affecting humans and animals that include sporadic and genetic Creutzfeldt-Jakob disease, sporadic (considered a subtype of sporadic CJD) and familial fatal insomnia, Gerstmann-Sträussler-Scheinker disease, Kuru disease and variant CJD in humans; bovine spongiform encephalopathy (“mad cow disease”) in cattle; scrapie in sheep; and the chronic wasting disease in mule deer and elk. The etiology of these diseases was elusive for many years and subject to diverse hypotheses. Fragments of nucleic acids, polysaccharides, or a protein have all been discussed as possible etiological agents, until Stanley Prusiner introduced the term “prion” to name the infectious proteinaceous particle and detailed the protein hypothesis [10,11], which, although initially greeted with skepticism, allowed him to win the Nobel Prize in Physiology or Medicine in 1997.
For a long time, the diagnosis was possible only post-mortem by showing the spongiform modifications induced in the brain, but in recent years, more sophisticated laboratory evaluations and brain imaging studies can aid in diagnosing the disease earlier [12]. Nonetheless, the diagnosis is often delayed by several months from the onset of the first symptoms [13], much to the distress of patients, who witness the rapid decline of their health and of their family members.
In our manuscript, we review the pathophysiology, clinical picture, diagnostic criteria, and differential diagnosis of CJD, highlighting recent therapeutic strategies evaluated in preclinical studies.

2. Sporadic Creutzfeldt-Jakob Disease—Overview of the Current Knowledge on Etiology, Pathogenesis, and Clinical Picture

2.1. Etiology

Currently, the etiology of CJD is considered a prion, or proteinaceous infectious particle (PrP), that takes on conformational changes and tends to spread in the central nervous system (CNS), leading to histopathologic changes.
The protein PrP is normally produced by all vertebrates cells and is highly conserved in mammals [14]. The human PrP gene (PRNP) is found on chromosome 20 and encodes a 253 amino acid protein. After removal of the C- and N-terminal signal peptides, the mature 208 amino acid protein (PrP23-231) is guided by chaperones into the endoplasmic reticulum and Golgi apparatus to fold and undergo post-translational modifications such as attachments of glycans or the addition of a glycophosphatidylinositol anchor to the C-terminus [15]. The cellular prion protein (PrPC) is expressed in most tissues of the body but mostly in the CNS, particularly in neurons [16]. Its functions are still elusive [17]; roles in cell adhesion and signaling, in copper metabolism [18], or the binding and transport of nucleic acids have been suggested [19]. Recently, it has been shown that PrPC interacts with and modulates a large number of membrane receptors (especially glutamate receptors), ion channels, and amino acid transporters [20,21]. Due to various reasons, the normal host protein undergoes a templated polymerization, igniting a series of pathogenic cascades and activating astrocytes and microglia, culminating in histopathological spongiform changes and rapid neuronal loss.

2.2. Pathogenesis

The key events triggering PrPSc formation are still incompletely elucidated. External factors (oxidative stress, inflammation, or age), as well as pathogenic PRNP mutations, may all be involved [22], followed by a failure of the cellular proteostasis machinery in clearing the misfolded protein [23]. Nuclear magnetic resonance imaging studies in recombinant mouse PrP protein have shown that normal cellular PrP is structurally about 40% alpha helix and 3% beta-sheet, while analyses performed on the altered protein from sheep with scrapie revealed a significantly altered conformation, with 45% beta-sheet and 30% alpha helix [24], making the aggregated protein resistant to enzymatic digestion. Genetic factors (several polymorphisms of the PRNP gene) might promote the conversion of PrPC into PrPSc [25].
In the case of orally or transcutaneously transmitted prion diseases, it is still debated how prions reach the CNS. One possibility would be hematogenous spread via immune cells and lymphoid organs [26], as demonstrated by the transmission of bovine spongiform encephalopathy by blood transfusion in sheep [27]. PrPC is expressed on cell surfaces of T lymphocytes [28], macrophages, natural killer cells, regulatory T cells, and dendritic cells [29] and has important roles in the organization of lymphoid tissues, such as lymph nodes and the spleen [30]. When PrPSc infects the host, it uses the immune cells to replicate, converts PrPC to PrPSc, and accumulates [29,31]; after which, it spreads to the CNS via B cells [32], follicular dendritic cells [33], or other dendritic cell populations [34].
Another route for reaching the CNS would be from the gut via the parasympathetic fibers of the vagus nerve [35] following modifications of the gut microbiota, similar to Parkinson’s disease, where the abnormally folded α-synuclein may reach the CNS via parasympathetic fibers and spread in the CNS in a prion-like fashion [36].
Alternatively, infections with neurotropic strains of viruses, such as influenza viruses [37], SARS-CoV-2 [8,38], or vaccination against COVID-19 [9], may induce conformational changes in PrPC to form PrPSc, which then spreads to neighboring neurons through different mechanisms such as exosomes [39], receptor-mediated internalization [40], or nanotubes [41]. However, the expression of PrPC is required for prion formation and replication, as shown by the resistance to the disease of mice devoid of PrPC [42].
Growing evidence points toward seeding nucleation as the mechanism leading to prion aggregation, during which, PrPSc oligomers convert PrPC monomers and incorporate them into protofibrils and fibrils [43]. Fibril fragmentation can create new nucleation sites and, by incorporating new PrPC monomers, amplify the reaction [44]. PrPC being anchored by glycophosphatidylinositol (GPI) in the lipid rafts of the cellular membranes can act as a receptor and concentrate PrPSc molecules on cell surfaces, thereby compromising the cellular integrity and inducing neurotoxic effects [45]. In addition, during the conversion phase, PrPC twists along one side of the fibril while being membrane-anchored by GPI, leading to membrane distortion and cellular damage [46]. Internalized PrPSc aggregates can block the ubiquitin–proteasome system and activate the unfolded protein response signaling pathway [47], leading to impaired protein synthesis, with consequent synaptic dysfunction and neuronal loss, as well as the deacetylation and degradation of PIKfyve (a FYVE finger-containing phosphoinositide kinase), resulting in a reduction of phosphoinositide diphosphate levels. The latter causes impairment in the maturation of endosomes, leading to enlarged endosomes that may become intracellular vacuoles [48]. Following shedding from the plasma membrane, it is cleaved by ADAM10 (A disintegrin and metalloproteinase domain-containing protein 10), resulting in N- and C-terminal fragments that retain biological activity [25].
Both neurons and glial cells are involved in the pathological processes. Microglial cells attempt to clear PrPSc but also release inflammatory cytokines that ignite neuronal apoptosis [15], while astroglia is also activated and polarizes to the A1 neurotoxic phenotype, supplementally contributing to neuronal damage [49]. Even oligodendrocytes exhibit impaired energy metabolism and solute transport caused by altered gene transcription, as shown by the significantly reduced levels of mRNAs for galactosylceramidase, monocarboxylic acid transporter 1, and solute carrier family 2 member 1: glucose transporter member 1: GLUT1 [50].

2.3. Classification

Creutzfeldt-Jakob disease can be classified into [15]:
-
sporadic CJD, accounting for more than 80% of cases, thought to be due to unknown cellular events that lead to the conversion of PrPC to PrPSc,
-
genetic CJD, about 10–15% of cases, linked to a series of mutations in the PRNP gene transmitted in an autosomal-dominant pattern with variable penetrance [25], and
-
acquired CJD, caused by human-to-human transmission via surgical or medical procedures (corneal transplants, dural grafts, administration of human pituitary-derived gonadotrophins, and human-derived growth hormone) [51,52,53].
Another classification, at the histo-molecular level is based on the PRNP polymorphism at codon 129 (methionine—M or valine—V) and the physicochemical properties of the PrPSc strain after limited proteinase K digestion (type 1 cleaved at residue 82 and migrating at 21 kDa and type 2 cleaved at residue 97 and having a molecular mass of 19 kDa) [54,55]. Initially, six molecular groups were identified, each correlating with clinical and neuropathological phenotypes of sporadic CJD: MM1, MV1, VV1, MM2, MV2, and VV2. Subsequently, the MM2 group has been separated into a “cortical” and a “thalamic” variant (MM2C and MM2T, respectively), while, in the MV2 group, a subtype with cerebellar kuru-type amyloid plaques, termed the MV2C subtype, has been described [56]. A MM1 subtype showing widespread PrP-amyloid plaques in the white matter has been designated as p-MM1 [57], and because MM1 and MV1 are phenotypically virtually indistinguishable, they were merged into the MM(V)1 subtype [58]. MM1, VV2, and MV2 account for the large majority of cases [58]. Interestingly, PrPSc types 1 and 2 have been found to co-occur in the same brain specimens in about one-third of cases [58,59].

2.4. Neuropathology

The microscopic changes described in CJD consist of depositions of PrPSc aggregates, microglial activation, spongiform degeneration affecting various areas of the brain, neuronal loss, and astrocytic gliosis [58].

2.4.1. PrPSc Depositions

PrPSc deposits can be detected either isolated or in combination, due to the phenotypic heterogeneity of the disease.
  • Synaptic deposits are the most common ones, consisting of microgranular, diffuse PrPSc deposits identified in the cerebral cortex (mainly the occipital one) and in the molecular layer of the cerebellum.
  • Plaque-like deposits are rounded and well defined, described in the cerebral cortex, striatum, thalamus, granular layer of the cerebellar cortex, and cerebellar white matter, as well as, occasionally, in the cerebral white matter.
  • Perivacuolar deposits are found in the cerebral cortex, associated with large, confluent vacuoles.
  • Perineuronal deposits are seen in the pyramidal neurons of the fifth cortical layer and in the hippocampus, delineating the perikarya and dendrites of neurons.

2.4.2. Microglial Activation

Microglia are activated early in the disease course, attempting to clear the accumulated PrPSc but also favors neurodegeneration via neuroinflammation. The finding of numerous activated microglia resulting from the proliferation of resident cells early in the disease course in animal models was confirmed in human cases as well by showing widespread microgliosis in post-mortem sporadic CJD brains [60].

2.4.3. Spongiform Degeneration

Spongiform changes are characteristic features of CJD, although the vacuole size and distribution may vary, depending on disease subtype and stage. Three types of spongiform changes can be recognized [58]:
  • Small, round vacuoles 2–5 μm in diameter, located in the cerebral cortex, striatum, thalamus, and molecular layer of the cerebellar cortex, are most commonly found.
  • Large vacuoles, 15–20 μm in diameter, with a tendency to merge into grape-like structures, are often observed in the cerebral cortex.
  • Vacuoles of intermediate size are often seen in subcortical structures and the cerebellum.

2.4.4. Synaptic Loss

Normally, PrPC is concentrated in synaptic terminals, where it co-localizes with synaptophysin. As such, the conversion of PrPC into PrPSc contributes to synaptic dysfunction and loss. An increase in synaptic proteins, such as SNAP25, Rab3A, synaptotagmin 1, or clathrin, as a consequence of impaired synaptic vesicle release and re-uptake, has been convincingly demonstrated in CJD by several researchers [61,62].

2.4.5. Amyloid Plaques

About 10–15% of CJD patients have also compact amyloid plaques formed by PrPSc aggregates, located mainly in the cerebellar cortex, confined between the granular and Purkinje cell layers, although, in cases with a more prolonged disease course, these plaques may be found in the cerebral cortex as well [58]. They measure up to 30 μm, have a dense core surrounded by thin, amyloid radial bundles [63], and mainly characterize the MV2 subtype [64].

2.4.6. Secondary Tauopathy

A deposition of abnormal hyperphosphorylated tau aggregates has been documented in CJD as well, appearing as small, punctate, neuritic deposits (in about 16% of cases) or also associating with neuronal and glial tau pathology (84% of cases) [65]. Most often, tau pathology affects the VV2 and MV2K subtypes [66].

2.4.7. Astrocytic Gliosis

PrPSc accumulates in astrocytes early in the disease course, leading to their polarization towards the A1 phenotype. Reactive astrocytes have been found mainly in white matter but also in the thalamus, hypothalamus, and cerebellum of human CJD patients [67]. However, the findings of increased mRNA levels of markers of neuroprotective A2 astrocytes in the thalamus and hippocampus and of increased mRNAs for A1-specific markers in the hippocampus and cortex of prion-infected mouse brains suggest a heterogenous astrocytic response across different brain regions in prion diseases [68]. Moreover, different prion strains were associated with distinct patterns of astrocytic activation [69].
Research has shown that astrocytic activation precedes microglial activation and neurotransmission-associated disturbances [70], suggesting that, in the early stages of the disease, neuroprotective glial phenotypes prevail, while, in the later stages of prion infection, the neurotoxic ones are more widespread [71].
Although the role of glial cells in prion diseases is not yet fully understood and is the subject of active research [72], astrogliosis and neuronal loss are both observed in CJD, similar to other neurodegenerative diseases [73].

2.5. Clinical Picture

The diagnosis of sporadic CJD is challenging, because patients present with a wide range of non-specific symptoms and signs, but the disease is known as rapidly progressing dementia-associating visual and cerebellar signs, myoclonus, the patients develop rapidly akinetic mutism, and the mean survival is about 6 months [58].
The prodromal phase is usually overlooked, because patients exhibit behavioral changes, unspecified fear, eating abnormalities and weight loss, depressive mood changes, or may complain of sleeping disturbances [74,75].
Patients seek medical advice usually when cognitive functions start to decline, in the early stage of CJD. Cognitive impairment usually affects all domains. In a relatively frequent phenotype, previously known as the Heidenhain variant of CJD, visual symptoms are the presenting symptoms and consist of the blurring of vision and diplopia, later associated with complex visual hallucinations and progressing to cortical blindness [76]. In the phenotype known as the Brownell-Oppenheimer variant, cerebellar ataxia (mainly gait ataxia) appears first and may be the only neurological sign for several weeks [77]. More rarely, the first neurological signs may be a parkinsonian syndrome [78], hemiparesis mimicking a stroke [79], various types of epileptic seizures [80], or myoclonus, although the latter is considered to be a rather characteristic sign in CJD in later stages [81].
The disease progresses rapidly to significant cognitive impairment, associating other neurological signs with the presenting ones, such as dysarthria or loss of speech, and patients rather rapidly develop akinetic mutism [58]. Death occurs within weeks to months, although, in rare cases, survival may be possible for a few years [82].
With the identification of the molecular subtypes of CJD, it became clear that these subtypes have rather characteristic phenotypic presentations, pathology, and mean survival times, as shown in Table 1.

