KL-VS heterozygosity is associated with reduced tau accumulation and lower memory impairment in Alzheimer’s disease ===================================================================================================================== * Julia Neitzel * Nicolai Franzmeier * Anna Rubinski * Martin Dichgans * Matthias Brendel * Rainer Malik * Michael Ewers * for the Alzheimer’s Disease Neuroimaging Initiative (ADNI) ## ABSTRACT Klotho-VS heterozygosity (KL-VShet) is associated with reduced risk of Alzheimer’s disease (AD). However, whether KL-VShet is associated with lower levels of pathologic tau, i.e. the key AD pathology driving neurodegeneration and cognitive decline, is unknown. Here, we assessed the interaction between KL-VShet and levels of beta-amyloid, a key driver of tau pathology, on the levels of PET-assessed neurofibrillary tau in 354 controls and patients within the AD continuum. KL-VShet showed lower cross-sectional increase in tau-PET per unit increase in amyloid-PET when compared to that of non-carriers. This effect of KL-VShet on tau-PET showed a tendency to be stronger in Klotho mRNA-expressing brain regions mapped onto a gene expression atlas. KL-VShet was related to better memory functions and this association was mediated by lower tau-PET. Amyloid-PET levels did not differ between KL-VShet carriers versus non-carriers. Together, our findings provide evidence for a protective role of KL-VShet against tau pathology and tau-related memory impairments in elderly humans at risk of AD dementia. ## INTRODUCTION Klotho is a transmembrane protein that has been associated with enhanced longevity and better brain health in aging1,2. Klotho is expressed primarily in the kidney and brain, where it has been implicated in a number of vital cellular functions (for review see3). Loss-of-function mutations in transgenic mice are associated with reduced Klotho protein expression, accelerated aging phenotypes and dramatically shortened life span1,4. In humans, two variants in the KLOTHO gene (KL, 13q13.1), rs9536314 (F352V) and rs9527025 (C370S) form a functional haplotype. Carrying one copy, but not two copies of the KL-VS haplotype, referred to as KL-VS heterozygosity (KL-VShet), has been previously linked to increased Klotho levels in the blood5,6. KL-VShet occurs in about 20-25% of the population5 and is associated with higher cognitive performance across the adult life span5,7-9, larger fronto-temporal grey matter volume in cognitively normal individuals8 and lower mortality6,10. Together, these results suggest a crucial role of Klotho in the maintenance of cognitive abilities and brain integrity during aging. Beyond the protective role of Klotho in normal aging, recent studies suggest an association between Klotho and reduced risk of Alzheimer’s disease (AD)11, the most frequent cause of dementia in the elderly12. A recent meta-analysis reported that KL-VShet was associated with reduced AD dementia risk and cognitive decline in elderly individuals carrying the ApoE ε4 allele13, i.e. the strongest genetic risk factor for AD dementia possibly through elevated levels of primary AD pathology including cortical beta-amyloid (Aβ) aggregation14,15. Importantly, KL-VShet was associated with reduced biomarker levels of Aβ deposition in ApoE ε4 carriers16 suggesting that KL-VShet may directly alter levels of primary AD pathology. Yet, an open question is whether Klotho is associated with altered levels of fibrillary tangles containing pathologic tau, i.e. the key driver of disease progression in AD17. In the presence of Aβ deposition, i.e. the earliest primary pathology in AD18,19, neurofibrillary tangles spread from the medial temporal lobe to higher cortical areas20. The progressive development of neurofibrillary tangles in the presence of Aβ pathology is closely associated with grey matter atrophy21-23 and cognitive worsening20,24-26 and is more predictive of such alterations than Aβ27. Due to the high clinical relevance of tau pathology, it is pivotal to understand whether the KL-VShet variant attenuates the accumulation of neurofibrillary tangles at a given level of Aβ deposition, and thus cognitive decline in AD. Studies in mouse models of Aβ and accelerated aging reported that enhancing Klotho expression was associated with reduced Aβ burden and phosphorylated tau11,28, although conflicting results were reported as well29. However, these mouse models fail to develop neurofibrillary tangles in the presence of Aβ and thus only incompletely recapitulate AD-specific tau pathology in humans. Here, we examined whether KL-VShet attenuates the association between higher Aβ and higher fibrillar tau assessed via positron emission tomography (PET) in a group of 354 elderly asymptomatic and symptomatic individuals recruited within a large North American multicenter study on AD30. We found the KL-VShet variant to be associated with an attenuated increase in regional tau-PET at pathological levels of global amyloid-PET, suggesting that KL-VShet was protective against AD-related increase in neurofibrillary tangles. This effect was particularly pronounced in ApoE ε4 carriers. The strength of the KL-VShet effect on region-specific tau-PET levels showed a trend to be correlated with the regional expression pattern of KL in the brain31,32 supporting the notion that the KL-VShet variant modulates regional accumulation of tau pathology. Importantly, KL-VShet was associated with higher memory performance and this effect was mediated by reduced tau-PET levels in KL-VShet carriers with elevated amyloid-PET burden. For Aβ, we did not find the previously reported protective effect of KL-VShet against Aβ pathology in the current sample, but confirmed this effect in a larger sample including all individuals with amyloid-PET but not necessarily tau-PET assessment available indicating that the effect size of KL-VShet on Aβ was smaller than that on tau pathology. Overall, we show a protective role of KL-VShet against the development of tau pathology and thus cognitive decline, suggesting that Klotho could be an attractive treatment target to slow the progression of AD. ## RESULTS Detailed sample characteristics are presented in **Table 1**. Among the 354 participants (213 CN, 111 MCI, 30 ADD), there were 92 KL-VShet carriers and 262 non-carriers. Demographics (age, sex, education) or ApoE ε4 status did not differ between