Prion-Related Diseases

Back

Background

The prion diseases are a large group of related neurodegenerative conditions, which affect both animals and humans.[1] Included are Creutzfeldt-Jakob disease (CJD) and Gerstmann-Sträussler-Scheinker (GSS) in humans, bovine spongiform encephalopathy (BSE, or "mad cow disease") in cattle, chronic wasting disease (CWD) in mule deer and elk, and scrapie in sheep. These diseases all have long incubation periods but are typically rapidly progressive once clinical symptoms begin. All prion diseases are fatal, with no effective form of treatment currently; however, increased understanding of their pathogenesis has recently led to the promise of effective therapeutic interventions in the near future.

Prion diseases are unique in that they can be inherited, they can occur sporadically, or they can be infectious. The infectious agent in the prion disease is composed mainly or entirely of an abnormal conformation of a host-encoded glycoprotein called the prion protein. The replication of prions involves the recruitment of the normally expressed prion protein, which has mainly an alpha-helical structure, into a disease-specific conformation that is rich in beta-sheet.[2]

The first of these diseases to be described was scrapie, a disease of sheep recognized for over 250 years. This illness, manifested by hyperexcitability, itching, and ataxia, leads to paralysis and death. It is called scrapie because of the tendency of affected animals to rub against the fences of their pens in order to stay upright, reflecting their cerebellar dysfunction. The transmission of this disease was demonstrated first in 1943 when a population of Scottish sheep was accidentally inoculated against a common virus using a formalin extract of lymphoid tissue from an animal with scrapie.[3] Accidental transmission of prions is a recurrent event in the history of these agents and is related to their unusual biophysical properties.

For related information, see Medscape Reference article Variant Creutzfeldt-Jakob Disease and Bovine Spongiform Encephalopathy.

Pathophysiology

A unifying feature of all the prionoses is their neuropathology. These illnesses tend to affect the gray matter of the central nervous system (CNS), producing neuronal loss, gliosis, and characteristic spongiform change. The latter is a vacuolation of the neuropil, and to a variable degree, of the neurons, shown below.



View Image

Prion-related diseases. Spongiform change in prion disease. This section shows mild parenchymal vacuolation and prominent reactive astrocytosis.

In addition, plaques with the typical staining properties of amyloid (eg, apple-green birefringence after Congo Red staining when viewed under polarized light) are observed in many of these conditions. In approximately 10% of patients with CJD, amyloid is present in the cerebellum or in the cerebral hemispheres. All cases of GSS are associated with multicentric cerebellar plaques. These amyloid plaques are immunoreactive with antibodies to the prion protein and do not immunoreact with antibodies to other amyloidogenic proteins, such as the amyloid-beta (which is deposited in Alzheimer disease).

Etiology of PrP-related diseases

Highly divergent hypotheses have been put forward regarding the makeup of the prions, including that they consist of nucleic acid only or protein only, are lacking both protein and nucleic acid, or are a polysaccharide. The most widely accepted hypothesis, first described by Griffith[4] and more explicitly detailed by Stanley Prusiner, MD, is the protein only hypothesis.[5] Prusiner introduced the term prion to indicate that scrapie is related to a proteinaceous infectious particle (PrP).[6]

This hypothesis was initially greeted with great skepticism in the scientific community; now it represents the current dogma, and Prusiner won the 1998 Noble Prize for Science. This hypothesis suggests that prions contain no nucleic acid and are referred to as PrPSc. The latter represents a conformationally modified form of a normal cellular PrPC, which is a normal host protein found on the surface of many cells, in particular neurons. PrPSc, when introduced into normal healthy cells, causes the conversion of PrPC into PrPSc, initiating a self-perpetuating vicious cycle.[5]

Other hypotheses for prion have included the virino hypothesis.[7] This hypothesis suggests that the infectious agent consists of a nucleic acid with host-derived PrPSc serving as a coat. The latter would explain the lack of an immunological and inflammatory response, while the presence of a nucleic acid provides an explanation for the numerous strains of scrapie, each with distinctive features. Other investigators have also suggested that the scrapie agent is a conventional virus with highly atypical properties. However, despite extensive searches, no nucleic acid associated with prion infection has been detected so far.

The protein-only hypothesis of prion propagation proposes the existence of an infectious agent composed solely of protein.[6] Recent reports claim that apart from the rare prion diseases, prion-like transmission of altered proteins may occur in several human diseases of the brain and other organs.[8, 9, 10]

Cell biology of prions - Normal cellular function of PrP

The human PrP gene (PRNP) is found on chromosome 20 and encodes a protein of 253 amino acids. PrPC is a glycosylphosphatidylinositol-anchored cell-surface glycoprotein; some have speculated that it may have a role in cell adhesion or signaling processes, but its exact cellular function remains unknown. The N-terminal region of PrP contains a segment of 5 repeats of an 8–amino acid sequence (ie, octapeptide repeat region) that contains a high-affinity binding site for copper ions; hence, PrP may have a role in copper transport or metabolism. Recent evidence suggests that copper imbalance is an early change during prion infections.[11] The function(s) of PrPC is likely to be of some importance because PrP is highly conserved among mammals and is found in all vertebrates.[12, 13] Also, prionlike proteins called PSI and URE3 are expressed in yeast.[14]

PrP is found in most tissues of the body but is expressed at highest levels in the CNS, in particular in neurons. PrP is also expressed widely on cells of the immune system. PrP knockout mice, which are engineered not to express the PrP gene, show no obvious pathological phenotype.[15] However, these mice have been shown to have abnormalities in synaptic physiology[16] and in circadian rhythms and sleep.[17]

The secondary structure of PrPC was first elucidated by nuclear magnetic resonance (NMR) imaging using recombinant mouse PrP protein.[18] More recently, this has been achieved using recombinant hamster and human PrP.[19, 20, 21] These studies have shown that PrPC is about 40% alpha-helix and about 3% beta-sheet. No high-resolution structural studies, such as NMR imaging, have been performed on PrPSc because it is highly insoluble and aggregated, which are properties that prevent use of these techniques. However, less exact structural methods such as circular dichroism and Fourier transform infrared spectroscopy have shown PrPSc to contain about 45% beta-sheet and 30% alpha-helix.[22, 23] This high beta-sheet content correlates with PrPSc resistance to enzymatic digestion and infectivity.

Prion strains and the species barrier

Many lines of evidence support the protein only hypothesis of prion propagation; however, a difficulty is the existence of several distinct isolates or strains of prions that can be stably passaged among inbred mice of the same genotype.[24] The existence of strains suggests that PrPSc could adopt multiple distinct pathological conformations. Strains are defined by the production of distinct patterns of incubation time, distributions of CNS involvement, and the pattern of proteolytic cleavage of PrPSc following proteinase K (PK) digestion.[5, 25] For example, at least 14 significantly different scrapie strains have been isolated from natural sheep scrapie by passage into mice.[24, 26]

The best studied are the two strains of transmissible mink encephalopathy (TME) called hyper (HY) and drowsy (DY).[27, 28] The truncated DY PrPSc fragments (PrP27-30) migrate 1-2 kd faster than similar preparations of HY because sites of PK cleavage differ and the two strains differ in terms of beta-sheet content.[28, 27]

Parchi et al defined 2 distinct types of sporadic CJD based on the analysis of PrP codon 129, which encodes either a valine or a methionine, and by the pattern of sodium dodecyl sulfate-polyacrylamide (SDS-PAGE) migration of the PrP27-30. Type 1 sporadic CJD has a molecular weight of the deglycosylated PrP27-30 of about 21 kd, while type 2 has a mobility of about 19 kd.[29] Collinge et al reported 2 further types related to infectious CJD.[30] These distinct types of sporadic CJD appear to have slightly different beta-sheet content that correlates with the degree of resistance to proteinase K digestion of each strain.[22]

Each strain of prion has characteristic range of infectivity. For example the 263K strain is pathogenic for hamsters but does not infect mice.[31] This effect is called a species barrier and is related to PrPSc being an effective template for homologous PrPC and a poor template for heterologous PrPC; hence, mouse PrPSc can effectively convert mouse PrPC, but it is a very poor template for human or hamster PrPC. This species barrier is not absolute, as is illustrated by the emergence of new variant CJD (vCJD).

The structure and folding properties of the cellular prion protein are well characterized, and, although its precise function remains enigmatic, constitutive knockout of protein expression in mice produces apparently healthy animals that are fully resistant to prion infection. In addition, data show that neuronal knockout of the gene encoding for prion protein during established brain infection leads to reversal of pathology and behavioral deficits.[32]

How prions reach the CNS

Prion diseases are transmitted naturally by peripheral routes, either orally or transcutaneously; hence, how prions are able to reach the CNS is an important issue. Although the prion diseases are neurological conditions, critical events in their pathogenesis take place in restricted sites out of the nervous system, especially in peripheral lymphoid organs.[33]

Lymphoid organs have long been known to be involved in the early stages of prion diseases.[34, 35, 36] In particular, the spleen and lymph nodes have been demonstrated to be the first sites of PrPSc replication after infection by peripheral routes, and they are also affected significantly following intracerebral challenge. Their importance for neuroinvasion after peripheral inoculation was suggested by studies showing that splenectomy and other methods that reduce peripheral lymphoid structures delay clinical manifestations.[35]

The hematogenic spread of prions to the CNS is suggested by experiments that show BSE to be transmissible from sheep to sheep by blood transfusion.[37] Three cases of vCJD infection associated with blood transfusion have also been observed (Health Protection Agency, Variant CJD and blood products). All received nonleucodepleted red blood cells.

The first case developed vCJD in 2003, 6.5 years following transfusion from a donor who developed vCJD 3.5 years following donation. The second patient died of causes unrelated to vCJD in 2004, 5 years following the transfusion. At autopsy, this individual had abnormal prion protein in the spleen and cervical lymph node but not in the brain, and other pathological features of vCJD were not observed. The donor developed symptoms of vCJD 18 months after his donation. The third patient developed vCJD in 2006, about 8 years following transfusion from a donor who was diagnosed with vCJD about 20 months after donation.

Hematogenic neuroinvasion has been shown to be dependent on the presence of B lymphocytes.[38] However, because expression of the cellular prion protein by B cells is not required for neuroinvasion, some have suggested that their main function is to allow maintenance of follicular dendritic cells.[39] However, more recent studies suggest that neuroinvasion is possible in the absence of both B cells and follicular dendritic cells.[40] Other studies have implicated the distinct CD11c+ dendritic cell population in prion neuroinvasion.[22, 41]

In addition to hematogenous spread, prions can reach the brain via the parasympathetic vagus nerve.[42] Hence, following intraperitoneal delivery of prions, disease can be delayed by sympathectomy or can be accelerated by sympathetic hyperinnervation of lymphoreticular organs.[43]

Which of these two routes for neuroinvasion is most important remains unclear; it may be scrapie strain–dependent. However, a more complete understanding of these stages and the cells involved in prion spread from the periphery may allow for development of a pharmacological gatekeeper that can be used to stop the movement of infectivity.

Epidemiology

Frequency

United States

The most common prion disease is CJD, with a uniform incidence of approximately 1 case per million population both in the United States and internationally. Familial forms of prion diseases, such as GSS and fatal familial insomnia (FFI), are much more rare. About 10% of cases of CJD are familial, with an autosomal dominant pattern of inheritance linked to mutations in the PRNP gene.