2.6. Evaluation

2.6.1. Laboratory Studies

Initial workup should include laboratory tests for dementia, such as serum chemistry panel, liver enzymes, ammonium levels, complete blood count, erythrocyte sedimentation rate (with blood cultures if any infection suspected), evaluation of the thyroid function, tests for neurosyphilis, and measurement of the B12 and folate levels, as well as serum levels of anti-thyroperoxidase antibodies to rule out Hashimoto encephalopathy.
If the patient’s history or general status and symptoms might be compatible with a paraneoplastic syndrome, specific antibodies should be searched (as detailed in the Section 2.8.
Routine CSF analysis is usually unrevealing, showing only a mild increase in the total protein levels in about one-third of cases [58].
A series of protein markers identified in the CSF have rather high sensitivity and specificity, being valuable aids in the diagnosis of CJD:
-
The family of 14-3-3 proteins was the first CSF biomarker to be used in diagnosing CJD. They are cytosolic proteins with regulatory functions, released into the CSF during neuronal damage. Although they may be increased in other conditions associated with neuronal destruction as well, the increase in 14-3-3 protein in the CSF shows an 87% sensitivity and 66% specificity if assessed semi-quantitatively by Western blotting, while assessment with ELISA increases the specificity to 84% [84].
-
The microtubule-associated protein tau (total tau—t-tau) as a marker of neuroaxonal degeneration is also largely used as a surrogate marker for the pre-mortem diagnosis of CJD [58,84], although it also increases in Alzheimer’s disease (AD) [85]. The cut-offs for differential diagnosis between CJD and AD are still not set, ranging between >1072 pg/mL [86] and >1583 pg/mL [87]. Because phosphorylated tau (p-tau) shows a less prominent increase in the CSF in CJD as compared to AD, the t-tau/p-tau ratio (cut-off < 0.075) [88] adds to the sensitivity (around 96%) and specificity (98–100%) [88]. In addition, the t-tau levels may suggest specific subtypes (being highest in the MM1, MV1, and VV2 types) [89] and could provide information related to prognosis and life expectancy [90].
-
Alpha-synuclein also shows significant increases, even more pronounced than in Parkinson’s disease or dementia with Lewy bodies [91] (proposed cut-off values being > 3300 pg/mL) [91], with a sensitivity of around 90% [92].
-
Neurofilament light (Nfl), a small subunit of a membrane cytoskeleton polymer, increases significantly in several neurodegenerative diseases, CJD included [93]. The proposed cut-off is 7000 pg/mL [94], with a sensitivity of around 85% [92].
-
YKL-40 (also known as chitinase 3-like 1), a marker of neuroinflammation, also shows an increase above the proposed cut-off of 315 ng/mL [95] in the CSF of patients with CJD, its sensitivity being around 76% [92], along with other markers of glial activation, such as CHIT-1 or GFAP (glial fibrillary acidic protein) [96,97].
-
Other biomarkers have been studied in recent years and shown to increase in sporadic CJD, such as neurogranin (related to synaptic plasticity) [98], ubiquitin (a marker of neuritic damage and neuroinflammation) [99], and calmodulin [100], as well as mitochondrial malate dehydrogenase 1 (a marker of mitochondrial dysfunction and oxidative stress) [101].
Because the assessment of these fluid biomarkers is invasive, requiring a lumbar puncture, the value of plasma biomarkers has been studied, but, unfortunately, they have a lower diagnostic accuracy. Increased plasma tau has a higher diagnostic accuracy compared to increases in plasma Nfl [102], but both are inferior to the evaluation of CSF total tau and 14-3-3 protein. Plasma microRNA studies have emerged as complementary tests in recent years [103].
Cell-free conversion assays based on the self-replicating ability of PrPSc can significantly aid in the diagnosis of prion disease. At the beginning of the third millennium, the protein misfolding cyclic amplification (PMCA) technique was used [104], requiring the mixing of suspected infected material homogenates (as source of PrPSc) with normal brain homogenate (source of PrPC), followed by repeated steps of incubation (allowing aggregate growth) and sonication (which fragmented the aggregates into smaller seeds to promote aggregate growth in the next incubation step). In the end, proteinase K (PK) treatment eliminated PrPC, and Western blotting revealed the residual PrPSc [105].
In 2008, a new technique, quaking-induced conversion (QuIC), was described [106], based on mixing bacterially expressed folded recombinant PrP with brain homogenates, followed by periods of orbital shaking and rest at controlled temperatures. Read-outs are performed by Western blotting after PK digestion. The method allowed for the detection of very low concentrations of PrPSc but was time-consuming.
A huge step forward in the early diagnosis of CJD was the development of the real-time quaking-induced conversion (RT-QuIC) assay, which indirectly detects PrPSc in the CSF, nasal fluids, or brain homogenate in concentrations as low as 10−13–10−15 g [107]. Supplementing the conversion medium with thioflavin T (ThT) allows for reading by using real-time ThT fluorescence measurements, which makes the assay faster and less expensive. The method has been integrated since 2017 into the International CJD Surveillance Network diagnostic criteria for CJD [108]. It has a high sensitivity (80–96%), although it tends to be lower in the MM1 subtype and the genetic forms of the disease, and a 99–100% specificity [105] but is expensive and less standardized compared to other surrogate biomarkers, such as 14-3-3 protein or t-tau [96].
RT-QuIC can also be performed in samples obtained from nasal brushing, which is less invasive than a lumbar puncture, with similar sensitivity and specificity (97% and 100%, respectively) [109]. However, since only preliminary data are available regarding the use of this diagnostic tool, and because a lumbar puncture would be needed for the screening of other neurodegenerative diseases, it is questionable whether RT-QuIC from nasal mucosa brushing samples will replace CSF RT-QuIC [96].

2.6.2. Imagistic Studies

The preferred imaging modality for suspected CJD is a MRI. Initially, the signal abnormalities are subtle but become more pronounced as the disease progresses and is associated with rapidly progressing cerebral atrophy.
At first, due to the low sensitivity of conventional MRI sequences in detecting cortical signal abnormalities, only abnormal signals in the caudate and putamen were discussed as potential diagnostic features [110]. In 2009, cortical signal hyperintensities in at least two lobes, except the frontal lobes on diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR), were added to the diagnostic criteria [111], while, in 2020, Bizzi and coworkers proposed the inclusion of signal abnormalities in the frontal cortex as well [112] while pointing out that the cingulate gyrus, insula, hippocampus, and cerebellum should be excluded, because they can be spontaneously hyperintense on the DWI of healthy individuals.
Specifically, abnormal signals appear usually bilaterally, but sometimes asymmetrically, in the [112,113]:
-
Cerebral cortex, affecting most commonly the insula, cingulate gyrus, or superior frontal gyrus; commonly the cuneus, precuneus, medial and/or inferior frontal gyri, occipital gyri, angular and supramarginal gyri, or superior parietal lobule (depending also on the clinical variant); and less commonly the medial and superior temporal gyri or the precentral and postcentral gyri
-
Basal ganglia, most commonly in the putamen and striatum but also in the thalamus
Depending on the phenotypic variants, the first signs may be detected [113]:
-
In the cerebellum, in the Brownell-Oppenheimer variant,
-
In the parietooccipital cortex in the Heidenhain variant,
-
In the putamen and thalamus in the Stern-Garcin variant, with extrapyramidal features from onset.
The most sensitive sequences are diffusion-weighted images (DWIs) [114], showing hyperintensities in the characteristic regions mentioned above, due to a combination of diffusion restriction (reduced diffusion of water caused by compartmentalization within vacuoles) or deposition of prion protein that restricts the free diffusion of water and to a high T2 signal that “shines through” to the DWI imaging (T2 shines through) [115]. The resultant signs are cortical ribboning, the pulvinar sign (hyperintensities in the thalamic pulvinar nuclei), and the hockey stick sign (hyperintensity of the medial thalamus) [116], previously thought to be more characteristic of the variant CJD. With disease progression, DWI signal intensity abnormalities may fade, likely due to neuronal loss and increased atrophy [117].
T2/FLAIR hyperintensities are more subtle than DWI changes and may be absent early in the disease course but are usually present in the same areas as DWI signal changes [115].
The ADC (apparent diffusion coefficient) aspect varies depending on the timing in the course of the disease. Initially, low values may be seen even before the DWI changes to be later replaced with normal intensity signals [115], probably related to neuronal loss and associated with prominent atrophy.
T1 sequences may, on occasion, show a high signal in the globus pallidus, while no abnormal enhancement can be detected after gadolinium administration [114].
The overall sensitivity of MRI abnormalities ranges between 69 and 92% [12,25,118], depending also on the expertise of the neuroradiologist. It can also suggest subtypes: basal ganglia hyperintensities are detected most often in VV2 and MV2 subtypes [12], while cortical ribboning characterizes mainly the MM subtypes [118].

2.6.3. Electroencephalography (EEG)

The EEG abnormalities depend on the disease stage. Initially, there may be only a non-specific, diffuse slowing background, later replaced by the characteristic symmetrical, generalized, bi-, triphasic, or mixed periodic sharp-wave complexes occurring with a frequency of about 1 cycle/second that are included in the World Health Organization diagnostic criteria. However, these findings tend to disappear in the late stages of CJD [119]. Their sensitivity ranges between 64 and 67%, with a 74–94% specificity [120]. In addition, other EEG abnormalities, such as convulsive and non-convulsive status epilepticus, have been reported in the literature [121,122], in which the EEG aspect may be difficult to discriminate from CJD but which responds to antiepileptic medication [25].

2.6.4. Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT)

PET using 18fluorine-fluorodeoxyglucose (18F-FDG) and other radiotracers can reveal cortical and subcortical hypometabolic areas and may suggest the presence of a specific CJD subtype but fails to differentiate between sporadic and genetic forms of CJD [123], while SPECT may identify hypoperfusion in the brain regions that exhibit DWI hyperintensities as well [124]. Although these methods could allow for further refinement of the imaging studies, they have not been incorporated into the diagnostic criteria and are just being explored.

2.7. Diagnostic Criteria

The World Health Organization proposed the following criteria for diagnosing sporadic Creutzfeldt-Jakob disease [125]:
(a)
Possible CJD:
  • progressive dementia
  • EEG atypical or not known
  • duration <2 years
  • At least two out of the following four clinical features:
    -
    myoclonus
    -
    visual or cerebellar disturbance
    -
    pyramidal/extrapyramidal dysfunction
    -
    akinetic mutism
(b)
Probable CJD: (in the absence of an alternative diagnosis from routine investigation)
  • progressive dementia
  • at least two of the following four clinical features:
    -
    myoclonus
    -
    visual or cerebellar disturbance
    -
    pyramidal/extrapyramidal dysfunction
    -
    akinetic mutism
  • a typical EEG, whatever the clinical duration of the disease
  • a positive 14-3-3 assay for CSF and a clinical duration to death <2 years
(c)
Confirmed (definite) CJD:
  • neuropathological confirmation
  • confirmation of protease-resistant prion protein (PrP) (immunocytochemistry or Western blot)
  • Presence of scrapie-associated fibrils
The European and UK criteria have incorporated characteristic MRI findings since 2009 [111] and were further refined in the subsequent updated guidelines. Currently, the 2017 revised diagnostic criteria for sporadic Creutzfeldt-Jakob disease [126] define:
A.
Definite sporadic CJD:
  • Progressive neurological syndrome
  • Neuropathologically or immunohistochemically or biochemically confirmed
B.
Probable sporadic CJD:
1.
Rapidly progressive cognitive impairment
  • Two of the following:
    -
    myoclonus
    -
    visual or cerebellar symptoms/signs
    -
    pyramidal or extrapyramidal features
    -
    akinetic mutism
  • and a typical EEG showing generalized periodic complexes
2.
Rapidly progressive cognitive impairment
  • Two of the following:
    -
    myoclonus
    -
    visual or cerebellar symptoms/signs
    -
    pyramidal or extrapyramidal features
    -
    akinetic mutism
  • and a typical MRI brain scan, showing a high signal in the caudate/putamen or at least two cortical regions (temporal, parietal, or occipital) on either the DWI or FLAIR
3.
Rapidly progressive cognitive impairment
  • Two of the following:
    -
    myoclonus
    -
    visual or cerebellar symptoms/signs
    -
    pyramidal or extrapyramidal features
    -
    akinetic mutism
  • and positive CSF 14-3-3 protein
  • and without routine investigations indicating an alternative diagnosis
4.
Progressive neurological syndrome and positive RT-QuIC in CSF or other tissues
C.
Possiblesporadic CJD:
  • Rapidly progressive cognitive impairment
  • Two of the following:
    -
    myoclonus
    -
    visual or cerebellar symptoms/signs
    -
    pyramidal or extrapyramidal features
    -
    akinetic mutism
and duration < 2 years
and the absence of a positive result for any of the four tests that would classify the case as “probable”
and without routine investigations indicating an alternative diagnosis
These criteria have a 92% sensitivity for detecting probable sporadic CJD, rising to almost 98% when all investigations are performed [12]. However, not every patient can undergo extensive evaluation, and biomarker testing has inherent limitations [127], which is why alternative diagnoses should not be ruled out easily [128].

2.8. Differential Diagnosis

2.8.1. Clinical Differential Diagnosis

Because the clinical presentation of CJD is quite heterogeneous, the differential diagnosis is exhaustive, and every effort should be made to identify the conditions mimicking CJD that are amenable to treatment.
(A) Other neurodegenerative diseases, such as Alzheimer’s disease, frontotemporal dementia, dementia with Lewy bodies, corticobasal degeneration, and multiple system atrophy, usually do not evolve as rapidly as CJD, although, on occasion, these conditions may exhibit an accelerated course. In addition, drug-related adverse events may lead to symptoms that might suggest CJD (tricyclic antidepressants or lithium leading to myoclonus and antiepileptics causing ataxia), or infections, metabolic disturbances, or various toxins may cause delirium in patients already cognitively impaired by an underlying neurodegenerative condition and mimic a rapid progression [129]. This is why a thorough history regarding recent changes in drugs and lab tests to monitor for metabolic disturbances or infections is advocated. Nevertheless, these conditions may be more similar to CJD than previously thought, since research has convincingly shown that the characteristic abnormally folded pathological protein propagates from cell to cell in a prion-like manner [36,130,131], and the possibility of iatrogenic transmission of Alzheimer’s diseases has been shown recently [132].
(B) Cerebrovascular conditions, either strategically placed stroke, multiple infarcts, or dural arteriovenous fistulas, and sinus thrombosis, as well as hypertensive encephalopathy, can cause rapidly progressing dementia, but usually, imaging may indicate the correct diagnosis.
(C) Infections of the central nervous system (herpes simplex encephalitis or Japanese encephalitis) usually occur in younger patients and are associated with fever and nuchal stiffness, as well as acute onset. Tertiary-stage neurosyphilis, with an increasing incidence in many developed countries [133], may also cause ataxia, movement disorders, and even psychosis, but serologic testing confirms the diagnosis. Lyme disease, caused by the Borrelia burgdorferi spirochete, also affects skin, joints, heart, and peripheral nervous system, aside from causing encephalomyelitis [129]. Opportunistic CNS infections in immunocompromised patients (toxoplasmosis in HIV-infected patients, cryptococcal meningitis, or progressive multifocal leukoencephalopathy caused by reactivation of the JC virus), as well as HIV-associated neurocognitive disorder, may also present as rapid progressive dementias in a considerable percentage of patients [129].
(D) Many toxic-metabolic etiologies of rapid progressive dementias are reversible if proper treatment is started early. Lithium may induce ataxia, tremor, myoclonus, or cognitive disturbances, even if the serum levels are within therapeutic ranges in patients with infections, dehydration, or renal impairment [134]. Lead poisoning, especially exposure to organic lead (gasoline additive), may adversely affect the limbic system and cause agitation, hallucinations, and altered sleep [135]. Thiamine deficiency (Wernicke-Korsakoff) syndrome should always be considered, not only in alcoholic patients but in every condition with the potential of causing malnutrition, such as gastric bypass surgery or repeated vomiting [136]. Some of the characteristic MRI findings may overlap those seen in CJD (detailed below), but urgent thiamine treatment can reverse the symptoms. Hypoglycemia, especially if accompanied by seizures, may result in a clinical picture compatible with CJD, and even MRI studies can show restricted diffusion in the cortex and basal ganglia but with relative sparing of the thalamus [137]. Hepatic encephalopathy can occur in both acute and chronic liver failure and manifest with a wide spectrum of neuropsychiatric symptoms, as well as movement disorders [138]. Especially, acute forms can show even overlapping MRI abnormalities, such as FLAIR and DWI cortical ribboning (usually sparing the perirolandic and occipital areas), but usually associated white matter hyperintensities in the internal capsule and subcortical white matter [139]. Rapid shifts in sodium can lead to extrapontine myelinolysis, presenting with encephalopathy and extrapyramidal syndromes, while the T2-weighted and DWI imaging may show hyperintensities in the basal ganglia, features resembling those seen in CJD [129], but these abnormalities usually disappear over weeks to months.
(E) A series of rare malignant conditions may pose serious challenges in the differential diagnosis of CJD. Primary CNS lymphoma, a diffuse B-cell non-Hodgkin’s lymphoma with an aggressive course and rising incidence [140], leads to a rapidly progressive cognitive decline, but the MRI shows usually enhancing lesions in contact with the cerebrospinal fluid [141]. Intravascular B-cell lymphoma is an even rarer condition and notoriously difficult to diagnose, leading to seizures, rapid cognitive decline, and upper and lower motor neuron symptoms. Again, imaging findings rather suggest a CNS vasculitis, with multiple T2/FLAIR hyperintensities in the white matter. Since the prominent neuronal damage may cause increases in 14-3-3 protein, RT-QuIC may help differentiate these cases from CJD, although the prognosis is significantly worse for the CNS lymphomas [142].
(F) Immune-mediated encephalitis. A series of antibody-mediated inflammatory disorders of the CNS, occurring either as paraneoplastic syndromes or as idiopathic conditions, may closely mimic CJD but respond very well to immune therapies. As such, every effort should be directed at identifying them [141]. While, in paraneoplastic syndromes, the antibodies are usually directed against intracellular antigens, more recently, a series of autoimmune encephalopathies with antibodies directed against cell-surface neuronal receptors or synaptic proteins [143] have been described, which are associated with underlying neoplasms in a variable proportion of cases. The onset is usually with psychiatric disturbances, but associated movement disorders (such as facial-brachial dystonic seizures) and the finding of CSF pleocytosis and MRI features suggestive of encephalitis, with a propensity for involvement of the limbic lobe, may aid in diagnosis [143]. Serum and CSF should be evaluated for antibody titers, although these may be initially negative in up to 50% of cases [144]. Table 2 provides an overview of the antibodies described in a series of autoimmune encephalopathies and their targets, as well as the incidence of underlying cancers and the most common neoplasia type.

