Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Inherited dementias

...Read more
Inherited dementias Peter Hedera, MD a , R. Scott Turner, MD, PhD b, * a Department of Neurology, Vanderbilt University, Nashville, Tennesse b Cognitive Disorders Section, Department of Neurology, University of Michigan Health System, and Veterans Affairs Medical Center Geriatric Research Education and Clinical Center, Ann Arbor, Michigan Familial Alzheimer’s disease and related amyloid angiopathies Although age-related progressive cognitive decline has been known since antiquity, a case report by Alois Alzheimer described the neuropathology associated with a ‘‘peculiar’’ dementing syndrome [1]. Dr. Alzheimer reported the 5-year clinical course of a 51-year old woman with progressive dementia and autopsy findings of neuronal loss, neurofibrillary tangles, and miliary amyloid plaques found by light microscopic examination of Bielshowsky silver-stained brain sections. Thus, he was the first to suggest a link, perhaps causal, between this dementing disease and the abnormal pro- teinaceous aggregates in brain. Or were they merely ‘‘tombstone’’ epipheno- mena? The debate continues. Alzheimer’s disease (AD) now affects approximately 2–3% of individuals at age 65, with an approximate doubling of incidence for every 5 years of age afterward. The prevalence of AD in one study approaches 50% of those over age 85 [24]. Although AD is the most common etiology of progressive dementia in the elderly in the U.S., it is not inevitable with aging, and ‘‘esca- pees’’ warrant further epidemiologic and genetic study. In 1990, there were an estimated 4 million people in the U.S. with AD. Because of an expanding population and increasing life expectancy the number of affected individuals is projected to increase to 14 million in the U.S. in 2050. The Diagnostic and Statistical Manual, 4th edition (DSM-IV), or the National Institute of Neu- rologic, Communicative Disorders and Stroke-AD and Related Disorders Association (NINCDS-ADRDA) criteria are used for the clinical diagnosis Neurol Clin N Am 20 (2002) 779–808 This work was supported by the Department of Veterans Affairs Geriatrics Research Education and Clinical Center and Grant No. P50 AG08671 from the National Institutes of Health. * Corresponding author. VAMC GRECC, 2215 Fuller Road, Ann Arbor, MI 48105. E-mail address: raymondt@umich.edu (R. Scott Turner). 0733-8619/02/$ - see front matter Ó 2002, Elsevier Science (USA). All rights reserved. PII:S0733-8619(01)00020-2
of AD [20,64]. These criteria are similar and require a gradually progressive dementia severe enough to impair social or occupational functioning with other etiologies excluded [54]. By definition, dementia requires a decline in memory and at least one other cognitive domain—visuospatial skills, lan- guage and calculation, praxis, gnosis, or frontal and executive function. A diagnosis of definite AD requires light microscopic examination of brain tis- sue sections (by autopsy, or rarely by brain biopsy). Thus, only possible and probable AD are routinely diagnosed in the clinic. Possible AD is diagnosed when uncertainty arises from an additional secondary etiology of dementia or the dementia has an atypical onset, course, or presentation. Diagnostic accuracies, compared to autopsy, for possible and probable AD by NINCDS-ADRDA criteria are approximately 50–60% and 80–90%, respec- tively, in specialized centers. The Khachaturian pathologic criteria for AD require that the density of amyloid plaques and neurofibrillary tangles in brain sections exceed a given threshold that increases with age [52]. In contrast, the Consortium to Estab- lish a Registry for Alzheimer’s Disease (CERAD) criteria focus exclusively on the density of amyloid plaques (the sine qua non pathologic marker of AD), in brain sections compared to given (published) high-power micro- scopic fields. In part because neurofibrillary tangles are not specific to AD, they were not considered essential to the diagnosis [68]. The recent ‘‘Reagan criteria’’ for AD, however, require both amyloid plaques and neu- rofibrillary tangles in multiple brain regions, and declare all such pathology abnormal [98]. These criteria incorporate the CERAD plaque density scale as well as Braak and Braak [5] staging of the density and distribution of pathologic abnormalities found in AD brain [5,68]. Despite numerous other profound neuropathologic changes in brain (neuronal and synaptic loss, gliosis, inflammation, cholinergic deficits, microvascular amyloid angio- pathy, oxidative damage, mitochondrial dysfunction, etc) the mainstay of pathologic diagnosis remains silver staining of brain sections and light microscopic examination of the density and distribution of amyloid plaques and neurofibrillary tangles—in other words, methods used by Dr. Alzheimer in 1907. As documented by Braak and Braak [5], the neuropathology of AD is not random but affects entorhinal cortex and hippocampus followed by other limbic structures and neocortex [5]. This is consistent with the initial primarily amnestic clinical presentation of patients with AD followed my several years of progressive and gradual decline in multiple cognitive domains. As is true for all neurodegenerative diseases, the etiology of the selective vulnerability of different brain regions to AD pathologies remains obscure. The major risk factor for AD is aging. Even subjects with Down’s syn- drome and individuals with genetic polymorphisms or mutations that increase risk of AD require a degree of aging before signs and symptoms commence. It remains unknown, however, what specific factors associated with aging increase risk of AD. Having a first-degree relative with AD 780 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808
Neurol Clin N Am 20 (2002) 779–808 Inherited dementias Peter Hedera, MDa, R. Scott Turner, MD, PhDb,* a Department of Neurology, Vanderbilt University, Nashville, Tennesse Cognitive Disorders Section, Department of Neurology, University of Michigan Health System, and Veterans Affairs Medical Center Geriatric Research Education and Clinical Center, Ann Arbor, Michigan b Familial Alzheimer’s disease and related amyloid angiopathies Although age-related progressive cognitive decline has been known since antiquity, a case report by Alois Alzheimer described the neuropathology associated with a ‘‘peculiar’’ dementing syndrome [1]. Dr. Alzheimer reported the 5-year clinical course of a 51-year old woman with progressive dementia and autopsy findings of neuronal loss, neurofibrillary tangles, and miliary amyloid plaques found by light microscopic examination of Bielshowsky silver-stained brain sections. Thus, he was the first to suggest a link, perhaps causal, between this dementing disease and the abnormal proteinaceous aggregates in brain. Or were they merely ‘‘tombstone’’ epiphenomena? The debate continues. Alzheimer’s disease (AD) now affects approximately 2–3% of individuals at age 65, with an approximate doubling of incidence for every 5 years of age afterward. The prevalence of AD in one study approaches 50% of those over age 85 [24]. Although AD is the most common etiology of progressive dementia in the elderly in the U.S., it is not inevitable with aging, and ‘‘escapees’’ warrant further epidemiologic and genetic study. In 1990, there were an estimated 4 million people in the U.S. with AD. Because of an expanding population and increasing life expectancy the number of affected individuals is projected to increase to 14 million in the U.S. in 2050. The Diagnostic and Statistical Manual, 4th edition (DSM-IV), or the National Institute of Neurologic, Communicative Disorders and Stroke-AD and Related Disorders Association (NINCDS-ADRDA) criteria are used for the clinical diagnosis This work was supported by the Department of Veterans Affairs Geriatrics Research Education and Clinical Center and Grant No. P50 AG08671 from the National Institutes of Health. * Corresponding author. VAMC GRECC, 2215 Fuller Road, Ann Arbor, MI 48105. E-mail address: raymondt@umich.edu (R. Scott Turner). 0733-8619/02/$ - see front matter  2002, Elsevier Science (USA). All rights reserved. PII: S 0 7 3 3 - 8 6 1 9 ( 0 1 ) 0 0 0 2 0 - 2 780 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 of AD [20,64]. These criteria are similar and require a gradually progressive dementia severe enough to impair social or occupational functioning with other etiologies excluded [54]. By definition, dementia requires a decline in memory and at least one other cognitive domain—visuospatial skills, language and calculation, praxis, gnosis, or frontal and executive function. A diagnosis of definite AD requires light microscopic examination of brain tissue sections (by autopsy, or rarely by brain biopsy). Thus, only possible and probable AD are routinely diagnosed in the clinic. Possible AD is diagnosed when uncertainty arises from an additional secondary etiology of dementia or the dementia has an atypical onset, course, or presentation. Diagnostic accuracies, compared to autopsy, for possible and probable AD by NINCDS-ADRDA criteria are approximately 50–60% and 80–90%, respectively, in specialized centers. The Khachaturian pathologic criteria for AD require that the density of amyloid plaques and neurofibrillary tangles in brain sections exceed a given threshold that increases with age [52]. In contrast, the Consortium to Establish a Registry for Alzheimer’s Disease (CERAD) criteria focus exclusively on the density of amyloid plaques (the sine qua non pathologic marker of AD), in brain sections compared to given (published) high-power microscopic fields. In part because neurofibrillary tangles are not specific to AD, they were not considered essential to the diagnosis [68]. The recent ‘‘Reagan criteria’’ for AD, however, require both amyloid plaques and neurofibrillary tangles in multiple brain regions, and declare all such pathology abnormal [98]. These criteria incorporate the CERAD plaque density scale as well as Braak and Braak [5] staging of the density and distribution of pathologic abnormalities found in AD brain [5,68]. Despite numerous other profound neuropathologic changes in brain (neuronal and synaptic loss, gliosis, inflammation, cholinergic deficits, microvascular amyloid angiopathy, oxidative damage, mitochondrial dysfunction, etc) the mainstay of pathologic diagnosis remains silver staining of brain sections and light microscopic examination of the density and distribution of amyloid plaques and neurofibrillary tangles—in other words, methods used by Dr. Alzheimer in 1907. As documented by Braak and Braak [5], the neuropathology of AD is not random but affects entorhinal cortex and hippocampus followed by other limbic structures and neocortex [5]. This is consistent with the initial primarily amnestic clinical presentation of patients with AD followed my several years of progressive and gradual decline in multiple cognitive domains. As is true for all neurodegenerative diseases, the etiology of the selective vulnerability of different brain regions to AD pathologies remains obscure. The major risk factor for AD is aging. Even subjects with Down’s syndrome and individuals with genetic polymorphisms or mutations that increase risk of AD require a degree of aging before signs and symptoms commence. It remains unknown, however, what specific factors associated with aging increase risk of AD. Having a first-degree relative with AD P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 781 increases one’s risk of developing AD approximately 2- to 4-fold, and this risk grows higher with increasing numbers of affected first-degree relatives. These familial risks clearly implicate genetic factors in AD