KL-VShet carrier and non-carrier groups (all p > 0.05). Continuous values of global amyloid-PET uptake did not differ between KL-VShet carriers versus non-carriers (t(174.74) = 1.40, p = 0.163). View this table: [Table 1.](http://medrxiv.org/content/early/2020/07/31/2020.07.29.20164434/T1) Table 1. sample characteristics ### KL-VS heterozygosity attenuates effects of amyloid on tau accumulation In our main analysis, we tested the hypothesis that KL-VShet modifies the association between Aβ and tau pathology (both assessed by continuous measures of PET uptake). In a region-of-interest (ROI) based analysis we focused on tau-PET in the inferior temporal cortex (i.e. ROI of early Aβ-related tau pathology20,24,33-35) and whole-brain tau-PET levels. The results of a linear regression analysis showed a significant KL-VShet *x* amyloid-PET interaction effect on tau-PET levels in both the inferior temporal ROI (standardized beta = −0.57, p = 0.018, effect size measured by Cohen’s f = 0.128) and the global ROI (beta = −0.54, p = 0.030, Cohen’s f = 0.118). For both tau-PET ROIs, the increase in tau-PET as a function of rising global amyloid-PET was attenuated in KL-VShet carriers versus non-carriers (**Figure 1a, b**). This result suggests that the KL-VShet variant was protective against Aβ-associated accumulation of tau pathology. All analyses were controlled for main effects of the interaction terms, age, sex, diagnosis, education and ApoE ε4 carrier status. To determine whether our findings were driven by clinical diagnosis, we specifically repeated the analyses in 111 MCI patients (21 KL-VShet carriers and 90 non-carriers) and found comparable KL-VShet *x* amyloid-PET interaction effects on tau-PET uptake (inferior temporal ROI: beta = -1.06, p = 0.021, Cohen’s f = 0.230; global ROI: beta = -1.01, p = 0.025, Cohen’s f = 0.224; **eFigure 1 in Supplement**). Previous studies have reported an ApoE-ε4-genotype dependent effect of KL-VShet on amyloid PET16. Hence, we additionally explored whether ApoE ε4 carriers showed a stronger protective effect of KL-VShet against tau accumulation than ApoE ε4 non-carriers, controlling for age, sex, education, diagnosis and global amyloid-PET levels in the regression analyses. We found KL-VShet to be associated with lower tau-PET levels in ApoE ε4 carriers for the global ROI (beta = −0.15, p = 0.032) and at trend level for the inferior temporal ROI (beta = −0.12, p = 0.080), whereas we found no main effect of KL-VShet in ApoE ε4 non-carriers (global ROI: beta = 0.02, p = 0.711; inferior temporal ROI: beta=0.01, p=0.817; **eFigure 2 in Supplement**). Tested in the whole sample, the KL-VShet *x* ApoE ε4 interaction effect on tau-PET levels was not significant (global ROI: beta = −0.09, p = 0.135; inferior temporal ROI: beta = −0.09, p = 0.108). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/07/31/2020.07.29.20164434/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2020/07/31/2020.07.29.20164434/F1) Figure 1. Association between KL-VS heterozygosity, amyloid- and tau-PET Scatterplots display the relationship between global amyloid-PET levels and tau-PET-levels measured **a** in inferior temporal gyri and **b** globally in neocortical areas as a function of KL-VShet variant. Blue and grey colour indicate individuals with heterozygous or non-heterozygous KL-VS alleles. Statistics of the KL-VShet *x* amyloid-PET interaction effect on tau-PET uptake were derived from multiple linear regression analyses, controlled for the main effects of KL-VShet and amyloid-PET levels as well as age, sex, diagnosis, education and ApoE ε4 carrier status. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/07/31/2020.07.29.20164434/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2020/07/31/2020.07.29.20164434/F2) Figure 2. Spatial patterns of KL-VShet-related attenuation of tau-PET and Klotho mRNA expression. a Surface mapping of the interaction effect between KL-VShet and amyloid-PET levels on tau-PET accumulation within 34 left-hemispheric regions of the Desikan-Kiliany atlas. Yellow colours indicate higher t-values reflective of a stronger interaction effect (all t-values inverted for illustration purpose). **b** Thresholded spatial map color-code only regions with a significant (p<0.05) KL-VShet *x* amyloid-PET interaction effect. **c** Surface mapping of median KL mRNA expression (i.e., log2 derived from the Allen Brain Atlas) within the identical 34 atlas regions. Yellow colours indicate higher KL mRNA expression. **d** Thresholded spatial maps restricted to regions falling above the 75th percentile of KL mRNA expression. **e** Scatterplot depicting the positive association between ROI-based KL mRNA expression and KL-VShet *x* amyloid-PET interaction effect on regional tau-PET uptake. The solid grey line together with the shaded areas illustrate least-squares regression line ±95% confidence interval. ### Spatial match between KL mRNA expression and the effect of KL-VS heterozygosity on tau-PET In order to estimate the spatial overlap between regions of KL gene expression and regions for which the KL-VShet *x* amyloid-PET interaction on tau-PET was significant, we obtained whole-brain mRNA expression levels of KL generated by post-mortem microarray assessments of six healthy brain donors and subsequently mapped to the Allen Brain Atlas31,32. We computed median scores of log2 mRNA expression of KL across the six donors within 34 left-hemispheric regions of the Freesurfer-based Desikan–Killiany brain atlas36. We focused on the left hemisphere, since all donors had microarray assessment available for the left hemisphere and only two donors had assessment for the right hemisphere. Furthermore, we estimated the KL-VShet *x* amyloid-PET interaction effect on tau-PET levels within the same 34 brain-atlas regions using the aforementioned regression model. Surface mapping of both the KL-VShet *x* amyloid-PET interaction effect (which were all in the same direction) and KL mRNA expression is displayed in **Figure 2a-d**. Spatial correlation analysis revealed an association at trend level (r = 0.34, p = 0.049; **Figure 2e**). This result suggests that regions with higher KL mRNA expression levels were more likely to display a stronger protective effect of KL-VShet on Aβ-related increase in tau-PET. Visual inspection of the thresholded spatial maps indicated that those areas showing