2.8.2. Imagistic Differential Diagnosis

Several other conditions may show resembling imagistic abnormalities, as shown in Table 3.

3. Therapeutic Attempts and Future Perspectives

Currently, there are no treatments approved for prion disease. Therapeutic strategies focus on maintaining quality of life and on symptomatic treatment.
Because sensory stimuli (light, noise, touch, or movement) may cause distress to patients with visual hallucinations or precipitate myoclonus, the patient may benefit from being placed in a quiet environment, with soft lighting, and the number of visitors should be limited [174].
Treatment of neuropsychiatric symptoms with benzodiazepines in patients already exhibiting ataxia may be a concern and place them at risk for falls; hence, selective serotonin or norepinephrine reuptake inhibitors (SSRIs and SNRIs) and trazodone should be preferred. In patients with extrapyramidal symptoms, typical antipsychotics should be avoided to minimize the risk of exacerbation. If hallucinations are distressing to the patient, an atypical antipsychotic (quetiapine) could be tried in low doses [174].
The caregiver burden should not be overlooked, as they are often worried by the genetic etiology, lack of education regarding transmissibility, and are unfamiliar with the disease progression [25,175].

3.1. Assessed Pharmacological Agents

A series of pharmacological agents have undergone formal clinical trials but with negative results.
-
Flupirtine, a triaminopyridine shown in vitro to protect neurons from apoptosis caused by amyloid beta peptides and prion protein fragments, was evaluated in a study focusing on cognitive decline in patients with CJD. Although the rate of progression of dementia was slowed in the 13 patients randomized to flupirtine compared to the 15 patients randomized to the placebo, the overall survival rate was not influenced [25,176].
-
Quinacrine (300 mg/day), an antimalarial drug that supposedly could prevent the conversion of PrP to disease-associated protein forms, was assessed in two clinical trials: an open-label trial in the UK (PRION1) and a double-blind, placebo-controlled, stratified-randomization treatment trial in the US [177]. Unfortunately, no difference in mortality rates could be demonstrated in either trial [178,179], despite a transient improvement in symptoms at the beginning of the treatment [180].
-
Pentosan polysulfate has poor penetrance across the blood–brain barrier and had to be delivered via intraventricular injection. Although the survival was prolonged in observational studies, no symptomatic clinical benefit was discerned [181], and the aggressive delivery procedure was followed by many complications, mainly subdural effusions [182].
-
Doxycycline, a tetracycline antibiotic with good blood–brain barrier penetration and shown to inhibit the aggregation of PrP proteins and reverse the protease resistance of PrPSc [183], had promising results in animal experiments [184], but in the clinical setting, it also failed to prolong survival [185], except when administered in the early stages [186].

3.2. Future Directions

Our increasing understanding of the disease at the molecular level has highlighted novel therapeutic directions. Considering the crucial role of the abnormally folded PrPSc in CJD pathogenesis, pharmacologically stabilizing the folded PrPC and preventing its conversion to the disease-associated isoform could interfere with the pathogenic cascades [187].

3.2.1. Active Immunization

Neuroinflammation has been increasingly shown to be involved in the pathogenesis of various neurodegenerative diseases [44,188,189], CJD included. As such, the possibility of immunization against CJD has been evaluated.
The conversion of PrPC to PrPSc most likely occurs on the plasma membrane, and the misfolded protein enters the cytoplasm through endocytosis. T cells are activated only via simultaneous activation of the T-cell receptor (TCR) and CD28. To prevent excessive activation, T cells express CTLA-4 (cytotoxic T lymphocyte-associated antigen 4), which competes with CD28 in the process of T-cell activation, as does PD1 (programmed cell death 1). Prions promote CTLA-4 expression, thereby inducing immune tolerance [187].
To overcome immune tolerance, several approaches have been pursued:
-
The administration of truncated, dimeric, or cross-linked PrP peptides has been shown to elicit strong immune responses in mice [190].
-
DNA vaccines encoding specific PrP sequences can enhance the immune response [191], although their efficacy in preventing prion diseases is questionable [192].
-
Using bacterial or viral vectors may bypass immune tolerance, as does mucosal vaccination [193].
-
Finally, the identification of exposed regions of the misfolded protein allowed for the development of vaccines targeting these regions [194], with robust immune responses but, again, with less clear efficacy [195].
However, the widespread expression of PrP under physiological conditions and some toxicological concerns raised by these vaccines makes the overall applicability of these strategies questionable [25].

3.2.2. Monoclonal Antibodies

Therapeutic strategies aiming at reducing the pathogenic PrPSc could lead to strain selection. Mice treated with quinacrine had transiently reduced levels of PrPSc; after which, the latter recovered rapidly, possibly because the drug reduced certain subsets of conformers, but other prion protein conformations continued to spread [196].
The use of anti-PrP monoclonal antibodies was supported by both in vitro and in animal experiments [197,198], leading to a first clinical trial with an intravenous humanized monoclonal antibody to cellular prion protein (PRN100) in six CJD patients compared to historical untreated patients [199]. Although well tolerated and effective in clearing the abnormal protein (as shown by the report of the two autopsied cases), no influence on the clinical progression could be demonstrated [199], a scenario very similar to the results of the studies with the anti-Aβ antibodies lecanemab, donanemab, and aducanumab for Alzheimer’s disease [200]. Moreover, some anti-PrP antibodies proved neurotoxic due to the formation of a R208-H140 hydrogen bond (the H-latch) in the antibody molecule. Using PrP sequences unable to form this bond can neutralize the neurotoxicity [201].

3.2.3. Immune Modulation

As shown before, a series of immune checkpoints, such as PD-1, CTLA-4, or LAG-3 (lymphocyte activation gene 3), are involved in inducing immune tolerance and prevent the body’s own immune system from clearing the abnormal protein. Immune checkpoint inhibitors are already used in certain subtypes of cancer [202], and attempts have been made to use them in CJD as well. However, neither a PD-1 nor LAG-3 blockade had a significant influence on PrPSc deposition or the disease course in animal experiments [203,204], suggesting that these molecules will not reproduce in neurodegenerative diseases the success seen in cancer [205].

3.2.4. Gene Therapy

Gene therapy could be a promising alternative for carriers of the PRNP mutation, in which symptoms occur in late adulthood and early therapeutic intervention could delay (or even prevent) the progression of the disease.
Antisense oligonucleotides (ASOs) are single-stranded synthetic nucleotides that bind to a target mRNA and prevent the transcription of the target protein via RNase H degradation of the RNA–ASO complex [206]. ASOs directed against the PRNP gene administered intraventricularly in mouse models have been shown to reduce the deposition of PrPSc and extend survival [207] with no significant side effects. As such, prophylactic ASO treatment in carriers of the PRNP gene would be a viable alternative to prevent the onset of the disease [208].
Alternatively, RNA interference (RNAi) uses microRNAs (miRNAs), short interfering RNAs (siRNAs), or short hairpin RNAs (shRNAs) to “silence” the target gene [205]. Lentiviral-mediated RNAi was shown to reduce the expression of the pathogenic protein and prolong survival in mice [209].
PrP-Fc2 is a highly soluble and stable PrP dimer able to inhibit PrPSc replication. Lentiviral-mediated transfer of PrP-Fc2 to the brains of prion-infected mice has significantly extended survival rates, showing the potential of being developed as a prophylactic approach [210]. However, extensive research is still needed before translating these genetic therapies into clinical trials [187].

3.2.5. Targeted Protein Degradation Therapies

Targeted protein degradation strategies are appealing in most neurodegenerative diseases with the accumulation of abnormal and misfolded proteins, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and CJD. They use small molecules to increase ubiquitin–proteasome system-mediated protein degradation with proteasome-targeting chimeras (PROTACs) [211] or enhance lysosome-dependent protein degradation with lysosome-targeting chimeras (LYTACs) [212], autophagy-targeted chimeras (AUTACs) [213], or chaperone-mediated autophagy (CMA) [214]. Of the CMA-based degraders, the most studied is heat shock protein 70 (HSP70), which has been found to decrease with age, thereby increasing the risk for neurodegenerative diseases in the elderly [215]. The lack of HSP70 accelerates the progression of prion disease [216], while its overexpression has a neuroprotective effect [217]. However, issues relating to off-target effects, limited solubility and permeability across the blood–brain barrier, or metabolic instability must be improved before the clinical translation of these strategies [187].

3.2.6. Stem Cell Therapies

Stem cells can be used to repair neurons damaged by the misfolded prions. They can be classified into embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and neuronal stem cells (NSCs) [218]. Intracerebral transplantation of fetal neural stem cells in animal models of prion disease prolonged both the incubation period and survival time [219], while nasal delivery of MSCs reduced neuroinflammation without impacting survival [220]. Despite exciting new developments in the area of stem cell research [221], clinical translation is still hampered by ethical issues and the risk of immune rejection and tumorigenesis [218].

4. Concluding Remarks: Challenges in the Quest for a Cure for Prion Diseases

Despite significant efforts in this area, there is currently no available therapy for prion diseases. As in most neurodegenerative diseases, patient studies miss the earliest stages of disease development, limiting opportunities for experimental interventions. In research on animal models, difficulties arise from an inability to recreate human-specific processes. The development of induced pluripotent stem cell (iPSC) technology has expanded the range of available human tissue models by enabling the design of three-dimensional structures (cerebral organoids) that can be modeled to reproduce the various subtypes of CJD [222].
Specifically, in prion disease, research is hampered by a series of factors, such as:
  • The low incidence of prion disease (about 1–2/1 million persons or 5/1 million persons aged over 65) poses serious difficulties in designing clinical trials. This could be overcome by optimizing the exchange of information between researchers and clinicians and sharing diagnostic tools and protocols, thereby facilitating data collection and recruitment of subjects [177].
  • The first clinical signs appear late in the disease course when degeneration is already quite advanced. In addition, the clinical presentation being unspecific, diagnosing CJD can be quite challenging and time-consuming, aided by the rapid, sometimes fulminant course to akinetic mutism and death in a few months.
  • We still lack specific markers that could be used as primary endpoints in clinical trials. To date, most of the trials were considered successful if they prolonged life, but the disease duration varies naturally in the various genetic subtypes. Moreover, given the expanding area of stem cell and organ/tissue transplantation, the need for testing donors with reliable markers for asymptomatic prion disease is even more stringent to avoid iatrogenic transmission.
  • From animal experiments, we know that a series of compound-resistant PrPSc strains can develop following the administration of a therapeutic compound [223]. Two hypotheses have tried to explain this phenomenon: (1) the “cloud” hypothesis, which posits that a prion strain contains a mixture of conformational variants from which the best-suited one in a specific environment thrives and becomes the dominant one while the others are progressively eliminated from the cloud over multiple propagation cycles [224], and (2) the “deformed templating” hypothesis, which postulates that prion conversion is not always fully faithful, leading to subsequent generations of prions with heterogeneous conformations. The new conformation(s) may possess a selective advantage in a specific environment and ultimately may become the dominant conformational variant [225]. As such, in vitro models must consist of cells capable of being infected by a wide range of PrPSc strains.
Nonetheless, although in the early stages, the novel therapeutic targets highlighted by research findings and the transition from externally administered drugs to recruiting immune cells to combat the disease show promise for the future. In our opinion, gene therapy (ASOs and RNA interference) would be applicable in patients with the familial form of CJD who can undergo genetic testing if a family member has been diagnosed with the disease, and vaccines could apply to persons with occupational exposure to prion diseases or potentially also to caregivers of patients with CJD [226], while sporadic cases would benefit from enhanced protein degradation strategies or intrabody approaches [131]. It is worth remembering that cancer therapies were barely effective half a century ago, whereas, today, their efficiency has increased significantly [187].

