Mutations
MAPT N279K
Quick Links
Overview
Pathogenicity: Frontotemporal Dementia : Pathogenic
Clinical
Phenotype: Frontotemporal Dementia
Position: (GRCh38/hg38):Chr17:46010324 T>G
Position: (GRCh37/hg19):Chr17:44087690 T>G
dbSNP ID: rs63750756
Coding/Non-Coding: Coding
DNA
Change: Substitution
Expected RNA
Consequence: Splicing Alteration
Expected Protein
Consequence: Isoform Shift; Missense
Codon
Change: AAT to AAG
Reference
Isoform: Tau Isoform Tau-F (441 aa)
Genomic
Region: Exon 10
Research
Models: 1
Findings
Among MAPT mutations, the N279K is one of the most frequent causes of familial frontotemporal dementia (FTD). The mutation has been identified in more than a dozen families worldwide, with data suggesting autosomal dominant inheritance with full penetrance. N297K is a very rare variant in the general population; indeed, it is absent from the gnomAD variant database (v.4.1.0, Apr 2024). Phenotypic presentation varies, but affected individuals generally develop symptoms of dementia, parkinsonism, and progressive supranuclear palsy (PSP). Individuals with the N279K mutation typically have a parkinsonism-predominant phenotype, consistent with the pallido-ponto-nigral degeneration (PPND) subtype of FTD. Personality changes, behavioral changes, and dementia also occur, but they may be less prominent and/or seen later in the course of the disease (Tsuboi et al., 2002).
One of the largest N279K kindreds is from North America, known as the PPND family, and has been longitudinally studied since 1987 (Caviness et al., 2003). This family comprises at least 57 affected people across eight generations. Individuals in this family develop a particularly aggressive form of disease characterized by progressive parkinsonism with dystonia, rigidity, bradykinesia, postural instability, dementia, ocular motility abnormalities, pyramidal tract dysfunction, frontal lobe release signs, personality changes, perseverative vocalizations, and urinary incontinence (Wszolek et al., 1992; Markopoulou et al., 2016). Olfactory dysfunction also appears prior to symptom onset, and develops in an age-dependent and irreversible manner; however, severe olfactory dysfunction is not fully penetrant (Markopoulou et al., 2016; Arvanitakis et al., 2007). Sleep impairment is also prominent in affected N279K-carrying individuals, as demonstrated by a reduced total sleep time and increased sleep latency, among other parameters, compared to unaffected family members (Spector et al., 2011). Mutation carriers in the PPND family have an average age of onset of 43 years and disease duration of 8 years (Markopoulou et al., 2016).
The N279K mutation has also been detected throughout the world beyond North America, including in more than 10 Japanese families (Arima et al., 2000; Yasuda et al. 1999; Tsuboi et al., 2002; Ogaki et al., 2011; Ogaki et al., 2012; Ikeda et al., 2019; Oka et al., 2020; Shimura et al., 2005). In at least three of these families, the clinical syndromes resembled PSP (Ogaki et al., 2011; Ogaki et al., 2012). In one of the largest of these families, family B, the reported pedigree indicates nine individuals over three generations affected by parkinsonism (n=7) or dementia with parkinsonism (n=2). The N279K mutation was present in three affected carriers (two with parkinsonism with dementia, one with parkinsonism without dementia) and apparently absent in a relative who remained healthy into her 60s, well past the family's mean age of onset which ranged from 42 to 46 years of age. Although the authors imply this latter individual did not carry the mutation, her genotype was not explicitly reported. All three affected carriers had parkinsonism as the initial symptom. Symptoms included bradykinesia, rigidity, postural tremors, gaze palsy, and motor impairments.
In another family, two individuals with parkinsonism- and one with dementia-predominant disease were reported, all of whom first presented symptoms early in the fourth decade (Oka et al., 2020). Of note, the patient with dementia-predominant disease clinically presented with bvFTD, and PSP was pathologically confirmed at death, 6 years after onset. All three individuals exhibited personality changes as well as bradykinesia, rigidity, and postural instability. Although they had affected family members, their mutational status had not been confirmed.
The N279K mutation was also identified in a large Chinese family with autosomal-dominant parkinsonism (Yang et al., 2015). The reported pedigree shows 21 affected family members over four generations. Prior to the identification of their mutation, affected family members were misdiagnosed with Parkinson’s disease due to their prominent parkinsonian symptoms (e.g., rigidity, hypokinesia, postural instability, and tremor). The diagnosis was later corrected to FTD with parkinsonian features. Disease in this family developed early (onset ranged from 39 to 48 years of age), and progressed rapidly (duration varied from 4 to 11 years). In addition to parkinsonism, personality changes were prominent early in the disease, including apathy, irritability, and disinhibition, and worsened as the disease progressed. Some family members were affected by supranuclear palsy, and dementia was common, although cognitive decline occurred subsequent to parkinsonism. Thirty-nine family members were genotyped (five affected, 34 unaffected) and the N279K mutation appeared to segregate with the presence of disease, but the ages of the unaffected non-carriers were not reported.
Beyond this extensive family study, N279K has been identified in others from China (Tang et al., 2016, Wu et al., 2018, Nan et al., 2024). Of note, segregation with disease was described in at least one of these studies in which a female patient from a FTD-parkinsonism family was found to carry N279K (Tang et al., 2016). She was initially misdiagnosed with Parkinson's disease based on tremor, bradykinesia, and rigidity at age 40, but by her second admission at age 44, she exhibited behavioral changes and increased irritability, leading to a revised diagnosis of FTD-parkinsonism. Two of her three siblings were affected, including a deceased brother and a living sister with similar symptoms who also carries N279K. At the time of publication, her other living sister and their father were unaffected and did not carry the mutation.
N279K has also been detected in South America. A 45-year-old Brazilian male was described with the behavioral variant of FTD (bvFTD) and PSP characterized by axial-predominant parkinsonian features and eye-movement abnormalities (Takada et al., 2016). This individual had four siblings who had died due to disease suggestive of PSP.
Reports of patients carrying N279K have also been described in Europe. An Irish 36-year-old female patient with the N279K mutation was diagnosed with PPND after being misdiagnosed with Parkinson’s disease by three specialists (O’Dowd et al., 2012). Her father (genetics not reported) had been diagnosed with Parkinson’s disease, although he had a poor response to levodopa therapy. Her initial symptoms included slowness of movement while swimming, reduced facial expressions, and insomnia, but by 44 years of age she required full nursing care and her symptoms included akinesia, aphasia, apraxia of eyelid opening, dysphagia, and cachexia. Her 42-year-old sister, also an N279K carrier, also exhibited a lack of facial expression. Italian (Soliveri et al., 2003) and French (Delisle et al., 1999) families with the N279K mutation have also been reported and include patients with dementia and PSP.
In a large international, retrospective cohort study, 44 individuals from 17 families with the N279K mutation were identified in the Frontotemporal Dementia Prevention Initiative and the published literature, including 22 with parkinsonism and 12 with dementia not otherwise specified (Moore et al., 2020). The mean age of disease onset was 43.8 years (n=36), the mean age at death was 52.4 years (n=38), and the mean disease duration was 6.5 years (n=31). No other information is provided about these families, and it is unclear whether it overlaps with the families described above. Of note, this study included both confirmed mutation carriers, as well as family members who were assumed to be carriers based on their clinical phenotype.
In sum, N279K is a common mutation underlying familial FTD, characterized by early onset and rapid progression. It presents globally across groups of diverse ancestries, with a predominant parkinsonism phenotype, and is often initially misdiagnosed as Parkinson's disease.
Neuropathology
Autopsy findings have revealed severe neuronal loss with gliosis in the frontal and temporal lobes, substantia nigra, pontine tegmentum, and globus pallidus, with some loss also in the caudate and putamen (Slowinski et al., 2007; Oka et al., 2020). MRI of a female Chinese Han patient also revealed bilateral frontal and temporal lobe atrophy (Nan et al., 2024). Notably absent from patients with the N279K mutation are plaques, tangles, Lewy bodies, and amyloid bodies (Wszolek et al., 1992).