pathogenesis. However, several environmental factors also increase the risk of developing AD. For example, a low level of education or severe head trauma with loss of consciousness both increase risk. Conversely, advanced education may be protective. For unclear reasons, female gender also increases risk of AD and this is hypothesized to be due to a lack of post-menopausal estrogen [96,103]. Recently, a resurgence of studies in risk factors for stroke, such as hypertension, diabetes mellitus, smoking, hypercholesterolemia, and possibly hyperhomocysteinemia, suggest that they may also increase risk of AD. Whether these factors act directly on AD pathogenic mechanisms, or indirectly by vascular compromise, or both, remains unclear [56]. Down’s syndrome (trisomy 21), including translocation Down’s (21q), is clearly linked to AD [25]. The high prevalence of progressive dementia in subjects with Down’s syndrome led to autopsy observations of typical AD pathology, including neurofibrillary tangles and amyloid plaques, in aging brain. However, the onset of dementia occurs in the third to fifth decade of life and neuropathology is found even earlier [87]. The mechanism may be a gene dosage effect since amyloid precursor protein (APP) is encoded on chromosome 21q. Cells of Down’s syndrome patients express about 1.5 times the normal amount of APP, and thus secrete higher levels of Ab peptides derived from APP by the proteolytic enzymes termed b-secretase and c-secretase. Ab peptides, including Ab40 and Ab42, are intrinsically insoluble and are the major component of amyloid plaques in AD brain. Like many other human diseases, the identification and analysis of probands and pedigrees with rare genetic forms of a common sporadic disease proved instructive with regards to disease causation and potential treatment strategies. Thus, much has been learned about the possible cause of AD by the study of rare patients with early-onset familial AD (OMIM # 104311) [87]. Other than a pedigree analysis showing an early-onset (presenile) highly-penetrant autosomal dominant pattern of inheritance, these genetic forms of AD are similar, both clinically and pathologically, to the overwhelming majority (>95%) of patients with sporadic (senile) AD. Thus, proposed pathogenic mechanisms of familial AD may be extrapolated to sporadic AD. The first gene mutations linked to familial AD were missense mutations in the APP gene on chromosome 21. Not coincidentally, missense mutations in APP cluster near the two proteolytic cleavage sites that release Ab from APP (Fig. 1). The location of these APP mutations immediately suggested a pathologic mechanism favoring amyloidogenic (producing Ab) over nonamyloidogenic APP catabolism (a toxic gain of function). For example, a double missense mutation in APP (K670N/M671L, in the 770 isoform numbering system) near the b-secretase cleavage site increases both Ab40 and Ab42 generation. In contrast, any one of several single missense mutations 782 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 Fig. 1. Amyloid precursor protein (APP) processing. APP is a transmembrane protein that may be cleaved either by a- and c-secretases to release p3 and a large amino-terminal ectodomain (the nonamyloidogenic pathway) or by b- and c-secretases to release 4 kD Ab peptides including Ab40 and Ab42 and a large amino-terminal ectodomain (the amyloidogenic pathway). Ab is the major component of amyloid plaques in AD brain. Missense mutations causing familial AD are clustered adjacent to b - or c-secretase cleavage sites and alter APP processing to favor Ab42 generation. Mutations linked to amyloid angiopathy and dementia cluster near the a-secretase cleavage site. in APP near the c-secretase site (T714I, V715M, I716V, V717I, G, F, or L, and L723P) increases only Ab42 secretion. In vitro studies reveal that Ab42 is even more spontaneously amyloidogenic than Ab40, again suggesting a disease mechanism. The identification of these APP mutations led to the notion that amyloid deposition in brain parenchyma causes AD—the amyloid hypothesis of AD (Fig. 2)—and allowed the generation of transgenic mice that develop AD-like pathologies. In fact, these transgenic mice exhibit age-dependent behavioral decline in learning and memory and progressive amyloid deposition in brain [29,45]. However, they do not develop neurofibrillary tangles, neuronal loss, or loss of synaptic cholinergic markers, making them at best a partial AD-like model of human disease [30]. Pathogenic mutations within the Ab sequence generally result in a different phenotype than AD, and present clinically with a combination of progressive dementia and lobar hemorrhagic or microvascular ischemic strokes. The pattern of inheritance is autosomal dominant. Pathologically, they are characterized by massive amyloid accumulation within blood vessel walls in addition to parenchymal amyloid deposition as seen in AD. Pathogenic missense mutations are known within the Ab sequence at positions P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 783 Fig. 2. The amyloid cascade hypothesis of Alzheimer’s disease (AD). Ab, derived from APP, forms insoluble neuritic plaques (NP) in brain, leading to neuronal morbidity and mortality, and neurofibrillary tangles (NFT). A ghost tangle remains when a neuron containing a NFT dies. In this model, genetic risk factors for AD (Down’s syndrome, mutations in APP, presenilin-1 (PS-1), or presenilin-2 (PS-2), and the apolipoprotein E4 (ApoE4) polymorphism) are located proximally, implying their causal role in AD pathogenesis. The mechanisms whereby age, female gender, and head trauma increase risk are obscure. In parallel with these pathologic events in brain, cognitively normal elderly individuals become progressively amnestic and demented for several years until death. Current drug treatments for AD such as acetylcholinesterase inhibitors and perhaps antioxidants (vitamin E) act on putative distal targets, allowing only modest, symptomatic, and temporary clinical benefit. Novel potentially disease-modifying treatments in clinical trials may prevent or treat AD by targeting more proximate causal events. These approaches include developing b- or c-secretase inhibitors to retard Ab generation, metal chelation to prevent Ab/amyloid deposition, and immunization to promote Ab/amyloid clearance. 692, 693, and 694 near the a-secretase cleavage site (Fig. 1). Because these mutations are intrinsic to Ab, they may also alter its propensity to form insoluble amyloid fibrils and shorter protofibrils. The APP A692G (Flemish) mutation promotes Ab production but retards its fibrillogenesis; this mutation leads to a combination of microvascular amyloidopathy and AD pathology, and presents with dementia and cerebral hemorrhages. The APP E693Q (Dutch) and APP D694N (Iowa) mutations promote amyloid fibril formation from Ab and also present clinically with dementia and lobar hemorrhages; the APP E693K (Italian) mutation presents similarly, possibly by a similar mechanism. Finally, the APP E693G (Arctic) mutation retards Ab40 and Ab42 production but enhances protofibril formation and presents clinically as AD, suggesting that protofibril formation from Ab may be the unifying event in AD pathogenesis [39]. Like all amyloidopathies, the b-pleated sheet conformation of Ab in amyloid plaques and blood vessels results in their fluorescence with thioflavin-S staining as well as apple-green 784 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 birefringence in Congo red-stained sections visualized with polarized light. A cautionary note: hereditary cerebral congophilic angiopathies with dementia are not limited to mutations in the Ab sequence of APP, but include other amyloidogenic proteins such as cystatin C and transthyretin. Most familial AD does not have an APP mutation, implicating other affected genes. By study of these pedigrees, mutations were identified in presenilin-1 (for ‘‘presenile’’, or onset less than 60–65 years of age) on chromosome 14 or the homologous gene presenilin-2 on chromosome 1 (see www.alzforum.org for a complete list of familial AD mutations). In fact, of the rare early-onset familial forms of AD, the most commonly found mutation is of presenilin-1. The identification of these mutants provided a test for the amyloid hypothesis of AD. Would they alter APP metabolism and Ab generation? Again, studies of cells in culture, samples (skin fibroblasts, serum, etc.) taken from affected patients, and transgenic mice reveal that all presenilin mutations increase Ab42 generation from APP (a toxic gain of function) [86]. Double transgenic mice expressing both human mutant APP and human mutant presenilin-1 exhibit markedly accelerated amyloid deposition in brain compared to transgenic mice carrying only human mutant APP. Thus, in common to all known familial AD mutations is an increased production of Ab42 from APP. However, presenilin mutations may have other detrimental effects promoting AD, for example, by increasing neuronal apoptosis. Another major advance in the genetics of AD was the surprising discovery of the link between Apolipoprotein E (ApoE) polymorphisms on chromosome 19 and sporadic late-onset AD [94]. ApoE is synthesized in the liver and plays a role in lipid and cholesterol transport in lipoprotein particles in blood. In the brain ApoE is secreted by glia, with receptors on neurons. The function of ApoE in the brain is unclear, but it may be involved in central lipid and cholesterol metabolism. Three ApoE polymorphisms—2, 3, and 4—result in six possible genotypes. These polymorphisms differ by only one or two amino acid residues at positions 112 and 158 (out of 299). The gene frequency in the population is 3 > 4 > 2. Having either one or two apoE4 alleles increases the risk of developing late-onset AD and lowers the average age of onset with a gene dosage effect. In other words, the hierarchy of individual risk is ApoE4/4 > ApoE4/x > ApoEx/x. The ApoE2 allele is slightly protective, and ApoE3 is intermediate in risk. Genetic risks may be additive, for example in ApoE4-positive individuals with APP mutations, but the ApoE allele has no impact on the most aggressive earliest-onset form of AD found in individuals with presenilin-1 mutations. The mechanisms whereby ApoE polymorphisms affect AD risk are unknown. ApoE does not influence APP metabolism; rather, in vivo and in vitro evidence suggests that ApoE4 promotes the formation of insoluble fibrillar amyloid from soluble Ab peptides. For example, double transgenic mice have been developed expressing mutant human APP and either human ApoE3 or ApoE4; similar to humans, mice expressing human ApoE4 develop P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 785 a greater amyloid burden in brain. Murine ApoE knockout lines expressing human mutant APP develop no amyloid plaques, again suggesting a role for ApoE in amyloidogenesis. However, additional mechanisms whereby ApoE4 increases risk of AD have not been excluded. Other than ApoE, genetic linkages to the sporadic late-onset form of AD remain controversial. For example, an increased risk of developing lateonset AD is also reported with polymorphisms in the a2-macroglobulin (a2-M) and low-density lipoprotein-related receptor protein (LRP) genes– both on chromosome 12. Perhaps not coincidentally, a2-M and ApoE, both ligands for Ab and the LRP, may all play a role in Ab clearance or deposition in brain. A locus on chromosome 10 may also increase the risk of sporadic late-onset AD. New genetic linkages for increased risk of AD will no doubt be found in the future and this effort will be aided by bioinformatics—analysis of massive databases of human genome sequences, with single nucleotide polymorphisms (SNPs) and