International

As of February 2006, 159 cases of definite or probable vCJD have been reported in the United Kingdom of which 153 persons have died (see The National Creutzfeldt-Jakob Disease Surveillance Unit). Whether these patients represent the beginning of a growing epidemic (such as that which occurred with BSE) or whether the number of cases will remain relatively low remains unclear. The first confirmed 3 cases were reported in 1995, with numbers of subsequent cases remaining relatively stable between 1996 and 2004 (9-28 cases per year). Only 5 cases were confirmed in 2005.

Two populations are disproportionally affected by CJD: Libyan-born Israelis and some populations in restricted areas of Slovakia where the incidence of CJD is 60-100 times greater than expected. These clusters were postulated to be related to dietary exposure of the scrapie agent; however, this was not supported by case-controlled studies. These local high rates of CJD are linked to a high prevalence of codon 200 mutations in the PRNP gene.

Mortality/Morbidity

Prion-related diseases are relentlessly progressive and invariably lead to death.

Race

Sporadic CJD occurs throughout the world in people of all races and typically has similar features.

Sex

No sex preponderance is known in prion diseases, with some rare exceptions. For example, women had a greater tendency than men to develop kuru because it was part of the ritual cannibalism for women to eat the brains (and neural tissue has the highest dose of PrPSc).

Age

The mean age of onset of sporadic CJD is 62 years. The incidence of sporadic CJD is about 1 case per million population; however, among individuals aged 60-74 years, the incidence is 5 cases per million population.[46] The age range can be broad; cases have been reported in people as young as 17 years and as old as 83 years.[47, 48]

vCJD occurs in younger patients, with a mean age of onset of 28 years.

Familial CJD, GSS, and FFI have mean ages of onset ranging from 45-49 years.

History

Several different forms of prion disease exist (see Table 1 below). The first human prionosis to be described is called kuru.[49, 50, 51] This is an illness of the Fore people living in the highlands of New Guinea that is thought to be linked to ritualistic cannibalism. Presumably, this illness originated with the consumption of an initial patient with sporadic CJD. Kuru was once the major cause of death among Fore women; however, the disease has virtually disappeared with the end of cannibalistic rituals. Similar to scrapie, patients clinically present with difficulty walking and they develop progressive signs of cerebellar dysfunction. Death occurs approximately 1 year following onset of symptoms.

The neuropathology of kuru, in common with all prionoses to a variable extent, includes widespread spongiform change and astrocytosis, as well as neuronal loss affecting the cerebral hemispheres and cerebellum. More intraneuronal vacuolation is observed in kuru compared to CJD (see below). In about 70% of cases, amyloid plaques are found, with amyloid deposition being a common, but not invariable, accompaniment of the prionoses. Gajdusek's detailed description of this illness led Hadlow to suggest that kuru might be the human representation of scrapie.[4] This in turn inspired Gajdusek and his team to test whether kuru was also transmissible. In 1966, they first showed kuru was transmissible to chimpanzees, after a long incubation.[52] Gajdusek was awarded the Noble Prize in 1976 for this work.

Table 1. Prion-Related Diseases, Hosts, and Mechanism of Transmission



View Table

See Table

By far the most common human prion disease is CJD, accounting for about 85% of all human prion disease. CJD was initially described by Jacob in 1921[53] ; ironically, the case reported by Creutzfeldt a year earlier is probably unrelated to the disease that carries his name. Clinically, CJD is characterized by a rapidly progressive dementia associated with myoclonic jerks, as well as a variable constellation of pyramidal, extrapyramidal, and cerebellar signs. The EEG findings typically show distinctive changes of high-voltage slow (1-2 Hz) and sharp wave complexes on an increasingly slow and low-voltage background. CJD is found throughout the world, with an incidence of about 1 case per million population. In addition to extensive cortical spongiosis, gliosis, and neuronal loss, 10% of CJD cases have amyloid plaques.[5] Ten percent of cases of CJD are familial, with an autosomal dominant pattern of inheritance linked to mutations in the PrP gene.[54]

Creutzfeldt-Jakob disease

Sporadic CJD is characterized by a rapidly progressive multifocal neurological dysfunction, myoclonic jerks, a terminal state of global severe cognitive impairment, and death in about 8 months.

About 40% of patients with sporadic CJD present with rapidly progressive cognitive impairment, 40% with cerebellar dysfunction, and the remaining 20% with a combination of both.

The clinical picture rapidly expands to include behavioral abnormalities, higher cortical dysfunction, cortical visual abnormalities, cerebellar dysfunction, and both pyramidal and extrapyramidal signs.

Almost all patients with sporadic CJD develop myoclonic jerks that involve either the entire body or a limb. These myoclonic jerks can occur spontaneously or can be precipitated by auditory or tactile stimulation.

During the course of sporadic CJD, most patients develop a characteristic picture on EEG with periodic or pseudoperiodic paroxysms of sharp waves or spikes on a slow background. These periodic complexes have a sensitivity and specificity of 67% and 87% respectively on a single EEG. However, if repeated recordings are obtained, more then 90% of patients show periodic EEG abnormalities.[55]

When evaluating a patient for possible sporadic CJD, the clinician should be guided by published case definitions; they are as follows:

Gerstmann-Sträussler-Scheinker disease

Gerstmann-Sträussler-Scheinker disease was described in a large kindred in 1936.[56]

Patients with this illness present with a slowly progressive limb and truncal ataxia, as well as dementia. Death occurs 3–8 years following presentation.

The prominent involvement of the brainstem often leads to symptoms suggestive of olivopontocerebellar degeneration. The pattern of inheritance is autosomal dominant and is caused by mutations of the PrP gene. The neuropathology of GSS is remarkable in that extensive and invariable amyloid deposition occurs, in addition to the typical spongiform change, gliosis, and neuronal loss. Interestingly, in several kindreds of GSS, extensive neurofibrillary tangle (NFT) formation is found.[57] NFTs are an essential feature of Alzheimer disease, but are also observed in other neurodegenerative conditions.

Another variation of autosomal dominantly inherited human prionosis has been termed prion protein congophilic angiopathy (ie, prion protein cerebral amyloid angiopathy [PrP-CAA]), which is characterized by cerebral vessel amyloid deposition and the presence of NFT.[58] Cerebral amyloid angiopathy (CAA) is also an essential feature of Alzheimer disease. Both these variants of prionoses further link the pathogenesis of Alzheimer disease and the prion-related diseases.

Fatal familial insomnia

Patients with FFI present with intractable insomnia, dysautonomia (ie, hyperthermia, hypertension, tachycardia, tachypnea, hyperhydrosis), dementia, and motor paralysis; however, the phenotypic expression is very variable even within the same family.[59] The age of onset is also variable, ranging from 18-60 years. Once symptoms begin, the course ranges from 6 months to 3 years. Because of the diversity of clinical presentations of this disorder, genotyping is very important for definitive diagnosis. Neuropathologically, marked atrophy of the anterior ventral and mediodorsal thalamic nuclei occurs because of neuronal loss and gliosis. Unlike other prionoses, spongiform change can be a minor feature or can be absent altogether.

All patients with FFI have a missense mutation at codon 178 of the PrP gene where Asn is replaced by Asp, coupled with a Met at the polymorphic codon 129.[60] The somewhat divergent clinical and neuropathological features of FFI, in comparison to other human prionoses, highlight the wide spectrum of disease associated with PrP dysfunction and suggest that other human illnesses have yet to be recognized as prionoses.

Fatal familial insomnia and Creutzfeldt-Jakob disease are associated with a D178N mutation of the PRNP gene located on chromosome 20. D178N mutation changes the aspartate to asparagine at codon 178. In this disease, the mutant chromosome encodes methionine in the polymorphism of codon 129. The cortex is spared but the thalamus is particularly susceptible to this type of prion disease; therefore, fatal insomnia is situated at the extreme end of a spectrum of prion diseases with frequent psychiatric presentations.[61]

There is an unusual incidence of this disease in Basque Country (Spain). Oliveros et al report a patient with postmortem diagnosis of fatal insomnia who had a phenotypic presentation of catatonia and they stress the importance of considering this disease in catatonia nonresponsive to ECT.[62]

Variant Creutzfeldt-Jakob disease

A recent epidemic of a new prionosis has occurred; BSE has led to more then 160,000 cattle deaths in the United Kingdom.[63] This new disease is thought to be caused by meat and bone meal dietary supplements to cattle that were contaminated with scrapie-infected sheep and other cattle with BSE. Extensive evidence suggests that BSE has also lead to a new type of CJD, called variant CJD (vCJD).[64] The first cases of vCJD were reported in 1995, when CJD was found in 2 British teenagers.[65, 66]

Only 4 cases of sporadic CJD have been reported previously among teenagers; the peak incidence of onset of sporadic CJD is in people aged 60-65 years. In addition to the early age, these cases had distinctive neuropathology that included so-called florid amyloid plaques, which are reminiscent of kuru-associated PrP amyloid plaques.[67, 68] Significantly, such florid amyloid plaques are also a feature of chronic wasting disease.[69]

As of February 2006, 159 cases of vCJD have been diagnosed in Great Britain (see The National Creutzfeldt-Jakob Disease Surveillance Unit). The latest numbers from other countries as of November 2005 are 15 in France, 3 from Ireland, 2 in the United States, and one each from Canada, Italy, Japan, Netherlands, Portugal, Saudi Arabia, and Spain (see Centers for Disease Control and Prevention, Variant Creutzfeldt-Jakob Disease). Both of the US cases, 1 of the 3 in Ireland, and the single cases from Canada and Japan were likely exposed while living in the UK. The emergence of vCJD has raised the specter of an epidemic of prion-related disease among the British population (and possibly a wider population) similar to that of BSE in cattle.

Physical

Physical signs and symptoms vary with the type of prion disease. vCJD differs from sporadic CJD in that psychiatric abnormalities and sensory symptoms are much more common at presentation of vCJD.

In 2009, Kahn et al reported a patient with inherited prion disease who sustained fractures that were successfully managed conservatively with unusual results, such as accelerated healing, akin to that seen in traumatic head injuries. Local growth factors, inflammatory cytokines, and endogenous bone morphogenic proteins have all been implicated in head injuries and the authors propose that similar factors may be responsible in prion disease, common to both conditions.[70]

Causes

Prion-related diseases are unique in that they can be related to infectious, sporadic, or familial causes (see Pathophysiology).

Infectious causes

Kuru, a form of prion disease, occurred among the Fore people of the Eastern Highlands of New Guinea and was related to ritualistic cannibalism. The disease is believed to have started with the ingestion of body parts of a patient with sporadic CJD, followed by a serial passage of the disease.

Many cases of iatrogenic CJD have been reported. In clinical practice, CJD has been transmitted by surgical instruments, EEG electrodes, corneal transplants, dura mater grafts, human pituitary-derived gonadotrophins, and human-derived growth hormone. Concerns that vCJD could be transmitted by blood transfusion have been borne out with 3 documented case.

vCJD in humans is presumed to have been caused by ingestion of beef products contaminated with BSE. BSE is presumed to have started because of the practice of supplementing the diets of calves and dairy cows with meat and bone products. These meat and bone products are thought to have been contaminated with scrapie material (from sheep) and/or with material from cattle with a sporadic form of bovine prion disease.

