Unmasking selective path integration deficits in Alzheimer's disease risk carriers

Anne Bierbrauer, Lukas Kunz, Carlos A Gomes, Maike Luhmann, Lorena Deuker, Stephan Getzmann, Edmund Wascher, Patrick D Gajewski, Jan G Hengstler, Marina Fernandez-Alvarez, Mercedes Atienza, Davide M Cammisuli, Francesco Bonatti, Carlo Pruneti, Antonio Percesepe, Youssef Bellaali, Bernard Hanseeuw, Bryan A Strange, Jose L Cantero, Nikolai Axmacher, Anne Bierbrauer, Lukas Kunz, Carlos A Gomes, Maike Luhmann, Lorena Deuker, Stephan Getzmann, Edmund Wascher, Patrick D Gajewski, Jan G Hengstler, Marina Fernandez-Alvarez, Mercedes Atienza, Davide M Cammisuli, Francesco Bonatti, Carlo Pruneti, Antonio Percesepe, Youssef Bellaali, Bernard Hanseeuw, Bryan A Strange, Jose L Cantero, Nikolai Axmacher

Abstract

Alzheimer's disease (AD) manifests with progressive memory loss and spatial disorientation. Neuropathological studies suggest early AD pathology in the entorhinal cortex (EC) of young adults at genetic risk for AD (APOE ε4-carriers). Because the EC harbors grid cells, a likely neural substrate of path integration (PI), we examined PI performance in APOE ε4-carriers during a virtual navigation task. We report a selective impairment in APOE ε4-carriers specifically when recruitment of compensatory navigational strategies via supportive spatial cues was disabled. A separate fMRI study revealed that PI performance was associated with the strength of entorhinal grid-like representations when no compensatory strategies were available, suggesting grid cell dysfunction as a mechanistic explanation for PI deficits in APOE ε4-carriers. Furthermore, posterior cingulate/retrosplenial cortex was involved in the recruitment of compensatory navigational strategies via supportive spatial cues. Our results provide evidence for selective PI deficits in AD risk carriers, decades before potential disease onset.

Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).