haplotypes, of large populations (such as the Icelandic study). This information will shed further light on the pathogenesis of AD, and every genetic linkage identified will be a test of the amyloid hypothesis. In favor of the amyloid hypothesis of AD is the evidence that Down’s syndrome, APP and presenilin mutations, and the ApoE4 polymorphism either promote Ab generation, especially Ab42, or its deposition in brain as amyloid. In fact, immunohistochemical stains reveal that early Ab deposits in aging brain are primarily Ab42. These pre-amyloid deposits (diffuse plaques) may evolve into mature neuritic plaques, and thus have been likened to fatty streaks that develop into atherosclerotic plaques in blood vessels. These diffuse plaques, unlike neuritic plaques, are thought to be benign because they are not surrounded by dystrophic neurites (swollen and deformed axonal and dendritic neuronal processes) and reactive gliosis (microglial and astrocytic) and are not associated with clinical dementia [87]. Evidence against the amyloid hypothesis of AD is the poor correlation of neuritic plaque burden, compared to neurofibrillary tangle density or synaptic loss, to clinical dementia, and weak evidence for linkage to other putative downstream pathologies, such as apoptosis, oxidative injury, inflammation, and mitochondrial dysfunction. For example, Ab peptides and fibrils are thought to be neurotoxic, but in vivo evidence is only suggestive, and mechanisms, perhaps involving elevated intracellular Ca2þ levels, are unclear. Also, there is poor linkage of amyloid plaques to the other major hallmark neuropathology of AD—neurofibrillary tangles that are composed primarily of phosphorylated tau. Two recent studies, however, including one with human mutant tau and APP double transgenic mice, support the notion that Ab pathology precedes and accelerates tau pathology [37,60]. Amyloid may be necessary but not sufficient to cause AD; neurofibrillary tangle formation, neuronal and synaptic loss, and other pathologies, albeit downstream of amyloid deposition, may be equally important to induce clinical dementia. 786 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 Presenilin-1 testing is commercially available (see www.athenadiagnostics. com) and may be diagnostic for AD in the rare patient with early-onset dementia (usually age 30–55) and a pedigree showing an autosomal-dominant pattern of inheritance. Because more than 60 different mutations in presenilin-1 are known (almost all single missense mutations) the entire presenilin-1 gene is sequenced, and this sometimes uncovers novel mutations. As is true in general for genetic markers of adult-onset diseases, presenilin-1 testing should not be obtained in minors, and with consenting symptomatic adults (or their legal guardian) testing must be obtained only if genetic counseling is available. Prenatal screening and testing of asymptomatic individuals in affected pedigrees raise ethical concerns. ApoE genotyping is commercially available but its application should be limited to research studies since this information adds little to the predictive value of clinical diagnosis, adds cost, and requires follow-up genetic counseling for the patient and family members [99]. Individuals with ApoE4 may not necessarily develop AD with age and many patients with AD do not carry the ApoE4 allele. Due to their rarity, genetic testing for APP and presenilin-2 mutations are not commercially available. In the late 1970s, a profound cholinergic deficit was discovered in human AD cerebral cortex. The source of this deficit is loss of cholinergic neurons in the basal forebrain (nucleus basalis of Meynert) that project to the hippocampus and neocortex and play a role in learning and memory [17]. These observations led to the hypothesis that supplementation of central cholinergic systems may be an effective treatment strategy for AD. In fact, the only drugs with proven efficacy in the treatment of cognitive deficits in patients with AD are acetylcholinesterase inhibitors [23,63]. The first of these medications approved by the U.S. Food and Drug Administration was tacrine (Cognex, approved 1993) [53]. However, this drug is limited by its Q.I.D. dosing and titration, side effects (especially nausea, vomiting, diarrhea, and hepatotoxicity), and requirement for serum alanine aminotransferase monitoring. Thus, newer acetylcholinesterase inhibitors without hepatotoxicity—donepezil (Aricept, Pfizer, NY approved 1996), rivastigmine (Exelon, Novartis, Basel, Switzerland approved 2000), and galantamine (Reminyl, Jaussen, Titusville, NJ approved 2001)—have eclipsed tacrine (Cognex, Pfizer, NY) [82]. There are few direct comparison trials of these drugs, but efficacy in improving or maintaining outcome measures over time appears comparable. Other central cholinergic supplementation strategies (cholinergic precursors, muscarinic cholinergic receptor agonists, etc) failed to prove efficacy. Other medications have questionable efficacy for the treatment of AD. Clinical trials of Ginkgo biloba extract in patients with mixed dementias are inconclusive. The antioxidants a-tocopherol (vitamin E) and selegiline (Deprenyl) delay functional decline and death in AD patients [84]. No cognitive benefits were found, but these were secondary endpoint measures in this study. There was no additive effect of the two compounds, but either was superior to placebo. P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 787 Despite flaws in this study, vitamin E (1000 IU B.I.D.) is routinely recommended to patients with AD. Although epidemiologic and pilot data are promising, treatment trials with estrogens have mostly shown no beneficial cognitive effect in postmenopausal women with AD. Likewise, despite a considerable inflammatory response to amyloid in brain and promising pilot studies of older nonsteroidal anti-inflammatory inhibitors (NSAIDs), drugs inhibiting cyclooxygenase-2 specifically (COX-2 inhibitors) have also proven disappointing in AD patients. Prednisone treatment is also without cognitive benefit in AD patients. Since retrospective epidemiologic studies appear promising, ‘‘statins’’ that lower serum cholesterol levels are being explored for potential benefit in prevention or treatment of AD. A phase II trial of atorvastatin (Lipitor) is in progress. Some clinical trials are focusing on more proximate events with the hypothesis that treatment of AD patients may be ‘‘too little, too late’’. Thus, some trials are enrolling individuals with minimal cognitive impairment (MCI), a pre-dementia syndrome with high risk of conversion to dementia [73]. For example, treatment with estrogen in post-menopausal women, donepezil (Aricept), or vitamin E, is being studied in patients with MCI for potential efficacy in delaying or preventing the onset of dementia and AD. The exact neuropsychometric boundaries between normal aging, MCI, and dementia, however, are controversial. To date, all treatments for AD are thought to be symptomatic only with no beneficial effect on underlying progressive disease processes. In support of this notion, cessation of donepezil (Aricept) results in acute loss of clinical benefits [82]. Construction of the amyloid cascade hypothesis and recent identification of the b- and c-secretases responsible for the release of Ab from APP have sparked an intense search for inhibitors of these protease enzymes as potential treatments for AD. For example, c-secretase inhibitors are now being studied in Phase II clinical trials. There is strong evidence to suggest that presenilins either are c-secretases or an important component of the c-secretase complex [87]. Unfortunately, these proteases are not specific to APP and have other important normal functions. For example, c-secretase cleaves both APP and Notch, a protein essential to normal mammalian development and adult processes such as hematopoesis. Underscoring its importance in normal development, homozygous presenilin-1 knockout mice are lethal in utero (and resemble Notch knock-out mice). A c-secretase inhibitor may therefore have dose-limiting toxic side effects. Another promising approach is inhibition of b-secretase, since mice with this gene (BACE-1) knocked out are viable and appear normal. Inhibitors of b-secretase are in the preclinical phases of investigation. Because of gliosis and humoral inflammatory responses to amyloid in AD brain tissue, it was hypothesized that immunization of human APP transgenic mice with Ab may exacerbate disease. In contrast, immunized mice develop little or no amyloid deposition, indicating a novel therapeutic strategy [85]. The mechanism of action remains unclear but immunization may 788 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 promote Ab and amyloid clearance by anti-Ab immunoglobulin (IgG) complexes and phagocytic cells (microglia) in brain; peripheral immune-mediated Ab clearance (a ‘‘sink’’) has not been excluded. Immunization not only prevents but also removes established amyloid plaques in human APP transgenic mouse brain. In support of the amyloid hypothesis, immunization prevents both plaque deposition in transgenic mouse brain and behavioral decline in learning and memory tasks. A Phase II clinical trial of Ab42 vaccination in AD patients with mild to moderate dementia was halted due to encephalitic complications. An alternate strategy with orally administered Clioquinol, (Novartis, Basel, Switzerland) a metal chelator, also prevents age-dependent amyloid plaque deposition in transgenic APP mouse brain [13]. Ab peptides bind selectively to Cu2þ and Zn2þ and thus, chelators of these ions solubilize Ab amyloid deposits in AD brain. A phase II clinical trial of Clioquinol (with vitamin B12) is in preparation. Finally, gene therapy for AD faces enormous barriers, such as access to the central nervous system, limited duration and extent of gene expression, and detrimental host responses to the vector or gene product. Nevertheless, a phase I clinical trial of nerve growth factor (NGF)-expressing fibroblasts injected into the brain of AD patients is underway. NGF promotes survival of neurons, including cholinergic neurons in the basal forebrain. AD research has become a model of how biochemical pathology and molecular genetics have quickly brought us to the threshold of safe and effective disease-modifying therapies. After decades of anosagnosia writ large and therapeutic nihilism, the clinical identification of rare patients and pedigrees with familial AD allowed the development of transgenic animal models of disease to test hypotheses and potential therapies. The challenge remains, however, to return to the clinic with new treatments for familial and sporadic AD based on our recent advances in understanding of this devastating neurodegenerative disorder. Frontotemporal dementias and related tauopathies Dr. Arnold Pick reported several patients with aphasia and dementia who had circumscribed brain atrophy on autopsy [74]. Postmortem analysis of these patients performed by Dr. Alzheimer showed that plaques similar to those seen in his now famous case of a ‘‘peculiar’’ dementing syndrome were absent in Dr. Pick’s cases [1,2]. Moreover, Alzheimer was the first to demonstrate the presence of argyrophilic inclusions and swollen achromatic neurons in this condition that became known as Pick’s disease. Progressive dementia with early personality changes, disinhibition, limited speech output leading to aphasia, and with pathologic changes consisting of focal fronto-temporal atrophy, argyrophilic intraneuronal inclusions (also known as Pick’s bodies) and achromatic neurons (Pick’s cells) has become a P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 789 prototype of