both a significant KL-VShet *x* amyloid-PET interaction effect (**Figure 2b**) and high KL mRNA expression (log2 > 75th percentile) (**Figure 2d**) were specifically located within the mesio-temporal and inferior temporal brain regions and the posterior cingulum. ### Tau mediates the beneficial effect of KL-VS heterozygosity on memory Next, we assessed whether KL-VShet is beneficial for memory functions via lowering tau pathology. Because the interaction effect of KL-VShet *x* amyloid-PET on tau-PET levels showed that the protective role of KL-VShet against tau accumulation was stronger at higher levels of amyloid-PET, we restricted our analysis to participants who displayed elevated global amyloid-PET levels. The optimal amyloid-PET SUVR threshold for sample selection was identified in our data as SUVR > 1.0 including 67 KL-VShet carriers and 191 non-carriers (see Methods for more details on sample selection). We used mediation analysis with 10,000 bootstrapping iterations in order to test whether KL-VShet is associated with better memory in individuals with elevated Aβ burden and whether this effect is mediated via reduced global tau-PET levels. Memory performance was measured by an established composite score based on participant’s results across multiple different memory tests (ADNI-MEM)37. The mediation analysis was controlled for age, sex, education, diagnosis, ApoE ε4 status and global amyloid-PET levels. Supporting our hypothesis, we found KL-VShet to be associated with higher ADNI-MEM scores (beta = 0.12, p = 0.029, Cohen’s f = 0.137, **Figure 3**) and that this relationship was significantly mediated by lower tau-PET levels (bootstrapped average causal mediation effect: beta = 0.05, 95% confidence interval = 0.01 - 0.10, p = 0.008). This result indicates that, in individuals with an elevated amyloid burden, KL-VShet carriers showed less impaired episodic-memory abilities when compared to KL-VShet non-carriers due to lower tau-PET levels in KL-VShet carriers. A path model of the mediation analysis is shown in **Figure 4**. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/07/31/2020.07.29.20164434/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2020/07/31/2020.07.29.20164434/F3) Figure 3. KL-VS heterozygosity effect on memory in individuals with elevated amyloid-PET burden Boxplot shows memory performance as a function of KL-VShet variant in individuals with a global amyloid-PET SUVR > 1.0. Blue and grey colour indicates individuals with heterozygous (N = 67) or non-heterozygous KL-VS alleles (N = 191). Memory was measured by an established composite score, ADNI-MEM, based on test performance across multiple different memory tests37. Statistical result of the main effect of KL-VShet on memory was derived from multiple linear regression analysis, controlled for age, sex, diagnosis, education and ApoE ε4 carrier status. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/07/31/2020.07.29.20164434/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2020/07/31/2020.07.29.20164434/F4) Figure 4. Lower Tau-PET levels mediate beneficial effect of KL-VS heterozygosity on memory in individuals with elevated amyloid-PET burden Path diagram of the mediation model (assessed only in individuals with global amyloid-PET SUVR > 1.0, N = 258), showing that the association between KL-VShet and better memory performance is mediated via lower global tau-PET uptake. Memory is measured by ADNI-MEM, i.e. an established memory composite score37. Path-weights are displayed as standardized beta values. All paths are controlled for age, sex, diagnosis, education, ApoE ε4 carrier status and continuous global amyloid-PET levels. Effects are reported at *p<0.05, **p<0.01 and \***|p<0.001. Significance of the indirect effect was determined using bootstrapping with 10,000 iterations as implemented in the *mediation* package in *R*. In order to address the question of whether KL-VShet exerts a beneficial influence on memory via lowered neurofibrillary tau in the whole sample without stratification based on amyloid-PET levels, we estimated the interaction effect between KL-VShet and amyloid-PET on ADNI-MEM scores, with and without controlling for global tau-PET levels as a covariate. We reasoned that if the association between KL-VShet on cognition is mediated by tau-PET, the interaction between KL-VShet and amyloid-PET on cognition should be diminished when controlling for tau-PET. Without controlling tau-PET levels, we found a borderline significant KL-VShet *x* amyloid-PET interaction effect on memory functions (beta = 0.42, p = 0.049, Cohen’s f = 0.107). Specifically, we observed that individuals with high amyloid-PET burden showed better cognitive performance when being KL-VShet carriers compared to non-carriers (**eFigure 3 in Supplement**). As hypothesized, the KL-VShet *x* amyloid-PET interaction on memory no longer reached significance level when global tau-PET levels were controlled for (beta = 0.31, p = 0.139). ### Confirmation of the protective effect of KL-VShet against Aβ accumulation in a larger sample A recent study found a protective influence of KL-VShet on longitudinal amyloid-PET in cognitively unimpaired ApoE ε4 carriers aged between 60 and 80 years, but not in ApoE ε4 non-carriers or older participants13. In contrast to this earlier report, we did not find an age- or ApoE ε4-dependent KL-VShet effect on cross-sectional amyloid-PET levels acquired at the time of tau-PET assessment in the current sample (KL-VShet *x* age interaction: beta = 0.02, p = 0.969, KL-VShet *x* ApoE ε4 interaction: beta = 0.02, p = 0.744; KL-VShet *x* age *x* ApoE ε4 interaction: beta = 0.39, p = 0.574; **eFigure 4 in Supplement**). However, more subtle effects may have been overlooked in the current more restricted sample of individuals undergoing both amyloid- and tau-PET. Therefore, in a supplementary analysis we included all participants with amyloid-PET (N=959) from ADNI, regardless of whether or not they underwent tau-PET assessment. We found a significant KL-VShet *x* age interaction effect one amyloid-PET which demonstrated that KL-VShet carriers in the lower age range (< 80 years) displayed lower amyloid-PET levels than non-carriers (beta=0.62, p=0.033, Cohen’s f = 0.069, **eFigure 5a in Supplement**). As observed earlier, the beneficial effect of KL-VShet on amyloid-PET levels was driven by ApoE ε4 