Your activity: 12 p.v.

Frontotemporal dementia: Epidemiology, pathology, and pathogenesis

Frontotemporal dementia: Epidemiology, pathology, and pathogenesis
Author:
Suzee E Lee, MD
Section Editor:
Steven T DeKosky, MD, FAAN, FACP, FANA
Deputy Editor:
Janet L Wilterdink, MD
Literature review current through: Dec 2022. | This topic last updated: Sep 22, 2021.

INTRODUCTION AND TERMINOLOGY — Frontotemporal dementia (FTD) is a clinically and neuropathologically heterogeneous disorder characterized by disturbances in behavior, personality, and language accompanied by focal degeneration of the frontal and/or temporal lobes.

FTD serves as an umbrella term for several clinical syndromes, including behavioral variant FTD (bvFTD), semantic variant primary progressive aphasia (svPPA), and nonfluent variant primary progressive aphasia (nfvPPA).

The term "frontotemporal lobar degeneration" (FTLD) applies to the pathological diagnoses associated with the clinical FTD spectrum. This pathologic entity can be subdivided according to the nature of the characteristic cytoplasmic or nuclear protein inclusions that are observed histopathologically and believed to be neurotoxic.

Each clinical FTD syndrome may be associated with different neuropathologies, which poses a significant challenge to the development and testing of therapies designed to target specific protein dysfunction in neurodegeneration. Moreover, studies reveal the significant neuropathological and genetic overlap between motor neuron disease and FTD.

This topic will review the epidemiology, pathogenesis, and pathology of FTD. The clinical manifestations, diagnosis, and treatment of FTD are discussed separately. (See "Frontotemporal dementia: Clinical features and diagnosis" and "Frontotemporal dementia: Treatment".)

EPIDEMIOLOGY — FTD is one of the more common causes of early-onset dementia and occurs at similar frequency to Alzheimer disease (AD) in patients younger than 65 years. A systematic review of 26 studies spanning Europe, Asia, North America, and South America found that, on average, FTD accounted for 2.7 percent of diagnoses among patients with dementia 65 years and older. For patients with dementia younger than 65 years, FTD accounted for 10.2 percent of diagnoses [1]. It has been suggested that the estimated incidence and prevalence of FTD are underreported, since many patients present with behavioral changes and are likely to be referred to psychiatric services [2].

The mean age of onset of FTD is approximately 58 years [3-8]. The reported age of onset has ranged from 20 to 80 years, although onset before 40 or after 75 years of age is unusual.

In most case series, the proportion of males and females with FTD is roughly equal [1], but some series have reported a male preponderance [2-5,8,9].

GENETIC FACTORS — FTD is highly heritable. Studies suggest that 30 to 50 percent of patients with FTD have at least one relative with dementia [10-12], with an autosomal-dominant inheritance pattern observed in approximately 10 to 25 percent of patients with FTD [4,5,7,8,10,13].

Patients with behavioral variant FTD (bvFTD), particularly when associated with motor neuron disease, are most likely to have an inherited condition; in one series, more than half of such patients demonstrated a family history consistent with autosomal-dominant inheritance [10].

The most common disease-causing genetic mutations include those in microtubule-associated protein tau (MAPT), the granulin precursor (GRN) gene, and a hexanucleotide expansion in chromosome 9 open reading frame 72 (C9orf72). Among patients with FTD who carry a genetic mutation, over 80 percent have a mutation in one of these three genes [14]. Mutations in these three genes account for approximately 15 percent of patients with a family history of dementia [15,16].

Mutations in other genes, including valosin-containing protein (VCP), charged multivesicular body protein 2B (CHMP2B), and TANK binding kinase 1 (TBK1), are rare [15,16]. The co-occurrence of two evidently pathogenic mutations in the same patient (eg, C9orf72 expansion plus a mutation in either GRN or MAPT) [17] and homozygous MAPT [18,19], GRN [20], and C9orf72 [21] expansion carriers have been reported.

MAPT mutations — Over 60 pathogenic autosomal-dominant mutations in MAPT have been identified [22]. MAPT mutations typically cause bvFTD with or without parkinsonism [23]. Some MAPT mutation carriers with parkinsonism also meet criteria for progressive supranuclear palsy (PSP) or corticobasal degeneration (CBD) [24]. Patients with semantic variant primary progressive aphasia (svPPA), PSP, and CBD have been reported but are less common. Mutations that do not affect the splicing of exon 10 in MAPT are typically associated with the bvFTD syndrome, while mutations that affect exon 10 splicing and increase the ratio of four repeat (4R) tau to three repeat (3R) tau more frequently cause bvFTD with PSP or parkinsonism [25-27]. The R406W mutation may present with an amnestic Alzheimer disease (AD)-like syndrome. (See 'FTLD-tau' below and 'Clinicopathological correlations' below.)

Compared with other frontotemporal lobar degeneration (FTLD) genetic mutations, MAPT mutation carriers have an earlier age of symptom onset, typically between the ages of 40 and 60 years [22]. In contrast to sporadic bvFTD, MAPT mutation carriers also may show early memory impairment with corresponding mesial temporal lobe atrophy [28,29].

At autopsy, patients with FTLD due to MAPT mutations invariably feature FTLD-tau pathology, yet the mechanism by which MAPT gene mutations result in FTLD is not well understood.