Author Contributions

Conceptualization, M.C.J.; investigation, A.J. and V.S.N.; original draft preparation, R.G.D. and V.O.H.; review and editing, M.C.J.; supervision, A.J. and V.S.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were generated during the writing of this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ironside, W.J. Creutzfeldt-Jakob disease: The story so far. Proc. R. Coll. Physicians Edinb. 1998, 28, 143–149. [Google Scholar] [CrossRef] [PubMed]
  2. Watson, N.; Brandel, J.P.; Green, A.; Hermann, P.; Ladogana, A.; Lindsay, T.; Mackenzie, J.; Pocchiari, M.; Smith, C.; Zerr, I.; et al. The importance of ongoing international surveillance for Creutzfeldt-Jakob disease. Nat. Rev. Neurol. 2021, 17, 362–379. [Google Scholar] [CrossRef] [PubMed]
  3. The National CJD Research & Surveillance Unit. 31st Annual Report 2022. Creutzfeldt-Jakob Disease Surveillance in the UK. Available online: https://www.cjd.ed.ac.uk/sites/default/files/report31.pdf (accessed on 18 March 2024).
  4. Denouel, A.; Brandel, J.P.; Peckeu-Abboud, L.; Seilhean, D.; Bouaziz-Amar, E.; Quadrio, I.; Oudart, J.B.; Lehmann, S.; Bellecave, P.; Laplanche, J.L.; et al. Prospective 25-year surveillance of prion diseases in France, 1992 to 2016: A slow waning of epidemics and an increase in observed sporadic forms. Euro Surveill. 2023, 28, 2300101. [Google Scholar] [CrossRef] [PubMed]
  5. Maddox, R.A.; Person, M.K.; Blevins, J.E.; Abrams, J.Y.; Appleby, B.S.; Schonberger, L.B.; Belay, E.D. Prion disease incidence in the United States: 2003–2015. Neurology 2020, 94, e153–e157. [Google Scholar] [CrossRef]
  6. Sun, Y.; Liu, C.C.; Fan, L.Y.; Huang, C.T.; Chen, T.F.; Lu, C.J.; Guo, W.Y.; Chang, Y.C.; Chiu, M.J. Incidence of and Mortality Due to Human Prion Diseases in Taiwan: A Prospective 20-Year Nationwide Surveillance Study from 1998 to 2017. Clin. Epidemiol. 2020, 12, 1073–1081. [Google Scholar] [CrossRef]
  7. Kim, Y.C.; Jeong, B.H. Creutzfeldt-Jakob Disease Incidence, South Korea, 2001–2019. Emerg. Infect. Dis. 2022, 28, 1863–1866. [Google Scholar] [CrossRef]
  8. Bernardini, A.; Gigli, G.L.; Janes, F.; Pellitteri, G.; Ciardi, C.; Fabris, M.; Valente, M. Creutzfeldt-Jakob disease after COVID-19: Infection-induced prion protein misfolding? A case report. Prion 2022, 16, 78–83. [Google Scholar] [CrossRef]
  9. Attia, A.M.M.A.; Badr, B.A.; Albalaihad, R.; Alharshan, R.; Almukhaitah, A.A. Creutzfeldt-Jakob Disease After the Second Dosage of The Novel Pfizer-Biontech Messenger Ribonucleic Acid (Mrna) COVID-19 Vaccination: A Case Report. HIV Nurs. 2023, 23, 743–747. [Google Scholar]
  10. Prusiner, S.B.; Scott, M.R.; DeArmond, S.J.; Cohen, F.E. Prion protein biology. Cell 1998, 93, 337–348. [Google Scholar] [CrossRef]
  11. Prusiner, S.B. Novel proteinaceous infectious particles cause scrapie. Science 1982, 216, 136–144. [Google Scholar] [CrossRef]
  12. Watson, N.; Hermann, P.; Ladogana, A.; Denouel, A.; Baiardi, S.; Colaizzo, E.; Giaccone, G.; Glatzel, M.; Green, A.J.E.; Haïk, S.; et al. Validation of Revised International Creutzfeldt-Jakob Disease Surveillance Network Diagnostic Criteria for Sporadic Creutzfeldt-Jakob Disease. JAMA Netw. Open 2022, 5, e2146319. [Google Scholar] [CrossRef] [PubMed]
  13. Paterson, R.W.; Torres-Chae, C.C.; Kuo, A.L.; Ando, T.; Nguyen, E.A.; Wong, K.; DeArmond, S.J.; Haman, A.; Garcia, P.; Johnson, D.Y.; et al. Differential diagnosis of Jakob-Creutzfeldt disease. Arch. Neurol. 2012, 69, 1578–1582. [Google Scholar] [CrossRef] [PubMed]
  14. Windl, O.; Dempster, M.; Estibeiro, P.; Lathe, R. A candidate marsupial PrP gene reveals two domains conserved in mammalian PrP proteins. Gene 1995, 159, 181–186. [Google Scholar] [CrossRef] [PubMed]
  15. Baiardi, S.; Mammana, A.; Capellari, S.; Parchi, P. Human prion disease: Molecular pathogenesis, and possible therapeutic targets and strategies. Exp. Opin. Therap. Targets 2023, 27, 1271–1284. [Google Scholar] [CrossRef]
  16. Acevedo-Morantes, C.Y.; Wille, H. The structure of human prions: From biology to structural models—Considerations and pitfalls. Viruses 2014, 6, 3875–3892. [Google Scholar] [CrossRef]
  17. Mahabadi, H.M.; Taghibiglou, C. cellular prion protein (PrPc): Putative interacting partners and consequences of the interaction. Int. J. Mol. Sci. 2020, 21, 7058. [Google Scholar] [CrossRef]
  18. Thackray, A.M.; Knight, R.; Haswell, S.J.; Bujdoso, R.; Brown, D.R. Metal imbalance and compromised antioxidant function are early changes in prion disease. Biochem. J. 2002, 362, 253–258. [Google Scholar] [CrossRef]
  19. Lathe, R.; Darlix, J.L. Prion protein PrP nucleic acid binding and mobilization implicates retroelements as the replicative component of transmissible spongiform encephalopathy. Arch. Virol. 2020, 165, 535–556. [Google Scholar] [CrossRef]
  20. Wulf, M.A.; Senatore, A.; Aguzzi, A. The biological function of the cellular prion protein: An update. BMC. Biol. 2017, 15, 34. [Google Scholar] [CrossRef]
  21. Baral, P.K.; Yin, J.; Aguzzi, A.; James, M.G. Transition of the prion protein from a structured cellular form (PrPC) to the infectious scrapie agent (PrPSc). Prot. Sci. 2019, 28, 2055–2063. [Google Scholar] [CrossRef]
  22. Sweeney, P.; Park, H.; Baumann, M.; Dunlop, J.; Frydman, J.; Kopito, R.; McCampbell, A.; Leblanc, G.; Venkateswaran, A.; Nurmi, A.; et al. Protein misfolding in neurodegenerative diseases: Implications and strategies. Transl. Neurodegener. 2017, 6, 6. [Google Scholar] [CrossRef] [PubMed]
  23. Yedidia, Y.; Horonchik, L.; Tzaban, S.; Yanai, A.; Taraboulos, A. Proteasomes and ubiquitin are involved in the turnover of the wild-type prion protein. EMBO J. 2001, 20, 5383–5391. [Google Scholar] [CrossRef] [PubMed]
  24. Pan, K.M.; Baldwin, M.; Nguyen, J.; Gasset, M.; Serban, A.; Groth, D.; Mehlhorn, I.; Huang, Z.; Fletterick, R.J.; Cohen, F.E.; et al. Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins. Proc. Natl. Acad. Sci. USA 1993, 90, 10962–10966. [Google Scholar] [CrossRef] [PubMed]
  25. Zerr, I.; Ladogana, A.; Mead, S.; Hermann, P.; Forloni, G.; Appleby, B.S. Creutzfeldt-Jakob disease and other prion diseases. Nat. Rev. Dis. Primers 2024, 10, 14. [Google Scholar] [CrossRef]
  26. Kimberlin, R.H.; Walker, C.A. Pathogenesis of mouse scrapie: Dynamics of agent replication in spleen, spinal cord and brain after infection by different routes. J. Comp. Pathol. 1979, 89, 551–562. [Google Scholar] [CrossRef]
  27. Houston, F.; Foster, J.D.; Chong, A.; Hunter, N.; Bostock, C.J. Transmission of BSE by blood transfusion in sheep. Lancet 2000, 356, 999–1000. [Google Scholar] [CrossRef]
  28. Mattei, V.; Garofalo, T.; Misasi, R.; Circella, A.; Manganelli, V.; Lucania, G.; Pavan, A.; Sorice, M. Prion protein is a component of the multimolecular signaling complex involved in T cell activation. FEBS. Lett. 2004, 560, 14–18. [Google Scholar] [CrossRef]
  29. Cha, S.; Kim, M.Y. The role of cellular prion protein in immune system. BMB Rep. 2023, 56, 645–650. [Google Scholar] [CrossRef]
  30. Kim, S.; Han, S.; Kim, T.; Nam, J.; Kim, Y.S.; Choi, E.K.; Kim, M.Y. Prolonged follicular helper T cell responses in ME7 scrapie-infected mice. Prion 2018, 12, 109–116. [Google Scholar] [CrossRef]
  31. Zhang, B.; She, P.; Yin, X.; Dai, Y.; Ding, M.; Cui, L. Expression and functions of cellular prion proteins in immunocytes. Scand. J. Immunol. 2020, 91, e12854. [Google Scholar] [CrossRef]
  32. Klein, M.A.; Frigg, R.; Flechsig, E.; Raeber, A.J.; Kalinke, U.; Bluethmann, H.; Bootz, F.; Suter, M.; Zinkernagel, R.M.; Aguzzi, A. A crucial role for B cells in neuroinvasive scrapie. Nature 1997, 390, 687–690. [Google Scholar] [CrossRef] [PubMed]
  33. Montrasio, F.; Frigg, R.; Glatzel, M.; Klein, M.A.; Mackay, F.; Aguzzi, A.; Weissmann, C. Impaired prion replication in spleens of mice lacking functional follicular dendritic cells. Science 2000, 288, 1257–1259. [Google Scholar] [CrossRef] [PubMed]
  34. Harischandra, D.S.; Kondru, N.; Martin, D.P.; Kanthasamy, A.; Jin, H.; Anantharam, V.; Kanthasamy, A.G. Role of proteolytic activation of protein kinase Cd in the pathogenesis of prion disease. Prion 2014, 8, 143–153. [Google Scholar] [CrossRef] [PubMed]
  35. Kong, Y.; Chen, Z.; Feng, X.; Zuo, Y.; Zhang, J. Gut microbiota and metabolome in sporadic Creutzfeldt-Jakob disease. J. Neurol. 2023, 270, 6021–6032. [Google Scholar] [CrossRef] [PubMed]
  36. Jurcau, A.; Andronie-Cioara, F.L.; Nistor-Cseppento, D.C.; Pascalau, N.; Rus, M.; Vasca, E.; Jurcau, M.C. The Involvement of Neuroinflammation in the Onset and Progression of Parkinson’s Disease. Int. J. Mol. Sci. 2023, 24, 14582. [Google Scholar] [CrossRef]
  37. Hara, H.; Sakaguchi, S. Virus Infection, Genetic Mutations, and Prion Infection in Prion Protein Conversion. Int. J. Mol. Sci. 2021, 22, 12439. [Google Scholar] [CrossRef]
  38. Leccese, D.; Cornacchini, S.; Nacmias, B.; Sorbi, S.; Bessi, V. Creutzfeldt-Jakob Disease in a Patient with Previous COVID-19 Infection: “The Virus Caused the Derangement in My Brain”. J. Alzheimers Dis. Rep. 2023, 7, 129–134. [Google Scholar] [CrossRef]
  39. Fevrier, B.; Vilette, D.; Archer, F.; Loew, D.; Faigle, W.; Vidal, M.; Laude, H.; Raposo, G. Cells release prions in association with exosomes. Proc. Natl. Acad. Sci. USA 2004, 101, 9683–9688. [Google Scholar] [CrossRef]
  40. Aguzzi, A.; Akk, L. Cell biology of prions and prionoids: A status report. Trends Cell Biol. 2016, 26, 40–51. [Google Scholar] [CrossRef]
  41. Gousset, K.; Schiff, E.; Langevin, C.; Marijanovic, Z.; Caputo, A.; Browman, D.T.; Chenouard, N.; de Chaumont, F.; Martino, A.; Enninga, J.; et al. Prions hijack tunnelling nanotubes for intercellular spread. Nat. Cell Biol. 2009, 11, 328–336. [Google Scholar] [CrossRef]
  42. Bueler, H.; Aguzzi, A.; Sailer, A.; Greiner, R.A.; Autenried, P.; Aguet, M.; Weissmann, C. Mice devoid of PrP are resistant to scrapie. Cell 1993, 73, 1339–1347. [Google Scholar] [CrossRef]
  43. Cox, B.; Ness, F.; Tuite, M. Analysis of the generation and segregation of propagons: Entities that propagate the [PSI+] prion in yeast. Genetics 2003, 165, 23–33. [Google Scholar] [PubMed]
  44. Scheckel, C.; Aguzzi, A. Prions, prionoids and protein misfolding disorders. Nat. Rev. Genet. 2018, 19, 405–418. [Google Scholar] [CrossRef] [PubMed]
  45. Ntt, L.; Wu, B.; Harris, D.A. Prion neurotoxicity. Brain Pathol. 2019, 29, 263–277. [Google Scholar]
  46. Kraus, A.; Hoyt, F.; Schwartz, C.L.; Hansen, B.; Artikis, E.; Hughson, A.G.; Raymond, G.J.; Race, B.; Baron, G.S.; Caughey, B. High-resolution structure and strain comparison of infectious mammalian prions. Mol. Cell 2021, 81, 4540–4551. [Google Scholar] [CrossRef] [PubMed]
  47. Thellung, S.; Corsaro, A.; Dellacasagrande, I.; Nizzari, M.; Zambito, M.; Florio, T. Proteostasis unbalance in prion diseases: Mechanisms of neurodegeneration and therapeutic targets. Front. Neurosci. 2022, 16, 966019. [Google Scholar] [CrossRef]
  48. Lakkaraju, A.K.K.; Frontzek, K.; Lemes, E.; Herrmann, U.; Losa, M.; Marpakwar, R.; Aguzzi, A. Loss of PIKfyve drives the spongiform degeneration in prion diseases. EMBO Mol. Med. 2021, 13, e14714. [Google Scholar] [CrossRef]
  49. Ugalde, C.L.; Lewis, V.; Stehmann, C.; McLean, C.A.; Lawson, V.A.; Collins, S.J.; Hill, A.F. Markers of A1 astrocytes stratify to molecular sub-types in sporadic Creutzfeldt-Jakob disease brain. Brain Commun. 2020, 2, fcaa029. [Google Scholar] [CrossRef]
  50. Andres Benito, P.; Dominguez Gonzalez, M.; Ferrer, I. Altered gene transcription linked to astrocytes and oligodendrocytes in frontal cortex in Creutzfeldt-Jakob disease. Prion 2018, 12, 216–225. [Google Scholar] [CrossRef]
  51. Martheswaran, T.; Desautels, J.D.; Moshirfar, M.; Shmunes, K.M.; Ronquillo, Y.C.; Hoopes, P.C. A Contemporary Risk Analysis of Iatrogenic Transmission of Creutzfeldt-Jakob Disease (CJD) via Corneal Transplantation in the United States. Ophthalmol. Ther. 2020, 9, 465–483. [Google Scholar] [CrossRef]
  52. Hamaguchi, T.; Sakai, K.; Kobayashi, A.; Kitamoto, T.; Ae, R.; Nakamura, Y.; Sanjo, N.; Arai, K.; Koide, M.; Katada, F.; et al. Characterization of Sporadic Creutzfeldt-Jakob Disease and History of Neurosurgery to Identify Potential Iatrogenic Cases. Emerg. Infect. Dis. 2020, 26, 1140–1146. [Google Scholar] [CrossRef] [PubMed]
  53. Douet, J.Y.; Huor, A.; Cassard, H.; Lugan, S.; Aron, N.; Mesic, C.; Vilette, D.; Barrio, T.; Streichenberger, N.; Perret-Liaudet, A.; et al. Prion strains associated with iatrogenic CJD in French and UK human growth hormone recipients. Acta Neuropathol. Commun. 2021, 9, 145. [Google Scholar] [CrossRef] [PubMed]
  54. Baiardi, S.; Rossi, M.; Mammana, A.; Appleby, B.S.; Barria, M.A.; Calì, I.; Gambetti, P.; Gelpi, E.; Giese, A.; Ghetti, B.; et al. Phenotypic diversity of genetic Creutzfeldt-Jakob disease: A histo-molecular-based classification. Acta Neuropathol. 2021, 142, 707–728. [Google Scholar] [CrossRef] [PubMed]
  55. Parchi, P.; Zou, W.; Wang, W.; Brown, P.; Capellari, S.; Ghetti, B.; Kopp, N.; Schulz-Schaeffer, W.J.; Kretzschmar, H.A.; Head, M.W.; et al. Genetic influence on the structural variations of the abnormal prion protein. Proc. Natl. Acad. Sci. USA 2000, 97, 10168–10172. [Google Scholar] [CrossRef]