fronto-temporal dementia of Pick’s type. However, it soon became apparent that the majority of patients with similar clinical features and brain atrophy limited to the frontal and temporal lobes did not have the pathologic hallmarks of Pick’s disease; histologic examination demonstrated gliosis and spongiosis in the fronto-temporal cortex. One of us (RST) reported the pathologic findings of three cases of progressive non-fluent aphasia to be this more frequent pathology termed ‘‘dementia lacking distinctive histopathology’’—neuronal loss and gliosis without neurofibrillary tangles, Lewy bodies, or Pick bodies [102]. There is considerable confusion in the literature regarding the nosology of this type of dementia. The Lund and Manchester research groups proposed clinical and pathologic criteria for this type of dementing disorder; they also suggested the term frontotemporal dementia (FTD) [97]. Even though FTD is a clinically and pathologically a heterogeneous group of dementing disorders, it is the preferred nomenclature at present (OMIM #600274). Many patients with FTD have affected first-degree relatives and a few large pedigrees with an autosomal dominant mode of inheritance have been identified. Several terms describing this condition have been used and linkage to chromosome 17q was first established in a pedigree with a dementing disorder designated as disinhibition-dementia-parkinsonism-amyotrophy complex [107]. Additional families have been mapped to 17q21.1-q21.3 suggesting genetic homogeneity of this condition [26]. The gene coding for microtubule associated protein tau (MAPT) is located within the region of chromosome 17q that was shared by all definitely linked families with FTD. Neuropathologic examination demonstrated the presence of taupositive intraneuronal inclusions in several familial cases and thus MAPT gene was a leading candidate as a cause of FTD. Identification of mutations in this gene has confirmed the link between tau and FTD [47,76]. For a complete list of tau mutations and polymorphisms linked to neurodegenerative disease, see www.alzforum.org. The exact prevalence of FTD has not been established but it is probably the third most common type of dementia after AD and dementia with Lewy bodies. FTD may account for up to 20% of all neurodegenerative dementia cases [69]. However, many patients are probably misdiagnosed. The fraction of patients with FTD diagnosed as probable AD varied from 9% in the Consortium to Establish Registry for Alzheimer’s Disease (CERAD) autopsy data to 15% in another series of demented patients [31,55]. A clinical survey using the Lund-Manchester diagnostic criteria estimated the frequency of FTD in patients with onset of dementia before the age of 65 years at 12% [83]. The prevalence of FTD increases with age similar to AD and Stevens et al, estimated the prevalence of FTD at 1.2:1,000,000 in the third decade, 3.4:1,000,000 in the fourth decade, 10.7:1,000,000 in the fifth and 28:1,000,000 in the sixth decade [93]. Similar to other neurodegenerative dementias, FTD has an insidious onset and progressive course. Personality changes with early loss of social 790 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 and personal awareness, disinhibition with socially inappropriate behavior, easy distractibility with impersistence, and perserverative or utilization behavior are typical heralding features of FTD [69,97]. Spontaneous speech becomes limited with frequent sterotypical speech and repetition of certain phrases; echolalia is also common. Patients with prominent signs of disinhibition may have increased verbosity but stereotypy of speech is typically present. Early behavioral symptoms may also include alcoholism, depression, or schizoaffective disorder and thus create difficulties in diagnosis early in the disease course. Personality changes and psychiatric symptoms may precede frank dementia by many years. Initially, short-term memory is spared together with visuospatial skills in contrast to AD; deficits in attention, problem solving skills, planning and other executive functions can be detected during neuropsychological evaluation [69,97]. However, the difference between AD and FTD in advanced stages of dementia are less prominent and both conditions can be difficult to distinguish. The presence of frontal cortical atrophy on brain MRI and frontal lobe hypoperfusion detected by SPECT or PET also support the diagnosis of FTD [69]. Two distinctive neurobehavioral syndromes have been described in patients with focal brain atrophy. An isolated deficit in expressive language with relatively preserved perception was designated as primary progressive aphasia (PPA) while severe naming and word comprehension with fluent and grammatically correct language was designated as semantic dementia (SD) [67,89,102]. Occasionally, patients from the same family may exhibit different behavioral phenotypes further supporting the notion that FTD, PPA and SD are related and reflect the differences in anatomical distribution of focal brain atrophy [69]. Frontal dementia can be associated with motor abnormalities, especially in familial cases. The concurrence of extrapyramidal syndrome with bradykinesia, rigidity, and postural instability with dementia is the most common; tremor is typically absent and hypokinetic-rigid syndrome is not levodopa responsive. Lower motor neuron dysfunction with amyotrophy can be also seen in some patients. Some families linked to chromosome 17q were originally classified based on the most conspicuous clinical signs as pallido-ponto-nigral degeneration or disinhibition-dementia-parkinsonism-amyotrophy complex [106,107]. The majority of patients with FTD have a ‘‘presenile’’ (ie, less than 65 years) onset between 50 and 60 years [93,106]. Early age of onset is also part of the proposed diagnostic criteria [69,97]. However, patients with the onset of FTD in the seventh decade have also been reported and ‘‘senile’’ (ie, more than 65 years) onset of frontal type dementia does not exclude the diagnosis of FTD [14,58]. The clinical picture of familial and sporadic cases of FTD is almost identical. Even though some studies suggest that familial cases of FTD have onset of disease ten years earlier than sporadic FTD cases (60.9  10.6 years and 72.3  8.5 years, respectively) [106], this was not confirmed in other case series of FTD [93]. P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 791 Genetic studies of patients with FTD identified several pedigrees with a clear autosomal dominant mode of inheritance. Wilhelmsen et al [107], were the first to identify linkage to a group of markers on chromosome 17q in a pedigree that they classified as disinhibition-dementia-parkinsonism-amyotrophy complex [107]. Several other pedigrees were subsequently linked to the same region. A consensus conference held in Ann Arbor, MI in 1996 identified 15 pedigrees linked to 17q21.1-q21.3 [26]. The association of an extrapyramidal syndrome with FTD was also emphasized and all families with proven linkage to chromosome 17q were designated as frontotemporal dementia and parkinsonism [26]. Analysis of clinical features of affected subjects from these families revealed that they are very similar to diagnostic criteria proposed by the Lund and Manchester research groups, further supporting the link between FTD and frontotemporal dementia and parkinsonism. There is evidence for genetic heterogeneity of FTD as a second locus on chromosome 3p11.1-q11.2 was identified in some families with autosomal dominant frontal lobe dementia [8]. The proportion of affected subjects with a positive family history of FTD is between 40% to 60% [38,93]. It is also intriguing that most patients who meet the neuropathologic criteria for Pick’s disease in the strictest sense (with Pick bodies and Pick cells) are sporadic cases. Mutations in the MAPT gene in some families linked to the FTD locus on chromosome 17q21 were identified in 1998 [47,76]. Missence and splice site mutations in this gene cosegregated with the disease thus confirming the role of tau in FTD [47,76,92]. However, there are also several FTD families with definitive linkage to 17q21 and without detected mutations in the MAPT gene; it is likely that mutations in intronic sequences or in regulatory regions of the gene disrupt the expression of tau and its biochemical properties [47,76]. Tau belongs to the microtubule associated protein (MAP) family and it contains several microtubule binding domains [10]. It is primarily found in neurons where it plays a role in microtubule assembly and stability, and may also participate in axonal extension and maintenance. The gene coding for tau has 16 exons. Tau expression is highly regulated by alternative splicing of exons 2, 3 and 10, resulting in the presence of six different isoforms, ranging from 352 to 441 amino acids, in adult human brain [10]. Each of these isoforms is likely to have different functions as their expression varies in different stages of development. Depending on whether exon 10 is excluded or included, tau has either 3 (3R tau) or 4 (4R tau) microtubule binding repeats [10]. In adult human brain the amount of 3Rtau/4Rtau is approximately equal. The identification of different types of mutation in the MAPT gene provided an important insight into the variability of pathologic changes seen in kindreds with chromosome 17-linked FTD. Several mutations affecting the exon 10 5¢ splice site have been identified [47,92]. The majority of splice site mutations lead to disruption of normal regulation of tau expression and 792 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 preferable incorporation of exon 10. This results in higher proportion of 4R tau and the functional consequence of this mutation; 4R tau binds microtubules with a higher affinity than the 3R isoform [32]. Families with this type of mutations have prominent tau pathology with mostly 4R isoforms. Missence mutations outside exon 10 alter the biochemical properties of tau, resulting in decreased microtubule binding capacity or decreased rates of tau-stimulated microtubule polymerization [41]. Tau-positive inclusions in families with this type of mutation contain both 3R tau and 4R tau isoforms. The neuropathologic classification based on the presence and ration of various isoforms of tau has been proposed [65]. Prominent tau pathology is not limited to FTD and can be seen in other neurodegenerative dementias, frequently referred to as tauopathies (see Display Box) [57]; the MPPT gene is a leading candidate gene for these disorders. Progressive supranuclear palsy (PSP), characterized by supranuclear gaze paralysis, hypokinetic-rigid syndrome with prominent postural instability and dementia is considered a sporadic disorder, though familial occurrence has been also reported [111]. Patients with PSP have a significant homozygous overrepresentation of a polymorphic dinucleotide repeat in intron 9 in the MAPT gene, called the A0 allele compared to control subjects [16]. No mutations in the MAPT gene have been found in PSP and this suggests that A0 is a low-penetrance PSP-susceptibility allele. Hyperphosphorylated tau in the neurofibrillary tangles in PSP contains only isoforms with exon 10, i.e., tau with 4 microtubule binding repeats [88]. Similarly, overrepresentation of 4R tau in tau-positive neuronal inclusions is associated with corticobasal degeneration (CBD) [65]. CBD is a sporadic disorder; aphasia, apraxia, and alien hand phenomenon combined with an asymmetric extrapyramidal syndrome are key diagnostic features of CBD [80]. Box 1. Neurodegenerative conditions with prominent tau abnormalities [58] Frontotemporal dementia (including Pick’s disease) Alzheimer disease Corticobasal