Nonetheless, although tau tangles are not typical, they do occur (Al-Dalahmah et al., 2024), and other types of tau inclusions have been described in neurons and glia in a variety of brain regions, including the cortex, basal ganglia, hippocampus, brain-stem nucleipons, and spinal cord white matter (Slowinski et al., 2007; Oka et al., 2020). Tau pathology includes pretangles, threads, grain-like granules, and coiled bodies (Slowinski et al., 2007). Moreover, tau pathology was also present in sleep-related brain regions, which correlated with the sleep dysfunction observed in some affected individuals (Spector et al., 2011). Inclusions consisted primarily of four-repeat (4R), not three-repeat (3R), isoforms of tau, consistent with PSP (Delisle et al., 1999; Ogaki et al., 2012). In some cases, substantial tau deposition was also observed in the medial temporal cortices and upper and lower motor neurons with accompanying corticospinal tract degeneration (Arima et al., 2000).
As the disease progresses from preclinical to symptomatic stages, imaging studies have provided insights into brain changes associated with the N279K mutation. Three siblings from the PPND kindred were examined at different disease stages (preclinical, age 43; prodromal, age 45; and mild FTD, age 51) for striatal dopamine transporter (DAT) binding using 123I-FP-CIT SPECT, and scores were inversely correlated with disease progression (Miyagawa et al., 2024). Notably, DAT expression was found to be reduced already in presymptomatic stages, which is in line with a finding in presymptomatic mutation carriers from a Chinese Han family using a different radioligand to detect DAT (Wu et al., 2018). All PPND family siblings eventually developed bvFTD and died between the ages of 52 and 55 (Miyagawa et al., 2024). At autopsy, frontotemporal degeneration was observed in all three individuals, showing extensive neuronal and glial 4R tau pathology along with neuronal loss and gliosis in the substantia nigra. An earlier study using the same imaging method to assess DAT had similar findings, showing that two affected N279K carriers who were diagnosed with bvFTD with parkinsonism had lower DAT density in the caudate nucleus and anterior putamen compared to control individuals and also when compared to patients with Parkinson’s disease (Takeshige et al., 2018).
Tau pathology associated with N279K appears to be distinct from that seen in Alzheimer's disease (AD). The PET imaging ligand 18F-AV-1451 (also known as flortaucipir or T807) was used to evaluate AD–type tau aggregates in the temporal pole in five individuals with the N279K mutation, including one asymptomatic participant and four participants with FTD with parkinsonism and PPND (Jones et al., 2018). While the signal was lower than in patients with AD and lower than in those with MAPT mutations outside of exon 10, it was similar to that in clinically healthy individuals.
The role of microglial activation in the early stages of disease has been examined. In three presymptomatic N279K gene carriers (aged 38-41 years) from the large PPND family, increased microglial activation was detected in the frontal, occipital, and posterior cingulate cortices by PET imaging compared to healthy controls (Miyoshi et al., 2010). Low dopamine synthesis rates were also observed by PET in the putamen, and MRI further revealed hippocampal atrophy.
Metabolic changes in the brain are another aspect explored through imaging in N279K mutation carriers. In the Irish patient with PPND described above, FDG-PET revealed hypometabolism in the mesial temporal lobes and increased T2 signal on fluid attenuated inversion recovery MRI in the same regions 5 years after symptom onset (O’Dowd et al., 2012).
In sum, neuropathological studies of patients carrying the N279K mutation reveal a pattern of severe neuronal loss, distinctive 4R tau pathology without typical AD features, and notable changes in brain metabolism and microglial activation, as evidenced by various imaging techniques across different disease stages.
Biological Effect
Tau splicing
N279K is located in an exonic splicing enhancer (D'Souza et al., 1999; Rodriguez-Martin et al., 2009). Similar to many intronic mutations in MAPT, the N279K mutation affects exon 10 splicing in vitro, resulting in more frequent incorporation of exon 10 into transcripts and the relative over-production of the 4R tau isoform (Delisle et al., 1999; Hasegawa et al., 1999). Indeed, in frontal cortex and cerebellum samples from patients with PPND, the 4R-to-3R ratio was greater than that in control brain samples (Hong et al., 1998). This occurs through the strengthening of a poly-purine positive cis-element in exon 10, which promotes greater exon 10 inclusion during splicing (Hutton et al., 2001). Namely, the splicing factors Tra2β and SF2/ASF have a stronger affinity to the exonic splicing enhancer in the presence of N279K (Jiang et al., 2003; Kondo et al., 2004; D’Souza and Schellenberg et al., 2006; Rodriguez-Martin et al., 2009). N279K may also lead to greater exon 10 inclusion by reducing binding to the splicing regulator SRp30c, which under wild-type conditions inhibits splicing of exon 10 (Wang et al., 2005).
To examine the effects of this mutation on splicing, induced pluripotent stem cells (iPSCs) have been generated from fibroblasts of affected N279K carriers. iPSC-derived neuronal stem cells carrying the N279K mutation also produce more 4R tau than 3R tau (Wren et al., 2015); this was also replicated in another model of iPSC-derived neurons (Ehrlich et al., 2015; Al-Dalahmah et al., 2024).
Tau aggregation
Hexapeptide motifs within the tau protein are important for tau aggregation and conformation, and several are found in the microtubule binding region, including the PHF6∗ motif (VQIINK), which coincides with the location of N279K (Karikari et al., 2019; Karikari et al., 2020). These motifs regulate aggregation through tau-tau interactions, which facilitate a shift from a random coil to a cross β-sheet structure. The K18 fragment, which encompasses several hexapeptide motifs, was modified to express the N279K mutation in one study to evaluate tau conformation and aggregation in vitro (Karikari et al., 2019). Transmission electron microscopy findings showed that while the filament length did not differ from those produced by wild-type K18 fragments, N279K K18 fragments exhibited wider filaments. This indicated that the morphology of tau aggregates can be affected by the N279K mutation. Moreover, the conformation of N279K K18 fragments differed from that seen in WT K18 based on distinct immunostaining patterns to antibodies detecting different epitopes.
Despite these alterations in the structure of tau aggregates, N279K does not seem to fuel tau aggregation per se. In N279K-expressing HEK293T cells that were exposed to preformed wild-type K18 tau fibrils, which are aggregation-prone, tau did not accumulate, indicating a lack of seeding and tau inclusion formation (Strang et al., 2018). In fact, in one in vitro study using thioflavin S dye fluorescence to measure aggregation, an N279K tau fragment was found to aggregate at a rate even slower than that of wild-type tau (Yao et al., 2003); this was also true for mutated K18 fragments in another study, but not for full-length tau (Barghorn et al., 2000).
Tau turnover
This mutation also appears to disrupt tau turnover. In silico analyses predicted it diminishes cathepsin cleavage which was confirmed using in vitro protease assays (Sampognaro et al., 2023). Consistent with these findings, the lysosomal degradation of mutant tau in neuronal-like SH-SY5Y cells, as well as in neurons derived from iPSCs, was reduced.
In addition, when N279K K18 tau oligomers were applied to SH-SY5Y cells, they were internalized through endocytosis to a greater extent than wild-type K18 oligomers (Karikari et al., 2019). Nonetheless, application of mutant oligomers did not affect cell viability as measured by lactate dehydrogenase release, a proxy for cytotoxicity.
Tau localization
Tau is usually localized to axons and associated with microtubules, although it can be found in other cellular compartments including the plasma membrane and nucleus. In COS-7 cells carrying the N279K mutation, tau was found to localize in the nucleus to a greater extent than in wild-type cells using confocal microscopy and subcellular fractitionation plus western blot methods (Ritter et al., 2018). Despite this change in the subcellular distribution of mutant tau, cell viability remained the same. In iPSC-derived neurons, however, somatodendritic redistribution of phosphorylated tau was not observed, but neurite extension was impaired compared to control cells (Ehrlich et al., 2015). In another study of human iPSC-derived neurons, exogenous application of N279K mutant K18 oligomers, which led to internalization of tau, resulted in tau colocalization with the nuclear protein nucleolin; interestingly, this colocalization was generally not observed when SH-SY5Y cell were used (Karikari et al., 2019).
Tau phosphorylation
Tau phosphorylation was assessed in COS-7 cells, and compared to wild-type–expressing cells, those with the N279K mutation had increased phosphorylation at the AT8 epitope (S202, T205; Ritter et al., 2018). This hyperphosphorylation of AT8, as well as AT100 (T212, S214), has also been observed in N279K iPSC-derived neurons (Iovino et al., 2015). Increased tau phosphorylation and fragmentation was also observed in another study of iPSC-derived neurons from N279K carriers (Ehrlich et al., 2015; Al-Dalahmah et al., 2024). Corresponding to the increased phosphorylation, one study found a reduction in the binding affinity of N279K tau for protein phosphatase 2A, a major phosphatase of tau, via coimmunoprecipitation of transfected mouse NIH 3T3 fibroblasts (Goedert et al., 2000).