GRN mutations — Like MAPT, the gene encoding progranulin (GRN) also lies on chromosome 17. The function of progranulin is complex, with important roles as a growth factor in development, wound repair, neuroinflammation, autophagy, and lysosomal function [30], but its role in neurodegeneration remains unclear [31-34].

The age of symptom onset ranges widely (35 to 87 years), but on average, disease onset occurs later than in MAPT carriers [31,35,36].

Over 100 autosomal-dominant mutations in GRN have been associated with FTLD [22]. All known mutations result in a truncated progranulin mRNA that undergoes nonsense-mediated decay via the nonsense-mediated surveillance system, which results in a haploinsufficiency of functional progranulin protein [37,38]. Progranulin haploinsufficiency results in FTLD-TDP pathology, but the mechanism by which this occurs remains unknown.

GRN mutation carriers typically develop bvFTD, nonfluent variant primary progressive aphasia (nfvPPA), or CBD, and patients with an amnestic AD-like syndrome have been described [39]. In some patients with GRN-associated FTLD, structural magnetic resonance imaging (MRI) brain scans demonstrate extensive white matter lesions, in addition to the more typical pattern of focal frontal, temporal, and parietal lobe atrophy [40,41]. (See 'FTLD-TDP' below and 'Clinicopathological correlations' below.)

C9orf72 expansion — A noncoding hexanucleotide repeat on chromosome 9 within C9orf72 is the most common genetic cause of familial FTD and amyotrophic lateral sclerosis (ALS), representing approximately 12 and 25 percent of familial cases, respectively [42,43]. The C9orf72 expansion has also been identified in patients with sporadic ALS and FTD. Compared with sporadic ALS, ALS due to C9orf72 is associated with earlier disease onset, bulbar disease, higher frequency of comorbid FTD, and shorter age-matched median survival. (See "Familial amyotrophic lateral sclerosis", section on 'C9ORF72 gene'.)

While the predominant clinical presentation of C9orf72 expansion carriers is bvFTD, ALS, or both (FTD-MND), patients with svPPA and nfvPPA have been reported [44-46].

Possible pathophysiologic mechanisms include haploinsufficiency of C9orf72 protein, the development of repeat RNA foci translated from the expanded repeat, and abnormal dipeptide-repeat (DPR) proteins translated from the repeat mRNA [47]. Defects in nucleocytoplasmic transport through the nuclear pore may also contribute to disease [48-51]. Repeat length appears highly variable across brain regions and other tissues, including blood, even within an individual [52,53]. Studies are mixed, with most showing no clear associations of repeat length with cognitive impairment [53], clinical syndrome (eg, FTD versus ALS), age of onset [52], or age at death [54], although longer repeats may be associated with longer disease duration in FTD, but not ALS [54], or shorter survival [52]. C9orf72 expansions result in FTLD-TDP pathology. (See 'FTLD-TDP' below.)

Other genetic mutations — Rare genetic variants associated with FTD include mutations in VCP, CHMP2B, TBK1, TAR DNA-binding protein (TARDBP; the gene that encodes TDP-43), sequestosome 1 (SQSTM1), triggering receptor expressed on myeloid cells 2 (TREM2), and coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) [55,56]. VCP is associated with bvFTD, inclusion body myositis, Paget bone disease, and familial ALS [57-59]. Mutations in TARDBP, fused in sarcoma (FUS), and ubiquilin 2 (UBQLN2) are linked to familial ALS but are rarely associated with FTD clinical presentations. (See "Familial amyotrophic lateral sclerosis", section on 'ALS14 (VCP gene)' and "Familial amyotrophic lateral sclerosis", section on 'ALS10 (TARDBP gene)' and "Familial amyotrophic lateral sclerosis", section on 'ALS6 (FUS gene)'.)

SHARED PATHOLOGICAL FEATURES — The gross pathology of all frontotemporal lobar degeneration (FTLD) subtypes reveals degeneration in the frontal and/or temporal lobes. Cortical and/or basal ganglia atrophy may be symmetric or asymmetric, and substantia nigra depigmentation emerges in a subset of cases. While behavioral variant FTD (bvFTD) typically targets the fronto-insula and the anterior cingulate cortex, specific atrophy patterns may be associated with underlying neuropathology [60], although the prediction of an individual's underlying pathology based on atrophy patterns generally remains tenuous.

Microscopically, most FTLD cases demonstrate microvacuolation and neuronal loss accompanied by swollen neurons, loss of myelin, and astrocytic gliosis. In addition, abnormal protein inclusions are seen in the cytoplasm and/or nuclei of neuronal and/or glial cells. The constituents of these abnormal protein inclusions are known in most cases and are used to classify FTLD pathology into distinct subtypes. (See 'Pathological subtypes' below and 'Clinicopathological correlations' below.)

PATHOLOGICAL SUBTYPES — Nearly all frontotemporal lobar degeneration (FTLD) cases are classified into three major subtypes based on the composition of the protein inclusions and their morphology and anatomic distribution. These major categories of protein inclusions are tau, the TAR DNA-binding protein 43 (TDP-43), and FET proteins.