degeneration Gerstmann-Sträussler-Scheinker disease Progressive supranuclear palsy Amyotrophic lateral sclerosis-Parkinsonism-Dementia complex of Guam Postencephalitic parkinsonism Dementia pugilistica Niemann-Pick disease type C Subacute sclerosis panencephalitis Trisomy 21 (Down syndrome) P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 793 Recently, a linkage disequilibrium between polymorphism CA3662 in the MAPT gene and the risk for amyotrophic lateral sclerosis-parkinsonismdementia complex of Guam has been found [77]. However, linkage analysis of one large Chamorro pedigree excluded the FTD locus on chromosome 17q suggesting that CA3662 is a modifying factor for a yet unidentified gene causing amyotrophic lateral sclerosis-parkinsonism-dementia complex of Guam. Familial prion diseases Prion disorders are a group of diseases causing a progressive neurodegenerative process with severe dementia. Prions can affect both humans and many animal species. Prion disorders can be classified as sporadic, acquired and genetic (inherited) [15]. Familial Creutzfeldt-Jakob disease (CJD) (OMIM #1234000), Gerstmann-Sträussler-Scheinker disease (GSS) (OMIM #137440) and familial fatal insomnia (FFI) (OMIM #600072) are examples of genetic prion disorders that are reviewed here. Prion disorders are also known as transmissible spongiform encephalopathies because the brain extracts from affected humans or animals are infectious (see below). Infection by slow or atypical viruses has been proposed. However, a rather extensive search failed to identify the presence of nucleic acid associated with this infection. The term prion was coined by Dr. Stanley Prusiner who suggested that prions are a novel class of infectious particles lacking either RNA or DNA [78]. The ‘‘protein infectious agent’’ or prion theory has now gained considerable support. The elucidation of pathogenesis of inherited prion disorders was one of the crucial pieces in the prion puzzle. Prions disorders have a unique position among other inherited neurologic disorders because they are caused both by different germline mutations and by infection, as they are transmissible by inoculation. The incidence of CJD has been relatively constant during the last few decades at approximately 1:1,000,000 [48]. Inherited prion disorders, especially familial CJD account for only 10–15% of all prion cases [15]. Several isolated population groups have been known to have a particularly high incidence of CJD: Chile, the Orava and Lucenec regions in Slovakia, and Libyan-Tunisian Jews are a few examples of regions and ethnic groups with an increased incidence of CJD [34]. This was attributed to various environmental factors, such as consumption of lightly cooked sheep brains and eyes by Israeli Jews born in Libya and Tunisia, who have an annual incidence of CJD between 2.3 and 3.5 per million [43]. However, detection of a germline mutation in the prion protein gene (PRNP) (see below) confirmed that the familial occurrence of CJD accounts for the increased frequency in these ethnic groups. CJD is best characterized as a rapidly progressive dementia with myoclonus [48]. Even though this description is principally true for both sporadic and familial patients, familial CJD cases may differ in some clinical aspects 794 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 that can create diagnostic difficulties. The age of disease onset in sporadic CJD is between 45 and 75 years, with the peak in the sixth decade in the majority of patients. The course of CJD is brutally fast and more than 80% of patients without a family history die within one year of onset. In general, patients with inherited forms of CJD tend to have an earlier age of onset and longer course. Moreover, there is a correlation between genotype (ie, type of mutation in the PRNP gene) and phenotype (see below). The first symptoms of CJD are quite non-specific and almost one third of the patients complain of fatigue and insomnia [6,48]. Memory complaints are prominent in another third of patients with a rapid progression to dementia. The most challenging diagnostic dilemmas arise in patients with less typical presentation, such as early focal neurologic symptoms and signs, including visual loss, aphasia, or ataxia. For instance ataxia is a heralding symptom in almost 15% patients. Ataxia was especially prominent in kuru, another form of prion disease seen in the Fore tribes of Papua New Guinea. Kuru also played an important role in our understanding of prion biology after the discovery that the disease was transmitted by consuming brains of affected persons during cannibalistic rituals [28]. CJD is rapidly progressive and patients become profoundly demented. Startle myoclonus is present in more than 80% of all patients, even though it may become less prominent in later stages of the disease. However, myoclonus is less common in familial cases and only approximately one half of inherited forms of CJD have prominent myoclonus. Pyramidal, extrapyramidal and cerebellar symptoms are also typical; lower motor neuron signs, visual disturbance or cortical blindness and choreoathetosis are less common and again, may cause diagnostic problems and delay the correct diagnosis. Akinetic mutism can be seen in terminal stages. The differences between clinical presentation were the basis for a clinical classification recognizing six main subtypes; however, there is no evidence that these subtypes have a different pathogenesis [108]. CJD can be relatively easily recognized in patients with rapidly progressive dementia and startle myoclonus. The diagnosis can be further supported by periodic discharges on electroencephalogram (EEG). Generalized, bisynchronous 1–2 cycle/sec discharges superimposed on slower background can be seen in the majority of patients. Repeat EEG recordings can increase the diagnostic yield if previous studies were negative. EEG discharges can be absent in some mutations in the PRNP gene (see below). Analysis of the 14-3-3 protein from the cerebrospinal fluid (CSF) is another useful diagnostic test [46]. This protein is found normally but is elevated in patients with CJD. However, elevation of the 14-3-3 protein in the CSF is not specific for prion disorders. Other conditions including encephalitis and acute ischemic stroke are associated with higher CSF level of 14-3-3. However, if only patients with progressive dementia and normal cell count in the CSF are selected, the sensitivity and specificity for the diagnosis of CJD is more than 95% [46]. This is true only for sporadic cases as many patients P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 795 with familial CJD have normal levels of the 14-3-3 protein. Tissue examination remains the gold standard for the diagnosis of CJD. Brain biopsy is sometimes necessary to establish the diagnosis in patients with an atypical clinical course [9]. CJD is also referred to as a spongiform encephalopathy because of prominent spongiform vacuolization of the gray matter, accompanied by gliosis and neuronal loss. Accumulation of insoluble prion protein (PrPSc indicating the scrapie isoform of the protein, also called PrPCJD) (see also below) in plaques is present in approximately 15% patients with sporadic CJD [9]. Immunostaining with antibodies against PrPSc reveals a widespread reactivity in synapses and around the vacuoles. PrPSc is derived from the cellular isoform PrPC (indicating the cellular isoform of the protein) that is encoded by the PRNP gene mapped to chromosome 20p12 [81]. The whole coding region consists of two exons and only the second exon is translated into a 253 amino acid protein. The gene also contains an unstable region of five variant tandem octapeptide coding repeats between codons 51 and 91 [15]. The first mutation in the PRNP gene was a 144 bp insertion in the open reading frame of the gene detected in a family with a typical CJD; however, the identical mutation was also detected in an unrelated family with a lateonset dementia and without any pathologic hallmarks of a spongiform encephalopathy [71]. This example indicates that the clinical phenotype of human prion disorders can be variable resulting in their underdiagnosis. The most common mutation, accounting for more than 70% of all familial cases is the glu200-to-lys mutation (E200K). This mutation is present in Libyan Jews and Slovaks with familial CJD [34,43]. A haplotype analysis confirmed that this mutation originated independently in these ethnic groups. The clinical picture of familial CJD due to E200K mutations resembles the course of sporadic cases and the average age of onset is 55  8 years; myoclonus and typical EEG changes are also common. The penetrance of this mutation is not complete (ie, not every individual who inherited the mutation will develop the disease) and is estimated around 60% to 80%; it also increases with the age [34,43]. Another common mutation in familial CJD is asp178-to-asn (D178N); this mutation is always associated with homozygosity for valine on codon 129 (see below) [35]. In general, these patients have an earlier age of onset (46  7 years) and a prolonged disease duration. EEG changes are absent in the vast majority of patients with this mutation [7]. More than 10 repeats in the region of the gene with octapeptide repeats is also associated with CJD [33]. These patients can manifest the disease as early in their twenties and the longest known disease duration in this type of mutation was 13 years. Individuals with sporadic CJD have no detectable mutations in the PRNP gene. However, certain polymorphic changes are more commonly associated with CJD, especially due to iatrogenic infection and the recently reported ‘‘new variant’’ of CJD (nvCJD) associated with the outbreak of bovine 796 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 spongiform encephalopathy (mad cow disease) in the United Kingdom [15]. The most relevant polymorphism is on codon 129 where either methionine or valine is present. Data from the Caucasian population show that half of individuals are heterozygous (51%) at residue 129; homozygosity for methionine is found in 38% and for valine in 11% [72]. Patients who developed CJD after iatrogenic transmission (human growth hormone, corneal transplants, dural grafts) are typically homozygotes for valine [15]. Homozygosity for methionine is found in the majority of sporadic cases of CJD. Moreover, all reported patients with nvCJD were homozygous for methionine [15,109]. Patients with nvCJD were atypically young with the mean age of onset 29 years. Emotional lability and dysesthesias are heralding symptoms followed by ataxia; dementia is a later finding in nvCJD [109]. Postmortem analysis demonstrated prominent amyloid plaques in the cortex and cerebellum. Similar histological findings were reported in kuru, a prion disease transmitted by cannibalism. Polymorphism on codon 129 also modifies the course of familial CJD. Individuals who inherited a PRNP mutation and are heterozygotes on codon 129 have later onset of disease that those who are homozygous either for valine or methionine [15]. GSS is a rare familial disorder with an autosomal mode of inheritance. Only a handful of families are known and the estimated prevalence is 2–5 per 100 million. GSS typically presents with progressive ataxia between the third and fourth decades, followed by the development of spasticity with hyporeflexia, extrapyramidal and lower motor neuron symptoms; dementia is usually a later sign [18]. The clinical presentation may vary even among the affected members from the same family. Pathologic hallmark of GSS are multicentric amyloid plaques that are positive for PrP protein. These plaques are most commonly found in