Protein phosphorylation
Apart from tau phosphorylation, protein phosphorylation more broadly is affected by the N279K mutation. In a proteomic analysis of the hippocampus in an N279K-expressing transgenic mouse model, phosphorylation of calreticulin and tubulin β4 was increased, while decreases were seen in other proteins, including heat shock cognate 71 kDa protein and tubulin β2 (Takano et al., 2009).
Microtubule binding
Across multiple studies, microtubule-binding assays revealed that recombinant N279K tau did not differ from wild-type tau in microtubule assembly, including lag time, rate of assembly, or maximal amount of microtubule polymers formed (D'Souza et al., 1999; also see Hasegawa et al., 1999 and Hong et al., 1998). However, one report found that N279K mutant tau had reduced microtubule assembly and binding compared with wild-type tau, possibly because the recombinant preparation (htau40) used in this study (i.e., the full-length tau) may have differed from those used in the other studies (Barghorn et al., 2000).
Metabolic abnormalities
Several lines of evidence suggest that the N279K mutation is associated with metabolic dysfunction. In a study using liquid chromatography–mass spectrometry, metabolic abnormalities were found in two lines of iPSC-derived human dopamine neurons carrying the N279K mutation compared to two wild-type cell lines, as observed by an increased abundance of oxidized phospholipids, suggesting that the mutation is associated with increased oxidative stress (Bradford et al., 2024). This is further supported by molecular assays demonstrating increased vulnerability to oxidative stress and activation of the unfolded protein response in iPSC-derived neurons from patients with the N279K mutation (Ehrlich et al., 2015; Al-Dalahmah et al., 2024). Moreover, using mass spectrometry and metabolomics analyses, higher levels of ATP were observed in these neurons as well as dysregulation in the mitochondrial electron transport chain, pyruvate metabolism, and amino acid metabolism (Al-Dalahmah et al., 2024). Extending these observations, in human iPSC–derived glutamatergic cortical neurons, mass spectrometry revealed that levels of proteins related to reactive oxygen species (ROS) regulation were increased in N279K cells versus controls, suggesting the involvement of oxidative stress pathways (Korn et al., 2023). This was further corroborated using live cell imaging, which showed that the rate of ROS production was higher in mutant cells. Crucially, this study showed these changes in neuronal oxidative stress were also associated with higher rates of cell death and increased expression of apoptosis markers (Korn et al., 2023).
The impact of the N279K mutation on cellular stress and apoptosis has been further explored in different model systems. For instance, in rat hippocampal neurons, transfection of a fragmented tau (tau151-421) carrying the N279K mutation potentiated apoptosis, but when the full-length N279K tau was transfected, no differences in apoptotic cells were seen compared to wild-type tau (Fasulo et al., 2005). Nonetheless, another report demonstrated that SH-SY5Y cells transfected with N279K tau were more vulnerable to apoptosis following serum withdrawal than those transfected with wild-type tau, seemingly through disrupted calcium homestasis (Furukawa et al., 2000). In addition, iPSC-derived neural stem cells show signs of cellular stress, including the accumulation of stress granules and vesicle trafficking deficits (Wren et al., 2015). Also of note, an N279K mouse model exhibited increased activated caspase-3 immunoreactivity compared to non-transgenic controls in neurons and astrocytes throughout the brain, pointing to the presence of ongoing apoptosis as a consequence of the mutation (Dawson et al., 2007). Together, these findings underscore N279K’s role in inducing significant metabolic disruptions, particularly through oxidative stress, which may contribute to the neurodegenerative processes observed in affected individuals.
Mitochondrial dysfunction
In human iPSC–derived glutamatergic cortical neurons, mitochondrial hyperpolarization, reduced mitochondrial mass, and increased mitochondrial fission were observed compared to a control line (Korn et al., 2023). Moreover, iPSC-derived neurons from patients with the N279K mutation exhibited impaired mitophagy and altered mitochondrial function (Al-Dalahmah et al., 2024). For example, compared to the control cell line, N279K-positive iPSC-derived neurons had an increased basal oxygen consumption rate. Another study using iPSC-derived neurons from an N279K carrier found that anterograde mitochondrial axonal transport was impaired compared to that in control cells (Iovino et al., 2015).
Astrocytic dysfunction
Astrocyte function was examined in an iPSC model of neural progenitor cells derived from a patient carrying the N279K mutation (Hallmann et al., 2017). While astrocyte differentiation per se was not perturbed, the astrocytes were larger and expressed greater levels of the 4R isoform of tau. Moreover, these iPSC-derived astrocytes were more vulnerable to oxidative stress as determined by the increase in cell death and lactate dehydrogenase release following rotenone application, as well as an increase in protein ubiquitination based on western blot analysis, pointing to altered protein degradation. Mutant iPSC-derived astrocytes also imparted dysfunction to previously healthy neurons in co-culture settings, namely an increase in vulnerability to oxidative stress, indicating the potential of the N279K mutation to induce non-cell autonomous effects.
Gene expression alterations
Investigations into the molecular mechanisms of the N279K mutation have included analyses of gene expression alterations. For instance, substantia nigra samples from patients carrying the N279K mutation were assessed via RNA microarrays (Al-Dalahmah et al., 2024). Differential expression was observed in genes related to microglia, spliceosome, and immune activation pathways. This study also conducted an in-depth analysis using single nucleus RNA sequencing to reveal cell type–specific alterations in gene expression compared to control samples.
Microarray has also been used to evaluate the transcriptome of iPSC-derived neurons from patients with the N279K mutation (Ehrlich et al., 2015). Gene expression profiles were found to be altered (e.g., increased expression of LOC100128252 and MAGEH1) compared to control cells, and these changes were also found in post-mortem samples from patients with FTD.
Moreover, in the iPSC-derived N279K astrocytes introduced above, transcriptome analysis revealed disease-associated changes when compared to control iPSC-derived astrocytes (Hallmann et al., 2017). Functional annotation uncovered that affected genes were related to plasma membrane, multicellular organism development, neurological system processes, and synaptic organization and transmission.
Several in silico algorithms, including Mutation Taster, SIFT, Provean, and PolyPhen-2, have predicted this variant is damaging (Nan et al., 2024). Moreover, its PHRED-scaled CADD score which integrates diverse information in silico, was above 20 (26.5), suggesting a deleterious effect.
Research Models
To explore the effects of the N279K mutation, several research models have been developed. A transgenic mouse model overexpressing a human tau with the N279K mutation was created to investigate clinical and neuropathological outcomes (Taniguchi et al., 2005; Takano et al., 2009; Takenokuchi et al., 2010; Chiba et al., 2012). Building on this, another transgenic model using a human tau minigene carrying N279K was developed to study exon 10 splicing (Dawson et al., 2007; Schoch et al., 2016; Jul 2016 news). More recently, the MAPT(H1.0*N279K)-GR mouse model was introduced, where the mouse Mapt-containing region is replaced with the mutated human H1 haplotype carrying the N279K mutation (Benzow et al., 2024; Nov 2022 news).
Moving to human cell models, several iPSC lines have been established from patients with the N279K mutation, allowing for detailed mechanistic studies at the cellular and molecular levels (Ehrlich et al., 2015; Iovino et al., 2015; Wren et al., 2015; Korn et al., 2023; Al-Dalahmah et al., 2024; Grigor’eva et al., 2024). These have been differentiated into neurons, as well as astrocytes (e.g., Hallmann et al., 2017), and genetically modified to express biosensor transgenes (Nadtochy et al., 2025). Additionally, N279K mutant human iPSC models using CRISPR have been created, including triple MAPT mutation lines (Szabo et al., 2023; García-León et al., 2018; D'Brant et al., 2024 preprint).
With regard to primary cell lines, patient-derived primary fibroblasts carrying the N279K mutation are available for request from the Coriell repository (Wray et al., 2012).
Lastly, to facilitate recombinant protein studies, a plasmids library has been developed containing N279K mutant full-length and K18 fragment (i.e., the four-repeat microtubule-binding region) tau, including cysteine-modified variants (Karikari et al., 2020).
Last Updated: 07 Apr 2025
References
News Citations
- Tipping the Balance Toward Four-Repeat Tau Exacerbates Toxicity in Mice
- Cornucopia: LOADs of New Mouse Models Available
Research Models Citations
Paper Citations
- Taniguchi T, Doe N, Matsuyama S, Kitamura Y, Mori H, Saito N, Tanaka C. Transgenic mice expressing mutant (N279K) human tau show mutation dependent cognitive deficits without neurofibrillary tangle formation. FEBS Lett. 2005 Oct 24;579(25):5704-12. Epub 2005 Oct 5 PubMed.