  56. Iwasaki, Y. The Braak hypothesis in prion disease with a focus on Creutzfeldt-Jakob disease. Neuropathology 2020, 40, 436–449. [Google Scholar] [CrossRef]
  57. Rossi, M.; Saverioni, D.; Di Barri, M.; Baiardi, S.; Lemstra, A.W.; Pirisinu, L.; Capellari, S.; Rozemuller, A.; Nonno, R.; Parchi, P. Atypical Creutzfeldt-Jakob disease with atypical PrP-amyloid plaques in white matter: Molecular characterization and transmission to bank voles show the m1 strain signature. Acta Neuropathol. Commun. 2017, 5, 87. [Google Scholar] [CrossRef]
  58. Zerr, I.; Parchi, P. Sporadic Creutzfeldt-Jakob disease. In Handbook of Clinical Neurology. Human Prion Diseases; 3rd series; Pocchiari, M., Manson, J., Eds.; Elsevier: Oxford, UK, 2018; Volume 153, pp. 155–174. [Google Scholar]
  59. Cali, I.; Puoti, G.; Smucny, J.; Curtiss, P.M.; Cracco, L.; Kitamoto, T.; Occhipinti, R.; Cohen, M.L.; Appleby, B.S.; Gambetti, P. Co-existence of PrPD types 1 and 2 in sporadic Creutzfeldt-Jakob disease of the VV subgroup: Phenotypic and prion protein characteristics. Sci. Rep. 2020, 10, 1503. [Google Scholar] [CrossRef]
  60. Franceschini, A.; Strammiello, R.; Capellari, S.; Giese, A.; Parchi, P. Regional pattern of microgliosis in sporadic Creutzfeldt-Jakob disease in relation to phenotypic variants and disease progression. Neuropathol. Appl. Neurobiol. 2018, 44, 574–589. [Google Scholar] [CrossRef]
  61. Wang, C.; Zhao, D.; Shah, S.Z.A.; Yang, W.; Li, C.; Yang, L. Proteome Analysis of Potential Synaptic Vesicle Cycle Biomarkers in the Cerebrospinal Fluid of Patients with Sporadic Creutzfeldt-Jakob Disease. Mol. Neurobiol. 2017, 54, 5177–5191. [Google Scholar] [CrossRef]
  62. Bentivenga, G.M.; Baiardi, S.; Mastrangelo, A.; Zenesini, C.; Mammana, A.; Polischi, B.; Capellari, S.; Parchi, P. Diagnostic and prognostic value of cerebrospinal fluid SNAP-25 and neurogranin in Creutzfeldt-Jakob disease in a clinical setting cohort of rapidly progressive dementias. Alzheimers. Res. Ther. 2023, 15, 150. [Google Scholar] [CrossRef]
  63. Jankovska, N.; Olejar, T.; Matej, R. Extracellular Amyloid Deposits in Alzheimer’s and Creutzfeldt–Jakob Disease: Similar Behavior of Different Proteins? Int. J. Mol. Sci. 2021, 22, 7. [Google Scholar] [CrossRef] [PubMed]
  64. Liberski, P.P. Amyloid plaques in transmissible spongiform encephalopathies (prion diseases). Folia Neuropathol. 2004, 42, 109–119. [Google Scholar] [PubMed]
  65. Kovacs, G.G.; Rahimi, J.; Ströbel, T.; Lutz, M.I.; Regelsberger, G.; Streichenberger, N.; Perret-Liaudet, A.; Höftberger, R.; Liberski, P.P.; Budka, H.; et al. Tau pathology in Creutzfeldt-Jakob disease revisited. Brain Pathol. 2017, 27, 332–344. [Google Scholar] [CrossRef]
  66. Lattanzio, F.; Abu-Rumeileh, S.; Franceschini, A.; Kai, H.; Amore, G.; Poggiolini, I.; Rossi, M.; Baiardi, S.; McGuire, L.; Ladogana, A.; et al. Prion-specific and surrogate CSF biomarkers in Creutzfeldt-Jakob disease: Diagnostic accuracy in relation to molecular subtypes and analysis of neuropathological correlates of p-tau and Aβ42 levels. Acta Neuropathol. 2017, 133, 559–578. [Google Scholar] [CrossRef] [PubMed]
  67. Tahir, W.; Thapa, S.; Schatzl, H.M. Astrocyte in prion disease: A double-edged sword. Neural Regen. Res. 2022, 17, 1659–1665. [Google Scholar]
  68. Makarava, N.; Chang, J.C.; Kushwaha, R.; Baskakov, I.V. Region-specific response of astrocytes to prion infection. Front. Neurosci. 2019, 13, 1048. [Google Scholar] [CrossRef]
  69. Bradford, B.M.; Wijaya, C.A.W.; Mabbott, N.A. Discrimination of prion strain targeting in the central nervous system via reactive astrocyte heterogeneity in CD44 expression. Front. Cell. Neurosci. 2019, 13, 411. [Google Scholar] [CrossRef]
  70. Makarava, N.; Chang, J.C.; Molesworth, K.; Baskakov, I.V. Region-specific glial homeostatic signature in prion diseases is replaced by a uniform neuroinflammation signature, common for brain regions and prion strains with different cell tropism. Neurobiol. Dis. 2020, 137, 104783. [Google Scholar] [CrossRef]
  71. Guijarro, I.M.; Garcés, M.; Andrés-Benito, P.; Marín, B.; Otero, A.; Barrio, T.; Carmona, M.; Ferrer, I.; Badiola, J.J.; Monzón, M. Assessment of glial activation response in the progress of natural scrapie after chronic dexamethasone treatment. Int. J. Mol. Sci. 2020, 21, 3231. [Google Scholar] [CrossRef]
  72. Smith, H.L.; Freeman, O.J.; Butcher, A.J.; Holmqvist, S.; Humoud, I.; Schätzl, T.; Hughes, D.T.; Verity, N.C.; Swinden, D.P.; Hayes, J.; et al. Astrocyte unfolded protein response induces a specific reactivity state that causes non-cell-autonomous neuronal degeneration. Neuron 2020, 105, 855–866. [Google Scholar] [CrossRef]
  73. Garcés, M.; Guijarro, M.I.; Vargas, A.; Badiola, J.J.; Monzón, M. Neuroglial patterns are shared by cerebella from prion and prion-like disorder affected patients. Mech. Ageing Dev. 2019, 184, 111176. [Google Scholar] [CrossRef] [PubMed]
  74. Yong, C.S.K.; Maniam, E.J.; Chang, C.W.L.; Lai, J.Y.; Ho, C.S.H. Case report: Creutzfeldt-Jakob disease presenting with anxiety symptoms in a COVID-19 post-infection patient. Front. Neurol. 2023, 14, 1239576. [Google Scholar] [CrossRef] [PubMed]
  75. El Sammak, S.; Attarian, H.; Taype-Roberts, C.; Standlee, J. REM sleep behavior disorder as a remote precedent to Creutzfeldt-Jakob disease. Neurol. Sci. 2022, 43, 5647–5649. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, S.L.; Zhang, L.Y.; Zhang, S.F.; Zhang, M.Y.; Zhu, M.; Dong, Q.; Wang, Q.S.; Han, X. Clinical and prognostic features of Heidenhain variant of Creutzfeldt-Jakob disease: A retrospective case series study. Eur. J. Neurol. 2022, 29, 2412–2419. [Google Scholar] [CrossRef]
  77. Pandey, S.; Garg, R.K.; Holla, V.V.; Kumar, N. A Case of Subacute Progressive Cerebellar Ataxia: Brownell-Oppenheimer Variant of Sporadic Creutzfeldt-Jakob Disease. Neurol. India 2021, 69, 217–218. [Google Scholar] [CrossRef]
  78. Tang, S.; Dou, X.; Zhang, Y. 18F-FP-CIT PET/CT in a case of probable sporadic Creutzfeldt-Jakob disease with parkinsonism as initial symptom. Prion 2022, 16, 91–94. [Google Scholar] [CrossRef]
  79. Bae, M.J.; Kang, I.H.; Hur, Y.R.; Hwang, K.H. Sporadic Creutzfeldt-Jakob Syndrome Misdiagnosed as Recurrent Stroke: A Case Report. Ann. Geriatr. Med. Res. 2021, 25, 133–136. [Google Scholar] [CrossRef]
  80. Cunha, I.A.; Gomes, I.; Gens, H.; Guimarães, S.; Martins, A.I.; Bento, C. Sporadic Creutzfeldt-Jakob disease presenting as epilepsia partialis continua and non-ictal nystagmus. Eur. J. Neurol. 2021, 28, 2456–2459. [Google Scholar] [CrossRef]
  81. Murphy, C.; Kang, C.D.; Hanif, B. Rapid cognitive decline and myoclonus in a 52-year-old woman. Cleve. Clin. J. Med. 2021, 88, 572–583. [Google Scholar]
  82. Tejedor-Romero, L.; López-Cuadrado, T.; Almazán-Isla, J.; Calero, M.; García López, F.J.; de Pedro-Cuesta, J. Survival Patterns of Human Prion Diseases in Spain, 1998-2018: Clinical Phenotypes and Etiological Clues. Front. Neurosci. 2022, 15, 773727. [Google Scholar] [CrossRef]
  83. Parchi, P.; Strammiello, R.; Notari, S.; Giese, A.; Langeveld, J.P.; Ladogana, A.; Zerr, I.; Roncaroli, F.; Cras, P.; Ghetti, B.; et al. Incidence and spectrum of sporadic Creutzfeldt-Jakob disease variants with mixed phenotype and co-occurrence of PrPSc types: An updated classification. Acta Neuropathol. 2009, 118, 659–671. [Google Scholar] [CrossRef] [PubMed]
  84. Senesi, M.; Lewis, V.; Varghese, S.; Stehmann, C.; McGlade, A.; Doecke, J.D.; Ellett, L.; Sarros, S.; Fowler, C.J.; Masters, C.L.; et al. Diagnostic performance of CSF biomarkers in a well-characterized Australian cohort of sporadic Creutzfeldt-Jakob disease. Front. Neurol. 2023, 14, 1072952. [Google Scholar] [CrossRef] [PubMed]
  85. Jack, C.R.; Bennett, D.A., Jr.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef] [PubMed]
  86. Li, Q.X.; Varghese, S.; Sarros, S.; Stehmann, C.; Doecke, J.D.; Fowler, C.J.; Masters, C.L.; Collins, S.J. CSF Tau supplements 14-3-3 protein detection for sporadic Creutzfeldt-Jakob disease diagnosis while transitioning to next generation diagnostics. J. Clin. Neurosci. 2018, 50, 292–293. [Google Scholar] [CrossRef] [PubMed]
  87. Hermann, P.; Haller, P.; Goebel, S.; Bunck, T.; Schmidt, C.; Wiltfang, J.; Zerr, I. Total and Phosphorylated Cerebrospinal Fluid Tau in the Differential Diagnosis of Sporadic Creutzfeldt-Jakob Disease and Rapidly Progressive Alzheimer’s Disease. Viruses 2022, 14, 276. [Google Scholar] [CrossRef]
  88. Llorens, F.; Karch, A.; Golanska, E.; Schmitz, M.; Lange, P.; Sikorska, B.; Liberski, P.P.; Zerr, I. Cerebrospinal Fluid Biomarker-Based Diagnosis of Sporadic Creutzfeldt-Jakob Disease: A Validation Study for Previously Established Cutoffs. Dement. Geriatr. Cogn. Disord. 2017, 43, 71–80. [Google Scholar] [CrossRef]
  89. Gmitterová, K.; Heinemann, U.; Krasnianski, A.; Gawinecka, J.; Zerr, I. Cerebrospinal Fluid Markers in the Differentiation of Molecular Subtypes of Sporadic Creutzfeldt-Jakob Disease. Eur. J. Neurol. 2016, 23, 1126–1133. [Google Scholar] [CrossRef]
  90. Llorens, F.; Rübsamen, N.; Hermann, P.; Schmitz, M.; Villar-Piqué, A.; Goebel, S.; Karch, A.; Zerr, I. A Prognostic Model for Overall Survival in Sporadic Creutzfeldt-Jakob Disease. Alzheimer’s Dement. 2020, 16, 1438–1447. [Google Scholar] [CrossRef]
  91. Schmitz, M.; Villar-Piqué, A.; Llorens, F.; Gmitterová, K.; Hermann, P.; Varges, D.; Zafar, S.; Lingor, P.; Vanderstichele, H.; Demeyer, L.; et al. Cerebrospinal Fluid Total and Phosphorylated α-Synuclein in Patients with Creutzfeldt–Jakob Disease and Synucleinopathy. Mol. Neurobiol. 2018, 56, 3476–3483. [Google Scholar] [CrossRef]
  92. Llorens, F.; Villar-Piqué, A.; Hermann, P.; Schmitz, M.; Calero, O.; Stehmann, C.; Sarros, S.; Moda, F.; Ferrer, I.; Poleggi, A.; et al. Diagnostic Accuracy of Prion Disease Biomarkers in Iatrogenic Creutzfeldt-Jakob Disease. Biomolecules 2020, 10, 290. [Google Scholar] [CrossRef]
  93. Kanata, E.; Golanska, E.; Villar-Piqué, A.; Karsanidou, A.; Dafou, D.; Xanthopoulos, K.; Schmitz, M.; Ferrer, I.; Karch, A.; Sikorska, B.; et al. Cerebrospinal fluid neurofilament light in suspected sporadic Creutzfeldt-Jakob disease. J. Clin. Neurosci. 2019, 60, 124–127. [Google Scholar] [CrossRef] [PubMed]
  94. Zerr, I.; Schmitz, M.; Karch, A.; Villar-Piqué, A.; Kanata, E.; Golanska, E.; Díaz-Lucena, D.; Karsanidou, A.; Hermann, P.; Knipper, T.; et al. Cerebrospinal fluid neurofilament light levels in neurodegenerative dementia: Evaluation of diagnostic accuracy in the differential diagnosis of prion diseases. Alzheimer’s Dement. 2018, 14, 751–763. [Google Scholar] [CrossRef] [PubMed]
  95. Llorens, F.; Thüne, K.; Tahir, W.; Kanata, E.; Diaz-Lucena, D.; Xanthopoulos, K.; Kovatsi, E.; Pleschka, C.; Garcia-Esparcia, P.; Schmitz, M.; et al. YKL-40 in the brain and cerebrospinal fluid of neurodegenerative dementias. Mol. Neurodegener. 2017, 12, 83. [Google Scholar] [CrossRef] [PubMed]
  96. Altuna, M.; Ruiz, I.; Zelaya, M.V.; Mendioroz, M. Role of Biomarkers for the Diagnosis of Prion Diseases: A Narrative Review. Medicina 2022, 58, 473. [Google Scholar] [CrossRef]
  97. Mok, T.H.; Mead, S. Preclinical Biomarkers of Prion Infection and Neurodegeneration. Curr. Opin. Neurobiol. 2020, 61, 82–88. [Google Scholar] [CrossRef]
  98. Blennow, K.; Diaz-Lucena, D.; Zetterberg, H.; Villar-Pique, A.; Karch, A.; Vidal, E.; Hermann, P.; Schmitz, M.; Ferrer Abizanda, I.; Zerr, I.; et al. CSF Neurogranin as a Neuronal Damage Marker in CJD: A Comparative Study with AD. J. Neurol. Neurosurg. Psychiatry 2019, 90, 846–853. [Google Scholar] [CrossRef]
  99. Abu-Rumeileh, S.; Oeckl, P.; Baiardi, S.; Halbgebauer, S.; Steinacker, P.; Capellari, S.; Otto, M.; Parchi, P. CSF Ubiquitin Levels Are Higher in Alzheimer’s Disease than in Frontotemporal Dementia and Reflect the Molecular Subtype in Prion Disease. Biomolecules 2020, 10, 497. [Google Scholar] [CrossRef]
  100. Chen, C.; Hu, C.; Zhou, W.; Chen, J.; Shi, Q.; Xiao, K.; Wang, Y.; Dong, X.-P. Calmodulin Level Is Significantly Increased in the Cerebrospinal Fluid of Patients with Sporadic Creutzfeldt-Jakob Disease. Eur. J. Neurol. 2021, 28, 1134–1141. [Google Scholar] [CrossRef]
  101. Zerr, I.; Villar-Piqué, A.; Schmitz, V.E.; Poleggi, A.; Pocchiari, M.; Sánchez-Valle, R.; Calero, M.; Calero, O.; Baldeiras, I.; Santana, I.; et al. Evaluation of Human Cerebrospinal Fluid Malate Dehydrogenase 1 as a Marker in Genetic Prion Disease Patients. Biomolecules 2019, 9, 800. [Google Scholar] [CrossRef]
  102. Abu-Rumeileh, S.; Baiardi, S.; Ladogana, A.; Zenesini, C.; Bartoletti-Stella, A.; Poleggi, A.; Mammana, A.; Polischi, B.; Pocchiari, M.; Capellari, S.; et al. Comparison between plasma and cerebrospinal fluid biomarkers for the early diagnosis and association with survival in prion disease. J. Neurol. Neurosurg. Psychiatry 2020, 91, 1181–1188. [Google Scholar] [CrossRef]