Figures

Fig. 1. Experimental paradigm.
Fig. 1. Experimental paradigm.
(A) Participants performed a novel PI task (the Apple Game) in a virtual environment. The task comprised three subtasks that differed with regard to the presence or absence of supportive spatial cues: the PPI subtask without any supportive cue, the BPI subtask with a circular boundary, and the LPI subtask with an intramaze landmark (lighthouse) close to the center of the environment. (B) In each trial, participants collected a basket (start phase) and tried to remember its location (goal location). After navigating toward a variable number of trees (1 to 5; outgoing phase), which disappeared after having been reached, participants had to find their way back to the goal location (incoming phase). Last, they received feedback via different numbers of stars, depending on response accuracy. (C) Outgoing distance refers to the cumulated distance during the outgoing phase, and incoming distance refers to the Euclidean distance between retrieval location (tree with apple) and goal location (basket). (D) PI performance was assessed as the distance between the correct goal location and the response location (drop error). The drop error can be separated into the distance error (i.e., the difference between the retrieval-to-goal distance and the retrieval-to-response distance) and the rotation error (i.e., the difference between the retrieval-to-goal rotation and the retrieval-to-response rotation). (E) The behavioral task comprised 8 practice trials followed by 16 trials in each subtask (short version; in the long version, all subtasks were performed twice, resulting in 32 trials in each subtask). (F) The fMRI task consisted of up to nine practice trials during the structural scan, followed by two functional runs with six blocks of four trials each. See also fig. S1 and movie S1.
Fig. 2. Performance as a function of…
Fig. 2. Performance as a function of genotype, distances, and subtask.
(A) Performance (which is inversely related to drop error) is specifically impaired in risk carriers when no supportive spatial cues are available, i.e., in the PPI subtask. (B) Risk carriers benefit more from environmental landmarks and boundaries than controls. (A) and (B) depict results from model 1b; results from model 1a are statistically equivalent. (C) Incoming (but not outgoing) distance is more closely related to spatial memory performance in risk carriers than controls (model 1c). (D) Goal-to-landmark distance (but not goal-to-boundary distance) is more relevant in risk carriers than controls (models 2a and 2b). (E) Movement-to-landmark distance (but not movement-to-boundary distance) is significantly lower in risk carriers than controls (models 2c and 2d). Y axes show parameter estimates resulting from the different models; error bars, SEM; *P < 0.05, **P < 0.01, and ***P < 0.001. Control, APOE ε3/ε3-carriers; Risk, APOE ε3/ε4-carriers; PI, path integration; BPI, boundary-supported PI; LPI, landmark-supported PI; vm, virtual meters. See also figs. S1 to S3 and tables S1 and S2.
Fig. 3. Performance as a function of…
Fig. 3. Performance as a function of genotype, age, incoming distance, and EC gray matter volume.
(A) EC gray matter volume predicts performance only during PI with long incoming distances (middle to right panels) and only in risk carriers (model 3b). (B) In younger risk carriers, EC gray matter volume predicted performance during PI trials with long incoming distances. In older risk carriers, EC gray matter volume predicted performance during the majority of all trials (model 3b). The young group comprises subjects aged 18 to 28 years; the older group contains subjects aged 53 to 75 years (see age histogram in fig. S1C). As the models contained two continuous predictors, one of them (incoming distance) was discretized into quintiles for post hoc tests and graphical depiction. Thicker lines mark slopes that are significantly different from zero. Y axes show parameter estimates resulting from the different models; shaded areas, SEM. Control, APOE ε3/ε3-carriers; Risk, APOE ε3/ε4-carriers; % volume, percent of whole-brain volume. See also fig. S1 and tables S1 and S3.
Fig. 4. Performance as a function of…
Fig. 4. Performance as a function of genotype, distances, and subtask split by age groups.
The younger age group comprises subjects aged 18 to 41 (n = 163), and the older age group comprises subjects aged 42 to 75 (n = 104). (A) Performance (which is inversely related to drop error) is specifically impaired in older risk carriers when no supportive spatial cues are available, i.e., in the PPI subtask. (B) In older participants, risk carriers benefit more from environmental landmarks and boundaries than controls. (A) and (B) depict results from model 1b; results from model 1a are statistically equivalent. (C) In both age groups, neither incoming nor outgoing distance is more closely related to spatial memory performance in risk carriers than controls. (D) In the younger age group, goal-to-boundary distance is more relevant in risk carriers than in controls (models 2a and 2b). (E) In younger participants, movement-to-landmark distance (but not movement-to-boundary distance) is significantly lower in risk carriers than in controls (models 2c and 2d). Y axes show parameter estimates resulting from the different models; error bars, SEM. +P < 0.10, *P < 0.05, **P < 0.01, and ***P < 0.001. Control, APOE ε3/ε3-carriers; Risk, APOE ε3/ε4-carriers. See also figs. S1 to S3 and tables S1, S2, and S4.