- Takano M, Otani M, Sakai A, Kadoyama K, Matsuyama S, Matsumoto A, Takenokuchi M, Sumida M, Taniguchi T. Use of a phosphosensor dye in proteomic analysis of human mutant tau transgenic mice. Neuroreport. 2009 Dec 9;20(18):1648-53. PubMed.
- Takenokuchi M, Kadoyama K, Chiba S, Sumida M, Matsuyama S, Saigo K, Taniguchi T. SJLB mice develop tauopathy-induced parkinsonism. Neurosci Lett. 2010 Apr 12;473(3):182-5. Epub 2010 Feb 21 PubMed.
- Chiba S, Takada E, Tadokoro M, Taniguchi T, Kadoyama K, Takenokuchi M, Kato S, Suzuki N. Loss of dopaminoreceptive neuron causes L-dopa resistant parkinsonism in tauopathy. Neurobiol Aging. 2012 Oct;33(10):2491-505. Epub 2011 Dec 14 PubMed.
- Dawson HN, Cantillana V, Chen L, Vitek MP. The tau N279K exon 10 splicing mutation recapitulates frontotemporal dementia and parkinsonism linked to chromosome 17 tauopathy in a mouse model. J Neurosci. 2007 Aug 22;27(34):9155-68. PubMed.
- Schoch KM, DeVos SL, Miller RL, Chun SJ, Norrbom M, Wozniak DF, Dawson HN, Bennett CF, Rigo F, Miller TM. Increased 4R-Tau Induces Pathological Changes in a Human-Tau Mouse Model. Neuron. 2016 Jun 1;90(5):941-7. Epub 2016 May 19 PubMed.
- Benzow K, Karanjeet K, Oblak AL, Carter GW, Sasner M, Koob MD. Gene replacement-Alzheimer's disease (GR-AD): Modeling the genetics of human dementias in mice. Alzheimers Dement. 2024 Apr;20(4):3080-3087. Epub 2024 Feb 11 PubMed.
- Ehrlich M, Hallmann AL, Reinhardt P, Araúzo-Bravo MJ, Korr S, Röpke A, Psathaki OE, Ehling P, Meuth SG, Oblak AL, Murrell JR, Ghetti B, Zaehres H, Schöler HR, Sterneckert J, Kuhlmann T, Hargus G. Distinct Neurodegenerative Changes in an Induced Pluripotent Stem Cell Model of Frontotemporal Dementia Linked to Mutant TAU Protein. Stem Cell Reports. 2015 Jul 14;5(1):83-96. Epub 2015 Jul 2 PubMed.
- Iovino M, Agathou S, González-Rueda A, Del Castillo Velasco-Herrera M, Borroni B, Alberici A, Lynch T, O'Dowd S, Geti I, Gaffney D, Vallier L, Paulsen O, Káradóttir RT, Spillantini MG. Early maturation and distinct tau pathology in induced pluripotent stem cell-derived neurons from patients with MAPT mutations. Brain. 2015 Nov;138(Pt 11):3345-59. Epub 2015 Jul 27 PubMed.
- Wren MC, Zhao J, Liu CC, Murray ME, Atagi Y, Davis MD, Fu Y, Okano HJ, Ogaki K, Strongosky AJ, Tacik P, Rademakers R, Ross OA, Dickson DW, Wszolek ZK, Kanekiyo T, Bu G. Frontotemporal dementia-associated N279K tau mutant disrupts subcellular vesicle trafficking and induces cellular stress in iPSC-derived neural stem cells. Mol Neurodegener. 2015 Sep 15;10:46. PubMed.
- Korn L, Speicher AM, Schroeter CB, Gola L, Kaehne T, Engler A, Disse P, Fernández-Orth J, Csatári J, Naumann M, Seebohm G, Meuth SG, Schöler HR, Wiendl H, Kovac S, Pawlowski M. MAPT genotype-dependent mitochondrial aberration and ROS production trigger dysfunction and death in cortical neurons of patients with hereditary FTLD. Redox Biol. 2023 Feb;59:102597. Epub 2022 Dec 30 PubMed.
- Al-Dalahmah O, Lam M, McInvale JJ, Qu W, Nguyen T, Mun JY, Kwon S, Ifediora N, Mahajan A, Humala N, Winters T, Angeles E, Jakubiak KA, Kühn R, Kim YA, De Rosa MC, Doege CA, Paryani F, Flowers X, Dovas A, Mela A, Lu H, DeTure MA, Vonsattel JP, Wszolek ZK, Dickson DW, Kuhlmann T, Zaehres H, Schöler HR, Sproul AA, Siegelin MD, De Jager PL, Goldman JE, Menon V, Canoll P, Hargus G. Osteopontin drives neuroinflammation and cell loss in MAPT-N279K frontotemporal dementia patient neurons. Cell Stem Cell. 2024 May 2;31(5):676-693.e10. Epub 2024 Apr 15 PubMed.
- Grigor'eva EV, Malakhova AA, Yarkova ES, Minina JM, Vyatkin YV, Nadtochy JA, Khabarova EA, Rzaev JA, Medvedev SP, Zakian SM. Generation and characterization of two induced pluripotent stem cell lines (ICGi052-A and ICGi052-B) from a patient with frontotemporal dementia with parkinsonism-17 associated with the pathological variant c.2013T>G in the MAPT gene. Vavilovskii Zhurnal Genet Selektsii. 2024 Nov;28(7):679-687. PubMed.
- Hallmann AL, Araúzo-Bravo MJ, Mavrommatis L, Ehrlich M, Röpke A, Brockhaus J, Missler M, Sterneckert J, Schöler HR, Kuhlmann T, Zaehres H, Hargus G. Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. Sci Rep. 2017 Mar 3;7:42991. PubMed.
- Nadtochy JA, Medvedev SP, Grigor'eva EV, Pavlova SV, Minina JM, Chechushkov AV, Malakhova AA, Kovalenko LV, Zakian SM. Transgenic iPSC Lines with Genetically Encoded MitoTimer to Study Mitochondrial Biogenesis in Dopaminergic Neurons with Tauopathy. Biomedicines. 2025 Feb 21;13(3) PubMed.
- Szabo L, Grimm A, García-León JA, Verfaillie CM, Eckert A. Genetically Engineered Triple MAPT-Mutant Human-Induced Pluripotent Stem Cells (N279K, P301L, and E10+16 Mutations) Exhibit Impairments in Mitochondrial Bioenergetics and Dynamics. Cells. 2023 May 13;12(10) PubMed.
- García-León JA, Cabrera-Socorro A, Eggermont K, Swijsen A, Terryn J, Fazal R, Nami F, Ordovás L, Quiles A, Lluis F, Serneels L, Wierda K, Sierksma A, Kreir M, Pestana F, Van Damme P, De Strooper B, Thorrez L, Ebneth A, Verfaillie CM. Generation of a human induced pluripotent stem cell-based model for tauopathies combining three microtubule-associated protein TAU mutations which displays several phenotypes linked to neurodegeneration. Alzheimers Dement. 2018 Oct;14(10):1261-1280. Epub 2018 Jul 20 PubMed. Correction.
- D'Brant L, Rugenstein N, Na SK, Miller MJ, Czajka TF, Trudeau N, Fitz EM, Tomaszek L, Fisher ES, Mash ES, Joy S, Lotz S, Borden S, Stevens K, Goderie S, Wang Y, Bertucci T, Karch CM, Temple S, Butler D. Fully Human Bifunctional Intrabodies Achieve Graded Reduction of Intracellular Tau and Rescue Survival of MAPT Mutation iPSC-derived Neurons. 2024 Jun 01 10.1101/2024.05.28.596248 (version 1) bioRxiv.
- Wray S, Self M, NINDS Parkinson's Disease iPSC Consortium, NINDS Huntington's Disease iPSC Consortium, NINDS ALS iPSC Consortium, Lewis PA, Taanman JW, Ryan NS, Mahoney CJ, Liang Y, Devine MJ, Sheerin UM, Houlden H, Morris HR, Healy D, Marti-Masso JF, Preza E, Barker S, Sutherland M, Corriveau RA, D'Andrea M, Schapira AH, Uitti RJ, Guttman M, Opala G, Jasinska-Myga B, Puschmann A, Nilsson C, Espay AJ, Slawek J, Gutmann L, Boeve BF, Boylan K, Stoessl AJ, Ross OA, Maragakis NJ, Van Gerpen J, Gerstenhaber M, Gwinn K, Dawson TM, Isacson O, Marder KS, Clark LN, Przedborski SE, Finkbeiner S, Rothstein JD, Wszolek ZK, Rossor MN, Hardy J. Creation of an open-access, mutation-defined fibroblast resource for neurological disease research. PLoS One. 2012;7(8):e43099. Epub 2012 Aug 27 PubMed.