  103. Norsworthy, P.J.; Thompson, A.G.B.; Mok, T.H.; Guntoro, F.; Dabin, L.C.; Nihat, A.; Paterson, R.W.; Schott, J.M.; Collinge, J.; Mead, S.; et al. A Blood MiRNA Signature Associates with Sporadic Creutzfeldt-Jakob Disease Diagnosis. Nat. Commun. 2020, 11, 3960. [Google Scholar] [CrossRef] [PubMed]
  104. Saborio, G.P.; Permanne, B.; Soto, C. Sensitive detection of pathological prion protein by cyclic amplification of protein misfolding. Nature 2001, 411, 810–813. [Google Scholar] [CrossRef] [PubMed]
  105. Ascari, L.M.; Rocha, S.C.; Gonçalves, P.B.; Vieira, T.C.R.G.; Cordeiro, Y. Challenges and advances in antemortem diagnosis of human transmissible spongiform encephalopathies. Front. Bioeng. Biotechnol. 2020, 8, 585896. [Google Scholar] [CrossRef] [PubMed]
  106. Atarashi, R.; Wilham, J.M.; Christensen, L.; Hughson, A.G.; Moore, R.A.; Johnson, L.M.; Onwubiko, H.A.; Priola, S.A.; Caughey, B. Simplified ultrasensitive prion detection by recombinant PrP conversion with shaking. Nat. Methods 2008, 5, 211–212. [Google Scholar] [CrossRef]
  107. Wilham, J.M.; Orrú, C.D.; Bessen, R.A.; Atarashi, R.; Sano, K.; Race, B.; Meade-White, K.D.; Taubner, L.M.; Timmes, A.; Caughey, B. Rapid end-point quantitation of prion seeding activity with sensitivity comparable to bioassays. PLoS Pathog. 2010, 6, e1001217. [Google Scholar] [CrossRef]
  108. Figgie, M.P.J.; Appleby, B.S. Clinical Use of Improved Diagnostic Testing for Detection of Prion Disease. Viruses 2021, 13, 789. [Google Scholar] [CrossRef]
  109. Behaeghe, O.; Mangelschots, E.; De Vil, B.; Cras, P. A Systematic Review Comparing the Diagnostic Value of 14-3-3 Protein in the Cerebrospinal Fluid, RT-QuIC and RT-QuIC on Nasal Brushing in Sporadic Creutzfeldt-Jakob Disease. Acta Neurol. Belg. 2018, 118, 395–403. [Google Scholar] [CrossRef]
  110. Tschampa, H.J.; Kallenberg, K.; Urbach, H.; Meissner, B.; Nicolay, C.; Kretzschmar, H.A.; Knauth, M.; Zerr, I. MRI in the diagnosis of sporadic Creutzfeldt-Jakob disease: A study on inter-observer agreement. Brain 2005, 128, 2026–2033. [Google Scholar] [CrossRef]
  111. Zerr, I.; Kallenberg, K.; Summers, D.M.; Romero, C.; Taratuto, A.; Heinemann, U.; Breithaupt, M.; Varges, D.; Meissner, B.; Ladogana, A.; et al. Updated clinical diagnostic criteria for sporadic Creutzfeldt-Jakob disease. Brain 2009, 132, 2659–2668. [Google Scholar] [CrossRef]
  112. Bizzi, A.; Pascuzzo, R.; Blevins, J.; Grisoli, M.; Lodi, R.; Moscatelli, M.E.M.; Castelli, G.; Cohen, M.L.; Schonberger, L.B.; Foutz, A.; et al. Evaluation of a New Criterion for Detecting Prion Disease with Diffusion Magnetic Resonance Imaging. JAMA Neurol. 2020, 77, 1141–1149. [Google Scholar] [CrossRef]
  113. Fragoso, D.C.; Gonçalves Filho, A.L.; Pacheco, F.T.; Barros, B.R.; Aguiar Littig, I.; Nunes, R.H.; Maia Júnior, A.C.; da Rocha, A.J. Imaging of Creutzfeldt-Jakob Disease: Imaging Patterns and Their Differential Diagnosis. Radiographics 2017, 37, 234–257. [Google Scholar] [CrossRef] [PubMed]
  114. Sacco, S.; Paoletti, M.; Staffaroni, A.M.; Kang, H.; Rojas, J.; Marx, G.; Goh, S.Y.; Luisa Mandelli, M.; Allen, I.E.; Kramer, J.H.; et al. Multimodal MRI staging for tracking progression and clinical-imaging correlation in sporadic Creutzfeldt-Jakob disease. Neuroimage Clin. 2021, 30, 102523. [Google Scholar] [CrossRef] [PubMed]
  115. Gaillard, F.; Petrovic, A.; Bell, D. T2 Shine Through. Reference Article, Radiopaedia.org. Available online: https://radiopaedia.org/articles/t2-shine-through?lang=us (accessed on 18 April 2024).
  116. Manara, R.; Fragiacomo, F.; Ladogana, A.; Vaianella, L.; Camporese, G.; Zorzi, G.; Vicinanza, S.; Zanusso, G.; Pocchiari, M.; Cagnin, A. MRI abnormalities in Creutzfeldt-Jakob disease and other rapidly progressive dementia. J. Neurol. 2024, 271, 300–309. [Google Scholar] [CrossRef] [PubMed]
  117. Eisenmenger, L.; Porter, M.C.; Carswell, C.J.; Thompson, A.; Mead, S.; Rudge, P.; Collinge, J.; Brandner, S.; Jäger, H.R.; Hyare, H. Evolution of diffusion-weighted magnetic resonance imaging signal abnormality in sporadic Creutzfeldt-Jakob disease, with histopathological correlation. JAMA Neurol. 2016, 73, 7. [Google Scholar] [CrossRef]
  118. Rudge, P.; Hyare, H.; Green, A.; Collinge, J.; Mead, S. Imaging and CSF analyses effectively distinguish CJD from its mimics. J. Neurol. Neurosurg. Psychiatry 2018, 89, 461–466. [Google Scholar] [CrossRef]
  119. Ayyappan, S.; Seneviratne, U. Electroencephalographic changes in sporadic Creutzfeldt-Jakob disease and correlation with clinical stages: A retrospective analysis. J. Clin. Neurophysiol. 2014, 31, 586–593. [Google Scholar] [CrossRef]
  120. Hermann, P.; Laux, M.; Glatzel, M.; Matschke, J.; Knipper, T.; Goebel, S.; Treig, J.; Schulz-Schaefer, W.; Cramm, M.; Schmitz, M.; et al. Validation and utilization of amended diagnostic criteria in Creutzfeldt-Jakob disease surveillance. Neurology 2018, 91, e331–e338. [Google Scholar] [CrossRef]
  121. Bhatia, P.; Sonbol, M.; Jain, D.; Rincon-Flores, N.; Frontera, A. Status Epilepticus in Creutzfeldt-Jakob Disease: A Case Report of an Unusual Presentation. Cureus 2022, 14, e26470. [Google Scholar] [CrossRef]
  122. Fanella, M.; Valente, G.; Borrello, L.; Marinelli, F.; Bracaglia, M.; Di Marco, O.; Costanzo, F.; Apponi, F.; De Simone, R. Non-convulsive status epilepticus versus periodic EEG pattern in sporadic Creutzfeldt-Jakob disease: Two sides of the same coin? Int. J. Neurosci. 2023, 19, 1–5. [Google Scholar] [CrossRef]
  123. Mattoli, M.V.; Giancipoli, R.G.; Cocciolillo, F.; Calcagni, M.L.; Taralli, S. The Role of PET Imaging in Patients with Prion Disease: A Literature Review. Mol. Imaging Biol. 2024, 26, 195–212. [Google Scholar] [CrossRef]
  124. Chen, Z.; Kong, Y.; Zhang, J.; Chu, M.; Liu, L.; Xie, K.; Cui, Y.; Ye, H.; Li, J.; Wang, L.; et al. Toward an early clinical diagnosis of MM2-type sporadic Creutzfeldt-Jakob disease. Ann. Clin. Transl. Neurol. 2023, 10, 1209–1218. [Google Scholar] [CrossRef] [PubMed]
  125. World Health Organisation. Global Surveillance, Diagnosis and Therapy of Human Transmissible Spongiform Encephalopathies: Report of a WHO Consultation, Geneva, Switzerland, 9–11 February 1998. Available online: https://iris.who.int/handle/10665/65516 (accessed on 24 April 2024).
  126. Creutzfeldt-Jakob Disease International Surveillance Network. Diagnostic Criteria for Surveillance of CJD from 1 January 2017. Updated 2021. Available online: https://www.eurocjd.ed.ac.uk/node/833 (accessed on 25 April 2024).
  127. Green, A.J.E. RT-QuIC: A new test for sporadic CJD. Pract. Neurol. 2019, 19, 49–55. [Google Scholar] [CrossRef] [PubMed]
  128. Hermann, P.; Appleby, B.; Brandel, J.P.; Caughey, B.; Collins, S.; Geschwind, M.D.; Green, A.; Haïk, S.; Kovacs, G.G.; Ladogana, A.; et al. Biomarkers and diagnostic guidelines for sporadic Creutzfeldt-Jakob disease. Lancet Neurol. 2021, 20, 235–246. [Google Scholar] [CrossRef] [PubMed]
  129. Geschwind, M.D.; Murray, K. Differential diagnosis with other rapid progressive dementias in human prion diseases. In Handbook of Clinical Neurology; 3rd series; Pocchiari, M., Manson, J., Eds.; Elsevier: Oxford, UK, 2018; Volume 153, pp. 371–397. [Google Scholar]
  130. Jurcau, A.; Nunkoo, V.S. Tau-targeted therapy in Alzheimer’s disease: History and current state. In Frontiers in Clinical Drug Research; Ibarra Arias, J.J.J., Ed.; Bentham Science Publishers: Singapore, 2021; Volume 2, pp. 56–138. [Google Scholar]
  131. Jurcau, A.; Simion, A.; Jurcau, M.C. Emerging antibody-based therapies for Huntington’s disease: Current status and perspectives for future development. Expert Rev. Neurother. 2024, 24, 299–312. [Google Scholar] [CrossRef] [PubMed]
  132. Banerjee, G.; Farmer, S.F.; Hyare, H.; Jaunmuktane, Z.; Mead, S.; Ryan, N.S.; Schott, J.M.; Werring, D.J.; Rudge, P.; Collinge, J. Iatrogenic Alzheimer’s disease in recipients of cadaveric pituitary-derived growth hormone. Nat. Med. 2024, 30, 394–402. [Google Scholar] [CrossRef]
  133. Marra, C.M. Syphilis screening in neurology. JAMA Neurol. 2016, 21, 1714–1728. [Google Scholar] [CrossRef]
  134. Lin, C.R.; Kuo, S.H. Ataxias: Hereditary, Acquired, and Reversible Etiologies. Semin. Neurol. 2023, 43, 48–64. [Google Scholar] [CrossRef]
  135. Verity, M.A. Comparative observations in inorganic and organic lead neurotoxicity. Environ. Health Perspect. 1990, 89, 43–48. [Google Scholar] [CrossRef]
  136. Scalzo, S.J.; Bowden, S.C.; Ambrose, M.L.; Whelan, G.; Cook, M.J. Wernicke-Korsakoff syndrome not related to alcohol use: A systematic review. J. Neurol. Neurosurg. Psychiatry 2015, 86, 1362–1368. [Google Scholar] [CrossRef]
  137. Rosenbloom, M.H.; Tartaglia, M.C.; Forner, S.A.; Wong, K.K.; Kuo, A.; Johnson, D.Y.; Colacurcio, V.; Andrews, B.D.; Miller, B.L.; DeArmond, S.J.; et al. Metabolic disorders with clinical and radiologic features of sporadic Creutzfeldt-Jakob disease. Neurol. Clin. Pract. 2015, 5, 108–115. [Google Scholar] [CrossRef]
  138. Kimura, M.; Kimura, H.; Ishikawa, H.; Matsuo, H.; Takada, M.; Matsuo, K. Hepatic Encephalopathy Mimicking Creutzfeldt-Jakob Disease on Laboratory, Physiological, and Imaging Evaluations. Am. J. Case Rep. 2021, 22, e932958. [Google Scholar] [CrossRef] [PubMed]
  139. Sharma, P.; Eesa, M.; Scott, N.S. Toxic and acquired metabolic encephalopathies: MRI appearance. AJR Am. J. Roentgenol. 2009, 193, 879–886. [Google Scholar] [CrossRef] [PubMed]
  140. Farrall, A.L.; Smith, J.R. Changing Incidence and Survival of Primary Central Nervous System Lymphoma in Australia: A 33-Year National Population-Based Study. Cancers 2021, 13, 403. [Google Scholar] [CrossRef] [PubMed]
  141. Mead, S.; Rudge, P. CJD mimics and chameleons. Pract. Neurol. 2017, 17, 113–121. [Google Scholar] [CrossRef]
  142. Bentivenga, G.M.; Baiardi, S.; Righini, L.; Ladogana, A.; Capellari, S.; Sabattini, E.; Parchi, P. Rapidly progressive dementia due to intravascular lymphoma: A prion disease reference center experience. Eur. J. Neurol. 2024, 31, e16068. [Google Scholar] [CrossRef]
  143. Hermetter, C.; Fazekas, F.; Hochmeister, S. Systematic Review: Syndromes, Early Diagnosis, and Treatment in Autoimmune Encephalitis. Front. Neurol. 2018, 9, 706. [Google Scholar] [CrossRef]
  144. Lee, S.K.; Lee, S.T. The Laboratory Diagnosis of Autoimmune Encephalitis. J. Epilepsy Res. 2016, 6, 45–50. [Google Scholar] [CrossRef]
  145. Banks, S.A.; Sechi, E.; Flanagan, E.P. Autoimmune encephalopathies presenting as dementia of subacute onset and rapid progression. Ther. Adv. Neurol. Disord. 2021, 14, 1756286421998906. [Google Scholar] [CrossRef]
  146. Laurido-Soto, O.; Brier, M.R.; Simon, L.E.; McCullough, A.; Bucelli, R.C.; Day, G.S. Patient characteristics and outcome associations in AMPA receptor encephalitis. J. Neurol. 2019, 266, 450–460. [Google Scholar] [CrossRef]
  147. Li, H.; Chen, J.; Zhou, P.; Meng, Q. Analysis of characteristics of movement disorders in patients with anti-N-methyl-D-aspartate receptor encephalitis. Front. Neurol. 2024, 15, 1357697. [Google Scholar] [CrossRef]
  148. Hara, M.; Ariño, H.; Petit-Pedrol, M.; Sabater, L.; Titulaer, M.J.; Martinez-Hernandez, E.; Schreurs, M.W.; Rosenfeld, M.R.; Graus, F.; Dalmau, J. DPPX antibody-associated encephalitis: Main syndrome and antibody effects. Neurology 2017, 88, 1340–1348. [Google Scholar] [CrossRef] [PubMed]
  149. Maureille, A.; Fenouil, T.; Joubert, B.; Picard, G.; Rogemond, V.; Pinto, A.L.; Thomas, L.; Ducray, F.; Quadrio, I.; Psimaras, D.; et al. Isolated seizures are a common early feature of paraneoplastic anti-GABAB receptor encephalitis. J. Neurol. 2019, 266, 195–206. [Google Scholar] [CrossRef] [PubMed]
  150. Spatola, M.; Petit-Pedrol, M.; Simabukuro, M.M.; Armangue, T.; Castro, F.J.; Barcelo Artigues, M.I.; Julià Benique, M.R.; Benson, L.; Gorman, M.; Felipe, A.; et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017, 88, 1012–1020. [Google Scholar] [CrossRef] [PubMed]
  151. Smith, K.M.; Dubey, D.; Liebo, G.B.; Flanagan, E.P.; Britton, J.W. Clinical Course and Features of Seizures Associated with LGI1-Antibody Encephalitis. Neurology 2021, 97, e1141–e1149. [Google Scholar] [CrossRef]
  152. Boyko, M.; Au, K.L.K.; Casault, C.; de Robles, P.; Pfeffer, G. Systematic review of the clinical spectrum of CASPR2 antibody syndrome. J. Neurol. 2020, 267, 1137–1146. [Google Scholar] [CrossRef]
  153. Gaig, C.; Graus, F.; Compta, Y.; Högl, B.; Bataller, L.; Brüggemann, N.; Giordana, C.; Heidbreder, A.; Kotschet, K.; Lewerenz, J.; et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017, 88, 1736–1743. [Google Scholar] [CrossRef]
  154. Christ, M.; Müller, T.; Bien, C.; Hagen, T.; Naumann, M.; Bayas, A. Autoimmune encephalitis associated with antibodies against the metabotropic glutamate receptor type 1: Case report and review of the literature. Ther. Adv. Neurol. Disord. 2019, 12, 1756286419847418. [Google Scholar] [CrossRef]
  155. Spatola, M.; Sabater, L.; Planagumà, J.; Martínez-Hernandez, E.; Armangué, T.; Prüss, H.; Iizuka, T.; Caparó Oblitas, R.L.; Antoine, J.C.; Li, R.; et al. Encephalitis with mGluR5 antibodies: Symptoms and antibody effects. Neurology 2018, 90, e1964–e1972. [Google Scholar] [CrossRef]