Fig. 5. fMRI results: Neural determinants of…
Fig. 5. fMRI results: Neural determinants of integrated path, goal proximity, and subtask.
(A) Representation of integrated path in EC and HC: Deactivation during navigation at longer integrated paths during outgoing phase and activation during incoming phase. (B) Representation of goal proximity: Activation for navigation closer to the goal in HC in both trial phases and in EC during the outgoing phase. (C) Subtask dependence of integrated path representations: HC deactivation during navigation at longer integrated paths during the outgoing phase was modulated by subtask. During the incoming phase, we observed a statistical trend for a similar result in EC and HC. (D) Significantly higher activity in PC/RSC during the LPI as compared to the PPI subtask. (E) Confirmatory whole-brain analyses: (left) activation of visual areas during the BPI as compared to the PPI subtask and (right) higher PC/RSC activity in the LPI than in the PPI subtask. P values are FDR-corrected for the number of contrasts and for the number of ROIs in all ROI analyses (A, B, and D). For post hoc comparisons between subtasks in (C), P values are FDR-corrected for number of subtasks. *P < 0.05, **P < 0.01, and ***P < 0.001. Error bars (A to D), SEM. Statistical parametric maps in (E) are thresholded at P < 0.05, family-wise error (FWE)–corrected for whole brain (left) and small volume–corrected for PC/RSC (middle and right), and clusters are considered significant at P < 0.05, FWE-corrected. FDR, false discovery rate; a.u., arbitrary units. See also figs. S4 and S5 and table S5.
Fig. 6. Grid-like representations in pmEC.
Fig. 6. Grid-like representations in pmEC.
(A) Left: Schematic depiction of angular differences in 360° space (inner numbers) and in 60° space (outer numbers). We expected higher pattern similarity for angular differences of mod(α,60°) = 0° (rose, where α is the angular difference between two movement directions) as compared to angular differences of mod(α,60°) = 30° (gray). We expected the same result when excluding pattern similarities of the same heading direction (dark rose). Right: Movement directions of two exemplary fMRI volumes (blue arrows). In the first volume, the subject navigates at an angle of 0° with respect to the reference axis. In the second volume, the subject navigates at an angle of 30°. This results in an angular difference of 30° (in 360° and 60° spaces) and thus mod(α,60°) = 30° (compare to blue arrows on the left). (B) In bilateral and in right pmEC, pattern similarity for angular differences of mod(α,60°) = 0° was significantly higher than pattern similarity for angular differences of mod(α,60°) = 30°, suggesting a hexadirectional symmetry of pattern similarities (dark blue bars). Same result when removing movements with similar heading directions in 360° space (light blue bars). (C) Control analyses. Left: No evidence for fourfold, fivefold, sevenfold, or eightfold rotational symmetry of pattern similarity in bilateral or right pmEC. Right: No evidence for sixfold rotational symmetry of pattern similarity in alEC, HC, and PC/RSC. Error bars (B and C), SEM. *P < 0.05. mod, modulus; dir., direction; pmEC, posterior-medial entorhinal cortex; alEC, anterior-lateral entorhinal cortex; HC, hippocampus.
Fig. 7. Mechanistic model to predict PI…
Fig. 7. Mechanistic model to predict PI performance.
We aimed at predicting PI performance as a function of fMRI-based representations of spatial features in combination with subtask and incoming distance (model 6). (A) Integrated path representations in EC interacted with incoming distance in predicting PI performance: At higher incoming distances, stronger integrated path representations in EC were associated with better performance. (B) Goal proximity representations in HC interacted with subtask in predicting PI performance. In none of the subtasks was the prediction significant by itself. (C) GLRs in pmEC interacted with subtask in predicting PI performance: Only in PPI, higher GLRs in pmEC were associated with better performance. (D) Landmark representations in PC/RSC interacted with subtask and incoming distance in predicting PI performance. In none of the individual subtask by incoming distance combinations was the prediction significant by itself. As the model contained two continuous predictors, one of them (incoming distance) was discretized into quintiles for post hoc tests and for graphical depiction; only quintiles 1 and 5 are depicted (A and D). Y axes show parameter estimates for performance; shaded areas, SEM.