- Karikari TK, Keeling S, Hill E, Lantero Rodrı Guez J, Nagel DA, Becker B, Höglund K, Zetterberg H, Blennow K, Hill EJ, Moffat KG. Extensive Plasmid Library to Prepare Tau Protein Variants and Study Their Functional Biochemistry. ACS Chem Neurosci. 2020 Oct 7;11(19):3117-3129. Epub 2020 Sep 16 PubMed.
- Tsuboi Y, Uitti RJ, Delisle MB, Ferreira JJ, Brefel-Courbon C, Rascol O, Ghetti B, Murrell JR, Hutton M, Baker M, Wszolek ZK. Clinical features and disease haplotypes of individuals with the N279K tau gene mutation: a comparison of the pallidopontonigral degeneration kindred and a French family. Arch Neurol. 2002 Jun;59(6):943-50. PubMed.
- Caviness JN, Tsuboi Y, Wszolek ZK. Clinical-electrophysiological correlation of tremor and myoclonus in a kindred with the N279K tau mutation. Parkinsonism Relat Disord. 2003 Jan;9(3):151-7. PubMed.
- Wszolek ZK, Pfeiffer RF, Bhatt MH, Schelper RL, Cordes M, Snow BJ, Rodnitzky RL, Wolters EC, Arwert F, Calne DB. Rapidly progressive autosomal dominant parkinsonism and dementia with pallido-ponto-nigral degeneration. Ann Neurol. 1992 Sep;32(3):312-20. PubMed.
- Markopoulou K, Chase BA, Robowski P, Strongosky A, Narożańska E, Sitek EJ, Berdynski M, Barcikowska M, Baker MC, Rademakers R, Sławek J, Klein C, Hückelheim K, Kasten M, Wszolek ZK. Assessment of Olfactory Function in MAPT-Associated Neurodegenerative Disease Reveals Odor-Identification Irreproducibility as a Non-Disease-Specific, General Characteristic of Olfactory Dysfunction. PLoS One. 2016;11(11):e0165112. Epub 2016 Nov 17 PubMed.
- Arvanitakis Z, Witte RJ, Dickson DW, Tsuboi Y, Uitti RJ, Slowinski J, Hutton ML, Lin SC, Boeve BF, Cheshire WP, Pooley RA, Liss JM, Caviness JN, Strongosky AJ, Wszolek ZK. Clinical-pathologic study of biomarkers in FTDP-17 (PPND family with N279K tau mutation). Parkinsonism Relat Disord. 2007 May;13(4):230-9. Epub 2006 Dec 29 PubMed.
- Spector AR, Dugger BN, Wszolek ZK, Uitti RJ, Fredrickson P, Kaplan J, Boeve BF, Dickson DW, Strongosky A, Lin SC. Anatomy of disturbed sleep in pallido-ponto-nigral degeneration. Ann Neurol. 2011 Jun;69(6):1014-1025. PubMed.
- Arima K, Kowalska A, Hasegawa M, Mukoyama M, Watanabe R, Kawai M, Takahashi K, Iwatsubo T, Tabira T, Sunohara N. Two brothers with frontotemporal dementia and parkinsonism with an N279K mutation of the tau gene. Neurology. 2000 May 9;54(9):1787-95. PubMed.
- Yasuda M, Kawamata T, Komure O, Kuno S, D'Souza I, Poorkaj P, Kawai J, Tanimukai S, Yamamoto Y, Hasegawa H, Sasahara M, Hazama F, Schellenberg GD, Tanaka C. A mutation in the microtubule-associated protein tau in pallido-nigro-luysian degeneration. Neurology. 1999 Sep 11;53(4):864-8. PubMed.
- Tsuboi Y, Baker M, Hutton ML, Uitti RJ, Rascol O, Delisle MB, Soulages X, Murrell JR, Ghetti B, Yasuda M, Komure O, Kuno S, Arima K, Sunohara N, Kobayashi T, Mizuno Y, Wszolek ZK. Clinical and genetic studies of families with the tau N279K mutation (FTDP-17). Neurology. 2002 Dec 10;59(11):1791-3. PubMed.
- Ogaki K, Motoi Y, Li Y, Tomiyama H, Shimizu N, Takanashi M, Nakanishi A, Yokoyama K, Hattori N. Visual grasping in frontotemporal dementia and parkinsonism linked to chromosome 17 (microtubule-associated with protein tau): a comparison of N-Isopropyl-p-[(123)I]-iodoamphetamine brain perfusion single photon emission computed tomography analysis with . Mov Disord. 2011 Feb 15;26(3):561-3. PubMed.
- Ogaki K, Li Y, Takanashi M, Ishikawa KI, Kobayashi T, Nonaka T, Hasegawa M, Kishi M, Yoshino H, Funayama M, Tsukamoto T, Shioya K, Yokochi M, Imai H, Sasaki R, Kokubo Y, Kuzuhara S, Motoi Y, Tomiyama H, Hattori N. Analyses of the MAPT, PGRN, and C9orf72 mutations in Japanese patients with FTLD, PSP, and CBS. Parkinsonism Relat Disord. 2012 Jul 18; PubMed.
- Ikeda A, Shimada H, Nishioka K, Takanashi M, Hayashida A, Li Y, Yoshino H, Funayama M, Ueno Y, Hatano T, Sahara N, Suhara T, Higuchi M, Hattori N. Clinical heterogeneity of frontotemporal dementia and Parkinsonism linked to chromosome 17 caused by MAPT N279K mutation in relation to tau positron emission tomography features. Mov Disord. 2019 Apr;34(4):568-574. Epub 2019 Feb 17 PubMed.
- Oka Y, Saiki H, Hashimoto Y, Terada Y, Nakamura T, Ayaki T, Orimo S, Matsumoto S. Japanese Familial Cases of Frontotemporal Dementia and Parkinsonism with N279K Tau Gene Mutation. Mov Disord Clin Pract. 2021 Jan;8(1):126-132. Epub 2020 Nov 3 PubMed.
- Shimura H, Mori H, Komatsuzaki Y, Nakamura K, Takanashi M, Hattori N, Mizuno Y. [A 54-year-old man with familial parkinsonism, gaze palsy, and dementia]. No To Shinkei. 2005 Dec;57(12):1104-12. PubMed.
- Yang Y, Tang BS, Weng L, Li N, Shen L, Wang J, Zuo CT, Yan XX, Xia K, Guo JF. Genetic Identification Is Critical for the Diagnosis of Parkinsonism: A Chinese Pedigree with Early Onset of Parkinsonism. PLoS One. 2015;10(8):e0136245. Epub 2015 Aug 21 PubMed.
- Tang M, Gu X, Wei J, Jiao B, Zhou L, Zhou Y, Weng L, Yan X, Tang B, Xu J, Shen L. Analyses MAPT, GRN, and C9orf72 mutations in Chinese patients with frontotemporal dementia. Neurobiol Aging. 2016 Oct;46:235.e11-5. Epub 2016 May 20 PubMed.
- Wu L, Liu J, Feng X, Dong J, Qin W, Liu Y, Wang J, Lu J, Chen K, Wang Y, Jia J. 11C-CFT-PET in Presymptomatic FTDP-17: A Potential Biomarker Predicting Onset. J Alzheimers Dis. 2018;61(2):613-618. PubMed.
- Nan H, Kim YJ, Chu M, Li D, Li J, Jiang D, Wu Y, Ohtsuka T, Wu L. Genetic and clinical landscape of Chinese frontotemporal dementia: dominance of TBK1 and OPTN mutations. Alzheimers Res Ther. 2024 Jun 13;16(1):127. PubMed.
- Takada LT, Bahia VS, Guimarães HC, Costa TV, Vale TC, Rodriguez RD, Porto FH, Machado JC, Beato RG, Cesar KG, Smid J, Nascimento CF, Grinberg LT, Brucki SM, Maximino JR, Camargos ST, Chadi G, Caramelli P, Nitrini R. GRN and MAPT Mutations in 2 Frontotemporal Dementia Research Centers in Brazil. Alzheimer Dis Assoc Disord. 2016;30(4):310-317. PubMed.