  156. Swayne, A.; Tjoa, L.; Broadley, S.; Dionisio, S.; Gillis, D.; Jacobson, L.; Woodhall, M.R.; McNabb, A.; Schweitzer, D.; Tsang, B.; et al. Antiglycine receptor antibody related disease: A case series and literature review. Eur. J. Neurol. 2018, 25, 1290–1298. [Google Scholar] [CrossRef]
  157. Hansen, N.; Lange, C.; Maass, F.; Hassoun, L.; Bouter, C.; Stöcker, W.; Schott, B.H.; Wiltfang, J.; Fitzner, D. Mild Amnestic Cognitive Impairment and Depressive Symptoms in Autoimmune Encephalitis Associated with Serum Anti-Neurexin-3α Autoantibodies. Brain Sci. 2021, 11, 673. [Google Scholar] [CrossRef]
  158. Li, E.C.; Lai, Q.L.; Cai, M.T.; Fang, G.L.; Shen, C.H.; Ding, M.P.; Zhang, Y.X. Anti-adenylate kinase 5 encephalitis: Clinical characteristics, diagnosis, and management of this rare entity. J. Transl. Autoimmun. 2023, 7, 100218. [Google Scholar] [CrossRef] [PubMed]
  159. Sun, Y.; Qin, X.; Huang, D.; Zhou, Z.; Zhang, Y.; Wang, Q. Anti-amphiphysin encephalitis: Expanding the clinical spectrum. Front. Immunol. 2023, 14, 1084883. [Google Scholar] [CrossRef]
  160. Qi, J.; Maheshwari, M. ANNA-1-associated paraneoplastic limbic encephalitis in a patient with pelvic ganglioneuroblastoma. Semin. Roentgenol. 2023, 58, 3–5. [Google Scholar] [CrossRef] [PubMed]
  161. Gilligan, M.; McGuigan, C.; McKeon, A. Paraneoplastic Neurologic Disorders. Curr. Neurol. Neurosci. Rep. 2023, 23, 67–82. [Google Scholar] [CrossRef]
  162. Tsiortou, P.; Alexopoulos, H.; Dalakas, M.C. GAD antibody-spectrum disorders: Progress in clinical phenotypes, immunopathogenesis and therapeutic interventions. Ther. Adv. Neurol. Disord. 2021, 14, 17562864211003486. [Google Scholar] [CrossRef] [PubMed]
  163. Kimura, A.; Takekoshi, A.; Yoshikura, N.; Hayashi, Y.; Shimohata, T. Clinical characteristics of autoimmune GFAP astrocytopathy. J. Neuroimmunol. 2019, 332, 91–98. [Google Scholar] [CrossRef] [PubMed]
  164. Jarius, S.; Bräuninger, S.; Chung, H.Y.; Geis, C.; Haas, J.; Komorowski, L.; Wildemann, B.; Roth, C. Inositol 1,4,5-trisphosphate receptor type 1 autoantibody (ITPR1-IgG/anti-Sj)-associated autoimmune cerebellar ataxia, encephalitis and peripheral neuropathy: Review of the literature. J. Neuroinflamm. 2022, 19, 196. [Google Scholar] [CrossRef]
  165. Bien, C.G. Diagnosing autoimmune encephalitis based on clinical features and autoantibody findings. Expert Rev. Clin. Immunol. 2019, 15, 511–527. [Google Scholar] [CrossRef]
  166. Wang, S.; Hou, H.; Tang, Y.; Zhang, S.; Wang, G.; Guo, Z.; Zhu, L.; Wu, J. An overview on CV2/CRMP5 antibody-associated paraneoplastic neurological syndromes. Neural Regen. Res. 2023, 18, 2357–2364. [Google Scholar] [CrossRef]
  167. Hegde, A.N.; Mohan, S.; Lath, N.; Lim, C.C. Differential diagnosis for bilateral abnormalities of the basal ganglia and thalamus. RadioGraphics 2011, 31, 5–30. [Google Scholar] [CrossRef]
  168. Dalmau, J.; Rosenfeld, M.R. Autoimmune encephalitis update. Neuro-Oncology 2014, 16, 771–778. [Google Scholar] [CrossRef] [PubMed]
  169. Bulakbasi, N.; Kocaoglu, M. Central nervous system infections of herpesvirus family. Neuroimaging Clin. N. Am. 2008, 18, 53–84. [Google Scholar] [CrossRef] [PubMed]
  170. Cianfoni, A.; Caulo, M.; Cerase, A.; Della Marca, G.; Falcone, C.; Di Lella, G.M.; Gaudino, S.; Edwards, J.; Colosimo, C. Seizure-induced brain lesions: A wide spectrum of variably reversible MRI abnormalities. Eur. J. Radiol. 2013, 82, 1964–1972. [Google Scholar] [CrossRef] [PubMed]
  171. U-King-Im, J.M.; Yu, E.; Bartlett, E.; Soobrah, R.; Kucharczyk, W. Acute hyperammonemic encephalopathy in adults: Imaging findings. Am. J. Neuroradiol. 2011, 32, 413–418. [Google Scholar] [CrossRef]
  172. Appenzeller, S.; Schirmacher, A.; Halfter, H.; Bäumer, S.; Pendziviat, M.; Timmerman, V.; De Jonghe, P.; Fekete, K.; Stögbauer, F.; Lüdemann, P.; et al. Autosomal dominant striatal degeneration is caused by a mutation in the phosphodiesterase 8B gene. Am. J. Hum. Genet. 2010, 86, 83–87. [Google Scholar] [CrossRef]
  173. Collie, D.A.; Summers, D.M.; Sellar, R.J.; Ironside, J.W.; Cooper, S.; Zeidler, M.; Knight, R.; Will, R.G. Diagnosing variant Creutzfeldt-Jakob disease with the pulvinar sign: MR imaging findings in 86 neuropathologically confirmed cases. Am. J. Neuroradiol. 2003, 24, 1560–1569. [Google Scholar]
  174. Appleby, B.S.; Yobs, D.R. Symptomatic treatment, care, and support of CJD patients. Handb. Clin. Neurol. 2018, 153, 399–408. [Google Scholar]
  175. Uflacker, A.; Edmondson, M.C.; Onyike, C.U.; Appleby, B.S. Caregiver burden in atypical dementias: Comparing frontotemporal dementia, Creutzfeldt-Jakob disease, and Alzheimer’s disease. Int. Psychogeriatr. 2016, 28, 269–273. [Google Scholar] [CrossRef]
  176. Otto, M.; Cepek, L.; Ratzka, P.; Doehlinger, S.; Boekhoff, I.; Wiltfang, J.; Irle, E.; Pergande, G.; Ellers-Lenz, B.; Windl, O.; et al. Efficacy of flupirtine on cognitive function in patients with CJD: A double-blind study. Neurology 2004, 62, 714–718. [Google Scholar] [CrossRef]
  177. Forloni, G.; Roiter, I.; Tagliavini, F. Clinical trials of prion disease therapeutics. Curr. Opin. Pharmacol. 2019, 44, 53–60. [Google Scholar] [CrossRef]
  178. Collinge, J.; Gorham, M.; Hudson, F.; Kennedy, A.; Keogh, G.; Pal, S.; Rossor, M.; Rudge, P.; Siddique, D.; Spyer, M.; et al. Safety and efficacy of quinacrine in human prion disease (PRION-1 study): A patient-preference trial. Lancet Neurol. 2009, 8, 334–344. [Google Scholar] [CrossRef] [PubMed]
  179. Geschwind, M.D.; Kuo, A.L.; Wong, K.S.; Haman, A.; Devereux, G.; Raudabaugh, B.J.; Johnson, D.Y.; Torres-Chae, C.C.; Finley, R.; Garcia, P.; et al. Quinacrine treatment trial for sporadic Creutzfeldt-Jakob disease. Neurology 2013, 81, 2015–2023. [Google Scholar] [CrossRef] [PubMed]
  180. Bone, I.; Belton, L.; Walker, A.S.; Darbyshire, J. Intraventricular pentosan polysulphate in human prion diseases: An observational study in the UK. Eur. J. Neurol. 2008, 15, 458–464. [Google Scholar] [CrossRef] [PubMed]
  181. Nakajima, M.; Yamada, T.; Kusuhara, T.; Furukawa, H.; Takahashi, M.; Yamauchi, A.; Kataoka, Y. Results of quinacrine administration to patients with Creutzfeldt-Jakob disease. Dement. Geriatr. Cogn. Disord. 2004, 17, 158–163. [Google Scholar] [CrossRef] [PubMed]
  182. Tsuboi, Y.; Doh-Ura, K.; Yamada, T. Continuous intraventricular infusion of pentosan polysulfate: Clinical trial against prion diseases. Neuropathology 2009, 29, 632–636. [Google Scholar] [CrossRef]
  183. Tagliavini, F.; Forloni, G.; Colombo, L.; Rossi, G.; Girola, L.; Canciani, B.; Angeretti, N.; Giampaolo, L.; Peressini, E.; Awan, T.; et al. Tetracycline affects abnormal properties of synthetic PrP peptides and PrP(Sc) in vitro. J. Mol. Biol. 2000, 300, 1309–1322. [Google Scholar] [CrossRef]
  184. De Luigi, A.; Colombo, L.; Diomede, L.; Capobianco, R.; Mangieri, M.; Miccolo, C.; Limido, L.; Forloni, G.; Tagliavini, F.; Salmona, M. The efficacy of tetracyclines in peripheral and intracerebral prion infection. PLoS ONE 2008, 3, e1888. [Google Scholar] [CrossRef]
  185. Haïk, S.; Marcon, G.; Mallet, A.; Tettamanti, M.; Welaratne, A.; Giaccone, G.; Azimi, S.; Pietrini, V.; Fabreguettes, J.R.; Imperiale, D.; et al. Doxycycline in Creutzfeldt-Jakob disease: A phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014, 13, 150–158. [Google Scholar] [CrossRef]
  186. Varges, D.; Manthey, H.; Heinemann, U.; Ponto, C.; Schmitz, M.; Schulz-Schaeffer, W.J.; Krasnianski, A.; Breithaupt, M.; Fincke, F.; Kramer, K.; et al. Doxycycline in early CJD: A double-blinded randomised phase II and observational study. J. Neurol. Neurosurg. Psychiatry 2017, 88, 119–125. [Google Scholar] [CrossRef]
  187. Liu, F.; Lü, W.; Liu, L. New implications for prion diseases therapy and prophylaxis. Front. Mol. Neurosci. 2024, 17, 1324702. [Google Scholar] [CrossRef]
  188. Andronie-Cioara, F.L.; Ardelean, A.I.; Nistor-Cseppento, C.D.; Jurcau, A.; Jurcau, M.C.; Pascalau, N.; Marcu, F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer’s Disease Progression. Int. J. Mol. Sci. 2023, 24, 1869. [Google Scholar] [CrossRef] [PubMed]
  189. Jurcau, A. Molecular Pathophysiological Mechanisms in Huntington’s Disease. Biomedicines 2022, 10, 1432. [Google Scholar] [CrossRef] [PubMed]
  190. Bachy, V.; Ballerini, C.; Gourdain, P.; Prignon, A.; Iken, S.; Antoine, N.; Rosset, M.; Carnaud, C. Mouse vaccination with dendritic cells loaded with prion protein peptides overcomes tolerance and delays scrapie. J. Gen. Virol. 2010, 91, 809–820. [Google Scholar] [CrossRef]
  191. Han, Y.; Li, Y.; Song, J.; Wang, Y.; Shi, Q.; Chen, C.; Zhang, B.; Guo, Y.; Li, C.; Han, J.; et al. Immune responses in wild-type mice against prion proteins induced using a DNA prime-protein boost strategy. Biomed. Environ. Sci. 2011, 24, 523–529. [Google Scholar] [PubMed]
  192. Nitschke, C.; Flechsig, E.; van den Brandt, J.; Lindner, N.; Lührs, T.; Dittmer, U.; Klein, M.A. Immunisation strategies against prion diseases: Prime-boost immunisation with a PrP DNA vaccine containing foreign helper T-cell epitopes does not prevent mouse scrapie. Vet. Microbiol. 2007, 123, 367–376. [Google Scholar] [CrossRef]
  193. Taschuk, R.; Scruten, E.; Woodbury, M.; Cashman, N.; Potter, A.; Griebel, P.; Tikoo, S.K.; Napper, S. Induction of PrPSc-specific systemic and mucosal immune responses in white-tailed deer with an oral vaccine for chronic wasting disease. Prion 2017, 11, 368–380. [Google Scholar] [CrossRef]
  194. Taschuk, R.; Marciniuk, K.; Määttänen, P.; Madampage, C.; Hedlin, P.; Potter, A.; Lee, J.; Cashman, N.R.; Griebel, P.J.; Napper, S. Safety, specificity and immunogenicity of a PrP(Sc)-specific prion vaccine based on the YYR disease specific epitope. Prion 2014, 8, 51–59. [Google Scholar] [CrossRef]
  195. Ma, Y.; Ma, J. Immunotherapy against Prion Disease. Pathogens 2020, 9, 216. [Google Scholar] [CrossRef]
  196. Collinge, J. Mammalian prions and their wider relevance in neurodegenerative diseases. Nature 2016, 539, 217–226. [Google Scholar] [CrossRef]
  197. Peretz, D.; Williamson, R.A.; Kaneko, K.; Vergara, J.; Leclerc, E.; Schmitt-Ulms, G.; Mehlhorn, I.R.; Legname, G.; Wormald, M.R.; Rudd, P.M.; et al. Antibodies inhibit prion propagation and clear cell cultures of prion infectivity. Nature 2001, 412, 739–743. [Google Scholar] [CrossRef]
  198. Ohsawa, N.; Song, C.H.; Suzuki, A.; Furuoka, H.; Hasebe, R.; Horiuchi, M. Therapeutic effect of peripheral administration of an anti-prion protein antibody on mice infected with prions. Microbiol. Immunol. 2013, 57, 288–297. [Google Scholar] [CrossRef] [PubMed]
  199. Mead, S.; Khalili-Shirazi, A.; Potter, C.; Mok, T.; Nihat, A.; Hyare, H.; Canning, S.; Schmidt, C.; Campbell, T.; Darwent, L.; et al. Prion protein monoclonal antibody (PRN100) therapy for Creutzfeldt-Jakob disease: Evaluation of a first-in-human treatment programme. Lancet Neurol. 2022, 21, 342–354. [Google Scholar] [CrossRef] [PubMed]
  200. Terao, I.; Kodama, W. Comparative efficacy, tolerability and acceptability of donanemab, lecanemab, aducanumab and lithium on cognitive function in mild cognitive impairment and Alzheimer’s disease: A systematic review and network meta-analysis. Ageing Res. Rev. 2024, 94, 102203. [Google Scholar] [CrossRef] [PubMed]
  201. Frontzek, K.; Bardelli, M.; Senatore, A.; Henzi, A.; Reimann, R.R.; Bedir, S.; Marino, M.; Hussain, R.; Jurt, S.; Meisl, G.; et al. A conformational switch controlling the toxicity of the prion protein. Nat. Struct. Mol. Biol. 2022, 29, 831–840. [Google Scholar] [CrossRef] [PubMed]
  202. Fares, J.; Fares, M.Y.; Fares, Y. Immune checkpoint inhibitors: Advances and impact in neuro-oncology. Surg. Neurol. Int. 2019, 10, 9. [Google Scholar] [CrossRef]
  203. Obst, J.; Mancuso, R.; Simon, E.; Gomez-Nicola, D. PD-1 deficiency is not sufficient to induce myeloid mobilization to the brain or alter the inflammatory profile during chronic neurodegeneration. Brain Behav. Immun. 2018, 73, 708–716. [Google Scholar] [CrossRef]
  204. Liu, Y.; Sorce, S.; Nuvolone, M.; Domange, J.; Aguzzi, A. Lymphocyte activation gene 3 (Lag3) expression is increased in prion infections but does not modify disease progression. Sci. Rep. 2018, 8, 14600. [Google Scholar] [CrossRef]
  205. Li, B.; Chen, M.; Zhu, C. Neuroinflammation in Prion Disease. Int. J. Mol. Sci. 2021, 22, 2196. [Google Scholar] [CrossRef]
  206. Jurcau, A.; Jurcau, M.C. Therapeutic Strategies in Huntington’s Disease: From genetic Defect to Gene Therapy. Biomedicines 2022, 10, 1895. [Google Scholar] [CrossRef]
  207. Minikel, E.V.; Zhao, H.T.; Le, J.; O’Moore, J.; Pitstick, R.; Graffam, S.; Carlson, G.A.; Kavanaugh, M.P.; Kriz, J.; Kim, J.B.; et al. Prion protein lowering is a disease-modifying therapy across prion disease stages, strains and endpoints. Nucleic Acids Res. 2020, 48, 10615–10631. [Google Scholar] [CrossRef]
  208. Aguzzi, A.; Frontzek, K. New paradigms of clinical trial design for genetic prion diseases. Lancet Neurol. 2020, 19, 284–285. [Google Scholar] [CrossRef] [PubMed]