the cerebellum but the cerebral cortex is involved as well. The extent of spongiform vacuolization varies and may be absent. GSS can be divided into three types and the discovery of various mutations in the PRNP gene supports this clinical classification [42]. The ataxic form of GSS closely resembles patients originally reported by Gerstmann, Sträussler and Scheinker. Analysis of a large pedigree with GSS showed linkage to chromosome 20 and subsequently, the pro-to-leu mutation on codon 102 was found in affected individuals [22]. This mutation was also found in other unrelated families with the same phenotype. Another type of GSS is a variant with neurofibrillary tangles (NFT). Postmortem examination of patients from an Indiana kindred with autosomal dominant progressive dementia and extrapyramidal symptoms revealed prominent NFTs and amyloid plaques. NFTs were undistinguishable from those seen in AD; however, plaques did not contain b amyloid protein but were positive for PrP protein. These patients had the phe198-to-ser mutation in the PRNP gene [42]. The third subtype of GSS is telencephalic presenting with progressive dementia; mutation ala117-to-val cosegregated with the disease in these patients [44]. P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 797 FFI is another dominantly inherited prion disease where disruption of the sleep/wake cycle leads to progressive insomnia. Ataxia and autonomic dysfunction are also common. Insomnia progresses into a dream-like condition and later into coma. Pathologic changes are mostly limited to the anterior ventral and medial dorsal nuclei of the thalamus. It was surprising that patients with FFI had the D178N mutation because the same mutation is associated with familial CJD [66]. However, patients with FFI were homozygous for methionine on residue 129 while patients whose phenotype was consistent with CJD had valine on the same residue on both alleles in the PRNP gene [36]. A sporadic form of FFI without D178N mutation has been also reported. Transmission of kuru via brain homogenate after intracerebral inoculation to chimpanzees prompted a search for viral particles as a cause of spongiform encephalopathies [28]. Subsequent research failed to identify a virus and it was proposed that spongiform encephalopathies are transmitted by a protein infectious agent or prion [78]. This theory gained wide acceptance after isolation of a protease-resistant scrapie (prion disorder affecting sheep) peptide that was linked to infectivity. The sequence of PrPSc was identical to a wild type protein encoded by the PRNP gene that was later mapped to chromosome 20 [81]. PrPSc is protease resistant, insoluble and contains b pleated sheet, while PrPC is protease sensitive, soluble, and contains only a helix. Analysis of PrPC and PrPSc did not reveal any changes in the amino sequence or known covalent posttranslational modifications [79]. Current evidence suggests that scrapie prion protein is an abnormal isoform of host-produced PrPC protein. PrPSc is derived by conformational change during conversion of PrPC protein and this conversion does not require synthesis of new protein [79]. Germline mutations in the PRNP gene most likely result in synthesis of PrPC protein that is unstable and transforms into PrPSc. This also explains the fact that familial CJD and GSS can be transmitted by brain extracts. PrPSc in sporadic or iatrogenic cases probably functions as a template that promotes conversion of PrPC into PrPSc [79]. The normal function of PrPC remains unknown. Mouse knock-out lines that do not express PrPC have a relatively unremarkable phenotype [11]. PrPC is expressed in synapses and studies with hippocampal slices showed abnormalities in inhibitory synaptic transmission. However, these mice are resistant to scrapie after intracerebral inoculation and this has further supported the hypothesis that PrPSc is derived from PrPC [12]. British and Danish familial dementia Worster-Drought et al, reported a single family in which affected members present in the fifth decade with a spastic gait disorder followed by cerebellar ataxia [110]. Cognitive decline was heralded by apathy and memory 798 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 decline, followed by a progressive dementia. The mode of transmission was consistent with an autosomal dominant mode. Histologic examination showed a conspicuous congophilic angiopathy. Two additional families that shared a common ancestry with the original family were reported in 1990 [75]. Postmortem examination showed a widespread congophilic angiopathy and non-neuritic amyloid plaques in the cerebellum, hippocampus and amygdala. Neurofibrillary tangles were present in hippocampal neurons. The classification of the disorder was uncertain and it was suggested that this was a novel type cerebral amyloid angiopathy of a British type. This disorder was later designated as familial British dementia (FBD) (OMIM #176500) [75]. Analysis of amyloid material from the plaques from one typically affected patient with FBD identified previously unknown protein subunit [104]. This peptide was derived from a novel precursor protein designated as integral membrane protein 2B (ITM2B), which has a single transmembranespanning domain. The BRI gene coding this protein is mapped to chromosome 13q14. Sequencing of the BRI gene in affected individuals with FBD detected a single base substitution in the stop codon resulting in open reading frame that is 33 nucleotides longer. Release of the 34-carboxy-terminal peptide from the abnormal ITM2B protein results in generation of insoluble amyloid peptide that was present in the vessels and non-neuritic amyloid plaques in the brains of FBD subjects [104]. BRI gene was also analyzed in patients with previously reported Danish type of dementia (OMIM # 117300) [95]. Heralding symptom in this kindred was the development of cataracts and deafness; ataxia and dementia with psychotic features were observed in the fourth decade. These patients had a 10-bp duplication one residue before the stop codon of the BRI gene, again resulting in a production of longer precursor protein with the amyloid subunit produced from the last 34 C-terminal amino acids [105]. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) Several families with recurrent strokes leading to multi-infarct dementia have been reported before 1993. The disorder was transmitted in an autosomal dominant mode. Patients, who typically did not have traditional risk factors for stroke, experienced stepwise deterioration with multiple subcortical strokes causing progressive dementia in the majority of cases. White matter degeneration with cystic changes, resembling Binswanger disease, which was associated with nonamyloid angiopathy were pathologic hallmarks. The disorder was described using several terms, including hereditary multiinfarct dementia and subcortical arteriosclerotic encephalopathy [61,90, 91]. In 1993 Tournier-Lasserve et al., studied a large pedigree with subcortical strokes and dementia, and proposed the term Cerebral Autosomal P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 799 Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy, also designated as CADASIL (OMIM # 125310) [101]. They also demonstrated a tight linkage to the group of markers on chromosome 19q12. Subsequent research by the same group led to the discovery of mutations in the NOTCH-3 gene as a cause of CADASIL [49]. CADASIL are the first example of genetically determined multi-infarct dementia. No epidemiologic data about CADASIL are available at present and this entity has been considered a rare cause of dementia. More that 150 families of different ethnic backgrounds have been reported [18]; however, it is quite likely that this condition is often unrecognized and thus more common than previously thought. Cerebral ischemia is the most common manifestation of the disease, and transient ischemic attacks (TIA) or strokes are seen in more than 80% of all patients [21,112]. The typical age of stroke onset is in the fourth decade but patients in their twenties can suffer from strokes. Only a small fraction of patients have hypertension and no stroke risk factors can be identified in the majority of affected individuals. Strokes can result in various degrees of neurologic deficit and also cause dementia. Most strokes are classic lacunar strokes localized in the subcortical white matter or basal ganglia. Cognitive decline is the second most common presentation of CADASIL [19,21,112]. Typically, cognitive deficit occurs in a step-wise fashion, reflecting new ischemic events. However, many patients may present with a progressive deterioration of cognitive function without any obvious strokes or TIAs, resembling a neurodegenerative dementing process [40]. The prevalence of dementia in CADASIL is estimated between 30% to 50% in symptomatic patients, even though more patients have signs of mild cognitive deficit without meeting formal criteria for dementia [19,21,112]. The risk for dementia increases with age and the majority of patients meet DSMIV criteria for dementia in the fifth and sixth decades [20]. Cognitive complaints are the initial symptom in approximately 5% of cases. The pattern of dementia seen in patients affected by CADASIL falls into the category of subcortical dementia with deficits in attention, episodic memory and disturbance of visuospatial skills; aphasia and apraxia are typically absent. However, cognitive deficit is quite variable even within CADASIL families. Many patients may exhibit prominent frontal lobe signs with disinhibition, apathy and perseverations. Moreover, frequency of depression is also higher in these patients and up to 20% of all patients have severe depression. Mood disorder must be distinguished from irreversible cognitive decline. Dementia is commonly associated with a gait disorder, urinary incontinence and signs of pseudobulbar palsy [19]. Analysis of larger cohorts of CADASIL patients revealed additional clinical features seen in these patients. Migraines with aura (classic migraine) are particularly common and have been reported in almost half of all patients [21,112]. Most common neurologic symptoms associated with headaches are visual and sensory disturbances but hemiplegic attacks are also 800 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 quite common. Moreover, headaches are the first symptom in 40% of all patients. Thus, migraines and TIAs are the most common presenting symptom of this entity. There is a considerable intra- and interfamilial variability in frequency of migraines and several pedigrees without migraines have been reported. The frequency of migraines may increase prior to first ischemic events in some patients. Epilepsy, especially with the onset of seizures in thirties is also common and typically is the sequel of strokes. CADASIL is a progressive disorder with shortened life span in affected individuals. The average age at death varied between 57 to 61 years with the range from 28 to 76 years. Stroke and its complications were the most common cause of death followed by pneumonia [18]. Diagnosis of CADASIL still requires a high index of suspicion and it is very likely that this condition is largely underdiagnosed. Clinical presentation of CADASIL is non-specific and pleiotropic. The diagnosis can be established relatively easy in individuals from large kindreds with a positive family and individual history of strokes, headaches and dementia. However, the hereditary nature is often overlooked because many patients are not symptomatic before reaching age 50, and some family members can be misdiagnosed. This diagnosis should be considered in patients younger than 50 years without