- O'Dowd S, Murray B, Roberts K, Cummins G, Magennis B, Lynch T. Pallidopontonigral degeneration: a deceptive familial tauopathy. Mov Disord. 2012 Jun;27(7):817-9. PubMed.
- Soliveri P, Rossi G, Monza D, Tagliavini F, Piacentini S, Albanese A, Bugiani O, Girotti F. A case of dementia parkinsonism resembling progressive supranuclear palsy due to mutation in the tau protein gene. Arch Neurol. 2003 Oct;60(10):1454-6. PubMed.
- Delisle MB, Murrell JR, Richardson R, Trofatter JA, Rascol O, Soulages X, Mohr M, Calvas P, Ghetti B. A mutation at codon 279 (N279K) in exon 10 of the Tau gene causes a tauopathy with dementia and supranuclear palsy. Acta Neuropathol. 1999 Jul;98(1):62-77. PubMed.
- Moore KM, Nicholas J, Grossman M, McMillan CT, Irwin DJ, Massimo L, Van Deerlin VM, Warren JD, Fox NC, Rossor MN, Mead S, Bocchetta M, Boeve BF, Knopman DS, Graff-Radford NR, Forsberg LK, Rademakers R, Wszolek ZK, van Swieten JC, Jiskoot LC, Meeter LH, Dopper EG, Papma JM, Snowden JS, Saxon J, Jones M, Pickering-Brown S, Le Ber I, Camuzat A, Brice A, Caroppo P, Ghidoni R, Pievani M, Benussi L, Binetti G, Dickerson BC, Lucente D, Krivensky S, Graff C, Öijerstedt L, Fallström M, Thonberg H, Ghoshal N, Morris JC, Borroni B, Benussi A, Padovani A, Galimberti D, Scarpini E, Fumagalli GG, Mackenzie IR, Hsiung GR, Sengdy P, Boxer AL, Rosen H, Taylor JB, Synofzik M, Wilke C, Sulzer P, Hodges JR, Halliday G, Kwok J, Sanchez-Valle R, Lladó A, Borrego-Ecija S, Santana I, Almeida MR, Tábuas-Pereira M, Moreno F, Barandiaran M, Indakoetxea B, Levin J, Danek A, Rowe JB, Cope TE, Otto M, Anderl-Straub S, de Mendonça A, Maruta C, Masellis M, Black SE, Couratier P, Lautrette G, Huey ED, Sorbi S, Nacmias B, Laforce R Jr, Tremblay ML, Vandenberghe R, Damme PV, Rogalski EJ, Weintraub S, Gerhard A, Onyike CU, Ducharme S, Papageorgiou SG, Ng AS, Brodtmann A, Finger E, Guerreiro R, Bras J, Rohrer JD, FTD Prevention Initiative. Age at symptom onset and death and disease duration in genetic frontotemporal dementia: an international retrospective cohort study. Lancet Neurol. 2020 Feb;19(2):145-156. Epub 2019 Dec 3 PubMed.
- Slowinski J, Dominik J, Uitti RJ, Ahmed Z, Dickson DD, Wszolek ZK. Frontotemporal dementia and Parkinsonism linked to chromosome 17 with the N279K tau mutation. Neuropathology. 2007 Feb;27(1):73-80. PubMed.
- Miyagawa T, Vernon C, Przybelski SA, Min PH, Fields JA, Dickerson BC, Dickson DW, Kantarci K, Lowe VJ, Wszolek ZK, Boeve BF. Prominent loss of striatal dopamine transporter binding in frontotemporal lobar degeneration with the MAPT N279K mutation present as early as at prodromal stage without parkinsonism. Parkinsonism Relat Disord. 2024 Nov;128:107144. Epub 2024 Sep 8 PubMed.
- Takeshige H, Nakayama S, Nishioka K, Li Y, Motoi Y, Hattori N. Marked Reduction in the Striatal Dopamine Transporter Uptake During the Early Stage of Motor Symptoms in Patients with the MAPT N279K Mutation. Intern Med. 2018 Oct 15;57(20):3015-3019. Epub 2018 Jun 6 PubMed.
- Jones DT, Knopman DS, Graff-Radford J, Syrjanen JA, Senjem ML, Schwarz CG, Dheel C, Wszolek Z, Rademakers R, Kantarci K, Petersen RC, Jack CR Jr, Lowe VJ, Boeve BF. In vivo 18F-AV-1451 tau PET signal in MAPT mutation carriers varies by expected tau isoforms. Neurology. 2018 Mar 13;90(11):e947-e954. Epub 2018 Feb 9 PubMed.
- Miyoshi M, Shinotoh H, Wszolek ZK, Strongosky AJ, Shimada H, Arakawa R, Higuchi M, Ikoma Y, Yasuno F, Fukushi K, Irie T, Ito H, Suhara T. In vivo detection of neuropathologic changes in presymptomatic MAPT mutation carriers: a PET and MRI study. Parkinsonism Relat Disord. 2010 Jul;16(6):404-8. Epub 2010 May 10 PubMed.
- D'Souza I, Poorkaj P, Hong M, Nochlin D, Lee VM, Bird TD, Schellenberg GD. Missense and silent tau gene mutations cause frontotemporal dementia with parkinsonism-chromosome 17 type, by affecting multiple alternative RNA splicing regulatory elements. Proc Natl Acad Sci U S A. 1999 May 11;96(10):5598-603. PubMed.
- Rodriguez-Martin T, Anthony K, Garcia-Blanco MA, Mansfield SG, Anderton BH, Gallo JM. Correction of tau mis-splicing caused by FTDP-17 MAPT mutations by spliceosome-mediated RNA trans-splicing. Hum Mol Genet. 2009 Sep 1;18(17):3266-73. Epub 2009 Jun 4 PubMed.
- Hasegawa M, Smith MJ, Iijima M, Tabira T, Goedert M. FTDP-17 mutations N279K and S305N in tau produce increased splicing of exon 10. FEBS Lett. 1999 Jan 25;443(2):93-6. PubMed.
- Hong M, Zhukareva V, Vogelsberg-Ragaglia V, Wszolek Z, Reed L, Miller BI, Geschwind DH, Bird TD, McKeel D, Goate A, Morris JC, Wilhelmsen KC, Schellenberg GD, Trojanowski JQ, Lee VM. Mutation-specific functional impairments in distinct tau isoforms of hereditary FTDP-17. Science. 1998 Dec 4;282(5395):1914-7. PubMed.
- Hutton M. Missense and splice site mutations in tau associated with FTDP-17: multiple pathogenic mechanisms. Neurology. 2001 Jun;56(11 Suppl 4):S21-5. PubMed.
- Jiang Z, Tang H, Havlioglu N, Zhang X, Stamm S, Yan R, Wu JY. Mutations in tau gene exon 10 associated with FTDP-17 alter the activity of an exonic splicing enhancer to interact with Tra2 beta. J Biol Chem. 2003 May 23;278(21):18997-9007. Epub 2003 Mar 20 PubMed.
- Kondo S, Yamamoto N, Murakami T, Okumura M, Mayeda A, Imaizumi K. Tra2 beta, SF2/ASF and SRp30c modulate the function of an exonic splicing enhancer in exon 10 of tau pre-mRNA. Genes Cells. 2004 Feb;9(2):121-30. PubMed.
- D'Souza I, Schellenberg GD. Arginine/serine-rich protein interaction domain-dependent modulation of a tau exon 10 splicing enhancer: altered interactions and mechanisms for functionally antagonistic FTDP-17 mutations Delta280K AND N279K. J Biol Chem. 2006 Feb 3;281(5):2460-9. Epub 2005 Nov 23 PubMed.
- Wang Y, Wang J, Gao L, Lafyatis R, Stamm S, Andreadis A. Tau exons 2 and 10, which are misregulated in neurodegenerative diseases, are partly regulated by silencers which bind a SRp30c.SRp55 complex that either recruits or antagonizes htra2beta1. J Biol Chem. 2005 Apr 8;280(14):14230-9. Epub 2005 Feb 3 PubMed.
- Karikari TK, Nagel DA, Grainger A, Clarke-Bland C, Crowe J, Hill EJ, Moffat KG. Distinct Conformations, Aggregation and Cellular Internalization of Different Tau Strains. Front Cell Neurosci. 2019;13:296. Epub 2019 Jul 9 PubMed.
- Strang KH, Croft CL, Sorrentino ZA, Chakrabarty P, Golde TE, Giasson BI. Distinct differences in prion-like seeding and aggregation between Tau protein variants provide mechanistic insights into tauopathies. J Biol Chem. 2018 Feb 16;293(7):2408-2421. Epub 2017 Dec 19 PubMed.