A number of cases of apparent sporadic CJD have occurred in the United States among young individuals (< 30 y). The incidence of sporadic CJD among such young individuals has historically been about 1 case per billion population. In the years 1979-1996, 4 cases of sporadic CJD were reported in the United States among individuals younger than 30 years. In the years 1997-2000, 5 cases have occurred in the United States among young patients. Two of these individuals came from adjacent counties in Michigan (ages at onset were 26 and 28 y), and 3 cases occurred among individuals who were known hunters of deer and/or elk.[71]

Over the same period, a major outbreak of CWD occurred among the deer and elk populations in many western states, which has now spread to at least 10 states (see Chronic Wasting Disease Alliance). CWD is a form of prion disease that occurs naturally in the deer and elk population; however, the pathology has many similarities to BSE, including the presence of florid plaques.[69] Significantly, transmission studies of CWD PrPSc in the laboratory have shown that it can cross the species barrier from deer to human PrP at about the same efficiency as the BSE prion agent.[72] These observations have led to the speculation that limited transmission of CWD to humans has occurred recently in the United States.

Findings indicated that transgenic mice that express the deer/elk prion protein can be infected with intracerebral injection of muscle tissue from symptomatic cervids,[73] which raises concerns about infectivity of meat products from these animals. Also, a nonhuman primate developed prion disease after intracerebral injection with brain material from symptomatic deer.[74]

Familial causes

The cause of familial forms of prion disease is related to mutations in the PrP gene. A number of mutations in the PrP gene are linked to autosomal dominant forms of prion disease. The image below is a representation of the human PrP gene, PRNP.



View Image

Prion-related diseases. A representation of the human proteinaceous infectious particle, or PrP, gene. Mutations associated with inherited prionoses a....

A signal peptide of 22 amino acids (dotted area) is cleaved at the amino terminus (N-terminus) synthesis, and a further sequence at the carboxyl terminus (dotted area) is removed during the addition of a glycosyl-phosphatidylinositol anchor (GPI).

Mutations associated with inherited prionoses are shown above the gene, while polymorphisms are shown below the gene. A polymorphism at codon 129 (M versus V) is common in white populations, while a polymorphism at codon 219 (E versus K) is common in Japanese populations.

The locations of the 4 putative helical regions are indicated by the boxes labeled H1 through H4, corresponding to residues 144-154, 179-193, and 200-218, respectively.

The diagonal striped area represents the region of octarepeats, spanning residues 51-91. Octarepeats of 16, 32, 40, 48, 56, 64, or 72 amino acids at codons 67, 75, or 83 are indicated by the rectangle above the octarepeat region. These inserts are associated with familial CJD.

Mutations at codons 102, 105, and 117 have been associated with GSS, while mutations at codons 198 and 217 are found in pedigrees with GSS and NFTs.

PrP-CAA has been linked to a point mutation at codon 145 that results in a stop codon. Familial CJD has been associated with mutations at codons 178, 180, 200, 210, and 232.

Interestingly, kindreds with FFI have the same D178N mutation as 178 familial CJD kindreds; however, the FFI phenotype is associated with a Met at codon 129, whereas the mutated allele in 178 CJD patients has a Val at the polymorphic codon 129.

Sporadic causes

Sporadic CJD is the most common form of prion disease.

It probably arises as a spontaneous conformational change in PrPC to a PrPSc form. The PrPSc form is then self-propagating, inducing more PrPC to convert to the PrPSc form.

The risk of transmission depends on both the type of procedure and the type of tissue involved, with brain, spinal cord, and eye having the highest risk.

To study the association between medical procedures and sporadic Creutzfeldt-Jakob disease (sCJD), Hamaguchi et al analyzed medical procedures (any surgical procedure, neurosurgery, ophthalmic surgery, and blood transfusion) for patients registered by the CJD Surveillance Committee in Japan from 1999–2008. The study included 753 patients with sCJD and 210 controls and patients who underwent neurosurgical or ophthalmic surgical procedures at the same hospital. No evidence was found that prion disease was transmitted through the investigated medical procedures before the onset of sCJD. After the onset of sCJD, 4.5% of the patients with sCJD underwent operations, and no special precautions against transmission of prion diseases were taken. The authors have not identified patients with prion disease attributed to these operations and conclude that surgical procedures or blood transfusion has little effect on the incidence of sCJD.[75]

Transfusion transmission of the prion, the agent of variant Creutzfeldt-Jakob disease (vCJD), is now established. Subjects infected through food may transmit the disease through blood donations.

Laboratory Studies

Initial workup includes laboratory tests as for dementia. To rule out a toxic and/or metabolic encephalopathy, evaluate CBC count, serum chemistry panel, liver function test results, ammonium levels, and erythrocyte sedimentation rate. Perform blood cultures if any type of bacterial infection is suspected.

In addition, evaluate thyroid function, B-12 levels, and folate levels and perform tests for neurosyphilis. The rapid plasma reagent (RPR) test or Venereal Disease Research Laboratory (VDRL) test results may be negative in neurosyphilis; hence, if this diagnosis is suspected, perform a fluorescent treponemal antibody test. Perform HIV testing if any risk factors are present.

To rule out Hashimoto encephalopathy, serum levels of anti-thyroperoxidase antibodies (formally known as antimicrosomal antibody) should be determined.[78]

If a paraneoplastic syndrome is possible (ie, when a history of malignancy or imaging studies finding an occult neoplasm), then the presence of autoantibodies should be investigated. These are reviewed in Dropcho 2005. A partial list of possible paraneoplastic syndromes and their associated tumors and autoantibodies is shown in Table 2.

Table 2. Paraneoplastic Syndromes, Associated Tumors, and Autoantibodies



View Table

See Table

 

Imaging Studies

MRI is an important imaging test. MRI may show hyperintense signals in the cortical ribbon, basal ganglia, and the thalamus on diffusion-weighted images (DWI) and fluid-attenuated inversion recovery (FLAIR) images. See the image below. In a recent review, DWI and FLAIR imaging was found to be 91% sensitive, 95% specific, and 94% accurate for the diagnosis of sporadic CJD.[80]



View Image

Shows characteristic signal changes of an MRI taken from a patient with sporadic CJD, using diffusion-weighted imaging (DWI). An abnormal signal is sh....

Two characteristic radiological signs have been described. The "hockey stick" sign, which refers to increased signal in the putamen and head of the caudate nucleus resembling a hockey stick, and the "pulvinar" sign, which corresponds to a usually bilaterally increased signal in the pulvinar thalamic nuclei. The latter sign has been found especially in patients with vCJD.

In a few patients, positron emission tomography (PET) scan has been performed in which regional hypometabolism of glucose was noted that correlated with the neuropathologic lesions found at autopsy.

CJD presents with multifaceted clinical signs including ataxia, visual, pyramidal, myoclonus, limb apraxia, limb dystonia, sensory, parkinsonism, and corticobasal syndrome (CBS). In their effort to study the relationship between clinical pattern and cerebral glucose metabolism on [18F]fluoro-2-deoxy-D-glucose positron emission tomography (FDG-PET) in CJD, Renard et al. report hypometabolism in brain areas related to some specific signs (i.e., ataxia, visual signs, and CBS), in addition to lateralized frontal and parietal hypometabolism previously reported in CJD.[81]  

In addition to MRI and PET imaging to finding changes characteristic of prion disease, it is often prudent to obtain imaging (contrast CT scans) of the chest, abdomen, and pelvis in patients to rule out the possibility of a malignancy that could be producing a paraneoplastic syndrome.

Other Tests

EEG

During the course of sporadic CJD, most patients develop a characteristic finding on EEG with periodic or pseudoperiodic paroxysms of sharp waves or spikes on a slow background.

These periodic complexes have a sensitivity and specificity of 67% and 87%, respectively, on a single EEG. However, if repeated recordings are obtained, more then 90% of patients show periodic EEG abnormalities.

In vCJD, EEG does not show the typical changes observed in sporadic CJD, and findings often are normal.

Procedures

Perform lumbar puncture (LP) in all suspected cases.

Disease-associated prion proteins include both PrPC and PrPSc. To detect low levels of this disease marker in biological material with high sensitivity, reagents that can precipitate the disease-associated prion protein are vital. Rees et al report that among the range of minerals available for this purpose, silicon dioxide is unaffected by the PrPSc strain or host species and the method can be used to precipitate bovine BSE.[82] This method can also reliably concentrate protease-resistant ovine PrPSc (PrPres) and has increased detection sensitivity by more than 1,500-fold.

Different isolates from the same host species with transmissible spongiform encephalopathies (TSEs) may show different eletrophoretic profiles, reflecting the existence of different prion strains. Experimental transmission of these atypical cases to various transgenic mouse lines has led to the recognition of a novel scrapie strain in sheep and goats, called Nor98, and of 2 variant strains of spongiform encephalopathy in cattle.[83]

In this study, streptomycin demonstrated its efficiency to detect PrPres both in the central nervous system and in the lymphoid tissue without practical difficulty and with rapid preparation. Because of its ability to act as a good agent for PrPSc examination in different tissues, recovery of PrPSc in biological fluids using streptomycin should open further perspectives of applications in CJD diagnostics. Streptomycin effects in vivo might thus also be questioned.

Histologic Findings

See Pathophysiology.

Staging

Clinical diagnosis is sometimes difficult (particularly in atypical sCJD cases such as MM2, MV2, VV1, or VV2 types) according to 6 phenotypes of sCJD divided by codon 129 polymorphisms of PrP (methionine/valine) and type of infectious PrP by Western blotting.

Medical Care

Discontinue any medication that could impair memory or cause confusion.

A number of potential therapeutic interventions are under current development, as discussed in Medication.

The transmissible spongiform encephalopathies are rapidly progressive neurodegenerative diseases, and outcome is inexorably fatal. No treatments have proven efficacious. Chemotherapeutic approaches have focused on blocking the conversion of the normal form of prion protein (PrPC) to its abnormal counterpart (PrPres) either by directly binding PrPC or PrPres, or by redistributing, sequestering, or downregulating PrPC, thus preventing its conversion. Others aim to enhance the clearance of PrPres. Other targets include accessory molecules such as the laminin receptor precursor, which influences conversion (or cell-signaling molecules) that may be required for pathogenesis.[84]

Other promising therapeutic approaches aimed to block the production of PrPSc are based on PrP RNA interference, passive or active immunization, dominant negative inhibition of PrPSc formation, as well as inhibition of interactions between PrPSc and other cofactors. An alternative strategy consists of combining gene therapy with cell therapy.[85]

Evidence is accumulating that PrPC may act as a receptor for protein aggregates and transduce neurotoxic signals in more common neurodegenerative disorders, such as Alzheimer's disease.

A general consensus on the role of PrPC and its physiological function within the brain is yet to be established. Lowering PrPC levels in the brain is predicted to be a powerful therapeutic strategy for the treatment of prion disease and the precise reason for PrPC's existence continues to remain enigmatic. 

Miguelez-Rodriguez et al. report that the existence of copathology significantly prolongs survival in patients with rapidly progressive dementia due to CJD. To further understand the molecular mechanisms behind prion diseases they suggest the study of major neurodegenerative-associated proteins in brains with CJD.[86]  

Consultations

You may wish to consult with a neurologist and/or an infectious disease specialist.

Medication Summary

All prion diseases are fatal; no effective treatment is available. Patients are currently provided symptomatic treatment. Hence, some patients with CJD who develop seizures should be administered antiepileptic drugs, while those with extrapyramidal symptoms should be administered anti-Parkinson drugs.