significant cerebrovascular disease risk factors who suffer from strokes and have a history of migraines or subcortical dementia. Increased signal changes in the subcortical white matter (leukoaraiosis) and in the brainstem can be seen on T-2 weighted MR images, together with evidence of lacunar strokes in basal ganglia and centrum semiovale; cortical strokes are typically absent [100] (Fig. 3). The presence of signal changes in the external capsule has been suggested as a highly specific finding in CADASIL patients [70]. Signal hyperintensities can be detected even in presymptomatic CADASIL patients who inherited the mutation or affected individuals who have only history of migraines without TIAs or strokes [112]. MRI abnormalities are nevertheless nonspecific and the diagnosis cannot be solely based on neuroimaging criteria. Multiple sclerosis, systemic lupus erythematosus, mitochondrial encephalopathy with lactic acidosis, and Binswanger disease among others, must be distinguished from CADASIL. A definitive diagnosis of CADASIL should be supported by the presence of a mutation in the disease-causing gene or by a pathologic diagnosis demonstrating typical vascular changes. CADASIL is an arteriopathy with thickening of the media and lumen reduction of small and medium size penetrating arteries leading to multiple strokes [4]. Similar pathologic changes are present in the leptomeningeal vessels. The vascular thickening is caused by accumulation of eosinophilic material that is strongly periodic acid Schiff-positive and negative after staining for amyloid [4]. Electron microscopy is also characteristic because of the presence of granular osmiophilic granules located between degenerated smooth muscle cells or within the thickened basal lamina (Fig. 4) [4,100]. Even though clinical symptoms in CADASIL are limited to the central nervous system, similar vascular P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 801 Fig. 3. FLAIR MR imaging of the brain of a patients with CADASIL demonstrating subcortical strokes and leukoencephalopathy. Fig. 4. Electron microscopy from a skin biopsy of a CADASIL patient demonstrating osmiophilic granules. 802 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 changes are present in small vessels in multiple organs, including the skin. Evaluation of the vessels for pathologic hallmarks of CADASIL from skin biopsies has been suggested as a diagnostic test thus eliminating the need for more invasive procedures, including brain biopsy [62]. Analysis of skin biopsy, including electron microscopy confirmed that this test is both sensitive and specific for the diagnosis of CADASIL, even in early stages of the disease; however, the rate of false negative skin biopsies remains to be determined [62]. A linkage study performed on two unrelated families from northern France identified tight linkage to a group of microsatellite markers on chromosome 19q13.1 [101]. Subsequent genetic analysis of 13 additional kindreds with clinical and radiologic features of CADASIL confirmed linkage to this region and suggested that this familial stroke syndrome is genetically homogeneous. Joutel et al., identified a gene with a strong homology to a mouse Notch-3 gene and 90% of patients affected with CADASIL had mutation segregating with the disease [49]. Human Notch-3 contains 33 exons encoding for 2,321 amino acids. Notch genes are glycosylated transmembrane proteins and human Notch-3 gene contains 34 epidermal growth factor (EGF) domains in the extracellular part of the protein. Each of these EGF domains contains six highly conserved cysteine residues. Detected mutations are always missense mutations causing a gain or loss of a cysteine and thus resulting in an odd number of cysteine residues [49,50]; the only exception is one splice-site mutation that has the same consequence on the number of cysteine residues. Moreover, mutations are not distributed randomly and approximately 65% of all mutations are clustered within exons 3 and 4 encoding for first five EGF domains [50]. This facilitates screening for mutations. A genetic test for Notch-3 is available as a clinical test, even though some laboratories screen only exons 3 and 4. There is no compelling evidence that CADASIL is genetically heterogenous and the same phenotype can be caused by mutations in others genes. Japanese patients with strokes and subcortical dementia have been reported but this condition is inherited in an autosomal recessive mode; moreover, these patients also had prominent low back pain without any evidence of lumbar disk herniations [27]. How the mutation in the Notch-3 gene causes adult onset strokes and secondary dementia remains unknown. The Notch-3 gene belongs to the group of highly conserved Notch-LIN-12 genes that play an important role in intracellular signaling during development [3]. Notch-3 is highly expressed during gastrulation and in the developing central nervous system; however, postnatal expression appears to be limited to vascular smooth muscle cells [51]. The product of mutated Notch-3 gene contains unpaired cysteines and it is likely that they undergo abnormal protein conformation or abnormal cross-linking with other proteins. Two possible explanations how mutations in the Notch-3 gene may lead to the pathology seen in CADASIL are a Notch signaling defect or protein accumulation [51]. Analysis of brain P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 803 tissues obtained from patients with CADASIL revealed accumulation of a Notch-3 cleavage product, which includes an extracellular domain where all EGF domains are present at the cytoplasmatic membranes of vascular smooth muscle cells [51]. Surprisingly, the osmiophilic granules do not contain Notch-3 cleavage products. Thus, it is apparent that the smooth muscle cells in the vessels are affected by the mutations in the Notch-3 gene; however, it remains to be elucidated how this leads to vascular thrombosis. Therapy of patients with CADASIL is also controversial. Several authors have suggested antiplatelet agents based on the clinical experience with other types of strokes; however, there is no evidence based on controlled clinical trials to support this treatment. Increased incidence of microbleeds has been demonstrated in patients with CADASIL and it is plausible that antiplatelet agents or anticoagulation may increase the risk for bleeding [59]. Migraines are also common in CADASIL and triptans should be avoided because of possible vasoconstriction and increased risk of migrainous strokes. References [1] Alzheimer A. Über eine eigenartige Erkrankung der Hirnrinde. Allg Z Psychiatr 1907;64:146–8. [2] Alzheimer A. Uber eigenartige Krankheitsfalle des spateren Alters. Zeitchrift fur die Gesamte Neurologie und Psychiatrie 1911;4:356–8. [3] Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate control and signal integration in development. Science 1999;284:770–6. [4] Baudrimont M, Dubas F, Joutel A, Tournier-Lasserve E, Bousser M-G. Autosomal dominant leukoencephalopathy and subcortical ischemic stroke. A clinicopathological study. Stroke 1993;24:122–5. [5] Braak H, Braak E. Neuropathologic staging of Alzheimer related changes. Acta Neuropathol 1991;82:239–59. [6] Brown P, Cathala F, Castaigne P, Gajdusek DC. Creutzfeldt-Jakob disease: clinical analysis of a consecutive series of 230 neuropathologically verified cases. Ann Neurol 1986;20:597–602. [7] Brown P, Goldfarb LG, Kovanen J, et al. Phenotypic characteristics of familial Creutzfeldt-Jakob disease associated with the codon 178-asn PRNP mutation. Ann Neurol 1992;31:282–5. [8] Brown J, Ashworth A, Gydesen S, et al. Familial non-specific dementia maps to chromosome 3. Hum Mol Genet 1995;4:1625–8. [9] Budka H, Aguzzi A, Brown P, et al. Neuropathological diagnostic criteria for CreutzfeldtJakob disease (CJD) and other human spongiform encephalopathies (prion disease). Brain Pathol 1995;5:459–66. [10] Buee L, Bussiere T, Buee-Scherrer V, Delacourte A, Hof PR. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res Rev 2000;33:95–130. [11] Bueler H, Fischer M, Lang Y, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature 1992;356:577–82. [12] Bueuler H, Aguzzi A, Sailer A, et al. Mice devoid of PrP are resistant to scrapie. Cell 1993;73:1339–47. [13] Cherny RA, Atwood CS, Xilinas ME, et al. Treatment with a copper-zinc chelator markedly and rapidly inhibits b-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 2001;30:665–76. 804 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 [14] Chow TW, Miller BL, Hayashi VN, Geschwind DH. Inheritance of frontotemporal dementia. Arch Neurol 1999;56:817–22. [15] Collinge J. Prion diseases of humans and animals: Their causes and molecular basis. Annu Rev Neurosci 2001;24:519–50. [16] Conrad C, Andreanis A, Trojanowski JQ, et al. Genetic evidence for the involvement of tau in progressive supranuclear palsy. Ann Neurol 1997;41:277–81. [17] Davies P, Maloney AJF. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976;2:1403. [18] Davous P. CADASIL: a review with proposed diagnostic criteria. Eur J Neurol 1998; 5:219–33. [19] Desmond DW, Moroney JT, Lynch T, et al. The natural history of CADASIL. A pooled analysis of previously published cases. Stroke 1999;30:1230–3. [20] Diagnostic and Statistical Manual of Mental Disorders. (4th ed): American Psychiatric Association, Washington, D.C., 1994. [21] Dichgans M, Mayer M, Uttner I, et al. The phenotypic spectrum of CADASIL: Clinical findings in 102 cases. Ann Neurol 1998;44:731–9. [22] Doh-ura K, Tateishi J, Sasaki H, Kitamoto T, Sakaki Y. Pro-to-leu change at position 102 of prion protein is the most common but not the sole mutation related to GerstmannStraussler syndrome. Biochem Biophys Res Commun 1989;163:974–9. [23] Doody RS, Stevens JC, Beck C, et al. Practice parameter: Management of dementia (an evidence-based review). Neurology 2001;56:1154–66. [24] Evans DA, Funkenstein HH, Albert MS, et al. Prevalence of Alzheimer’s disease in a community population of older persons higher than previously reported. JAMA 1989; 262:2551–6. [25] Evenhuis HM. The natural history of dementia in Down’s syndrome. Arch Neurol 1990;47:263–7. [26] Foster NL, Wilhelmsen KC, Sima AAF, et al. Frontotemporal dementia and parkinsonism linked to chromosome 17: A consensus conference. Ann Neurol 1997;41:706–15. [27] Fukutake T, Hirayama K. Familial young-adult onset arteriosclerotic leukoencephalopathy with alopecia and lumbago without arterial hypertension. Eur Neurol 1995;35:69–79. [28] Gajdusek DC, Gibbs CJJ, Alpers MP. Experimental transmission of a kuru-like syndrome to chimpanzee. Nature 1966;209:794–6. [29] Games D, Adams D, Alessandrini R, et al. Alzheimer-type neuropathology in transgenic mice expressing V717F b-amyloid precursor protein. Nature 1995;373:523–7. [30] Gau J-T, Steinhilb ML, Kao T-C, et al. Stable b-secretase activity and presynaptic cholinergic function during progressive CNS amyloidogenesis in Tg2576 mice. Am J Pathol 2002;160:731–8. [31] Gearing M, Mirra SS, Hedreen JC, et al. The Consortium to Establish Registry for Alzheimer’s Disease (CERAD) Part X. Neuropathologic confirmation of the clinical diagnosis of Alzheimer’s disease. Neurology 1995;45:461–6. [32] Goedert M, Spillanti MG, Jakes R, Rutherford D, Crowther RA. Multiple isoforms of human microtubule-associated protein tau: sequence and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron 1989;3:519–26. [33] 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 Nat Acad Sci 1991;88:10926–30. [34] Goldfarb LG, Brown P, Mitrova E, et al. Creutzfeldt-Jakob disease associated with the PRNP codon 200LYS mutation: an analysis of 45 families. Eur J Epidemiol 1991;7:477–86. [35] Goldfarb LG, Brown P, Haltia M, et al. Creutzfeldt-Jakob disease cosegregates with the codon 178-asn PRNP mutation in families of European origin. Ann Neurol 1992;31:274–81. [36] 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;258:806–8. P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 805 [37] Gotz J, Chen F, van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301L tau transgenic mice induced by Ab42 fibrils. Science 2001;293:1491–5. [38] Gustafson L. Frontal lobe degeneration of non-Alzheimer type. Clinical picture and differential diagnosis. Arch Gerontol Geriatr 1987;6:209–23. [39] Haass C, Steiner H. Protofibrils, the unifying toxic molecule of neurodegenerative disorders? Nat Neurosci 2001;4:859–60. [40] Hedera P, Friedland RP. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy: Study of two American families with predominant dementia. J Neurol Sci 1997;146:27–33. [41] Hong M, Zhukareva V, Vogelsberg-Ragaglia V, et al. Mutation-specific functional impairments in distinct tau isoforms of hereditary FTDP-17. Science 1998;282:1914–7. [42] Hsiao K, Baker HF, Crow TJ, et al. Linkage of a prion protein missense variant to Gerstmann-Straussler syndrome. Nature 1989;338:342–5. [43] 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;324:1091–7. [44] Hsiao K, Dlouhy SR, Farlow MR, et al. Mutant prion proteins in Gerstmann-StrausslerScheinker disease with neurofibrillary tangles. Nat Genet 1992;1:68–71. [45] Hsiao K, Chapman P, Nilsen S, et al. Correlative memory deficits, Ab elevation and amyloid plaques in transgenic mice. Science 1996;274:99–102. [46] Hsich G, Kenny K, Gibbs Jr. CJ. The 14–3-3 brain protein in the cerebrospinal fluid as a marker for transmissible spongiform encephalopathies. N Engl J Med 1996;335:924–30. [47] Hutton M, Lendon CL, Rizzu P, et al. Association of missense and 5¢-splice site mutations in tau with the inherited dementia FTDP-17. Nature 1998;393:702–5. [48] Johnson RT, Gibbs Jr. CJ. Creutzfeldt-Jakob disease and related transmissible spongiform encephalopathies. N Engl J Med 1998;339:1994–2003. [49] Joutel A, Corpechot C, Ducros A, et al. Notch-3 mutations in CADASIL, a hereditary condition causing stroke and dementia. Nature 1996;383:707–10. [50] Joutel A, Vahedi K, Corpechot C, et al. Strong clustering and stereotypic nature of Notch-3 mutations in CADASIL patients. Lancet 1997;350:1511–5. [51] Joutel A, Andreux F, Gaulis S, et al. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest 2000;105:597–605. [52] Khachaturian ZS. Diagnosis of Alzheimer’s disease. Arch Neurol 1985;42:1097–105. [53] Knapp MJ, Knopman DS, Solomon PR, et al. A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer’s disease. JAMA 1994;271:985–91. [54] Knopman DS, DeKosky ST, Cummings JL, et al. Practice parameter: Diagnosis of dementia (an evidence-based review). Neurology 2001;56:1143–53. [55] Kosunen O, Soininen H, Paljarvi L, et al. Diagnostic accuracy of Alzheimer’s disease: a neuropathological study. Acta neuropathol 1996;91:185–93. [56] Kukull WA, Ganguli M. Epidemiology of dementia: concepts and overview. Neurol Clin 2000;18:923–50. [57] Lee VM-Y, Trojanowski JQ. Transgenic mouse model of tauopathies: Prospects for animal models of Pick’s disease. Neurology 2001;56(Suppl 4):S26–30. [58] Lendon CL, Lynch T, Norton J, et al. Hereditary dysphasic disinhibition dementia. A frontotemporal dementia linked to 17q21–22. Neurology 1998;50:1546–55. [59] Lesnik Oberstein SAJ, van den Boom R, van Buchem MA, et al. Cerebral microbleeds in CADASIL. Neurology 2001;57:1066–70. [60] Lewis J, Dickson DW, Lin W-L, et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 2001;293:187–491. [61] Mas JL, Dilouya A, de Recondo J. A familial disorder with subcortical strokes, dementia, and leukoencephalopathy. Neurology 1992;42:1015–20. [62] Mayer M, Straube A, Bruening R, et al. Muscle and skin biopsies are a sensitive diagnostic tool in the diagnosis of CADASIL. J Neurol 1999;246:526–32. [63] Mayeux R, Sano M. Treatment of Alzheimer’s Disease. N Engl J Med 1999;341:1670–9. 806 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 [64] McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 1984;34:939–44. [65] McKhann GM, Albert MS, Grossman M, Miller B, Dickson D, Trojanowski JQ. Clinical and pathological diagnosis of frontotemporal dementia. Report of the work group on frontotemporal dementia and Pick’s disease. Arch Neurol 2001;58:1803–9. [66] 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;326:444–9. [67] Mesulam M-M. Slowly progressive aphasia without generalized dementia. Ann Neurol 1982;11:592–8. [68] Mirra SS, Heyman A, McKeel D, et al. The consortium to establish a registry for Alzheimer’s disease (CERAD): Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology 1991;41:479–86. [69] Neary D, Snowden JS, Gustafson L, et al. Frontotemporal lobar degeneration. A consensus on clinical diagnostic criteria. Neurology 1998;51:1546–54. [70] O’Sullivan M, Jarosz JM, Martin RJ, et al. MRI hyperintensities of the temporal lobe and external capsule in patients with CADASIL. Neurology 2001;56:628–34. [71] Owen F, Poulter M, Lofthouse R, et al. Insertion in prion protein gene in familial Creutzfeldt-Jakob disease. Lancet 1989;I:51–2. [72] Owen F, Poulter M, Collinge J, Crow TJ. Codon 129 changes in the prion protein gene in Caucasians. Am J Hum Genet 1990;46:1215–6. [73] Petersen RC, Stevens JC, Ganguli M, et al. Practice parameter: Early detection of dementia: Mild cognitive impairment (an evidence-based review). Neurology 2001;56:1133–42. [74] Pick A. Uber die Beziehungen der senilen Hirnatrophie zur Aphasie. Prager Medizinische Wochenschrift 1892;17:165–7. [75] Plant GT, Revesz T, Barnard RO, Harding AE, Gautier-Smith PC. Familial cerebral amyloid angiopathy with nonneuritic amyloid plaque formation. Brain 1990;113: 721–47. [76] Poorkaj P, Bird T, Wijsman E, et al. Tau is a candidate gene for chromosome 17 frontotemporal dementia. Ann Neurol 1998;43:815–25. [77] Poorkaj P, Tsuang D, Wijsman E, et al. TAU is a susceptibility gene for amyotrophic lateral sclerosis-parkinsonism-dementia complex of Guam. Arch Neurol 2001;58:1871–8. [78] Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science 1982;216: 136–144. [79] Prusiner SB. Molecular biology of prion diseases. Science 1991;252:1515–22. [80] Riley DE, Lang AE, Lewis A, et al. Cortical-basal ganglionic degeneration. Neurology 1990;40:1203–12. [81] Robakis NK, Devine Gage EA, Jenkins EC, et al. Localization of a human gene homologous to the PrP gene on a short arm of chromosome 20 and detection of PrPrelated antigens in normal human brain. Biochem Biophys Res Commun 1986;140: 758–65. [82] Rogers SL, Friedhoff LT. The efficacy and safety of donepezil in patients with Alzheimer’s disease: results of a US Multicentre, Randomized, Double-Blind, Placebo-Controlled Trial. The Donepezil Study Group. Dementia 1996;7:293–303. [83] Rossor MN. Pick’s disease. A clinical overview. Neurology 2001;56(Suppl 4):S3–5. [84] Sano M, Ernesto C, Thomas RG, et al. A controlled trial of selegiline, a-tocopherol, or both as treatments for Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. N Engl J Med 1997;336:1216–22. [85] Schenk D, Barbour R, Dunn W, et al. Immunization with amyloid-b attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999;400:173–7. [86] Scheuner D, Eckman C, Jensen M, et al. Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nat Med 1996;2:864–70. P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 807 [87] Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev 2001;81:741–66. [88] Sergeant N, Wattez A, Delacourte A. Neurofibrillary degeneration in progressive supranuclear palsy and corticobasal degeneration: tau pathologies with exclusively ‘exon 10’ isoforms. J Neurochem 1999;72:1243–9. [89] Snowden JS, Goulding PJ, Neary D. Semantic dementia: a form of circumscribed atrophy. Behav Neurol 1989;2:167–82. [90] Sonninem V, Savontaus M-L. Hereditary multi-infarct dementia. Eur Neurol 1987; 27:209–15. [91] Sourander P. Walinder. Hereditary multi-infarct dementia. Morphological and clinical studies of a new disease. Acta Neuropathol 1977;39:247–54. [92] Spillanti MG, Murrell JR, Goedert M, et al. Mutation in the tau gene in familial multiple system tauopathy with presenile dementia. Proc Natl Acad Sci USA 1998;96:7737–41. [93] Stevens M, van Duijn CM, Kamphorts W, et al. Familial aggregation in frontotemporal dementia. Neurology 1998;50:1541–5. [94] Strittmatter WJ, Saunders AM, Schmechel D, et al. Apoliproprotein E—high avidity binding to b-amyloid and increased frequency of type-4 allele in late-onset Alzheimer’s disease. Proc Natl Acad Sci USA 1994;90:1977–81. [95] Stromgrem E, Dalby A, Dalby MA, Ranheim B. Cataracts, deafness, cerebellar ataxia, psychosis, and dementia – a new syndrome. Acta Neurol Scand 1970;46(suppl.43):261–2. [96] Tang MX, Jacobs D, Stern Y, et al. Effect of oestrogen during menopause on risk and age at onset of Alzheimer’s disease. Lancet 1996;348:429–32. [97] The Lund and Manchester Groups. Clinical and neuropathological criteria for frontotemporal dementia. J Neurol Neurosurg Psychiat 1994;57:416–8. [98] The National Institute on Aging and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathologic Assessment of Alzheimer’s Disease. Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. Neurobiol Aging 1997;18:S1–2. [99] The Ronald and Nancy Reagan Research Institute of the Alzheimer’s Association and the National Institute on Aging Working Group. Consensus report of the Working Group on: Molecular and biochemical markers of Alzheimer’s disease. Neurobiol Aging 1998;19:109–16. [100] Tournier-Lasserve E, Iba-Zizen M-T, Romero N, Bousser M-G. Autosomal dominant syndrome with stroke-like episodes and leukoencephalopathy. Stroke 1991;22:1297–302. [101] Tournier-Lasserve E, Joutel A, Melki J, et al. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy maps to chromosome 19q12. Nat Genet 1993;3:256–9. [102] Turner RS, Kenyon LC, Trojanowski JQ, Gonatas N, Grossman M. Clinical, neuroimaging, and pathologic features of progressive non-fluent aphasia. Ann Neurol 1996;39: 166–73. [103] Turner RS. Alzheimer’s disease in man and transgenic mice. Females at higher risk. Am J Pathol 2001;158:797–801. [104] Vidal R, Frangione B, Rostagno A, et al. A stop-codon mutation in the BRI gene associated with familial British dementia. Nature 1999;399:776–81. [105] Vidal R, Revesz T, Rostagno A, et al. A decamer duplication in the 3-prime region of the BRI gene originates an amyloid peptide that is associated with dementia in a Danish kindred. Proc Nat Acad Sci 2000;97:4920–5. [106] Wijker M, Wszolek ZK, Wolters ECH, et al. Localization of the gene for rapidly progressive autosomal dominant parkinsonism and dementia with pallido-ponto-nigral degeneration to chromosome 17q21. Hum Mol Genet 1996;5:151–4. [107] Wilhelmsen KC, Lynch T, Pavlov E, Higgins M, Hygaard TG. Localization of disinhibition-dementia-parkinsonism-amyotrophy complex to 17q21–22. Am J Hum Genet 1994;55:1159–65. [108] Will RG, Matthews WB. A retrospective study of Creutzfeldt-Jakob disease in England and Wales 1970–79. I: Clinical features. J Neurol Neurosurg Psych 1984;47:134–140. 808 P. Hedera, R.S. Turner / Neurol Clin N Am 20 (2002) 779–808 [109] Will RG, Zeidler M, Stewart GE, et al. Diagnosis of new variant Creutzfeldt-Jakob disease. Ann Neurol 2000;47:575–82. [110] Worster-Drought C, Hill TR, McMenemey WH. Familial presenile dementia with spastic paralysis. Brain 1933;14:27–34. [111] Yebenes de JG, Sarasa JL, Daniel SE, Lees AJ. Familial progressive supranuclear palsy. Description of a pedigree and review of the literature. Brain 1995;118:1095–103. [112] Chabriat H, Vahedi K, Iba-Zizen MT, et al. Clinical spectrum of CADASIL: a study of 7 families. Lancet 1995;346:934–9.