- Yao TM, Tomoo K, Ishida T, Hasegawa H, Sasaki M, Taniguchi T. Aggregation analysis of the microtubule binding domain in tau protein by spectroscopic methods. J Biochem. 2003 Jul;134(1):91-9. PubMed.
- Barghorn S, Zheng-Fischhöfer Q, Ackmann M, Biernat J, von Bergen M, Mandelkow EM, Mandelkow E. Structure, microtubule interactions, and paired helical filament aggregation by tau mutants of frontotemporal dementias. Biochemistry. 2000 Sep 26;39(38):11714-21. PubMed.
- Sampognaro PJ, Arya S, Knudsen GM, Gunderson EL, Sandoval-Perez A, Hodul M, Bowles K, Craik CS, Jacobson MP, Kao AW. Mutations in α-synuclein, TDP-43 and tau prolong protein half-life through diminished degradation by lysosomal proteases. Mol Neurodegener. 2023 May 2;18(1):29. PubMed. Correction.
- Ritter ML, Avila J, García-Escudero V, Hernández F, Pérez M. Frontotemporal Dementia-Associated N279K Tau Mutation Localizes at the Nuclear Compartment. Front Cell Neurosci. 2018;12:202. Epub 2018 Jul 12 PubMed.
- Goedert M, Satumtira S, Jakes R, Smith MJ, Kamibayashi C, White CL 3rd, Sontag E. Reduced binding of protein phosphatase 2A to tau protein with frontotemporal dementia and parkinsonism linked to chromosome 17 mutations. J Neurochem. 2000 Nov;75(5):2155-62. PubMed.
- Bradford X, Fernandes HJ, Snowden SG. Changes in Oxidised Phospholipids in Response to Oxidative Stress in Microtubule-Associated Protein Tau (MAPT) Mutant Dopamine Neurons. Antioxidants (Basel). 2024 Apr 24;13(5) PubMed.
- Fasulo L, Ugolini G, Cattaneo A. Apoptotic effect of caspase-3 cleaved tau in hippocampal neurons and its potentiation by tau FTDP-mutation N279K. J Alzheimers Dis. 2005 Feb;7(1):3-13. PubMed.
- Furukawa K, D'Souza I, Crudder CH, Onodera H, Itoyama Y, Poorkaj P, Bird TD, Schellenberg GD. Pro-apoptotic effects of tau mutations in chromosome 17 frontotemporal dementia and parkinsonism. Neuroreport. 2000 Jan 17;11(1):57-60. PubMed.
Further Reading
Papers
- Kawai J, Sasahara M, Hazama F, Kuno S, Komure O, Nomura S, Yamaguchi M. Pallidonigroluysian degeneration with iron deposition: a study of three autopsy cases. Acta Neuropathol. 1993;86(6):609-16. PubMed.
- Berger Z, Roder H, Hanna A, Carlson A, Rangachari V, Yue M, Wszolek Z, Ashe K, Knight J, Dickson D, Andorfer C, Rosenberry TL, Lewis J, Hutton M, Janus C. Accumulation of pathological tau species and memory loss in a conditional model of tauopathy. J Neurosci. 2007 Apr 4;27(14):3650-62. PubMed.
- McRae CA, Diem G, Yamazaki TG, Mitek A, Wszolek ZK. Interest in genetic testing in pallido-ponto-nigral degeneration (PPND): a family with frontotemporal dementia with Parkinsonism linked to chromosome 17. Eur J Neurol. 2001 Mar;8(2):179-83. PubMed.
- Cheshire WP, Tsuboi Y, Wszolek ZK. Physiologic assessment of autonomic dysfunction in pallidopontonigral degeneration with N279K mutation in the tau gene on chromosome 17. Auton Neurosci. 2002 Nov 29;102(1-2):71-7. PubMed.
- Arima K, Kowalska A, Hasegawa M, Mukoyama M, Watanabe R, Kawai M, Takahashi K, Iwatsubo T, Tabira T, Sunohara N. Two brothers with frontotemporal dementia and parkinsonism with an N279K mutation of the tau gene. Neurology. 2000 May 9;54(9):1787-95. PubMed.
- Yasuda M, Kawamata T, Komure O, Kuno S, D'Souza I, Poorkaj P, Kawai J, Tanimukai S, Yamamoto Y, Hasegawa H, Sasahara M, Hazama F, Schellenberg GD, Tanaka C. A mutation in the microtubule-associated protein tau in pallido-nigro-luysian degeneration. Neurology. 1999 Sep 11;53(4):864-8. PubMed.
- Chen Q, Boeve BF, Tosakulwong N, Lesnick T, Brushaber D, Dheel C, Fields J, Forsberg L, Gavrilova R, Gearhart D, Haley D, Gunter JL, Graff-Radford J, Jones D, Knopman D, Graff-Radford N, Kraft R, Lapid M, Rademakers R, Syrjanen J, Wszolek ZK, Rosen H, Boxer AL, Kantarci K. Frontal lobe 1H MR spectroscopy in asymptomatic and symptomatic MAPT mutation carriers. Neurology. 2019 Aug 20;93(8):e758-e765. Epub 2019 Jul 17 PubMed.
- Clark LN, Poorkaj P, Wszolek Z, Geschwind DH, Nasreddine ZS, Miller B, Li D, Payami H, Awert F, Markopoulou K, Andreadis A, D'Souza I, Lee VM, Reed L, Trojanowski JQ, Zhukareva V, Bird T, Schellenberg G, Wilhelmsen KC. Pathogenic implications of mutations in the tau gene in pallido-ponto-nigral degeneration and related neurodegenerative disorders linked to chromosome 17. Proc Natl Acad Sci U S A. 1998 Oct 27;95(22):13103-7. PubMed.
- Tsuboi Y, Baker M, Hutton ML, Uitti RJ, Rascol O, Delisle MB, Soulages X, Murrell JR, Ghetti B, Yasuda M, Komure O, Kuno S, Arima K, Sunohara N, Kobayashi T, Mizuno Y, Wszolek ZK. Clinical and genetic studies of families with the tau N279K mutation (FTDP-17). Neurology. 2002 Dec 10;59(11):1791-3. PubMed.
- Wszolek ZK, Lagerlund TD, Steg RE, McManis PG. Clinical neurophysiologic findings in patients with rapidly progressive familial parkinsonism and dementia with pallido-ponto-nigral degeneration. Electroencephalogr Clin Neurophysiol. 1998 Sep;107(3):213-22. PubMed.
- Reed LA, Schmidt ML, Wszolek ZK, Balin BJ, Soontornniyomkij V, Lee VM, Trojanowski JQ, Schelper RL. The neuropathology of a chromosome 17-linked autosomal dominant parkinsonism and dementia ("pallido-ponto-nigral degeneration"). J Neuropathol Exp Neurol. 1998 Jun;57(6):588-601. PubMed.
- Kowalska A, Asada T, Arima K, Kumakiri C, Kozubski W, Takahashi K, Tabira T. Genetic analysis in patients with familial and sporadic frontotemporal dementia: two tau mutations in only familial cases and no association with apolipoprotein epsilon4. Dement Geriatr Cogn Disord. 2001;12(6):387-92. PubMed.
- Baba Y, Tsuboi Y, Baker MC, Uitti RJ, Hutton ML, Dickson DW, Farrer M, Putzke JD, Woodruff BK, Ghetti B, Murrell JR, Boeve BF, Petersen RC, Verpillat P, Brice A, Delisle MB, Rascol O, Arima K, Dysken MW, Yasuda M, Kobayashi T, Sunohara N, Komure O, Kuno S, Sperfeld AD, Stoppe G, Kohlhase J, Pickering-Brown S, Neary D, Bugiani O, Wszolek ZK. The effect of tau genotype on clinical features in FTDP-17. Parkinsonism Relat Disord. 2005 Jun;11(4):205-8. PubMed.
- Chen Q, Boeve BF, Senjem M, Tosakulwong N, Lesnick TG, Brushaber D, Dheel C, Fields J, Forsberg L, Gavrilova R, Gearhart D, Graff-Radford J, Graff-Radford NR, Jack CR Jr, Jones DT, Knopman DS, Kremers WK, Lapid M, Rademakers R, Syrjanen J, Boxer AL, Rosen H, Wszolek ZK, Kantarci K, LEFFTDS Consortium. Rates of lobar atrophy in asymptomatic MAPT mutation carriers. Alzheimers Dement (N Y). 2019;5:338-346. Epub 2019 Jul 30 PubMed.