Together, FTLD-tau and FTLD-TDP account for approximately 90 percent of FTLD cases [61]. Approximately 50 percent of FTLD cases feature FTLD-tau pathology, while FTLD-TDP accounts for approximately 40 percent, and FTLD-FET accounts for 5 to 10 percent. FTLD in which inclusions are labeled only by the markers of the ubiquitin-proteasome system (UPS) is rare (<1 percent) and is found in patients carrying the rare CHMP2B mutation [62].

FTLD-tau — Tau protein is encoded by the MAPT gene on chromosome 17. Tau stabilizes microtubules, promotes microtubule assembly, and regulates axonal transport [63-65].

Tau protein is alternatively spliced into six isoforms, among which are a three amino acid sequence repeat isoform (3R) and a four repeat isoform (4R). In normal cells, 3R and 4R tau occur in equal proportions. In FTLD-tau pathology, the ratio of hyperphosphorylated 3R and 4R tau varies amongst pathological subtypes. FTLD-tau neuropathologies are thus subclassified by whether tau inclusions are predominantly composed of 3R or 4R tau, or a mixture of both. Pick disease is classified as a 3R tauopathy. FTLD due to a MAPT mutation and chronic traumatic encephalopathy feature both 3R and 4R tau inclusions. Corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), and less common pathologies such as argyrophilic grain disease and globular glial tauopathy (GGT) predominantly feature inclusions with the 4R species. Chronic traumatic encephalopathy, CBD, and PSP are discussed separately. (See "Sequelae of mild traumatic brain injury", section on 'Chronic traumatic encephalopathy' and "Corticobasal degeneration" and "Progressive supranuclear palsy (PSP): Clinical features and diagnosis".)

The earliest report of FTLD pathology described distinctive round, silver-staining inclusions named Pick bodies [66,67]. While the term "Pick disease" historically referred to both the clinical and pathological diagnosis, current FTLD nomenclature uses "Pick disease" to refer to the pathological diagnosis only. In Pick disease, Pick bodies develop in the granule cells of the dentate gyrus, the pyramidal neurons in the CA1 region of the hippocampus, and the pyramidal neurons in the frontal and temporal lobes. Pick bodies are spherical tau-positive neuronal cytoplasmic inclusions (NCIs) composed primarily of 3R tau and occur in association with achromatic ballooned neurons, or Pick cells [68]. Tau deposition in Pick disease progresses sequentially, beginning in the frontotemporal limbic/paralimbic and neocortical regions, then involving subcortical structures (eg, basal ganglia, locus ceruleus, raphe nuclei), primary motor cortex and precerebellar nuclei, and finally visual cortex [69]. Pick disease presents more commonly as behavioral variant FTD (bvFTD), although nonfluent variant primary progressive aphasia (nfvPPA), semantic variant primary progressive aphasia (svPPA), and CBD are also associated clinical syndromes [60,69,70].

Rare FTLD-tau pathologies include the pathology associated with MAPT mutation carriers, argyrophilic grain disease, and GGT.

FTLD caused by a MAPT mutation, also known as FTD with parkinsonism linked to chromosome 17 (FTDP-17), exhibits hyperphosphorylated tau inclusions in neurons and glia in cortical and subcortical regions. The characteristics of tau pathology vary between MAPT families, with some correlation between the specific MAPT mutation and pathological features [24]. Mutations that affect the splicing of exon 10 and increase the ratio of 4R to 3R tau result in a pathology reminiscent of PSP and CBD [25-27,71]. Mutations in other exons may lead to a predominance of either Pick bodies (3R tau) or Alzheimer disease (AD)-like neurofibrillary tangles with a mixture of 3R and 4R [71]. (See 'Genetic factors' above.)

Argyrophilic grain disease was first described as a pathological finding with small, spindle-shaped argyrophilic inclusions or "grains" consisting of 4R tau within medial temporal lobes and limbic regions, which may also be seen in other cortical regions and the lateral hypothalamus [72]. Argyrophilic grain disease may arise alone or along with other neuropathologies and has been seen in association with aging, amnestic mild cognitive impairment (MCI), AD-like dementia, and slowly progressive bvFTD [73-78]. Aside from slower disease progression, clinical features that may distinguish MCI due to argyrophilic grain disease versus AD have yet to be determined.

GGT (also reported as "sporadic multiple system tauopathy with dementia") is a rare 4R tau pathology characterized by widespread globular neuronal and glial tau-positive inclusions in gray and white matter [79]. The clinical presentations of GGT include bvFTD with or without parkinsonism, primary lateral sclerosis, or a combination of these two syndromes [80].

FTLD-TDP — TDP-43 protein is an RNA binding protein present in the nucleus of normal cells and involved in transcription regulation [81-84]. In FTLD-TDP, abnormally phosphorylated and ubiquitinated TDP-43 appears in various abnormal morphologies, including NCIs, neuronal intranuclear inclusions, dystrophic neurites, and glial cytoplasmic inclusions [81,83-85]. Based on the types of inclusions and their distribution throughout the brain, FTLD-TDP can be further subdivided into four types [62,86]:

Type A, with many compact NCIs and many short, thick dystrophic neurites, predominantly in layer 2 of the cortex

Type B, with moderate diffuse granular NCIs and few dystrophic neurites, in all layers of cortex

Type C, with few NCIs and many long, thick dystrophic neurites, in all layers of cortex

Type D, with few NCIs; many short, thick dystrophic neurites; and many lentiform neuronal intranuclear inclusions, in all layers of cortex

Type A is the most common subtype, accounting for approximately 40 to 50 percent of FTLD-TDP cases, followed by type B (approximately 30 percent), type C (up to 25 percent), and type D, which is rare [83,87]. The mechanisms through which TDP-43 protein abnormalities lead to neurodegeneration are a focus of intense research but are not yet understood.