A number of medications have been shown in experimental systems to be effective at preventing prion propagation. These have included Congo red and its analogs,[87, 88, 89, 90] anthracyclines[91] , amphotericin B and its analogs,[92, 93, 94, 95, 96] sulfated polyanions,[97, 98, 99] and tetrapyrroles.[100, 101] Some of these have been shown to delay the incubation times of animals infected with scrapie, but these agents have limitations in terms of toxic effects and/or unfavorable pharmacokinetic properties. Of these compounds, amphotericin B failed to ameliorate CJD in a single patient.[102]

In addition, tissue culture studies have shown that acridine and phenothiazine derivatives (eg, quinacrine, chlorpromazine) can inhibit the conversion of PrPC to PrPSc.[103] These types of drugs have been used in humans for many years as antimalarial and antipsychotic drugs; however, recent reports of anecdotal use of these agents in limited numbers of patients with sporadic CJD or vCJD have so far not supported their use,[104] and animal studies have also been negative.[105, 89, 106] An extensive clinical trial (PRION-1) on this approach was initiated in the UK in 2004 (see National Prion Clinic).

Another compound that is being tested in patients is pentosan polysulphate, which seemed promising based on an animal study.[89] This compound does not cross the blood-brain barrier and has been delivered by intraventricular administration to symptomatic patients. In the only reported vCDJ case study using this approach, no obvious side effects were observed, and the clinical symptoms appeared to be slightly attenuated, although brain atrophy progressed based on CT scans.[107]

The author and their colleagues have recently designed a number of compounds that interact with the PrPSc structure and act as beta-sheet breakers,[108, 109, 110] inhibiting the conformation of PrP associated with disease. These compounds were designed by first synthesizing a large number of different PrP homologous peptides. These were then screened for inhibitory activity on the conversion of PrPC to PrPSc using an in vitro system. That synthetic peptides corresponding to PrP residues 109-141 can reproduce some of the properties of PrPSc in vitro is well known.[111, 112, 113] The author's studies determined the ability of these various candidate beta-sheet breaker peptides to inhibit amyloidlike fibril formation of PrP109-141 using a fluorometric assay based on the fluorescence emission of thioflavine T.[114, 115] This assay showed that a 13-residue peptide (iPrP13) had the greatest beta-sheet–breaking capability. Using this peptide,the author and his colleagues were able to show that the proteinase K sensitivity of extracted mouse PrPSc and of human PrPSc extracted from patients with sporadic CJD or from patients with vCJD was increased in a concentration-dependent fashion by iPrP13.[108]

The in vivo effect of iPrP13 was tested by using mouse-adapted scrapie strain 139A. Incubation time assays were performed using 3 different 10-fold dilutions of extracted 139A PrPSc, in the presence or absence of an equimolar concentration of iPrP13. At each dilution, one group of mice was injected with untreated and nonincubated PrPSc, a second group was inoculated with PrPSc that was incubated for 48 hours alone, a third group was inoculated with PrPSc and nonincubated iPrP13, and a fourth group was inoculated with PrPSc and iPrP13 incubated for 48 hours. iPrP13 induced a substantial delay in the appearance of disease. These results suggest that beta-sheet breakers may have therapeutic potential in the prionoses.

This type of therapeutic approach, in which the disease-associated abnormal protein conformation is the target, currently is under extensive investigation for the prionoses as well as for other conformational disorders, such as Alzheimer disease.[116, 117]

Other more recent approaches include chelation therapy. Copper has been implicated in prion propagation,[118] and the authors have demonstrated that a chelator, D-penicillamine, which selectively chelates copper, delays the onset of prion disease in infected mice.[119] In vitro, copper enhanced the proteinase K resistance of the prion protein, which was counteracted by co-incubation with D-penicillamine. Overall, these findings indicate that copper levels can influence the conformational state of PrP, thereby enhancing its infectivity, and this effect can be attenuated by chelator-based therapy.

An additional therapeutic approach that may be of benefit for the prion diseases is immunological.[120] A number of recent reports have shown that immunization with alpha-helix peptides is highly successful at reducing cerebral amyloid accumulation, a key neuropathologic feature in Alzheimer disease, in transgenic mouse models of that disease.[121, 122, 123, 124]

Alpha-helix peptides are normal constituents of biological fluids such as blood and CSF at low concentrations, and they are the major component of the amyloid deposits that characterize Alzheimer disease. Passive immunization studies in the Alzheimer disease model mice suggest that an antibody-mediated clearance of alpha-helix peptides is critical for a therapeutic response.[125] The authors have extended this immunological approach to prion disease and suggest that this approach can be applied to all members of the extended category of neurodegenerative conformational diseases.[126, 127, 128, 129]

Interestingly, prior work has shown that vaccination with an attenuated CJD strain can prevent expression of a more virulent strain.[130] The authors have reported that vaccination with recombinant mouse prion protein (recPrP) delays the onset of prion disease in mice.[126] Vaccination was performed both prior to peripheral prion exposure and after exposure. A delay in disease onset was observed in both groups but was more effective in animals immunized prior to exposure. The increase in the incubation period closely correlated with the anti-PrP antibody titer. The mechanism of the delay with vaccination is not clear, but the correlation of the increased incubation time with antibody titer. The authors subsequently showed that passive immunization in mice using anti-PrP antibodies prolonged the incubation times, which suggests that humoral immunity is critical for a therapeutic response.[129]

Subsequent mouse studies using much higher doses of antibodies completely prevented symptoms of prion disease.[131] Antibody administration is unlikely to be used prophylactically in large populations because of high cost but can potentially be used in humans following accidental exposure.

The authors were recently able to prevent symptoms in about 30% of infected mice by administering a Salmonella -based prion vaccine,[127] which because of low cost has the potential to be used prophylactically in livestock and perhaps also in high-risk human populations. Antibody binding to PrPC and/or PrPSc may possibly interfere with PrPSc -mediated conversion of PrPC to PrPSc and thereby delay the onset of clinical symptoms. Recent in vitro studies support this view,[132, 133] and immunization with prion peptides of 20 amino acids has been shown to reduce the levels of PrPSc in scrapie-infected mouse tumors[134] without affecting PrPC levels. Hence, epitope mapping of the anti-PrP antibodies produced by immunization may provide insights on which portions of the prion molecule are important for prion replication.

The ultimate goal of such immunological approaches is for human testing; the recent problems with the phase II clinical trial of A-beta1-42 vaccination for Alzheimer disease highlight the difficulties of translating successful therapeutic approaches from mouse models to humans (in this trial of A-beta1-42, a significant number of patients developed encephalitis as a complication). A number of potential toxic side effects of vaccine-based approaches exist in humans; further animal and in vitro experimentation is required. One source of potential toxicity is from the immunogen that is used. In the authors' Alzheimer disease vaccine development studies, the A-beta sequence was altered, making it nonfibrillogenic and nontoxic while maintaining or increasing its immunogenicity, to overcome this source of toxicity.[135, 136]

Similar types of alterations are underway to limit any potential toxicity from using the native PrP sequence as an immunogen. The authors' in vivo findings serve as a starting point for the development of vaccine-based approaches for the prion diseases and suggest that prion-based immunization is promising as a potential therapy.

Further Outpatient Care

The rate of progression of prion diseases is rapid. Frequent follow-up care is necessary to assess the need for symptomatic treatments. If the patient develops seizures, parkinsonian features, or behavioral problems, appropriate pharmacological treatments can be administered.

Further Inpatient Care

The initial steps of the dementia workup (eg, LP) can be performed most speedily if the patient is admitted to the hospital.

If the diagnosis is not clear, inpatient care speeds up referral for a brain biopsy.

Deterrence/Prevention

Prion diseases may spread by iatrogenic means. Hence, take care not to reuse EEG and/or electromyography (EMG) needles, surgical instruments, and other tools that have been exposed to a patient with prion disease. The prion agent is remarkably resistant to inactivation; hence, routine sterilization procedures, such as autoclaving, are ineffective.

As the incidence of BSE in Europe continues to decline, iatrogenic transmission from person to person is considered a serious threat to public health.

Preventive measures include donor deferral policies, technologies for prion removal from labile blood components and prion detection in plasma, and establishing a sensitive and rapid reference assay able to confirm the positive results from any putative blood screening assay.[137]

Interestingly, passive immunization with antibodies against prion protein (PrP), a major component of the prion infectious agents, was shown to protect mice from infection, indicating the possibility of prion vaccines. However, PrP is a host protein; therefore, immune tolerance to PrP has hampered development of them.

In the absence of a large-scale screening test, it is impossible to establish the prevalence of infection in the blood donor population and transfused patients. This lack of a test also prevents specific screening of blood donations. Since leukoreduction is probably insufficient to totally eliminate the transfusion risk, recently developed prion-specific filters could be a solution.[138]

Complications

Any part of the CNS may be involved during the progression of prionoses; therefore, all types of CNS complications may be observed.

Prognosis

The prionoses are rapidly progressive. The median survival duration from the time of diagnosis to death varies from 8 months (as in sporadic CJD) to 60 months (as in GSS).

Patients with familial prion-related disease tend to have a longer course than those with sporadic disease.

Patient Education

For excellent patient education resources, visit eMedicineHealth's Brain and Nervous System Center. Also, see eMedicineHealth's patient education article Mad Cow Disease and Variant Creutzfeldt-Jakob Disease.

Author

Tarakad S Ramachandran, MBBS, MBA, MPH, FAAN, FACP, FAHA, FRCP, FRCPC, FRS, LRCP, MRCP, MRCS, Professor Emeritus of Neurology and Psychiatry, Clinical Professor of Medicine, Clinical Professor of Family Medicine, Clinical Professor of Neurosurgery, State University of New York Upstate Medical University; Neuroscience Director, Department of Neurology, Crouse Irving Memorial Hospital

Disclosure: Nothing to disclose.

Coauthor(s)

Arun Ramachandran, MD, State University of New York Upstate Medical University

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Florian P Thomas, MD, PhD, MA, MS, Chair, Neuroscience Institute and Department of Neurology, Director, National MS Society Multiple Sclerosis Center and Hereditary Neuropathy Foundation Center of Excellence, Hackensack University Medical Center; Founding Chair and Professor, Department of Neurology, Hackensack Meridian School of Medicine at Seton Hall University; Professor Emeritus, Department of Neurology, St Louis University School of Medicine; Editor-in-Chief, Journal of Spinal Cord Medicine

Disclosure: Nothing to disclose.

Chief Editor

Niranjan N Singh, MBBS, MD, DM, FAHS, FAANEM, Adjunct Associate Professor of Neurology, University of Missouri-Columbia School of Medicine; Medical Director of St Mary's Stroke Program, SSM Neurosciences Institute, SSM Health

Disclosure: Nothing to disclose.

Additional Contributors

Roberta J Seidman, MD, Associate Professor of Clinical Pathology, Stony Brook University School of Medicine; Director of Neuropathology, Department of Pathology, Stony Brook University Medical Center

Disclosure: Nothing to disclose.

Acknowledgements

The authors and editors of Medscape Reference gratefully acknowledge the contributions of previous authors Thomas Wisniewski, MD and Einar M Sigurdsson, PhD to the development and writing of this article.