carriers rather than non-carriers (**eFigure 5bc in Supplement**). See **eTable 1 in Supplement** for detailed sample characteristics of the larger ADNI amyloid-PET sample compared to the current ADNI tau-PET sample. Thus, our results in the larger sample are consistent with those from Belloy et al.’s analysis on the effect of KL-VShet on amyloid-PET stratified by age and ApoE genotype while also showing that the effect size of KL-VShet on tau-PET is stronger than that on amyloid-PET. ## DISCUSSION The heterozygous KLOTHO gene variant KL-VShet, has been previously associated with higher longevity and cognition performance in adulthood and reduced AD dementia risk13. We demonstrate that elderly KL-VShet carriers with elevated Aβ burden, i.e. the earliest primary AD pathology, exhibited lower tau-PET levels when compared to those in KL-VShet non-carriers. The KL-VShet variant was associated with better memory performance, and this relationship was mediated by lower tau-PET levels, suggesting that lower levels of pathologic tau in the KL-VShet carriers explain the protective association between KL-VShet and memory performance. Although our findings do not implicate a causative mechanism of Klotho in AD, we provide evidence for a protective role of KL-VShet against tau pathology which is the key AD brain alteration linked to cognitive impairment. To our knowledge, the current study is the first to date that evaluated the interaction between KL-VShet and Aβ on tau accumulation and cognitive decline in humans. There is a growing literature on protective genetic variants in AD38-40, but only a few studies have reported genetic variants to be associated with lower tau pathology in AD41. For the KL-VShet variant, previous studies reported an association with reduced Aβ accumulation in elderly ApoE ε4 risk-carriers13,16. We extend these previous findings by showing that the relationship between Aβ accumulation and fibrillar tau is modulated by KL-VShet, such that lower local and global tau-PET levels were observed per unit increase of global amyloid-PET burden in KL-VShet carriers when compared to those in non-carriers. This is important because Aβ deposition precedes the development of dementia symptoms by up to 20 years14, and is associated with enhanced production of tau and the development of neurotoxic neurofibrillary tangles42. The region showing the strongest interaction effect between KL-VShet and amyloid-PET on tau-PET was the inferior temporal gyrus (Figure 2a), a brain area that typically shows an early Aβ-related increase in tau-PET24 before elevated tau-PET levels extend to other higher cortical brain areas20. The protective association between KL-VShet and tau-PET was present selectively in participants with abnormally elevated levels of amyloid-PET and in ApoE ε4 carriers. Together, these results support the notion that KL-VShet is associated with an AD-related rather than age-related reduction of tau pathology. We found KL-VShet to be associated with better memory performance, mediated by the effect of KL-VShet on tau-PET. Our results are broadly consistent with those from studies on healthy aging, reporting a protective effect of KL-VShet on cognition5,8-10, and lower risk of conversion from cognitively normal to mild cognitive impairment or AD dementia in ApoE ε4 carriers13,16. Our findings suggest that the association between KL-VShet and lower neurofibrillary tau pathology is of central importance for the protective effect of KL-VShet against cognitive impairment. A previously reported absence of an association between KL-VShet and cognitive decline in asymptomatic participants with elevated levels of Aβ43 did not assess the presence of abnormal neurofibrillary tau, which may have hampered to detect an effect of KL-VShet on cognitive decline in subjects at risk of AD 33. Previous studies reported KL-VShet to be associated with lower amyloid-PET in ApoE ε4 carriers (but not in ApoE ε4 non-carriers), which was strongest in the age range between 60 and 80 years13,16. In our primary analysis we did not confirm age- or ApoE ε4-dependent effects of KL-VShet on tau-PET. By investigating a larger sample of all participants with available amyloid-PET regardless of the availability of tau-PET (N = 959), we were able to substantiate those earlier findings13,16. Specifically, we showed reduced amyloid-PET burden in younger KL-VShet carriers (< 80 years) and, in accordance with previous work, this association was mainly driven by ApoE ε4 carriers rather than non-carriers. Comparing effect sizes, Cohen’s f = 0.069 for the effect of KL-VShet on amyloid-PET versus f = 0.118 for the effect on tau-PET, strengthens the important role of changes in tau pathology for understanding the role of Klotho in AD. The mechanisms linking Klotho to tau pathology remain elusive. Klotho is a pleiotropic protein that has been implicated in multiple biological processes including insulin regulation4, growth factor functions, in particular of FGF2344, regulation of members of the redox system45, and calcium signaling46. One possibility of how the Klotho protein might be linked to reduced neurofibrillary tau is its involvement in autophagy47, a mechanism that is involved in the clearance of AD pathologies48. Lentiviral overexpression of Klotho protein in an APP-PS1 mouse model of Aβ deposition reduced Aβ plaque load in aged mice and rescued the impaired autophagy possibly by modulating the Akt/mTOR pathway11,49. Since APP-PS1 mice do not develop tau pathology, it remains, however, to be tested whether Klotho-induced autophagy reduces tau pathology. Those mechanistic explanations remain speculative at this point and the current work encourages future studies to investigate the mechanism that could underlie the protection Klotho exerts against the development of tau pathology. Our findings of the spatial correspondence between the strength of the effect of KL-VShet on regional tau-PET and the spatial distribution of Klotho mRNA suggests a local effect of Klotho on the development of fibrillar tau especially in temporal brain areas. The result of the spatial correlation analysis was at trend level and call for replication. Nevertheless, it is worth noting that alternative splicing of the human Klotho mRNA results in both a membrane-bound and a secreted transcript of Klotho1,4, indicating that Klotho may act both in a