  209. White, M.D.; Farmer, M.; Mirabile, I.; Brandner, S.; Collinge, J.; Mallucci, G.R. Single treatment with RNAi against prion protein rescues early neuronal dysfunction and prolongs survival in mice with prion disease. Proc. Natl. Acad. Sci. USA 2008, 105, 10238–10243. [Google Scholar] [CrossRef] [PubMed]
  210. Genoud, N.; Ott, D.; Braun, N.; Prinz, M.; Schwarz, P.; Suter, U.; Trono, D.; Aguzzi, A. Antiprion prophylaxis by gene transfer of a soluble prion antagonist. Am. J. Pathol. 2008, 172, 1287–1296. [Google Scholar] [CrossRef] [PubMed]
  211. Espargaró, A.; Sabate, R. Phosphorylation-driven aggregative proteins in neurodegenerative diseases: Implications and therapeutics. Neural Regen. Res. 2024, 19, 966–968. [Google Scholar] [CrossRef] [PubMed]
  212. Banik, S.M.; Pedram, K.; Wisnovsky, S.; Ahn, G.; Riley, N.M.; Bertozzi, C.R. Lysosome-targeting chimaeras for degradation of extracellular proteins. Nature 2020, 584, 291–297. [Google Scholar] [CrossRef]
  213. Kocak, M.; Ezazi Erdi, S.; Jorba, G.; Maestro, I.; Farrés, J.; Kirkin, V.; Martinez, A.; Pless, O. Targeting autophagy in disease: Established and new strategies. Autophagy 2022, 18, 473–495. [Google Scholar] [CrossRef]
  214. Bourdenx, M.; Martín-Segura, A.; Scrivo, A.; Rodriguez-Navarro, J.A.; Kaushik, S.; Tasset, I.; Diaz, A.; Storm, N.J.; Xin, Q.; Juste, Y.R.; et al. Chaperone-mediated autophagy prevents collapse of the neuronal metastable proteome. Cell 2021, 184, 2696–2714.e25. [Google Scholar] [CrossRef]
  215. Evgen’ev, M.; Bobkova, N.; Krasnov, G.; Garbuz, D.; Funikov, S.; Kudryavtseva, A.; Kulikov, A.; Samokhin, A.; Maltsev, A.; Nesterova, I. The Effect of Human HSP70 Administration on a Mouse Model of Alzheimer’s Disease Strongly Depends on Transgenicity and Age. J. Alzheimers. Dis. 2019, 67, 1391–1404. [Google Scholar] [CrossRef]
  216. Mays, C.E.; Armijo, E.; Morales, R.; Kramm, C.; Flores, A.; Tiwari, A.; Bian, J.; Telling, G.C.; Pandita, T.K.; Hunt, C.R.; et al. Prion disease is accelerated in mice lacking stress-induced heat shock protein 70 (HSP70). J. Biol. Chem. 2019, 294, 13619–13628. [Google Scholar] [CrossRef]
  217. Thackray, A.M.; Lam, B.; McNulty, E.E.; Nalls, A.V.; Mathiason, C.K.; Magadi, S.S.; Jackson, W.S.; Andréoletti, O.; Marrero-Winkens, C.; Schätzl, H.; et al. Clearance of variant Creutzfeldt-Jakob disease prions in vivo by the Hsp70 disaggregase system. Brain 2022, 145, 3236–3249. [Google Scholar] [CrossRef]
  218. Nistor-Cseppento, D.C.; Jurcau, M.C.; Jurcau, A.; Andronie-Cioara, F.L.; Marcu, F. Stem Cell- and Cell-Based Therapies for ischemic Stroke. Bioengineering 2022, 11, 717. [Google Scholar] [CrossRef] [PubMed]
  219. Relaño-Ginés, A.; Lehmann, S.; Bencsik, A.; Herva, M.E.; Torres, J.M.; Crozet, C.A. Stem cell therapy extends incubation and survival time in prion-infected mice in a time window-dependent manner. J. Infect. Dis. 2011, 204, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
  220. Hay, A.J.D.; Latham, A.S.; Mumford, G.; Hines, A.D.; Risen, S.; Gordon, E.; Siebenaler, C.; Gilberto, V.S.; Zabel, M.D.; Moreno, J.A. Intranasally delivered mesenchymal stromal cells decrease glial inflammation early in prion disease. Front. Neurosci. 2023, 17, 1158408. [Google Scholar] [CrossRef] [PubMed]
  221. Zayed, M.; Kook, S.-H.; Jeong, B.-H. Potential Therapeutic Use of Stem Cells for Prion Diseases. Cells 2023, 12, 2413. [Google Scholar] [CrossRef]
  222. Groveman, B.R.; Smith, A.; Williams, K.; Haigh, C.L. Cerebral organoids as a new model for prion disease. PLoS Pathog. 2021, 17, e1009747. [Google Scholar] [CrossRef]
  223. Krance, S.H.; Luke, R.; Shenouda, M.; Israwi, A.R.; Colpitts, S.J.; Darwish, L.; Strauss, M.; Watts, J.C. Cellular models for discovering prion disease therapeutics: Progress and challenges. J. Neurochem. 2020, 153, 139–289. [Google Scholar] [CrossRef]
  224. Collinge, J. Prion Strain Mutation and Selection. Science 2010, 328, 1111–1112. [Google Scholar] [CrossRef]
  225. Makarava, N.; Baskakov, I.V. The evolution of transmissible prions: The role of deformed templating. PLoS Path. 2013, 9, e1003759. [Google Scholar] [CrossRef]
  226. Pritzkow, S.; Ramirez, F.; Lyon, A.; Schulz, P.E.; Appleby, B.; Moda, F.; Ramirez, S.; Notari, S.; Gambetti, P.; Soto, C. Detection of prions in the urine of patients affected by sporadic Creutzfeldt-Jakob disease. Ann. Clin. Transl. Neurol. 2023, 10, 2316–2323. [Google Scholar] [CrossRef]
Table 1. Epidemiological, clinical, and neuropathological characteristics of the subtypes of sporadic CJD (adapted from references [54,57,83].
Table 1. Epidemiological, clinical, and neuropathological characteristics of the subtypes of sporadic CJD (adapted from references [54,57,83].
Subtype of Sporadic CJDMean Age of Onset (Years)Mean Disease Duration (Months)Neuropathological FeaturesCharacteristic Clinical Symptoms/Signs
MM(V)1694Spongiform changes in the cerebral (occipital) cortex, cerebellum, thalamus and striatumDementia, visual impairments, ataxia, myoclonus
VV2656–9Involvement of the cerebellum, basal ganglia, and diencephalon; spongiform changes often limited to the deep layers of the neocortexAtaxia, with dementia following later in the disease course
MV2K659–17Amyloid kuru plaques in the cerebellum, involvement of the basal gangliaAtaxia, extrapyramidal signs, dementia
MM2T42–5216–18Atrophy of the medial thalamus and inferior olive, patchy spongiform changes in the cortexAtaxia, double vision, sleep disturbances, psychiatric symptoms, followed by cognitive decline
MM2C61–6416–18Rapidly progressive dementiaSpongiform changes and PrP deposits in the neocortex
VV142–4418–21Dementia, followed by ataxia, extrapyramidal signsSpongiform changes in the neocortex, hippocampus, and striatum
Table 2. Characteristics of antibody-mediated autoimmune encephalopathies (adapted from ref. [143,145]).
Table 2. Characteristics of antibody-mediated autoimmune encephalopathies (adapted from ref. [143,145]).
Type of AntigenAutoantibody TargetIncidence of Cognitive ImpairmentClinical FeaturesPossibility of Underlying Cancer and Type of More Common NeoplasiaMRI Features (T2/FLAIR)Ref.
Surface antigensAMPA receptors100%LE, hyponatremia64%, small cell lung, thymomaHyperintensities in medial temporal lobes and/or cerebellum[146]
NMDA receptors90–100%LE, psychosis, facio-brachial dyskinesias40–60%, usually ovarian teratomaNormal aspect or non-specific regional changes[147]
DPPX80–100%Sleep disturbances, gastrointestinal symptoms (diarrhea)10%, hematologic malignanciesNormal or non-specific changes[148]
GABAB receptors80–100%LE, epileptic seizures40–60%, thymoma, bronchial carcinomaHyperintense signals in medial temporal lobes
[149]
GABAA receptors67%Refractory seizures, status epilepticus25–40%, thymomaHyperintense signals in multiple cortical and subcortical areas[150]
LGI1 receptors90–100%LE, facio-brachial dystonic seizures, myoclonus, hyponatremia,10%, thymoma, bronchial carcinomaHyperintensities in medial temporal lobes and basal ganglia[151]
CASPR240–80%LE, stiff person syndrome, ataxia10–20%, thymomaNormal aspect or hyperintensities in medial temporal lobes[152]
IgLON530–40%Sleep disturbances, ataxiarareNormal aspect[153]
mGLUR1rareCerebellar ataxiaRare cases of Hodgkin’s lymphomaNormal aspect or cerebellar atrophy[154]
mGLUR590%LE, seizures50%, Hodgkin’s lymphomaNormal aspect or hyperintensities in various brain regions[155]
GlyRα140–50%Stiff person syndrome, rigidity, myoclonus, seizures10%, thymomaNormal aspect or non-specific features[156]
Neurexin 3α40–50%Orofacial dyskinesiasunknownNormal aspect[157]
Intracellular antigensAK5100%LENot cancer-associatedTemporal lobe hyperintensities[158]
amphiphysin30%LE, peripheral neuropathy80%, small cell lung, breastNormal aspect or temporal lobe hyperintensities[159]
ANNA1 (Hu)10–20%LE, cerebellar ataxia, sensory neuronopathy80–90%, small cell lung carcinomaTemporal lobe hyperintensities[160]
ANNA2 (Ri)10–20%Ataxia, opsoclonus-myoclonus75%, small cell lung carcinoma, breast adenocarcinomaNon-specific features[161]
ANNA310–20%LE, cerebellar ataxia, peripheral neuropathy80–90%, small cell lung carcinomaNon-specific changes[161]
GAD3–5%LE, ataxia, seizures8%, small cell lung carcinomaAtrophy of temporal and frontal lobes[162]
GFAP15–60%Tremor, myoclonus, ataxia35%, teratomasHyperintensities in the posterior parts of the thalamus[163]
ITPR120%Cerebellar ataxia, seizures30–40%, breast cancerNormal aspect[164]
Ma260–70%LE, sleep disorders, narcolepsy90%, testicular neoplasiasTemporal lobe hyperintensities[165]
CRMP5 (CV2)30%LE, chorea, cerebellar ataxia, myelopathy, optic neuritis90%, small cell lung carcinomaNormal aspect or multiple hyperintensities in the temporal lobe, basal ganglia, thalamus and frontal lobe[166]
Abbreviations: AK5—adenylate cyclase 5; AMPAR—α-amino-3-hydroxy-5-methyl- 4-isoxazole- propionic acid receptor; ANA—anti-nuclear antibody; ANNA—anti-neuronal nuclear antibody; CASPR2—contactin-associated protein 2; CRMP5—collapsing response-mediator protein-5. DPPX—dipeptidyl aminopeptidase-like protein 6; GABAAR, γ-aminobutyric acid type-A receptor; GABABR, γ-aminobutyric acid type-B receptor; GAD—glutamic acid decarboxylase; GFAP—glial fibrillary acidic protein; GlyRα1, glycine receptor subunit alpha-1; IgLON5, immunoglobulin-like cell adhesion molecule IgLON family member 5; ITPR1—inositol trisphosphate receptor type 1; LE, limbic encephalitis; LGI1, leucine-rich glioma inactivated 1; mGluR1, metabotropic glutamate receptor 1; mGluR5, metabotropic glutamate receptor 5; NMDAR, N-methyl-D-aspartate receptor.
Table 3. Conditions with MRI features resembling those seen in CJD.
Table 3. Conditions with MRI features resembling those seen in CJD.
Main Brain Areas InvolvedConditionSimilar AspectsDifferencesSupplemental Clues to DiagnosisRef.
CortexSevere hypoxic-ischemic encephalopathyDWI, FLAIR and T2 hyperintensities in the cerebral cortex, hippocampus, basal ganglia
-
Involvement of the perirolandic areas
-
swelling of the affected areas
Acute onset following cardio-respiratory arrest, asphyxia, drowning[167]
Autoimmune encephalopathyDWI, FLAIR and T2 hyperintensities in the cortex, insula and cingulate areasFLAIR/T2 hyperintensities involving mainly hippocampus, amygdala, mesial temporal lobeSpecific autoantibodies[168]
Infectious encephalitis (herpetic encephalitis)Asymmetrical DWI and FLAIR/T2 hyperintensities in the cortex, mainly of the medial temporal lobe and orbitofrontal lobeT1 hyperintensity in the presence of necrosis and hemorrhageAcute onset, fever, stiff neck[169]
Postictal state following focal or generalized seizuresDWI, FLAIR/T2 hyperintensities in the hippocampus, neocortex, splenium of the corpus callosum, basal ganglia and thalamiAbnormalities are transient and disappear on subsequent imaging studies [170]
HyperammonemiaExtensive cortical signal abnormalities with restricted diffusion affecting mainly cingulate gyrus and insulaInvolvement of the perirolandic cortexIncreased serum ammonia[171]
Basal gangliaExtrapontine osmotic demyelinationBilateral symmetrical FLAIR/T2 hyperintensities in the globus pallidus, putamen, thalamusAbsent DWI abnormalitiesRapid correction of hypo- or hyperosmolar states[167]
Ebstein Barr virus encephalitisDWI/FLAIR/T2 hyperintensities in basal ganglia, cortex, and splenium of the corpus callosumAbnormalities are transient [169]
Autosomal dominant striatal degenerationDWI/FLAIR/T2 signal abnormalities in the striatum
-
Sparing of globus pallidus and thalamus
-
characteristic antero-posterior gradient in the putamen
Familial clustering[172]
ThalamusVariant CJDPulvinar and double hockey stick signsLack of cortical involvementYounger age of onset[173]
Wernicke encephalopathyFLAIR/T2 hyperintensities in the medial thalamus
-
Cortical involvement rare
-
signal abnormalities also in the periaqueductal region and mammillary bodies
Presence of conditions leading to thiamine deficiency[113]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jurcau, M.C.; Jurcau, A.; Diaconu, R.G.; Hogea, V.O.; Nunkoo, V.S. A Systematic Review of Sporadic Creutzfeldt-Jakob Disease: Pathogenesis, Diagnosis, and Therapeutic Attempts. Neurol. Int. 2024, 16, 1039-1065. https://doi.org/10.3390/neurolint16050079

AMA Style

Jurcau MC, Jurcau A, Diaconu RG, Hogea VO, Nunkoo VS. A Systematic Review of Sporadic Creutzfeldt-Jakob Disease: Pathogenesis, Diagnosis, and Therapeutic Attempts. Neurology International. 2024; 16(5):1039-1065. https://doi.org/10.3390/neurolint16050079

Chicago/Turabian Style

Jurcau, Maria Carolina, Anamaria Jurcau, Razvan Gabriel Diaconu, Vlad Octavian Hogea, and Vharoon Sharma Nunkoo. 2024. "A Systematic Review of Sporadic Creutzfeldt-Jakob Disease: Pathogenesis, Diagnosis, and Therapeutic Attempts" Neurology International 16, no. 5: 1039-1065. https://doi.org/10.3390/neurolint16050079

APA Style

Jurcau, M. C., Jurcau, A., Diaconu, R. G., Hogea, V. O., & Nunkoo, V. S. (2024). A Systematic Review of Sporadic Creutzfeldt-Jakob Disease: Pathogenesis, Diagnosis, and Therapeutic Attempts. Neurology International, 16(5), 1039-1065. https://doi.org/10.3390/neurolint16050079

Article Metrics

Back to TopTop