References

  1. Sadowski M, Verma A, Wisniewski T. Prion Diseases. Bradley WG, Daroff RB, Fenichel GM, Jankovic J, eds. Neurology in Clinial Practice. Philadelphia: Elsevier Inc; 2004. 1613-1630.
  2. Takada LT, Geschwind MD. Prion diseases. Semin Neurol. 2013 Sep. 33(4):348-56. [View Abstract]
  3. Gordon WS. Advances in veterinary research. Vet Rec. 1946. 58:518-525.
  4. Griffith JS, Hadlow WJ. Scrapie and kuru. Lancet. 1959. 11:289-290.
  5. Prusiner SB, Scott MR, DeArmond SJ, Cohen FE. Prion protein biology. Cell. 1998 May 1. 93(3):337-48. [View Abstract]
  6. Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. 1982 Apr 9. 216(4542):136-44. [View Abstract]
  7. Weissmann C. The Ninth Datta Lecture. Molecular biology of transmissible spongiform encephalopathies. FEBS Lett. 1996 Jun 24. 389(1):3-11. [View Abstract]
  8. Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009 Jul. 11(7):909-13. [View Abstract]
  9. Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem. 2009 May 8. 284(19):12845-52. [View Abstract]
  10. Ren PH, Lauckner JE, Kachirskaia I, Heuser JE, Melki R, Kopito RR. Cytoplasmic penetration and persistent infection of mammalian cells by polyglutamine aggregates. Nat Cell Biol. 2009 Feb. 11(2):219-25. [View Abstract]
  11. Thackray AM, Knight R, Haswell SJ, Bujdoso R, Brown DR. Metal imbalance and compromised antioxidant function are early changes in prion disease. Biochem J. 2002 Feb 15. 362:253-8. [View Abstract]
  12. Harris DA, Lele P, Snider WD. Localization of the mRNA for a chicken prion protein by in situ hybridization. Proc Natl Acad Sci U S A. 1993 May 1. 90(9):4309-13. [View Abstract]
  13. Windl O, Dempster M, Estibeiro P, Lathe R. A candidate marsupial PrP gene reveals two domains conserved in mammalian PrP proteins. Gene. 1995 Jul 4. 159(2):181-6. [View Abstract]
  14. Masison DC, Edskes HK, Maddelein ML, Taylor KL, Wickner RB. [URE3] and [PSI] are prions of yeast and evidence for new fungal prions. Curr Issues Mol Biol. 2000 Apr. 2(2):51-9. [View Abstract]
  15. Büeler H, Aguzzi A, Sailer A, et al. Mice devoid of PrP are resistant to scrapie. Cell. 1993 Jul 2. 73(7):1339-47. [View Abstract]
  16. Collinge J, Whittington MA, Sidle KC, et al. Prion protein is necessary for normal synaptic function. Nature. 1994 Jul 28. 370(6487):295-7. [View Abstract]
  17. Tobler I, Gaus SE, Deboer T, et al. Altered circadian activity rhythms and sleep in mice devoid of prion protein. Nature. 1996 Apr 18. 380(6575):639-42. [View Abstract]
  18. Riek R, Hornemann S, Wider G, Billeter M, Glockshuber R, Wuthrich K. NMR structure of the mouse prion protein domain PrP(121-321). Nature. 1996 Jul 11. 382(6587):180-2. [View Abstract]
  19. Hosszu LL, Baxter NJ, Jackson GS, et al. Structural mobility of the human prion protein probed by backbone hydrogen exchange. Nat Struct Biol. 1999 Aug. 6(8):740-3. [View Abstract]
  20. James TL, Liu H, Ulyanov NB, et al. Solution structure of a 142-residue recombinant prion protein corresponding to the infectious fragment of the scrapie isoform. Proc Natl Acad Sci U S A. 1997 Sep 16. 94(19):10086-91. [View Abstract]
  21. Knaus KJ, Morillas M, Swietnicki W, Malone M, Surewicz WK, Yee VC. Crystal structure of the human prion protein reveals a mechanism for oligomerization. Nat Struct Biol. 2001 Sep. 8(9):770-4. [View Abstract]
  22. Aucouturier P, Geissmann F, Damotte D, et al. Infected splenic dendritic cells are sufficient for prion transmission to the CNS in mouse scrapie. J Clin Invest. 2001 Sep. 108(5):703-8. [View Abstract]
  23. Pan KM, Baldwin M, Nguyen J, et al. Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci U S A. 1993 Dec 1. 90(23):10962-6. [View Abstract]
  24. Baron T. Identification of inter-species transmission of prion strains. J Neuropathol Exp Neurol. 2002 May. 61(5):377-83. [View Abstract]
  25. Kascsak RJ, Rubenstein R, Merz PA, et al. Immunological comparison of scrapie-associated fibrils isolated from animals infected with four different scrapie strains. J Virol. 1986 Sep. 59(3):676-83. [View Abstract]
  26. Carp RI, Rubenstein R. Diversity and significance of scrapie strains. Semin Virol. 1991. 2:203-13.
  27. Bessen RA, Marsh RF. Distinct PrP properties suggest the molecular basis of strain variation in transmissible mink encephalopathy. J Virol. 1994 Dec. 68(12):7859-68. [View Abstract]
  28. Caughey B, Raymond GJ, Bessen RA. Strain-dependent differences in beta-sheet conformations of abnormal prion protein. J Biol Chem. 1998 Nov 27. 273(48):32230-5. [View Abstract]
  29. Parchi P, Castellani R, Capellari S, et al. Molecular basis of phenotypic variability in sporadic Creutzfeldt-Jakob disease. Ann Neurol. 1996 Jun. 39(6):767-78. [View Abstract]
  30. Collinge J, Sidle KC, Meads J, Ironside J, Hill AF. Molecular analysis of prion strain variation and the aetiology of 'new variant' CJD. Nature. 1996 Oct 24. 383(6602):685-90. [View Abstract]
  31. Kimberlin RH, Walker CA. Evidence that the transmission of one source of scrapie agent to hamsters involves separation of agent strains from a mixture. J Gen Virol. 1978 Jun. 39(3):487-96. [View Abstract]
  32. Nicoll AJ, Collinge J. Preventing prion pathogenicity by targeting the cellular prion protein. Infect Disord Drug Targets. 2009 Feb. 9(1):48-57. [View Abstract]
  33. Aucouturier P, Carp RI, Carnaud C, Wisniewski T. Prion diseases and the immune system. Clin Immunol. 2000 Aug. 96(2):79-85. [View Abstract]
  34. Eklund CM, Kennedy RC, Hadlow WJ. Pathogenesis of scrapie virus infection in the mouse. J Infect Dis. 1967 Feb. 117(1):15-22. [View Abstract]
  35. Fraser H, Dickinson AG. Studies of the lymphoreticular system in the pathogenesis of scrapie: the role of spleen and thymus. J Comp Pathol. 1978 Oct. 88(4):563-73. [View Abstract]
  36. Kimberlin RH, Walker CA. Pathogenesis of mouse scrapie: dynamics of agent replication in spleen, spinal cord and brain after infection by different routes. J Comp Pathol. 1979 Oct. 89(4):551-62. [View Abstract]
  37. Houston F, Foster JD, Chong A, Hunter N, Bostock CJ. Transmission of BSE by blood transfusion in sheep. Lancet. 2000 Sep 16. 356(9234):999-1000. [View Abstract]
  38. Klein MA, Frigg R, Flechsig E, et al. A crucial role for B cells in neuroinvasive scrapie. Nature. 1997 Dec 18-25. 390(6661):687-90. [View Abstract]
  39. Montrasio F, Frigg R, Glatzel M, et al. Impaired prion replication in spleens of mice lacking functional follicular dendritic cells. Science. 2000 May 19. 288(5469):1257-9. [View Abstract]
  40. Shlomchik MJ, Radebold K, Duclos N, Manuelidis L. Neuroinvasion by a Creutzfeldt-Jakob disease agent in the absence of B cells and follicular dendritic cells. Proc Natl Acad Sci U S A. 2001 Jul 31. 98(16):9289-94. [View Abstract]
  41. Harischandra DS, Kondru N, Martin DP, Kanthasamy A, Jin H, Anantharam V, et al. Role of proteolytic activation of protein kinase Cd in the pathogenesis of prion disease. Prion. 2014 Jan-Feb. 8(1):143-53. [View Abstract]
  42. Beekes M, McBride PA, Baldauf E. Cerebral targeting indicates vagal spread of infection in hamsters fed with scrapie. J Gen Virol. 1998 Mar. 79 ( Pt 3):601-7. [View Abstract]
  43. Glatzel M, Heppner FL, Albers KM, Aguzzi A. Sympathetic innervation of lymphoreticular organs is rate limiting for prion neuroinvasion. Neuron. 2001 Jul 19. 31(1):25-34. [View Abstract]
  44. Meiner Z, Halimi M, Polakiewicz RD, Prusiner SB, Gabizon R. Presence of prion protein in peripheral tissues of Libyan Jews with Creutzfeldt-Jakob disease. Neurology. 1992 Jul. 42(7):1355-60. [View Abstract]
  45. Neufeld MY, Josiphov J, Korczyn AD. Demyelinating peripheral neuropathy in Creutzfeldt-Jakob disease. Muscle Nerve. 1992 Nov. 15(11):1234-9. [View Abstract]
  46. Holman RC, Khan AS, Belay ED, Schonberger LB. Creutzfeldt-Jakob disease in the United States, 1979-1994: using national mortality data to assess the possible occurrence of variant cases. Emerg Infect Dis. 1996 Oct-Dec. 2(4):333-7. [View Abstract]
  47. Masters CL, Harris JO, Gajdusek DC, Gibbs CJ Jr, Bernoulli C, Asher DM. Creutzfeldt-Jakob disease: patterns of worldwide occurrence and the significance of familial and sporadic clustering. Ann Neurol. 1979 Feb. 5(2):177-88. [View Abstract]
  48. Cathala F, Baron H. Clinical Aspects of Creutzfeldt-Jakob Disease. Prusiner SB, McKinley MP, eds. Prions: novel infectious pathogens causing scrapie and Creutzfeldt-Jakob disease. New York, NY: Academic Press; 1987. 467-509.
  49. Gajdusek DC, Zigas V. Degenerative disease of the central nervous system in New Guinea: the epidemic occurrence of "kuru" in the native population. N Engl J Med. 1957. 257:974-978.
  50. Gajdusek DC, Zigas V. Clinical, pathological and epidemiological study of an acute progressive degenerative disease of the central nervous system among natives of the eastern highlands of New Guinea. Am J Med. 1959. 26:442-469.
  51. Liberski PP, Sikorska B, Lindenbaum S, Goldfarb LG, McLean C, Hainfellner JA, et al. Kuru: Genes, Cannibals and Neuropathology. J Neuropathol Exp Neurol. 2012 Feb. 71(2):92-103. [View Abstract]
  52. Gajdusek DC, Gibbs CJ, Alpers M. Experimental transmission of a Kuru-like syndrome to chimpanzees. Nature. 1966 Feb 19. 209(5025):794-6. [View Abstract]
  53. Jacob A. Uber eigenaritge erkrankungen des zentral-nervensystems mit bemerkenswertem anatomischen befunde (spastische pseudosklerose-encephalomyelopathie mit disseminierten degenerationsherden). Z Gesamte Neurol Psychiatre. 1921. 64:147-228.
  54. Hofmann J, Wolf H, Grassmann A, Arndt V, Graham J, Vorberg I. Creutzfeldt-Jakob disease and mad cows: lessons learnt from yeast cells. Swiss Med Wkly. 2012 Jan 24. 142:[View Abstract]
  55. Chiofalo N, Fuentes A, Galvez S. Serial EEG findings in 27 cases of Creutzfeldt-Jakob disease. Arch Neurol. 1980 Mar. 37(3):143-5. [View Abstract]
  56. Gerstmann J, Straussler E, Scheinker I. Über eine eigenartige hereditar-familiare Erkrankung des Zentralnervensystems zugleich ein Beitrag zür frage des vorzeitigen kokalen Alterns. Z Neurol. 1936. 154:736-762.
  57. Ghetti B, Tagliavini F, Giaccone G, et al. Familial Gerstmann-Sträussler-Scheinker disease with neurofibrillary tangles. Mol Neurobiol. 1994 Feb. 8(1):41-8. [View Abstract]
  58. Ghetti B, Piccardo P, Spillantini MG, et al. Vascular variant of prion protein cerebral amyloidosis with tau-positive neurofibrillary tangles: the phenotype of the stop codon 145 mutation in PRNP. Proc Natl Acad Sci U S A. 1996 Jan 23. 93(2):744-8. [View Abstract]
  59. Medori R, Tritschler HJ, LeBlanc A, et al. Fatal familial insomnia, a prion disease with a mutation at codon 178 of the prion protein gene. N Engl J Med. 1992 Feb 13. 326(7):444-9. [View Abstract]
  60. Goldfarb LG, Brown P, Haltia M, et al. Creutzfeldt-Jakob disease cosegregates with the codon 178Asn PRNP mutation in families of European origin. Ann Neurol. 1992 Mar. 31(3):274-81. [View Abstract]
  61. Solvason HB, Harris B, Zeifert P, Flores BH, Hayward C. Psychological versus biological clinical interpretation: a patient with prion disease. Am J Psychiatry. 2002 Apr. 159(4):528-37. [View Abstract]
  62. Oliveros RG, Saracibar N, Gutierrez M, , Munon T, Gonzalez-Pinto A. Catatonia due to a prion familial disease. Schizophr Res. 2009 Mar. 108(1-3):309-10. [View Abstract]
  63. Collinge J. Human prion diseases and bovine spongiform encephalopathy (BSE). Hum Mol Genet. 1997. 6(10):1699-705. [View Abstract]
  64. Collinge J, Rossor M. A new variant of prion disease. Lancet. 1996 Apr 6. 347(9006):916-7. [View Abstract]