Several autosomal-dominant FTLD mutations result in FTLD-TDP pathology:

FTLD caused by mutations in GRN [37,88] is typically associated with FTLD-TDP type A pathology. (See 'GRN mutations' above.)

FTLD and amyotrophic lateral sclerosis (ALS) caused by the C9orf72 repeat expansion is associated with variable degrees of FTLD-TDP pathology [43,89] and also inclusions of dipeptide-repeat (DPR) proteins directly derived from the expanded repeat [90]. Most patients show TDP type B neuropathology, though some show features more consistent with type A or a mixture between types A and B. (See 'C9orf72 expansion' above.)

Inclusion body myositis with Paget disease of bone and FTD, caused by VCP gene mutations [91], results in type D pathology.

TDP-43 inclusions are also seen in most cases of ALS, in patients with and without clinical FTD symptomatology [92,93]. Based on the significant clinical and pathologic overlap among FTLD-TDP with or without motor neuron disease and TDP-positive ALS with or without dementia, it has been suggested that TDP-43 proteinopathies exist along a clinicopathologic spectrum of multisystem neurodegenerative diseases [94]. (See "Epidemiology and pathogenesis of amyotrophic lateral sclerosis", section on 'Pathology'.)

FTLD-FET — Mutations in the gene coding for FUS protein were originally identified as a cause of familial ALS. (See "Familial amyotrophic lateral sclerosis", section on 'ALS6 (FUS gene)'.)

Given the overlap between motor neuron disease and FTD, and FUS protein's functional homology with TDP-43, the involvement of FUS in FTD was explored, and most cases of FTLD with inclusions that were tau and TDP negative were found to stain positively for FUS protein [95]. FUS is a member of the FET protein family, which consists of other RNA- and DNA-binding proteins in Ewing sarcoma (EWS) and TATA-box binding protein associated factor 15 (TAF15) [96]. In addition to FUS, these pathological inclusions were also found to harbor EWS and TAF15 proteins [97]. FET proteins are also found in basophilic inclusion body disease (BIBD) and neuronal intermediate filament inclusion disease (NIFID) [98,99]. Together, FTLD-FET is estimated to account for 5 to 10 percent of all FTLD cases.

FTLD-UPS — FTLD cases that only label for markers of UPS are rare. Most FTLD-UPS cases are associated with FTD linked to chromosome 3 caused by mutations in CHMP2B. The neuropathology includes ubiquitin- and p62-positive granular neurocytoplasmic inclusions (that are negative for tau, TDP-43, and FUS proteins) [100,101].

CLINICOPATHOLOGICAL CORRELATIONS — Predicting the underlying pathology remains challenging for behavioral variant FTD (bvFTD), yet the specific primary progressive aphasia (PPA) subtypes do show stronger clinicopathological correlations.

In one series of bvFTD, 29 percent had FTLD-tau, 47 percent had FTLD-TDP, and 7 percent had FTLD-FUS (with 13 percent showing a primary diagnosis of Alzheimer disease [AD] and 3 percent with amyotrophic lateral sclerosis [ALS]) [60].

Semantic variant PPA (svPPA) is nearly always associated with FTLD-TDP type C (ranging from 75 to 100 percent across various cohorts) [102].

Nonfluent variant PPA (nfvPPA) is most commonly associated with a four repeat (4R) tau pathology, with some reports describing an association with TDP type A [102].

Logopenic variant PPA is usually caused by AD pathology, reported in up to 95 percent in one series [103].

Viewed from the pathological-clinical perspective:

FTLD-tau pathology is most commonly associated with bvFTD, nfvPPA, and corticobasal degeneration (CBD).

Within FTLD-TDP, common clinical syndromes for type A include bvFTD, nfvPPA, and CBD [86,104]. Type B mostly commonly appears as bvFTD with or without motor neuron disease. Type C nearly always presents as svPPA, and much less commonly bvFTD [104]. Type D, the rarest subtype, has only been identified in VCP mutation carriers. In one series, hallucinations in the initial years of the disease were related to TDP-43 pathology, including but not limited to C9orf72 repeat expansion carriers [105]. The presence of perseverative or compulsive behavior was more common in the type B and C histotypes.

FTLD-FUS is the most common of the FTLD-FET pathologies and is strongly associated with the bvFTD phenotype, usually in association with ALS.

SUMMARY

Terminology – Frontotemporal dementia (FTD) is a clinically and neuropathologically heterogeneous disorder characterized by disturbances in behavior, personality, and language accompanied by focal degeneration of the frontal and/or temporal lobes.

Frontotemporal lobar degeneration (FTLD) denotes the pathological diagnoses associated with the clinical FTD spectrum. In addition to behavioral variant FTD (bvFTD), nonfluent variant primary progressive aphasia (nfvPPA), and semantic variant primary progressive aphasia (svPPA), FTLD neuropathology is found in patients with motor neuron disease, corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP). (See 'Introduction and terminology' above.)