cell-autonomous manner and as a humoral factor. Therefore, differences in gene expression in Klotho in the brain and/or different circulating levels of Klotho linked to KL-VShet5 may influence the development of pathological tau11, but this link remains to be investigated. Several caveats should be considered when interpreting the current results. Firstly, the human KL gene consists of three polymorphic variants. We decided to focus on the KL-VS haplotype given the existing evidence of its beneficial influence on Aβ and cognition in both mice and humans5,8-10,13,50. While the second variant C1818T (rs564481) is located on the fourth exon and likely has no functional consequences itself, the third variant G395A (rs1207568) is located in the promotor region and may be a potential regulatory site of KL. The two latter variants appear more frequent in Asian populations, where they have been linked to cardiovascular risk factors51. Related to the current research question, an investigation across three independent cohorts of oldest old Danes found different polymorphic variants of KL, besides KL-VS, to be associated with better cognitive functions7. It has yet to be proven whether these other KL variants also support resilience in AD. Another caveat is that we did not measure Klotho protein levels in the serum or CSF. Circulating levels of Klotho decrease during aging52 and are associated with cognitive performance5 and grey matter volume53 in cognitively unimpaired individuals. In patients with AD, CSF levels of Klotho are reduced50, where the experimental reversal of reduced Klotho expression in transgenic mouse models exerted beneficial effects on Aβ and cognition8,11,28. While the KL-VShet variant has been associated with higher circulating levels of Klotho53, it remains to be investigated whether the association between KL-VShet and pathological tau are mediated by higher protein levels in the CSF and brain tissue. In summary, due to the high clinical relevance of tau accumulation and mixed results from trials on Aβ immunization54, strategies for targeting tau pathology are moving into focus55. Our findings revealed a protective effect of KL-VShet against tau accumulation which particularly manifested in at-risk individuals, where lower tau pathology was related to better cognitive functions. Our results encourage future studies on enhancing Klotho proteins levels as a therapeutic intervention to slow down the development of tau pathology and dementia in AD. ## MATERIALS & METHODS ### Sample 354 participants were selected from ADNI phase 3 ([ClinicalTrials.gov](http://ClinicalTrials.gov) ID: [NCT02854033](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT02854033&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom)) based on the availability of KL-VS and ApoE ε4 genotyping, T1-weighted MRI, 18F-AV1451 tau-PET and 18F-AV45 amyloid-PET. MR and PET imaging had to be acquired during the same study visit. The two single nucleotide polymorphisms (SNPs) for KL-VS (rs9536314 for F352V, rs9527025 for C370S) and ApoE (rs429358, rs7412) were genotyped using DNA extracted by Cogenics from a 3 mL aliquot of EDTA blood. Participants were assigned to the heterozygous KL-VS group when they carried 1, but not 2, copies of the KL-VS haplotype. ApoE ε4 carriers were defined as individuals carrying at least one ε4 allele. Clinical classification was performed by the ADNI centers, dividing participants into cognitively normal (CN, MMSE>24, CDR=0, non-depressed), mild cognitively impairment (MCI; MMSE>24, CDR=0.5, objective memory-loss on the education adjusted Wechsler Memory) or AD dementia (ADD; 19 0.8-1.2). The optimal amyloid-PET threshold was identified as global SUVR > 1.0 (**eFigure 6 in Supplement**). Significance of the mediation effect was determined using 10,000 bootstrapped iterations, where each path of the model was controlled for age, sex, diagnosis, education, ApoE ε4 carrier status and global amyloid-PET levels. In an alternative approach, we estimated the interaction effect between KL-VShet *x* amyloid-PET on cognitive functions. This analysis was performed in the entire sample. Importantly, the interaction analysis was once run without and once with controlling global tau-PET levels. We specifically hypothesized that if the beneficial effect of KL-VShet is dependent on lowering tau accumulation, then the interaction effect should be diminished in the tau-controlled analysis. Other covariates considered in the multiple regression models were age, sex, diagnosis, education and ApoE ε4 carrier status. Lastly, we performed an exploratory analysis with the aim to confirm previously observed age-dependent associations between KL-VShet and lower amyloid-PET burden in ApoE ε4 carriers13. For this purpose, we tested for a KL-VShet *x* age effect on global amyloid-PET levels in the current sample (N = 354) and in a larger ADNI sample (N = 959) including all participants with amyloid-PET assessment and KL-VS status (regardless of whether or not they underwent tau-PET assessment). Main effects of KL-VShet and age as well as sex, education and diagnosis were considered as covariates. In sensitivity analyses, we reran the interaction model in stratified groups of either ApoE ε4 carriers or non-carriers. All statistical analyses were conducted with *R* statistical software (version 3.6.1). P-values were considered significant when meeting a two-tailed alpha threshold of 0.05. ## Data availability The data that support the findings of this study were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) and are available from the ADNI database (adni.loni.usc.edu) upon registration and compliance with the data use agreement. A list including the anonymized participant identifiers of the currently used sample and the source file can be downloaded from the ADNI database ([http://adni.loni.usc.edu/](http://adni.loni.usc.edu/)). The R-script used for the current study can be obtained from the first author upon request. ## Data Availability The data that support the findings of this study were obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and are available from the ADNI database (adni.loni.usc.edu) upon registration and compliance with the data use agreement. A list including the anonymized participant identifiers of the currently used sample and the source file can be downloaded from the ADNI database ([http://adni.loni.usc.edu/](http://adni.loni.usc.edu/)). The R-script used for the current study can be obtained from the first author upon request. [http://adni.loni.usc.edu/](http://adni.loni.usc.edu/) ## Contributions J.N.: study concept and design, data processing, statistical analysis, interpretation of the results, and writing the manuscript. N.F.: critical revision of the manuscript. A.R.: data processing and critical revision of the manuscript. M.D.: critical revision of the manuscript. M.B.: critical revision of the manuscript. R.M.; .data processing and critical revision of the manuscript. M.E.: study concept and design, interpretation of the results, and writing the manuscript. ADNI provided all data used for this study. ## Acknowledgement Data used in preparation of this manuscript were obtained from the ADNI database (adni. loni.usc.edu). As such, the investigators within the ADNI study contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this paper. A complete listing of ADNI investigators can be found at: [http://adni.loni.usc.edu/wp-content/uploads/how\_to\_apply/ADNI\_Acknowledgement\_List](http://adni.loni.usc.edu/wp-content/uploads/how\_to_apply/ADNI_Acknowledgement_List). The study was funded by DAAD post-doc fellowship (to JN), grants from the Alzheimer Forschung Initiative (AFI, Grant 15035 to ME), Legerlotz Stiftung (to ME), LMUexcellent (to ME), Hertie Foundation for Clinical Neurosciences (to NF), LMU Förderung Forschung Lehre (Reg. 1032 to NF), European Union’s Horizon 2020 research and innovation programme (grant agreement No. 666881 [SVDs@target] and 667375 [CoSTREAM]; to MD) the DFG as part of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy – ID 390857198) and the CRC 1123 (B3) to MD). M.B. received speaker honoraria from GE healthcare and LMI and is an advisor of LMI. ADNI data collection and sharing for this project was funded by the ADNI (National Institutes of Health Grant U01 AG024904) and DOD ADNI (Department of Defense award number W81XWH-12-2-0012). ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging, and Bioengineering, and through contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Phar-maceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Phar-maceutical Research & Development LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Takeda Pharma-ceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health ([www.fnih.org](http://www.fnih.org)). * Received July 29, 2020. * Revision received July 29, 2020. * Accepted July 31, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. Kuro-o, M. et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390, 45–51 (1997). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/36285&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9363890&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997YE47700045&link_type=ISI) 2. Zhu, Z. et al. Klotho gene polymorphisms are associated with healthy aging and longevity: Evidence from a meta-analysis. Mech. Ageing. Dev. 178, 33–40 (2019). 3. Kuro-o, M. The Klotho proteins in health and disease. Nat. Rev. Nephrol. 15, 27–44 (2019). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30455427&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 4. Kurosu, H. et al. Suppression of aging in mice by the hormone Klotho. Science 309, 1829–1833 (2005). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzMDkvNTc0Mi8xODI5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDcvMzEvMjAyMC4wNy4yOS4yMDE2NDQzNC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 5. Dubal, D. B. et al. Life extension factor klotho enhances cognition. Cell Rep. 7, 1065–1076 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.celrep.2014.03.076&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24813892&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000336495700017&link_type=ISI) 6. Arking, D. E., Atzmon, G., Arking, A., Barzilai, N. & Dietz, H. C. Association between a functional variant of the KLOTHO gene and high-density lipoprotein cholesterol, blood pressure, stroke, and longevity. Circ. Res. 96, 412–418 (2005). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNpcmNyZXNhaGEiO3M6NToicmVzaWQiO3M6ODoiOTYvNC80MTIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNy8zMS8yMDIwLjA3LjI5LjIwMTY0NDM0LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 7. Mengel-From, J. et al. Genetic variants in KLOTHO associate with cognitive function in the oldest old group. J. Gerontol. A Biol. Sci. 71, 1151–1159 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/gerona/glv163&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26405063&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 8. Yokoyama, J. S. et al. Variation in longevity gene KLOTHO is associated with greater cortical volumes. Ann. Clin. Transl. Neur. 2, 215–230 (2015). 9. Deary, I. J. et al. KLOTHO genotype and cognitive ability in childhood and old age in the same individuals. Neurosc. Lett. 378, 22–27 (2005). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neulet.2004.12.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15763166&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000227863400005&link_type=ISI) 10. de Vries, C. F. et al. Klotho, APOEε4, cognitive ability, brain size, atrophy, and survival: a study in the Aberdeen Birth Cohort of 1936. Neurobiol. Aging 55, 91–98 (2017). 11. Zeng, C.-Y. et al. Lentiviral vector–mediated overexpression of Klotho in the brain improves Alzheimer’s disease–like pathology and cognitive deficits in mice. Neurobiol. Aging 78, 18–28 (2019). 12. Barker, W. W. et al. Relative frequencies of Alzheimer disease, Lewy body, vascular and frontotemporal dementia, and hippocampal sclerosis in the State of Florida Brain Bank. Alzheimer Dis. Assoc. Disord. 16, 203–212 (2002). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00002093-200210000-00001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12468894&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000179640800001&link_type=ISI) 13. Belloy, M. E. et al. Association of Klotho-VS Heterozygosity With Risk of Alzheimer Disease in Individuals Who Carry APOE4. JAMA Neurol. 77, 849–862 (2020). 14. Jansen, W. J. et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA 313, 1924–1938 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2015.4668&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25988462&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 15. Corder, E. H. et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261, 921–923 (1993). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIyNjEvNTEyMy85MjEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNy8zMS8yMDIwLjA3LjI5LjIwMTY0NDM0LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 16. Erickson, C. M. et al. KLOTHO heterozygosity attenuates APOE4-related amyloid burden in preclinical AD. Neurology 92, e1878–e1889 (2019). 17. Bejanin, A. et al. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 140, 3286–3300 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awx243&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29053874&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 18. Bateman, R. J. et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. NEJM 367, 795–804 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1202753&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22784036&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000308067400001&link_type=ISI) 19. Mattsson-Carlgren, N. et al. Abeta deposition is associated with increases in soluble and phosphorylated tau that precede a positive Tau PET in Alzheimer’s disease. Sci Adv 6, eaaz2387 (2020). [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo4OiJhZHZhbmNlcyI7czo1OiJyZXNpZCI7czoxMzoiNi8xNi9lYWF6MjM4NyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA3LzMxLzIwMjAuMDcuMjkuMjAxNjQ0MzQuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 20. Schöll, M. et al. PET imaging of tau deposition in the aging human brain. Neuron 89, 971–982 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuron.2016.01.028&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26938442&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 21. Harrison, T. M. et al. Longitudinal tau accumulation and atrophy in aging and alzheimer disease. Ann. Neurol. 85, 229–240 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.25406&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30597624&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 22. LaPoint, M. R. et al. The association between tau PET and retrospective cortical thinning in clinically normal elderly. Neuroimage 157, 612–622 (2017). 23. Hanseeuw, B. J. et al. Fluorodeoxyglucose metabolism associated with tau-amyloid interaction predicts memory decline. Ann. Neurol. 81, 583–596 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.24910&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28253546&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 24. Johnson, K. A. et al. Tau positron emission tomographic imaging in aging and early A lzheimer disease. Ann. Neurol 79, 110–119 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.24546&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26505746&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 25. Ossenkoppele, R. et al. Associations between tau, Aβ, and cortical thickness with cognition in Alzheimer disease. Neurology 92, e601–e612 (2019). 26. Aschenbrenner, A. J., Gordon, B. A., Benzinger, T. L., Morris, J. C. & Hassenstab, J. J. Influence of tau PET, amyloid PET, and hippocampal volume on cognition in Alzheimer disease. Neurology 91, e859–e866 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.0000000000006075&link_type=DOI) 27. La Joie, R. et al. Prospective longitudinal atrophy in Alzheimer’s disease correlates with the intensity and topography of baseline tau-PET. Sci Transl Med 12, eaau5732 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE1OiIxMi81MjQvZWFhdTU3MzIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wNy8zMS8yMDIwLjA3LjI5LjIwMTY0NDM0LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 28. Kuang, X. et al. Klotho upregulation contributes to the neuroprotection of ligustilide in an Alzheimer’s disease mouse model. Neurobiol. Aging. 35, 169–178 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neurobiolaging.2013.07.019&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23973442&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000326898400018&link_type=ISI) 29. Dubal, D. B. et al. Life extension factor klotho prevents mortality and enhances cognition in hAPP transgenic mice. J. Neurosci. 35, 2358–2371 (2015). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjk6IjM1LzYvMjM1OCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA3LzMxLzIwMjAuMDcuMjkuMjAxNjQ0MzQuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 30. Weiner, M. W. et al. The Alzheimer’s Disease Neuroimaging Initiative 3: Continued innovation for clinical trial improvement. Alzheimers Dement. 13, 561–571 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2016.10.006&link_type=DOI) 31. Hawrylycz, M. J. et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature 489, 391–399 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature11405&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22996553&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000308860900037&link_type=ISI) 32. Hawrylycz, M. et al. Canonical genetic signatures of the adult human brain. Nat. Neurosc. 18, 1832–1844 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nn.4171&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26571460&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 33. Pontecorvo, M. J. et al. Relationships between flortaucipir PET tau binding and amyloid burden, clinical diagnosis, age and cognition. Brain 140, 748–763 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/aww334&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28077397&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 34. Lockhart, S. N. et al. Amyloid and tau PET demonstrate region-specific associations in normal older people. Neuroimage 150, 191–199 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuroimage.2017.02.051&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28232190&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 35. Sepulcre, J. et al. In vivo tau, amyloid, and gray matter profiles in the aging brain. J. Neurosc. 36, 7364–7374 (2016). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjEwOiIzNi8yOC83MzY0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDcvMzEvMjAyMC4wNy4yOS4yMDE2NDQzNC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 36. French, L. & Paus, T. A FreeSurfer view of the cortical transcriptome generated from the Allen Human Brain Atlas. Front. Neurosc. 