  65. Bateman D, Hilton D, Love S, Zeidler M, Beck J, Collinge J. Sporadic Creutzfeldt-Jakob disease in a 18-year-old in the UK. Lancet. 1995 Oct 28. 346(8983):1155-6. [View Abstract]
  66. Britton TC, al-Sarraj S, Shaw C, Campbell T, Collinge J. Sporadic Creutzfeldt-Jakob disease in a 16-year-old in the UK. Lancet. 1995 Oct 28. 346(8983):1155. [View Abstract]
  67. Collee JG, Bradley R. BSE: a decade on--Part I. Lancet. 1997 Mar 1. 349(9052):636-41. [View Abstract]
  68. Will RG. Surveillance of prion disease in humans. HF Baker, Ridley RM, eds. Methods in Molecular Medicine: Prion Diseases. Totowa, NJ: Humana Press Inc; 1996. 119-137.
  69. Liberski PP, Guiroy DC, Williams ES, Walis A, Budka H. Deposition patterns of disease-associated prion protein in captive mule deer brains with chronic wasting disease. Acta Neuropathol. 2001 Nov. 102(5):496-500. [View Abstract]
  70. Khan RM, Gunaratne LA, Kinmont JC. Rapid fracture healing in a patient with inherited prion disease. Ann R Coll Surg Engl. 2009 Apr. 91(3):261-2. [View Abstract]
  71. Belay ED, Gambetti P, Schonberger LB, et al. Creutzfeldt-Jakob disease in unusually young patients who consumed venison. Arch Neurol. 2001 Oct. 58(10):1673-8. [View Abstract]
  72. Raymond GJ, Bossers A, Raymond LD, et al. Evidence of a molecular barrier limiting susceptibility of humans, cattle and sheep to chronic wasting disease. EMBO J. 2000 Sep 1. 19(17):4425-30. [View Abstract]
  73. Angers RC, Browning SR, Seward TS, et al. Prions in skeletal muscles of deer with chronic wasting disease. Science. 2006 Feb 24. 311(5764):1117. [View Abstract]
  74. Marsh RF, Kincaid AE, Bessen RA, Bartz JC. Interspecies transmission of chronic wasting disease prions to squirrel monkeys (Saimiri sciureus). J Virol. 2005 Nov. 79(21):13794-6. [View Abstract]
  75. Hamaguchi T, Noguchi-Shinohara M, Nozaki I, Nakamura Y, Sato T, Kitamoto T. Medical procedures and risk for sporadic Creutzfeldt-Jakob disease, Japan, 1999-2008. Emerg Infect Dis. 2009 Feb. 15(2):265-71. [View Abstract]
  76. Appleby BS, Rincon-Beardsley TD, Appleby KK, Crain BJ, Wallin MT. Initial diagnoses of patients ultimately diagnosed with prion disease. J Alzheimers Dis. 2014 Jan 1. 42(3):833-9. [View Abstract]
  77. Seipelt M, Zerr I, Nau R, et al. Hashimoto's encephalitis as a differential diagnosis of Creutzfeldt-Jakob disease. J Neurol Neurosurg Psychiatry. 1999 Feb. 66(2):172-6. [View Abstract]
  78. Castillo P, Woodruff B, Caselli R, et al. Steroid-responsive encephalopathy associated with autoimmune thyroiditis. Arch Neurol. 2006 Feb. 63(2):197-202. [View Abstract]
  79. Cossu G, Melis M, Molari A, et al. Creutzfeldt-Jakob disease associated with high titer of antithyroid autoantibodies: case report and literature review. Neurol Sci. 2003 Oct. 24(3):138-40. [View Abstract]
  80. Young GS, Geschwind MD, Fischbein NJ, et al. Diffusion-weighted and fluid-attenuated inversion recovery imaging in Creutzfeldt-Jakob disease: high sensitivity and specificity for diagnosis. AJNR Am J Neuroradiol. 2005 Jun-Jul. 26(6):1551-62. [View Abstract]
  81. Renard D, Castelnovo G, Collombier L, Thouvenot E, Boudousq V. FDG-PET in Creutzfeldt-Jakob disease: Analysis of clinical-PET correlation. Prion. 2017 Nov 2. 11 (6):440-453. [View Abstract]
  82. Rees HC, Maddison BC, Owen JP, Whitelam GC, Gough KC. Concentration of disease-associated prion protein with silicon dioxide. Mol Biotechnol. 2009 Mar. 41(3):254-62. [View Abstract]
  83. Laude H. Beringue V. Newly discovered forms of prion diseases in ruminants. [Review]. Pathologie Biologie. 2009. 57(2):117-26.
  84. Sim VL. Caughey B. Recent advances in prion chemotherapeutics. [Review]. Infectious Disorders - Drug Targets. 2009. 9(1):81-91.
  85. Relano-Gines A. Gabelle A. Lehmann S. Milhavet O. Crozet C. Gene and cell therapy for prion diseases. [Review]. Infectious Disorders - Drug Targets. 2009. 9(1):58-68.
  86. Miguelez-Rodriguez A, Santos-Juanes J, Vicente-Etxenausia I, Perez de Heredia-Goñi K, Garcia B, Quiros LM, et al. Brains with sporadic Creutzfeldt-Jakob disease and copathology showed a prolonged end-stage of disease. J Clin Pathol. 2018 May. 71 (5):446-450. [View Abstract]
  87. Caspi S, Halimi M, Yanai A, Sasson SB, Taraboulos A, Gabizon R. The anti-prion activity of Congo red. Putative mechanism. J Biol Chem. 1998 Feb 6. 273(6):3484-9. [View Abstract]
  88. Demaimay R, Harper J, Gordon H, Weaver D, Chesebro B, Caughey B. Structural aspects of Congo red as an inhibitor of protease-resistant prion protein formation. J Neurochem. 1998 Dec. 71(6):2534-41. [View Abstract]
  89. Doh-ura K, Ishikawa K, Murakami-Kubo I, et al. Treatment of transmissible spongiform encephalopathy by intraventricular drug infusion in animal models. J Virol. 2004 May. 78(10):4999-5006. [View Abstract]
  90. Demaimay R, Chesebro B, Caughey B. Inhibition of formation of protease-resistant prion protein by Trypan Blue, Sirius Red and other Congo Red analogs. Arch Virol Suppl. 2000. 277-83. [View Abstract]
  91. Tagliavini F, McArthur RA, Canciani B, et al. Effectiveness of anthracycline against experimental prion disease in Syrian hamsters. Science. 1997 May 16. 276(5315):1119-22. [View Abstract]
  92. Adjou KT, Demaimay R, Lasmezas CI, Seman M, Deslys JP, Dormont D. Differential effects of a new amphotericin B derivative, MS-8209, on mouse BSE and scrapie: implications for the mechanism of action of polyene antibiotics. Res Virol. 1996 Jul-Aug. 147(4):213-8. [View Abstract]
  93. Adjou KT, Demaimay R, Deslys JP, et al. MS-8209, a water-soluble amphotericin B derivative, affects both scrapie agent replication and PrPres accumulation in Syrian hamster scrapie. J Gen Virol. 1999 Apr. 80 ( Pt 4):1079-85. [View Abstract]
  94. Adjou KT, Privat N, Demart S, et al. MS-8209, an amphotericin B analogue, delays the appearance of spongiosis, astrogliosis and PrPres accumulation in the brain of scrapie-infected hamsters. J Comp Pathol. 2000 Jan. 122(1):3-8. [View Abstract]
  95. Mange A, Milhavet O, McMahon HE, Casanova D, Lehmann S. Effect of amphotericin B on wild-type and mutated prion proteins in cultured cells: putative mechanism of action in transmissible spongiform encephalopathies. J Neurochem. 2000 Feb. 74(2):754-62. [View Abstract]
  96. Mange A, Nishida N, Milhavet O, McMahon HE, Casanova D, Lehmann S. Amphotericin B inhibits the generation of the scrapie isoform of the prion protein in infected cultures. J Virol. 2000 Apr. 74(7):3135-40. [View Abstract]
  97. Farquhar C, Dickinson A, Bruce M. Prophylactic potential of pentosan polysulphate in transmissible spongiform encephalopathies. Lancet. 1999 Jan 9. 353(9147):117. [View Abstract]
  98. Ladogana A, Casaccia P, Ingrosso L, et al. Sulphate polyanions prolong the incubation period of scrapie-infected hamsters. J Gen Virol. 1992 Mar. 73 ( Pt 3):661-5. [View Abstract]
  99. Farquhar CF, Dickinson AG. Prolongation of scrapie incubation period by an injection of dextran sulphate 500 within the month before or after infection. J Gen Virol. 1986 Mar. 67 ( Pt 3):463-73. [View Abstract]
  100. Priola SA, Raines A, Caughey WS. Porphyrin and phthalocyanine antiscrapie compounds. Science. 2000 Feb 25. 287(5457):1503-6. [View Abstract]
  101. Priola SA, Raines A, Caughey W. Prophylactic and therapeutic effects of phthalocyanine tetrasulfonate in scrapie-infected mice. J Infect Dis. 2003 Sep 1. 188(5):699-705. [View Abstract]
  102. Masullo C, Macchi G, Xi YG, Pocchiari M. Failure to ameliorate Creutzfeldt-Jakob disease with amphotericin B therapy. J Infect Dis. 1992 Apr. 165(4):784-5. [View Abstract]
  103. Korth C, May BC, Cohen FE, Prusiner SB. Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease. Proc Natl Acad Sci U S A. 2001 Aug 14. 98(17):9836-41. [View Abstract]
  104. Nakajima M, Yamada T, Kusuhara T, et al. Results of quinacrine administration to patients with Creutzfeldt-Jakob disease. Dement Geriatr Cogn Disord. 2004. 17(3):158-63. [View Abstract]
  105. Barret A, Tagliavini F, Forloni G, et al. Evaluation of quinacrine treatment for prion diseases. J Virol. 2003 Aug. 77(15):8462-9. [View Abstract]
  106. Collins SJ, Lewis V, Brazier M, Hill AF, Fletcher A, Masters CL. Quinacrine does not prolong survival in a murine Creutzfeldt-Jakob disease model. Ann Neurol. 2002 Oct. 52(4):503-6. [View Abstract]
  107. Todd NV, Morrow J, Doh-ura K, et al. Cerebroventricular infusion of pentosan polysulphate in human variant Creutzfeldt-Jakob disease. J Infect. 2005 Jun. 50(5):394-6. [View Abstract]
  108. Soto C, Kascsak RJ, Saborio GP, et al. Reversion of prion protein conformational changes by synthetic beta-sheet breaker peptides. Lancet. 2000 Jan 15. 355(9199):192-7. [View Abstract]
  109. Wisniewski T, Aucouturier P, Soto C, Frangione B. The prionoses and other conformational disorders. Amyloid. 1998 Sep. 5(3):212-24. [View Abstract]
  110. Wisniewski T, Sigurdsson EM, Aucouturier P, et al. Conformation as a therapeutic target in the prionoses and other neurodegenerative conditions. Molecular and Cellular Pathology in Prion Disease. 2001.
  111. De Gioia L, Selvaggini C, Ghibaudi E, et al. Conformational polymorphism of the amyloidogenic and neurotoxic peptide homologous to residues 106-126 of the prion protein. J Biol Chem. 1994 Mar 18. 269(11):7859-62. [View Abstract]
  112. Nguyen J, Baldwin MA, Cohen FE, Prusiner SB. Prion protein peptides induce alpha-helix to beta-sheet conformational transitions. Biochemistry. 1995 Apr 4. 34(13):4186-92. [View Abstract]
  113. Zhang H, Kaneko K, Nguyen JT, et al. Conformational transitions in peptides containing two putative alpha-helices of the prion protein. J Mol Biol. 1995 Jul 21. 250(4):514-26. [View Abstract]
  114. Naiki H, Higuchi K, Nakakuki K, Takeda T. Kinetic analysis of amyloid fibril polymerization in vitro. Lab Invest. 1991 Jul. 65(1):104-10. [View Abstract]
  115. Wisniewski T, Castano EM, Golabek A, Vogel T, Frangione B. Acceleration of Alzheimer's fibril formation by apolipoprotein E in vitro. Am J Pathol. 1994 Nov. 145(5):1030-5. [View Abstract]
  116. Sigurdsson EM, Permanne B, Soto C, Wisniewski T, Frangione B. In vivo reversal of amyloid-beta lesions in rat brain. J Neuropathol Exp Neurol. 2000 Jan. 59(1):11-7. [View Abstract]
  117. Soto C, Sigurdsson EM, Morelli L, Kumar RA, Castano EM, Frangione B. Beta-sheet breaker peptides inhibit fibrillogenesis in a rat brain model of amyloidosis: implications for Alzheimer's therapy. Nat Med. 1998 Jul. 4(7):822-6. [View Abstract]
  118. Vidal R, Ghiso J, Wisniewski T, Frangione B. Alzheimer's presenilin 1 gene expression in platelets and megakaryocytes. Identification of a novel splice variant. FEBS Lett. 1996 Sep 9. 393(1):19-23. [View Abstract]
  119. Sigurdsson EM, Brown DR, Alim MA, Scholtzova H, Carp R, Meeker HC, et al. Copper chelation delays the onset of prion disease. J Biol Chem. 2003 Nov 21. 278(47):46199-202. [View Abstract]
  120. Sigurdsson EM. Immunotherapy for conformational diseases. Curr Pharm Des. 2006. 12(20):2569-85. [View Abstract]
  121. Schenk D, Barbour R, Dunn W, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature. 1999 Jul 8. 400(6740):173-7. [View Abstract]
  122. Weiner HL, Lemere CA, Maron R, et al. Nasal administration of amyloid-beta peptide decreases cerebral amyloid burden in a mouse model of Alzheimer's disease. Ann Neurol. 2000 Oct. 48(4):567-79. [View Abstract]
  123. Janus C, Pearson J, McLaurin J, et al. A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease. Nature. 2000 Dec 21-28. 408(6815):979-82. [View Abstract]