Epidemiology – FTD is a common cause of early-onset dementia and occurs at a similar frequency to Alzheimer disease (AD) in patients younger than 65 years. The mean age of onset is 58 years. (See 'Epidemiology' above.)

Genetic factors – Thirty to 50 percent of patients with FTD have at least one relative with dementia. The most common pathogenic genetic mutations include variants in MAPT, GRN, and the C9orf72 repeat expansion. (See 'Genetic factors' above.)

Pathology – The gross pathology of FTLD includes symmetric or asymmetric atrophy in the frontal and/or temporal lobes. Characteristic microscopic findings include microvacuolation and neuronal loss, accompanied by swollen neurons, loss of myelin, and astrocytic gliosis. In addition, abnormal protein inclusions are seen in the cytoplasm and/or nuclei of neuronal and/or glial cells. (See 'Shared pathological features' above.)

FTLD is subdivided according to the properties of the neuropathological inclusions. The majority of FTLD cases are found to harbor inclusions composed of hyperphosphorylated tau protein or the TDP-43 protein. FTLD-FET pathology is less common, accounting for only 5 to 10 percent of FTLD cases. (See 'Pathological subtypes' above.)

FTLD-tau includes several different pathological subtypes, including Pick disease, chronic traumatic encephalopathy, CBD, PSP, the pathology of MAPT mutation carriers, argyrophilic grain disease, and global glial tauopathy. (See 'FTLD-tau' above.)

In FTLD-TDP, abnormally phosphorylated and ubiquitinated TDP-43 appears as various morphologies. FTLD-TDP pathology is also found in patients with genetic mutations in GRN and VCP and the C9orf72 repeat expansion. (See 'FTLD-TDP' above.)

Clinicopathological correlations – Predicting the underlying pathology remains challenging for bvFTD; most show either FTLD-tau or FTLD-TDP neuropathology.

Specific variants of primary progressive aphasia (PPA) show strong associations with neuropathology. svPPA is nearly always associated with TDP type C. nfvPPA is most commonly associated with a four repeat (4R) tau pathology. The logopenic variant of PPA is usually caused by AD. (See 'Clinicopathological correlations' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Bruce L Miller, MD, who contributed to an earlier version of this topic review.