9, 323 (2015). 37. Crane, P. K. et al. Development and assessment of a composite score for memory in the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Brain Imaging Behav. 6, 502–516 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11682-012-9186-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22782295&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 38. Dumitrescu, L. et al. Genetic Variants and Functional Pathways Associated with Resilience to Alzheimer’s Disease. bioRxiv, 2020.2002.2019.954651, doi:10.1101/2020.02.19.954651 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4wMi4xOS45NTQ2NTF2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA3LzMxLzIwMjAuMDcuMjkuMjAxNjQ0MzQuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 39. Franzmeier, N. et al. The BDNF Val66Met SNP modulates the association between beta-amyloid and hippocampal disconnection in Alzheimer’s disease. Mol. Psychiatr., (2019). 40. Hohman, T. J., Dumitrescu, L., Cox, N. J. & Jefferson, A. L. Genetic resilience to amyloid related cognitive decline. Brain Imaging Behav. 11, 401–409 (2017). 41. Kleineidam, L. et al. PLCG2 protective variant p. P522R modulates Tau pathology and disease progression in patients with mild cognitive impairment. Acta Neuropathol. 139, 1025–1044 (2020). 42. Sato, C. et al. Tau Kinetics in Neurons and the Human Central Nervous System. Neuron 98, 861–864 (2018). 43. Porter, T. et al. Klotho allele status is not associated with Abeta and APOE epsilon4-related cognitive decline in preclinical Alzheimer’s disease. Neurobiol. Aging 76, 162–165 (2019). 44. Urakawa, I. et al. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature 444, 770–774 (2006). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature05315&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17086194&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000242581100063&link_type=ISI) 45. Zeldich, E. et al. The neuroprotective effect of Klotho is mediated via regulation of members of the redox system. J. Biol. 289, 24700–24715 (2014). 46. Chang, Q. et al. The b-Glucuronidase Klotho Hydrolyzes and Activates the TRPV5 Channel. Science 21, 490-493 (2005) 47. Fernandez, A. F. et al. Disruption of the beclin 1-BCL2 autophagy regulatory complex promotes longevity in mice. Nature 558, 136–140 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-018-0162-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29849149&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 48. Uddin, M. S. et al. Autophagy and Alzheimer’s Disease: From Molecular Mechanisms to Therapeutic Implications. Front. Aging Neurosc.i 10, 04 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fnagi.2018.00004&link_type=DOI) 49. Kuang, X. et al. Neuroprotective effect of ligustilide through induction of α-secretase processing of both APP and Klotho in a mouse model of Alzheimer’s disease. Front. Aaging Neurosc. 9, 353 (2017). 50. Semba, R. D. et al. Klotho in the cerebrospinal fluid of adults with and without Alzheimer’s disease. Neurosc. Lett. 558, 37–40 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neulet.2013.10.058&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24211693&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 51. Rhee, E. et al. Relationship between polymorphisms G395A in promoter and C1818T in exon 4 of the KLOTHO gene with glucose metabolism and cardiovascular risk factors in Korean women. J. Endocrinol. Invest. 29, 613–618 (2006). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF03344160&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16957409&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000240667700006&link_type=ISI) 52. Yamazaki, Y. et al. Establishment of sandwich ELISA for soluble alpha-Klotho measurement: Age-dependent change of soluble alpha-Klotho levels in healthy subjects. Biochem. Biophys. Res. Commun. 398, 513–518 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.bbrc.2010.06.110&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20599764&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000280867400031&link_type=ISI) 53. Yokoyama, J. S. et al. Systemic klotho is associated with KLOTHO variation and predicts intrinsic cortical connectivity in healthy human aging. Brain Imaging Behav. 11, 391–400 (2017). 54. Bachurin, S. O., Bovina, E. V. & Ustyugov, A. A. Drugs in clinical trials for Alzheimer’s disease: the major trends. Medicinal research reviews 37, 1186–1225 (2017). 55. Cole, M. A. & Seabrook, G. R. On the horizon—the value and promise of the global pipeline of Alzheimer’s disease therapeutics. Alzheimers Dement. 6, e12009 (2020).. 56. Desikan, R. S. et al. An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest. Neuroimage 31, 968–980 (2006). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuroimage.2006.01.021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16530430&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000238358500003&link_type=ISI) 57. Landau, S. M. et al. Amyloid deposition, hypometabolism, and longitudinal cognitive decline. Ann. Neurol. 72, 578–586 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.23650&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23109153&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 58. Maass, A. et al. Comparison of multiple tau-PET measures as biomarkers in aging and Alzheimer’s disease. Neuroimage 157, 448–463 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuroimage.2017.05.058&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28587897&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 59. Marquié, M. et al. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann. Neurol. 78, 787–800 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.24517&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26344059&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 60. Grothe, M. J. et al. Molecular properties underlying regional vulnerability to Alzheimer’s disease pathology. Brain 141, 2755–2771 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awy189&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30016411&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F07%2F31%2F2020.07.29.20164434.atom) 61. Franzmeier, N., Rubinski, A., Neitzel, J. & Ewers, M. The BIN1 rs744373 SNP is associated with increased tau-PET levels and impaired memory. Nat. Commun. 10, 1766 (2019).