  124. Morgan D, Diamond DM, Gottschall PE, et al. Arendash GW Ab peptide vaccination prevents memory loss in an animal model of Alzheimer's disease. Nature. 2001. 408:982-985.
  125. Bard F, Cannon C, Barbour R, et al. Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat Med. 2000 Aug. 6(8):916-9. [View Abstract]
  126. Sigurdsson EM, Brown DR, Daniels M, et al. Immunization delays the onset of prion disease in mice. Am J Pathol. 2002 Jul. 161(1):13-7. [View Abstract]
  127. Goni F, Knudsen E, Schreiber F, et al. Mucosal vaccination delays or prevents prion infection via an oral route. Neuroscience. 2005. 133(2):413-21. [View Abstract]
  128. Pankiewicz J, Sadowski M, Kascsak R, et al. Therapeutic mechanisms of anti-prion antibodies in a tissue culture model of prion infection. Soc Neurosci Abst. 2005.
  129. Sigurdsson EM, Sy MS, Li R, Scholtzova H, Kascsak RJ, Kascsak R, et al. Anti-prion antibodies for prophylaxis following prion exposure in mice. Neurosci Lett. 2003 Jan 23. 336(3):185-7. [View Abstract]
  130. Manuelidis L. Vaccination with an attenuated Creutzfeldt-Jakob disease strain prevents expression of a virulent agent. Proc Natl Acad Sci U S A. 1998 Mar 3. 95(5):2520-5. [View Abstract]
  131. White AR, Enever P, Tayebi M, et al. Monoclonal antibodies inhibit prion replication and delay the development of prion disease. Nature. 2003 Mar 6. 422(6927):80-3. [View Abstract]
  132. Enari M, Flechsig E, Weissmann C. Scrapie prion protein accumulation by scrapie-infected neuroblastoma cells abrogated by exposure to a prion protein antibody. Proc Natl Acad Sci U S A. 2001 Jul 31. 98(16):9295-9. [View Abstract]
  133. Peretz D, Williamson RA, Kaneko K, et al. Antibodies inhibit prion propagation and clear cell cultures of prion infectivity. Nature. 2001 Aug 16. 412(6848):739-43. [View Abstract]
  134. Souan L, Tal Y, Felling Y, Cohen IR, Taraboulos A, Mor F. Modulation of proteinase-K resistant prion protein by prion peptide immunization. Eur J Immunol. 2001 Aug. 31(8):2338-46. [View Abstract]
  135. Sigurdsson EM, Scholtzova H, Mehta PD, Frangione B, Wisniewski T. Immunization with a nontoxic/nonfibrillar amyloid-beta homologous peptide reduces Alzheimer's disease-associated pathology in transgenic mice. Am J Pathol. 2001 Aug. 159(2):439-47. [View Abstract]
  136. Sigurdsson EM, Knudsen E, Asuni A, Fitzer-Attas C, Sage D, Quartermain D, et al. An attenuated immune response is sufficient to enhance cognition in an Alzheimer's disease mouse model immunized with amyloid-beta derivatives. J Neurosci. 2004 Jul 14. 24(28):6277-82. [View Abstract]
  137. Coste J. Prowse C. Eglin R. Fang C. Subgroup on TSE. A report on transmissible spongiform encephalopathies and transfusion safety. [Review]. Vox Sanguinis. 2009. 96(4):284-291.
  138. Lefrere JJ, Hewitt P. From mad cows to sensible blood transfusion: the risk of prion transmission by labile blood components in the United Kingdom and in France. Transfusion. 2009 Apr. 49(4):797-812. [View Abstract]
  139. Brown P, Wolff A, Gajdusek DC. A simple and effective method for inactivating virus infectivity in formalin-fixed tissue samples from patients with Creutzfeldt-Jakob disease. Neurology. 1990 Jun. 40(6):887-90. [View Abstract]
  140. Ironside JW, Bell JE. The 'high-risk' neuropathological autopsy in AIDS and Creutzfeldt-Jakob disease: principles and practice. Neuropathol Appl Neurobiol. 1996 Oct. 22(5):388-93. [View Abstract]
  141. Aucouturier P, Kascsak RJ, Frangione B, Wisniewski T. Biochemical and conformational variability of human prion strains in sporadic Creutzfeldt-Jakob disease. Neurosci Lett. 1999 Oct 15. 274(1):33-6. [View Abstract]
  142. Brown P, Rau EH, Johnson BK, Bacote AE, Gibbs CJ Jr, Gajdusek DC. New studies on the heat resistance of hamster-adapted scrapie agent: threshold survival after ashing at 600 degrees C suggests an inorganic template of replication. Proc Natl Acad Sci U S A. 2000 Mar 28. 97(7):3418-21. [View Abstract]
  143. Budka H, Aguzzi A, Brown P, et al. Tissue handling in suspected Creutzfeldt-Jakob disease (CJD) and other human spongiform encephalopathies (prion diseases). Brain Pathol. 1995 Jul. 5(3):319-22. [View Abstract]
  144. Bueler H, Fischer M, Lang Y, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature. 1992 Apr 16. 356(6370):577-82. [View Abstract]
  145. Castilla J, Saa P, Soto C. Detection of prions in blood. Nat Med. 2005 Sep. 11(9):982-5. [View Abstract]
  146. Chang B, Cheng X, Pan T, et al. An ultra-sensitive assay for detecting prions in blood. Proc Natl Acad Sci (USA) in press. 2006.
  147. Citron M, Vigo-Pelfrey C, Teplow DB, et al. Excessive production of amyloid beta-protein by peripheral cells of symptomatic and presymptomatic patients carrying the Swedish familial Alzheimer disease mutation. Proc Natl Acad Sci U S A. 1994 Dec 6. 91(25):11993-7. [View Abstract]
  148. Collinge J, Palmer MS, Dryden AJ. Genetic predisposition to iatrogenic Creutzfeldt-Jakob disease. Lancet. 1991 Jun 15. 337(8755):1441-2. [View Abstract]
  149. Collins SJ, Lawson VA, Masters CL. Transmissible spongiform encephalopathies. Lancet. 2004 Jan 3. 363(9402):51-61. [View Abstract]
  150. Dlouhy SR, Hsiao K, Farlow MR, et al. Linkage of the Indiana kindred of Gerstmann-Sträussler-Scheinker disease to the prion protein gene. Nat Genet. 1992 Apr. 1(1):64-7. [View Abstract]
  151. Dominguez DI, De Strooper B, Annaert W. Secretases as therapeutic targets for the treatment of Alzheimer's disease. Amyloid. 2001 Jun. 8(2):124-42. [View Abstract]
  152. Dropcho EJ. Update on paraneoplastic syndromes. Curr Opin Neurol. 2005 Jun. 18(3):331-6. [View Abstract]
  153. Finkenstaedt M, Szudra A, Zerr I, et al. MR imaging of Creutzfeldt-Jakob disease. Radiology. 1996 Jun. 199(3):793-8. [View Abstract]
  154. Foster PR. Prions and blood products. Ann Med. 2000 Oct. 32(7):501-13. [View Abstract]
  155. Gabizon R, Rosenmann H, Meiner Z, et al. Mutation and polymorphism of the prion protein gene in Libyan Jews with Creutzfeldt-Jakob disease (CJD). Am J Hum Genet. 1993 Oct. 53(4):828-35. [View Abstract]
  156. Goldfarb LG, Brown P, Goldgaber D, et al. Creutzfeldt-Jakob disease and kuru patients lack a mutation consistently found in the Gerstmann-Sträussler-Scheinker syndrome. Exp Neurol. 1990 Jun. 108(3):247-50. [View Abstract]
  157. Goldfarb LG, Brown P, McCombie WR, et al. Transmissible familial Creutzfeldt-Jakob disease associated with five, seven, and eight extra octapeptide coding repeats in the PRNP gene. Proc Natl Acad Sci U S A. 1991 Dec 1. 88(23):10926-30. [View Abstract]
  158. Goldfarb LG, Haltia M, Brown P, et al. New mutation in scrapie amyloid precursor gene (at codon 178) in Finnish Creutzfeldt-Jakob kindred. Lancet. 1991 Feb 16. 337(8738):425. [View Abstract]
  159. Goldfarb LG, Mitrova E, Brown P, Toh BK, Gajdusek DC. Mutation in codon 200 of scrapie amyloid protein gene in two clusters of Creutzfeldt-Jakob disease in Slovakia. Lancet. 1990 Aug 25. 336(8713):514-5. [View Abstract]
  160. Goldfarb LG, Petersen RB, Tabaton M, et al. Fatal familial insomnia and familial Creutzfeldt-Jakob disease: disease phenotype determined by a DNA polymorphism. Science. 1992 Oct 30. 258(5083):806-8. [View Abstract]
  161. Goldgaber D, Goldfarb LG, Brown P, et al. Mutations in familial Creutzfeldt-Jakob disease and Gerstmann-Sträussler-Scheinker's syndrome. Exp Neurol. 1989 Nov. 106(2):204-6. [View Abstract]
  162. Haass C, Hung AY, Schlossmacher MG, Teplow DB, Selkoe DJ. beta-Amyloid peptide and a 3-kDa fragment are derived by distinct cellular mechanisms. J Biol Chem. 1993 Feb 15. 268(5):3021-4. [View Abstract]
  163. Head MW, Bunn TJ, Bishop MT, et al. Prion protein heterogeneity in sporadic but not variant Creutzfeldt-Jakob disease: UK cases 1991-2002. Ann Neurol. 2004 Jun. 55(6):851-9. [View Abstract]
  164. Hetz C, Soto C. Protein misfolding and disease: the case of prion disorders. Cell Mol Life Sci. 2003 Jan. 60(1):133-43. [View Abstract]
  165. Hill AF, Zeidler M, Ironside J, Collinge J. Diagnosis of new variant Creutzfeldt-Jakob disease by tonsil biopsy. Lancet. 1997 Jan 11. 349(9045):99-100. [View Abstract]
  166. Hsiao K, Baker HF, Crow TJ, et al. Linkage of a prion protein missense variant to Gerstmann-Sträussler syndrome. Nature. 1989 Mar 23. 338(6213):342-5. [View Abstract]
  167. Hsiao K, Dlouhy SR, Farlow MR, et al. Mutant prion proteins in Gerstmann-Sträussler-Scheinker disease with neurofibrillary tangles. Nat Genet. 1992 Apr. 1(1):68-71. [View Abstract]
  168. Hsiao K, Doh-ura K, KitamotoT. A prion protein amino acid substitution in ataxic Gerstmann-Straussler syndrome. Ann Neurol. 1989. 26:137.
  169. Hsiao K, Meiner Z, Kahana E, et al. Mutation of the prion protein in Libyan Jews with Creutzfeldt-Jakob disease. N Engl J Med. 1991 Apr 18. 324(16):1091-7. [View Abstract]
  170. Jeong JK, Lee JH, Moon JH, Lee YJ, Park SY. Melatonin-mediated ß-catenin activation protects neuron cells against prion protein-induced neurotoxicity. J Pineal Res. 2014 Nov. 57(4):427-34. [View Abstract]
  171. Kitamoto T, Ohta M, Doh-ura K, Hitoshi S, Terao Y, Tateishi J. Novel missense variants of prion protein in Creutzfeldt-Jakob disease or Gerstmann-Sträussler syndrome. Biochem Biophys Res Commun. 1993 Mar 15. 191(2):709-14. [View Abstract]
  172. Kovacs GG, Voigtlander T, Gelpi E, Budka H. Rationale for diagnosing human prion disease. World J Biol Psychiatry. 2004 Apr. 5(2):83-91. [View Abstract]
  173. Maness LM, Banks WA, Podlisny MB, Selkoe DJ, Kastin AJ. Passage of human amyloid beta-protein 1-40 across the murine blood-brain barrier. Life Sci. 1994. 55(21):1643-50. [View Abstract]
  174. Miyazono M, Kitamoto T, Doh-ura K, Iwaki T, Tateishi J. Creutzfeldt-Jakob disease with codon 129 polymorphism (valine): a comparative study of patients with codon 102 point mutation or without mutations. Acta Neuropathol. 1992. 84(4):349-54. [View Abstract]
  175. Nochlin D, Sumi SM, Bird TD, et al. Familial dementia with PrP-positive amyloid plaques: a variant of Gerstmann-Sträussler syndrome. Neurology. 1989 Jul. 39(7):910-8. [View Abstract]
  176. Owen F, Poulter M, Lofthouse R, et al. Insertion in prion protein gene in familial Creutzfeldt-Jakob disease. Lancet. 1989 Jan 7. 1(8628):51-2. [View Abstract]
  177. Owen F, Poulter M, Shah T, et al. An in-frame insertion in the prion protein gene in familial Creutzfeldt-Jakob disease. Brain Res Mol Brain Res. 1990 Apr. 7(3):273-6. [View Abstract]
  178. Quadrio I, Ugnon-Cafe S, Dupin M, Esposito G, Streichenberger N, Krolak-Salmon P. Rapid diagnosis of human prion disease using streptomycin with tonsil and brain tissues. Lab Invest. 2009 Apr. 89(4):406-13. [View Abstract]
  179. Ripoll L, Laplanche JL, Salzmann M, et al. A new point mutation in the prion protein gene at codon 210 in Creutzfeldt-Jakob disease. Neurology. 1993 Oct. 43(10):1934-8. [View Abstract]
  180. Sakaguchi S. Prospects for preventative vaccines against prion diseases. Protein & Peptide Letters. 2009. 16(3):260-70.
  181. Soto C, Saborio GP, Anderes L. Cyclic amplification of protein misfolding: application to prion-related disorders and beyond. Trends Neurosci. 2002 Aug. 25(8):390-4. [View Abstract]
  182. Weissmann C. Molecular genetics of transmissible spongiform encephalopathies. J Biol Chem. 1999 Jan 1. 274(1):3-6. [View Abstract]
  183. Will RG, Ironside JW, Zeidler M, et al. A new variant of Creutzfeldt-Jakob disease in the UK. Lancet. 1996 Apr 6. 347(9006):921-5. [View Abstract]