  1. Hogan DB, Jetté N, Fiest KM, et al. The Prevalence and Incidence of Frontotemporal Dementia: a Systematic Review. Can J Neurol Sci 2016; 43 Suppl 1:S96.
  2. Ratnavalli E, Brayne C, Dawson K, Hodges JR. The prevalence of frontotemporal dementia. Neurology 2002; 58:1615.
  3. Johnson JK, Diehl J, Mendez MF, et al. Frontotemporal lobar degeneration: demographic characteristics of 353 patients. Arch Neurol 2005; 62:925.
  4. Rosso SM, Donker Kaat L, Baks T, et al. Frontotemporal dementia in The Netherlands: patient characteristics and prevalence estimates from a population-based study. Brain 2003; 126:2016.
  5. Hodges JR, Davies RR, Xuereb JH, et al. Clinicopathological correlates in frontotemporal dementia. Ann Neurol 2004; 56:399.
  6. Grossman M. Frontotemporal dementia: a review. J Int Neuropsychol Soc 2002; 8:566.
  7. Hodges JR, Davies R, Xuereb J, et al. Survival in frontotemporal dementia. Neurology 2003; 61:349.
  8. Knibb JA, Xuereb JH, Patterson K, Hodges JR. Clinical and pathological characterization of progressive aphasia. Ann Neurol 2006; 59:156.
  9. Mesulam MM. Primary progressive aphasia. Ann Neurol 2001; 49:425.
  10. Goldman JS, Farmer JM, Wood EM, et al. Comparison of family histories in FTLD subtypes and related tauopathies. Neurology 2005; 65:1817.
  11. Binetti G, Nicosia F, Benussi L, et al. Prevalence of TAU mutations in an Italian clinical series of familial frontotemporal patients. Neurosci Lett 2003; 338:85.
  12. Bird T, Knopman D, VanSwieten J, et al. Epidemiology and genetics of frontotemporal dementia/Pick's disease. Ann Neurol 2003; 54 Suppl 5:S29.
  13. Le Ber I, Guedj E, Gabelle A, et al. Demographic, neurological and behavioural characteristics and brain perfusion SPECT in frontal variant of frontotemporal dementia. Brain 2006; 129:3051.
  14. Rademakers R, Neumann M, Mackenzie IR. Advances in understanding the molecular basis of frontotemporal dementia. Nat Rev Neurol 2012; 8:423.
  15. Sieben A, Van Langenhove T, Engelborghs S, et al. The genetics and neuropathology of frontotemporal lobar degeneration. Acta Neuropathol 2012; 124:353.
  16. Mignarri A, Battistini S, Tomai Pitinca ML, et al. Double trouble? Progranulin mutation and C9ORF72 repeat expansion in a case of primary non-fluent aphasia. J Neurol Sci 2014; 341:176.
  17. van Blitterswijk M, Baker MC, DeJesus-Hernandez M, et al. C9ORF72 repeat expansions in cases with previously identified pathogenic mutations. Neurology 2013; 81:1332.
  18. Ng AS, Sias AC, Pressman PS, et al. Young-onset frontotemporal dementia in a homozygous tau R406W mutation carrier. Ann Clin Transl Neurol 2015; 2:1124.
  19. Behnam M, Ghorbani F, Shin JH, et al. Homozygous MAPT R406W mutation causing FTDP phenotype: A unique instance of a unique mutation. Gene 2015; 570:150.
  20. Canafoglia L, Morbin M, Scaioli V, et al. Recurrent generalized seizures, visual loss, and palinopsia as phenotypic features of neuronal ceroid lipofuscinosis due to progranulin gene mutation. Epilepsia 2014; 55:e56.
  21. Fratta P, Poulter M, Lashley T, et al. Homozygosity for the C9orf72 GGGGCC repeat expansion in frontotemporal dementia. Acta Neuropathol 2013; 126:401.
  22. Greaves CV, Rohrer JD. An update on genetic frontotemporal dementia. J Neurol 2019; 266:2075.
  23. 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.
  24. Ghetti B, Wszolek ZK, Boeve BF, et al. Frontotemporal dementia and parkinsonism linked to chromosome 17. In: Neurodegeneration: The Molecular Pathology of Dementia and Movement Disorders, 2nd ed, Dickson DW, Weller RO (Eds), Wiley-Blackwell, 2011. p.110.
  25. Delisle MB, Murrell JR, Richardson R, et al. A mutation at codon 279 (N279K) in exon 10 of the Tau gene causes a tauopathy with dementia and supranuclear palsy. Acta Neuropathol 1999; 98:62.
  26. Iijima M, Tabira T, Poorkaj P, et al. A distinct familial presenile dementia with a novel missense mutation in the tau gene. Neuroreport 1999; 10:497.
  27. Skoglund L, Viitanen M, Kalimo H, et al. The tau S305S mutation causes frontotemporal dementia with parkinsonism. Eur J Neurol 2008; 15:156.
  28. Whitwell JL, Jack CR Jr, Boeve BF, et al. Voxel-based morphometry patterns of atrophy in FTLD with mutations in MAPT or PGRN. Neurology 2009; 72:813.
  29. Rohrer JD, Ridgway GR, Modat M, et al. Distinct profiles of brain atrophy in frontotemporal lobar degeneration caused by progranulin and tau mutations. Neuroimage 2010; 53:1070.
  30. Kao AW, McKay A, Singh PP, et al. Progranulin, lysosomal regulation and neurodegenerative disease. Nat Rev Neurosci 2017; 18:325.
  31. Bruni AC, Momeni P, Bernardi L, et al. Heterogeneity within a large kindred with frontotemporal dementia: a novel progranulin mutation. Neurology 2007; 69:140.
  32. He Z, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med (Berl) 2003; 81:600.
  33. Zhu J, Nathan C, Jin W, et al. Conversion of proepithelin to epithelins: roles of SLPI and elastase in host defense and wound repair. Cell 2002; 111:867.
  34. Chitramuthu BP, Bennett HPJ, Bateman A. Progranulin: a new avenue towards the understanding and treatment of neurodegenerative disease. Brain 2017; 140:3081.
  35. Pickering-Brown SM, Rollinson S, Du Plessis D, et al. Frequency and clinical characteristics of progranulin mutation carriers in the Manchester frontotemporal lobar degeneration cohort: comparison with patients with MAPT and no known mutations. Brain 2008; 131:721.
  36. Rizzu P, van Mil SE, Anar B, et al. CHMP2B mutations are not a cause of dementia in Dutch patients with familial and sporadic frontotemporal dementia. Am J Med Genet B Neuropsychiatr Genet 2006; 141B:944.