- Covello G, Siva K, Adami V, Denti MA. HCS-Splice: A High-Content Screening Method to Advance the Discovery of RNA Splicing-Modulating Therapeutics. Cells. 2023 Jul 28;12(15) PubMed.
- de Silva R, Lashley T, Strand C, Shiarli AM, Shi J, Tian J, Bailey KL, Davies P, Bigio EH, Arima K, Iseki E, Murayama S, Kretzschmar H, Neumann M, Lippa C, Halliday G, MacKenzie J, Ravid R, Dickson D, Wszolek Z, Iwatsubo T, Pickering-Brown SM, Holton J, Lees A, Revesz T, Mann DM. An immunohistochemical study of cases of sporadic and inherited frontotemporal lobar degeneration using 3R- and 4R-specific tau monoclonal antibodies. Acta Neuropathol. 2006 Apr;111(4):329-40. Epub 2006 Mar 22 PubMed.
- Deutschländer AB, Boeve BF, Rosen HJ, Boxer AL, Wszolek ZK, LEFFTDS Consortium. Tau Mutations as a Novel Risk Factor for Cancer-Letter. Cancer Res. 2018 Nov 15;78(22):6523-6524. Epub 2018 Oct 29 PubMed.
- Ferman TJ, McRae CA, Arvanitakis Z, Tsuboi Y, Vo A, Wszolek ZK. Early and pre-symptomatic neuropsychological dysfunction in the PPND family with the N279K tau mutation. Parkinsonism Relat Disord. 2003 Jun;9(5):265-70. PubMed.
- Frank AR, Wszolek ZK, Jack CR Jr, Boeve BF. Distinctive MRI findings in pallidopontonigral degeneration (PPND). Neurology. 2007 Feb 20;68(8):620-1. PubMed.
- Frucht SJ. Commentary on "Pallidopontonigral degeneration: a deceptive familial tauopathy". Mov Disord. 2012 Jun;27(7):820-1. PubMed.
- Kantarci K, Boeve BF, Wszolek ZK, Rademakers R, Whitwell JL, Baker MC, Senjem ML, Samikoglu AR, Knopman DS, Petersen RC, Jack CR Jr. MRS in presymptomatic MAPT mutation carriers: a potential biomarker for tau-mediated pathology. Neurology. 2010 Aug 31;75(9):771-8. PubMed.
- Kishore A, Wszolek ZK, Snow BJ, de la Fuente-Fernandez R, Arwert F, Wijker M, Schulzer M, Calne DB, Vingerhoets FJ. Presynaptic nigrostriatal function in genetically tested asymptomatic relatives from the pallido-ponto-nigral degeneration family. Neurology. 1996 Dec;47(6):1588-90. PubMed.
- Kumaran R, Kingsbury A, Coulter I, Lashley T, Williams D, de Silva R, Mann D, Revesz T, Lees A, Bandopadhyay R. DJ-1 (PARK7) is associated with 3R and 4R tau neuronal and glial inclusions in neurodegenerative disorders. Neurobiol Dis. 2007 Oct;28(1):122-32. Epub 2007 Jul 18 PubMed.
- Miklossy J, Qing H, Guo JP, Yu S, Wszolek ZK, Calne D, McGeer EG, McGeer PL. Lrrk2 and chronic inflammation are linked to pallido-ponto-nigral degeneration caused by the N279K tau mutation. Acta Neuropathol. 2007 Sep;114(3):243-54. Epub 2007 Jul 17 PubMed.
- Ono M, Sahara N, Kumata K, Ji B, Ni R, Koga S, Dickson DW, Trojanowski JQ, Lee VM, Yoshida M, Hozumi I, Yoshiyama Y, van Swieten JC, Nordberg A, Suhara T, Zhang MR, Higuchi M. Distinct binding of PET ligands PBB3 and AV-1451 to tau fibril strains in neurodegenerative tauopathies. Brain. 2017 Mar 1;140(3):764-780. PubMed.
- Shimura H, Mori H, Komatsuzaki Y, Nakamura K, Takanashi M, Hattori N, Mizuno Y. [A 54-year-old man with familial parkinsonism, gaze palsy, and dementia]. No To Shinkei. 2005 Dec;57(12):1104-12. PubMed.
- Slowinski JL, Schweitzer KJ, Imamura A, Uitti RJ, Strongosky AJ, Dickson DW, Broderick DF, Wszolek ZK. Brainstem atrophy on routine MR study in pallidopontonigral degeneration. J Neurol. 2009 May;256(5):827-9. Epub 2009 Mar 1 PubMed.
- Staffaroni AM, Goh SM, Cobigo Y, Ong E, Lee SE, Casaletto KB, Wolf A, Forsberg LK, Ghoshal N, Graff-Radford NR, Grossman M, Heuer HW, Hsiung GR, Kantarci K, Knopman DS, Kremers WK, Mackenzie IR, Miller BL, Pedraza O, Rascovsky K, Tartaglia MC, Wszolek ZK, Kramer JH, Kornak J, Boeve BF, Boxer AL, Rosen HJ, ARTFL-LEFFTDS Longitudinal Frontotemporal Lobar Degeneration Consortium. Rates of Brain Atrophy Across Disease Stages in Familial Frontotemporal Dementia Associated With MAPT, GRN, and C9orf72 Pathogenic Variants. JAMA Netw Open. 2020 Oct 1;3(10):e2022847. PubMed.
- Whitwell JL, Jack CR Jr, Boeve BF, Senjem ML, Baker M, Rademakers R, Ivnik RJ, Knopman DS, Wszolek ZK, Petersen RC, Josephs KA. Voxel-based morphometry patterns of atrophy in FTLD with mutations in MAPT or PGRN. Neurology. 2009 Mar 3;72(9):813-20. PubMed.
- Whitwell JL, Jack CR Jr, Boeve BF, Senjem ML, Baker M, Ivnik RJ, Knopman DS, Wszolek ZK, Petersen RC, Rademakers R, Josephs KA. Atrophy patterns in IVS10+16, IVS10+3, N279K, S305N, P301L, and V337M MAPT mutations. Neurology. 2009 Sep 29;73(13):1058-65. PubMed.
- Whitwell JL, Josephs KA, Avula R, Tosakulwong N, Weigand SD, Senjem ML, Vemuri P, Jones DT, Gunter JL, Baker M, Wszolek ZK, Knopman DS, Rademakers R, Petersen RC, Boeve BF, Jack CR Jr. Altered functional connectivity in asymptomatic MAPT subjects: a comparison to bvFTD. Neurology. 2011 Aug 30;77(9):866-74. Epub 2011 Aug 17 PubMed.
- Woodruff BK, Baba Y, Hutton ML, Wszolek ZK, Tsuboi Y, Kobayashi T, Ghetti B, Arima K, Yasuda M, Rascol O. Haplotype-phenotype correlations in kindreds with the N279K mutation in the tau gene. Arch Neurol. 2004 Aug;61(8):1327; author reply 1327. PubMed.
- Wszolek ZK, Uitti RJ, Hutton M. A mutation in the microtubule-associated protein tau in pallido-nigro-luysian degeneration. Neurology. 2000 May 23;54(10):2028-30. PubMed.
- Wszolek ZK, Tsuboi Y, Uitti RJ, Reed L. Two brothers with frontotemporal dementia and parkinsonism with an N279K mutation of the tau gene. Neurology. 2000 Dec 26;55(12):1939. PubMed.
- Wszolek ZK, Kardon RH, Wolters EC, Pfeiffer RF. Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17): PPND family. A longitudinal videotape demonstration. Mov Disord. 2001 Jul;16(4):756-60. PubMed.
Protein Diagram
Primary Papers
- Wszolek ZK, Pfeiffer RF, Bhatt MH, Schelper RL, Cordes M, Snow BJ, Rodnitzky RL, Wolters EC, Arwert F, Calne DB. Rapidly progressive autosomal dominant parkinsonism and dementia with pallido-ponto-nigral degeneration. Ann Neurol. 1992 Sep;32(3):312-20. PubMed.
Alzpedia
Disclaimer: Alzforum does not provide medical advice. The Content is for informational, educational, research and reference purposes only and is not intended to substitute for professional medical advice, diagnosis or treatment. Always seek advice from a qualified physician or health care professional about any medical concern, and do not disregard professional medical advice because of anything you may read on Alzforum.
Comments
No Available Comments
Make a Comment
To make a comment you must login or register.