Prion-related diseases. Spongiform change in prion disease. This section shows mild parenchymal vacuolation and prominent reactive astrocytosis.

Prion-related diseases. A representation of the human proteinaceous infectious particle, or PrP, gene. Mutations associated with inherited prionoses are shown above the gene, while polymorphisms are shown below the gene. A polymorphism at codon 129 (M versus V) is common in white populations, while a polymorphism at codon 219 (E versus K) is common in Japanese populations. The locations of the 4 putative helical regions, H1-H4, correspond to residues 109-122, 129-141, 178-191, and 202-218, respectively. This diagram does not illustrate all of the alpha-helical regions. A diagonal striped area represents the region of octarepeats, spanning residues 51-91. Octarepeats of 16, 32, 40, 48, 56, 64, or 72 amino acids at codons 67, 75, or 83 are indicated by the rectangle above the octarepeat region. These inserts are associated with familial Creutzfeldt-Jakob disease (CJD).

Shows characteristic signal changes of an MRI taken from a patient with sporadic CJD, using diffusion-weighted imaging (DWI). An abnormal signal is shown in both the basal ganglia (red arrows) and the cortical ribbon (yellow arrow).

Prion-related diseases. Spongiform change in prion disease. This section shows mild parenchymal vacuolation and prominent reactive astrocytosis.

Prion-related diseases. A representation of the human proteinaceous infectious particle, or PrP, gene. Mutations associated with inherited prionoses are shown above the gene, while polymorphisms are shown below the gene. A polymorphism at codon 129 (M versus V) is common in white populations, while a polymorphism at codon 219 (E versus K) is common in Japanese populations. The locations of the 4 putative helical regions, H1-H4, correspond to residues 109-122, 129-141, 178-191, and 202-218, respectively. This diagram does not illustrate all of the alpha-helical regions. A diagonal striped area represents the region of octarepeats, spanning residues 51-91. Octarepeats of 16, 32, 40, 48, 56, 64, or 72 amino acids at codons 67, 75, or 83 are indicated by the rectangle above the octarepeat region. These inserts are associated with familial Creutzfeldt-Jakob disease (CJD).

Shows characteristic signal changes of an MRI taken from a patient with sporadic CJD, using diffusion-weighted imaging (DWI). An abnormal signal is shown in both the basal ganglia (red arrows) and the cortical ribbon (yellow arrow).

Disease Host Mechanism
KuruHumanCannibalism
Sporadic CJDHumanSpontaneous PrPC to PrPSc conversion or somatic mutation
Iatrogenic CJDHumanInfection from prion-containing material, eg, dura mater, electrode
Familial CJDHumanMutations in the PrP gene
vCJDHumanInfection from BSE
GSSHumanMutations in the PrP gene
FFIHumanD178N mutation in the PrP gene, with M129 polymorphism
Sporadic fatal insomniaHumanSpontaneous PrPC to PrPSc conversion or somatic mutation
ScrapieSheepInfection in susceptible sheep
BSECattleInfection from contaminated food
TMEMinkInfection from sheep or cattle in food
CWDMule, deer, elkUnclear
Feline spongiform encephalopathyCatsInfection from contaminated food
Exotic ungulate encephalopathyNyala, oryx, kuduInfection from contaminated food
Clinical Syndrome Neoplasm Autoantibodies
Limbic encephalitisSmall cell lung carcinoma



Testicular/breast, thymoma



Anti-Hu, antiCV2,PCA-2, ANNA-3



Anti-Ma2 Anti-VGKC, anti-CV2



Cerebellar degenerationBreast, ovary, lung, othersAnti-Yo, anti-Ma, anti-Ri



Anti-Hu, anti-CV2



Opsoclonus myoclonusBreast, ovarian, small cell carcinoma of lung



Neuroblastoma



Anti-Ri, anti-Yo, Anti-Hu,



Anti-amphiphysin Anti-Hu