  37. Baker M, Mackenzie IR, Pickering-Brown SM, et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006; 442:916.
  38. Cruts M, Gijselinck I, van der Zee J, et al. Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 2006; 442:920.
  39. Le Ber I, Camuzat A, Hannequin D, et al. Phenotype variability in progranulin mutation carriers: a clinical, neuropsychological, imaging and genetic study. Brain 2008; 131:732.
  40. Caroppo P, Le Ber I, Camuzat A, et al. Extensive white matter involvement in patients with frontotemporal lobar degeneration: think progranulin. JAMA Neurol 2014; 71:1562.
  41. Sudre CH, Bocchetta M, Cash D, et al. White matter hyperintensities are seen only in GRN mutation carriers in the GENFI cohort. Neuroimage Clin 2017; 15:171.
  42. Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011; 72:257.
  43. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 2011; 72:245.
  44. Sha SJ, Takada LT, Rankin KP, et al. Frontotemporal dementia due to C9ORF72 mutations: clinical and imaging features. Neurology 2012; 79:1002.
  45. Simón-Sánchez J, Dopper EG, Cohn-Hokke PE, et al. The clinical and pathological phenotype of C9ORF72 hexanucleotide repeat expansions. Brain 2012; 135:723.
  46. Rohrer JD, Isaacs AM, Mizielinska S, et al. C9orf72 expansions in frontotemporal dementia and amyotrophic lateral sclerosis. Lancet Neurol 2015; 14:291.
  47. Gendron TF, Belzil VV, Zhang YJ, Petrucelli L. Mechanisms of toxicity in C9FTLD/ALS. Acta Neuropathol 2014; 127:359.
  48. Haeusler AR, Donnelly CJ, Periz G, et al. C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature 2014; 507:195.
  49. Zhang K, Donnelly CJ, Haeusler AR, et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 2015; 525:56.
  50. Freibaum BD, Lu Y, Lopez-Gonzalez R, et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 2015; 525:129.
  51. Jovičić A, Mertens J, Boeynaems S, et al. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat Neurosci 2015; 18:1226.
  52. van Blitterswijk M, DeJesus-Hernandez M, Niemantsverdriet E, et al. Association between repeat sizes and clinical and pathological characteristics in carriers of C9ORF72 repeat expansions (Xpansize-72): a cross-sectional cohort study. Lancet Neurol 2013; 12:978.
  53. Nordin A, Akimoto C, Wuolikainen A, et al. Extensive size variability of the GGGGCC expansion in C9orf72 in both neuronal and non-neuronal tissues in 18 patients with ALS or FTD. Hum Mol Genet 2015; 24:3133.
  54. Suh E, Lee EB, Neal D, et al. Semi-automated quantification of C9orf72 expansion size reveals inverse correlation between hexanucleotide repeat number and disease duration in frontotemporal degeneration. Acta Neuropathol 2015; 130:363.
  55. Deleon J, Miller BL. Frontotemporal dementia. Handb Clin Neurol 2018; 148:409.
  56. Häkkinen S, Chu SA, Lee SE. Neuroimaging in genetic frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 145:105063.
  57. Johnson JO, Mandrioli J, Benatar M, et al. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 2010; 68:857.
  58. Gijselinck I, Van Mossevelde S, van der Zee J, et al. Loss of TBK1 is a frequent cause of frontotemporal dementia in a Belgian cohort. Neurology 2015; 85:2116.
  59. Van Mossevelde S, van der Zee J, Gijselinck I, et al. Clinical features of TBK1 carriers compared with C9orf72, GRN and non-mutation carriers in a Belgian cohort. Brain 2016; 139:452.
  60. Perry DC, Brown JA, Possin KL, et al. Clinicopathological correlations in behavioural variant frontotemporal dementia. Brain 2017; 140:3329.
  61. Snowden J, Neary D, Mann D. Frontotemporal lobar degeneration: clinical and pathological relationships. Acta Neuropathol 2007; 114:31.
  62. Neumann M, Mackenzie IRA. Review: Neuropathology of non-tau frontotemporal lobar degeneration. Neuropathol Appl Neurobiol 2019; 45:19.
  63. Ebneth A, Godemann R, Stamer K, et al. Overexpression of tau protein inhibits kinesin-dependent trafficking of vesicles, mitochondria, and endoplasmic reticulum: implications for Alzheimer's disease. J Cell Biol 1998; 143:777.
  64. Sato-Harada R, Okabe S, Umeyama T, et al. Microtubule-associated proteins regulate microtubule function as the track for intracellular membrane organelle transports. Cell Struct Funct 1996; 21:283.
  65. Hirokawa N, Funakoshi T, Sato-Harada R, Kanai Y. Selective stabilization of tau in axons and microtubule-associated protein 2C in cell bodies and dendrites contributes to polarized localization of cytoskeletal proteins in mature neurons. J Cell Biol 1996; 132:667.
  66. Pick A. Ueber die Beiehungen der senilen Hirnatrophie zur Aphasie. Prager Medicinische Wochenschrift 1892; 17:165.
  67. Alzheimer A. Über eigenartige Krankheitsfälle des späteren Alters. Z Gesamte Neurol Psychiatr 1911; 4:356.
  68. Zhukareva V, Mann D, Pickering-Brown S, et al. Sporadic Pick's disease: a tauopathy characterized by a spectrum of pathological tau isoforms in gray and white matter. Ann Neurol 2002; 51:730.
  69. Irwin DJ, Brettschneider J, McMillan CT, et al. Deep clinical and neuropathological phenotyping of Pick disease. Ann Neurol 2016; 79:272.
  70. Graff-Radford NR, Damasio AR, Hyman BT, et al. Progressive aphasia in a patient with Pick's disease: a neuropsychological, radiologic, and anatomic study. Neurology 1990; 40:620.
  71. Mackenzie IR, Neumann M. Molecular neuropathology of frontotemporal dementia: insights into disease mechanisms from postmortem studies. J Neurochem 2016; 138 Suppl 1:54.
  72. Braak H, Braak E. Cortical and subcortical argyrophilic grains characterize a disease associated with adult onset dementia. Neuropathol Appl Neurobiol 1989; 15.
  73. Grinberg LT, Wang X, Wang C, et al. Argyrophilic grain disease differs from other tauopathies by lacking tau acetylation. Acta Neuropathol 2013; 125:581.
  74. Ishihara K, Araki S, Ihori N, et al. Argyrophilic grain disease presenting with frontotemporal dementia: a neuropsychological and pathological study of an autopsied case with presenile onset. Neuropathology 2005; 25:165.
  75. Tsuchiya K, Mitani K, Arai T, et al. Argyrophilic grain disease mimicking temporal Pick's disease: a clinical, radiological, and pathological study of an autopsy case with a clinical course of 15 years. Acta Neuropathol 2001; 102:195.
  76. Rodriguez RD, Suemoto CK, Molina M, et al. Argyrophilic Grain Disease: Demographics, Clinical, and Neuropathological Features From a Large Autopsy Study. J Neuropathol Exp Neurol 2016; 75:628.
  77. Ding ZT, Wang Y, Jiang YP, et al. Argyrophilic grain disease: frequency and neuropathology in centenarians. Acta Neuropathol 2006; 111:320.
  78. Jicha GA, Petersen RC, Knopman DS, et al. Argyrophilic grain disease in demented subjects presenting initially with amnestic mild cognitive impairment. J Neuropathol Exp Neurol 2006; 65:602.
  79. Cairns NJ, Bigio EH, Mackenzie IR, et al. Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol 2007; 114:5.
  80. Ahmed Z, Bigio EH, Budka H, et al. Globular glial tauopathies (GGT): consensus recommendations. Acta Neuropathol 2013; 126:537.
  81. Davidson Y, Kelley T, Mackenzie IR, et al. Ubiquitinated pathological lesions in frontotemporal lobar degeneration contain the TAR DNA-binding protein, TDP-43. Acta Neuropathol 2007; 113:521.
  82. Arai T, Hasegawa M, Akiyama H, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 2006; 351:602.
  83. Cairns NJ, Neumann M, Bigio EH, et al. TDP-43 in familial and sporadic frontotemporal lobar degeneration with ubiquitin inclusions. Am J Pathol 2007; 171:227.
  84. Neumann M, Mackenzie IR, Cairns NJ, et al. TDP-43 in the ubiquitin pathology of frontotemporal dementia with VCP gene mutations. J Neuropathol Exp Neurol 2007; 66:152.
  85. Sampathu DM, Neumann M, Kwong LK, et al. Pathological heterogeneity of frontotemporal lobar degeneration with ubiquitin-positive inclusions delineated by ubiquitin immunohistochemistry and novel monoclonal antibodies. Am J Pathol 2006; 169:1343.
  86. Mackenzie IR, Neumann M, Baborie A, et al. A harmonized classification system for FTLD-TDP pathology. Acta Neuropathol 2011; 122:111.
  87. Josephs KA, Hodges JR, Snowden JS, et al. Neuropathological background of phenotypical variability in frontotemporal dementia. Acta Neuropathol 2011; 122:137.
  88. Cruts M, Rademakers R, Gijselinck I, et al. Genomic architecture of human 17q21 linked to frontotemporal dementia uncovers a highly homologous family of low-copy repeats in the tau region. Hum Mol Genet 2005; 14:1753.
  89. Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2012; 72:257.
  90. Mori K, Weng SM, Arzberger T, et al. The C9orf72 GGGGCC Repeat Is Translated into Aggregating Dipeptide-Repeat Proteins in FTLD/ALS. Science 2013.
  91. Watts GD, Wymer J, Kovach MJ, et al. Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nat Genet 2004; 36:377.
  92. Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006; 314:130.
  93. Mackenzie IR, Bigio EH, Ince PG, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol 2007; 61:427.
  94. Geser F, Lee VM, Trojanowski JQ. Amyotrophic lateral sclerosis and frontotemporal lobar degeneration: a spectrum of TDP-43 proteinopathies. Neuropathology 2010; 30:103.
  95. Urwin H, Josephs KA, Rohrer JD, et al. FUS pathology defines the majority of tau- and TDP-43-negative frontotemporal lobar degeneration. Acta Neuropathol 2010; 120:33.
  96. Mackenzie IR, Neumann M. FET proteins in frontotemporal dementia and amyotrophic lateral sclerosis. Brain Res 2012; 1462:40.
  97. Neumann M, Bentmann E, Dormann D, et al. FET proteins TAF15 and EWS are selective markers that distinguish FTLD with FUS pathology from amyotrophic lateral sclerosis with FUS mutations. Brain 2011; 134:2595.
  98. Munoz DG, Neumann M, Kusaka H, et al. FUS pathology in basophilic inclusion body disease. Acta Neuropathol 2009; 118:617.
  99. Neumann M, Roeber S, Kretzschmar HA, et al. Abundant FUS-immunoreactive pathology in neuronal intermediate filament inclusion disease. Acta Neuropathol 2009; 118:605.
  100. Holm IE, Englund E, Mackenzie IR, et al. A reassessment of the neuropathology of frontotemporal dementia linked to chromosome 3. J Neuropathol Exp Neurol 2007; 66:884.
  101. Holm IE, Isaacs AM, Mackenzie IR. Absence of FUS-immunoreactive pathology in frontotemporal dementia linked to chromosome 3 (FTD-3) caused by mutation in the CHMP2B gene. Acta Neuropathol 2009; 118:719.
  102. Montembeault M, Brambati SM, Gorno-Tempini ML, Migliaccio R. Clinical, Anatomical, and Pathological Features in the Three Variants of Primary Progressive Aphasia: A Review. Front Neurol 2018; 9:692.
  103. Santos-Santos MA, Rabinovici GD, Iaccarino L, et al. Rates of Amyloid Imaging Positivity in Patients With Primary Progressive Aphasia. JAMA Neurol 2018; 75:342.
  104. Spinelli EG, Mandelli ML, Miller ZA, et al. Typical and atypical pathology in primary progressive aphasia variants. Ann Neurol 2017; 81:430.
  105. Scarioni M, Gami-Patel P, Timar Y, et al. Frontotemporal Dementia: Correlations Between Psychiatric Symptoms and Pathology. Ann Neurol 2020; 87:950.
Topic 14138 Version 20.0

References