References

    1. Coughlan G., Laczó J., Hort J., Minihane A.-M., Hornberger M., Spatial navigation deficits—Overlooked cognitive marker for preclinical Alzheimer disease? Nat. Rev. Neurol. 14, 496–506 (2018).
    1. Sperling R. A., Jack C. R. Jr., Aisen P. S., Testing the right target and right drug at the right stage. Sci. Transl. Med. 3, 111cm33 (2011).
    1. Corder E. H., Saunders A. M., Strittmatter W. J., Schmechel D. E., Gaskell P. C., Small G. W., Roses A. D., Haines J. L., Pericak-Vance M. A., Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261, 921–923 (1993).
    1. Kunz L., Schröder T. N., Lee H., Montag C., Lachmann B., Sariyska R., Reuter M., Stirnberg R., Stöcker T., Messing-Floeter P. C., Fell J., Doeller C. F., Axmacher N., Reduced grid-cell–like representations in adults at genetic risk for Alzheimer’s disease. Science 350, 430–433 (2015).
    1. Coughlan G., Coutrot A., Khondoker M., Minihane A.-M., Spiers H., Hornberger M., Toward personalized cognitive diagnostics of at-genetic-risk Alzheimer’s disease. Proc. Natl. Acad. Sci. U.S.A. 116, 9285–9292 (2019).
    1. Mondadori C. R. A., de Quervain D. J.-F., Buchmann A., Mustovic H., Wollmer M. A., Schmidt C. F., Boesiger P., Hock C., Nitsch R. M., Papassotiropoulos A., Henke K., Better memory and neural efficiency in young apolipoprotein E ε4 carriers. Cereb. Cortex 17, 1934–1947 (2007).
    1. Weissberger G. H., Nation D. A., Nguyen C. P., Bondi M. W., Han S. D., Meta-analysis of cognitive ability differences by apolipoprotein E genotype in young humans. Neurosci. Biobehav. Rev. 94, 49–58 (2018).
    1. Ghebremedhin E., Schultz C., Braak E., Braak H., High frequency of apolipoprotein E ϵ4 allele in young individuals with very mild Alzheimer’s disease-related neurofibrillary changes. Exp. Neurol. 153, 152–155 (1998).
    1. Hanseeuw B. J., Betensky R. A., Jacobs H. I. L., Schultz A. P., Sepulcre J., Becker J. A., Cosio D. M. O., Farrell M., Quiroz Y. T., Mormino E. C., Buckley R. F., Papp K. V., Amariglio R. A., Dewachter I., Ivanoiu A., Huijbers W., Hedden T., Marshall G. A., Chhatwal J. P., Rentz D. M., Sperling R. A., Johnson K., Association of amyloid and tau with cognition in preclinical Alzheimer disease: A longitudinal study. JAMA Neurol. 76, 915–924 (2019).
    1. Braak H., Thal D. R., Ghebremedhin E., Del Tredici K., Stages of the pathologic process in Alzheimer disease: Age categories from 1 to 100 years. J. Neuropathol. Exp. Neurol. 70, 960–969 (2011).
    1. Hafting T., Fyhn M., Molden S., Moser M.-B., Moser E. I., Microstructure of a spatial map in the entorhinal cortex. Nature 436, 801–806 (2005).
    1. Fu H., Rodriguez G. A., Herman M., Emrani S., Nahmani E., Barrett G., Figueroa H. Y., Goldberg E., Hussaini S. A., Duff K. E., Tau pathology induces excitatory neuron loss, grid cell dysfunction, and spatial memory deficits reminiscent of early Alzheimer’s disease. Neuron 93, 533–541.e5 (2017).
    1. Doeller C. F., Barry C., Burgess N., Evidence for grid cells in a human memory network. Nature 463, 657–661 (2010).
    1. Bellmund J. L. S., Deuker L., Schröder T. N., Doeller C. F., Grid-cell representations in mental simulation. eLife 5, e17089 (2016).
    1. Kunz L., Maidenbaum S., Chen D., Wang L., Jacobs J., Axmacher N., Mesoscopic neural representations in spatial navigation. Trend Cogn. Sci. 23, 615–630 (2019).
    1. Stangl M., Achtzehn J., Huber K., Dietrich C., Tempelmann C., Wolbers T., Compromised grid-cell-like representations in old age as a key mechanism to explain age-related navigational deficits. Curr. Biol. 28, 1108–1115.e6 (2018).
    1. Hardcastle K., Ganguli S., Giocomo L. M., Environmental boundaries as an error correction mechanism for grid cells. Neuron 86, 827–839 (2015).
    1. Burak Y., Fiete I. R., Accurate path integration in continuous attractor network models of grid cells. PLOS Comput. Biol. 5, e1000291 (2009).
    1. Gil M., Ancau M., Schlesiger M. I., Neitz A., Allen K., De Marco R. J., Monyer H., Impaired path integration in mice with disrupted grid cell firing. Nat. Neurosci. 21, 81–91 (2018).
    1. Howard L. R., Javadi A. H., Yu Y., Mill R. D., Morrison L. C., Knight R., Loftus M. M., Staskute L., Spiers H. J., The hippocampus and entorhinal cortex encode the path and Euclidean distances to goals during navigation. Curr. Biol. 24, 1331–1340 (2014).
    1. Epstein R. A., Patai E. Z., Julian J. B., Spiers H. J., The cognitive map in humans: Spatial navigation and beyond. Nat. Neurosci. 20, 1504–1513 (2017).
    1. Mitchell A. S., Czajkowski R., Zhang N., Jeffery K., Nelson A. J. D., Retrosplenial cortex and its role in spatial cognition. Brain Neurosci. Adv. 2, 2398212818757098 (2018).
    1. Folstein M. F., Folstein S. E., McHugh P. R., “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J. Psychiatr. Res. 12, 189–198 (1975).
    1. Miller J., Watrous A. J., Tsitsiklis M., Lee S. A., Sheth S. A., Schevon C. A., Smith E. H., Sperling M. R., Sharan A., Asadi-Pooya A. A., Worrell G. A., Meisenhelter S., Inman C. S., Davis K. A., Lega B., Wanda P. A., Das S. R., Stein J. M., Gorniak R., Jacobs J., Lateralized hippocampal oscillations underlie distinct aspects of human spatial memory and navigation. Nat. Commun. 9, 2423 (2018).
    1. Matura S., Prvulovic D., Jurcoane A., Hartmann D., Miller J., Scheibe M., O’Dwyer L., Oertel-Knöchel V., Knöchel C., Reinke B., Karakaya T., Fußer F., Pantel J., Differential effects of the ApoE4 genotype on brain structure and function. Neuroimage 89, 81–91 (2014).
    1. Shine J. P., Valdés-Herrera J. P., Hegarty M., Wolbers T., The human retrosplenial cortex and thalamus code head direction in a global reference frame. J. Neurosci. 36, 6371–6381 (2016).
    1. Destrieux C., Fischl B., Dale A., Halgren E., Automatic parcellation of human cortical gyri and sulci using standard anatomical nomenclature. Neuroimage 53, 1–15 (2010).
    1. Navarro Schröder T., Haak K. V., Jimenez N. I. Z., Beckmann C. F., Doeller C. F., Functional topography of the human entorhinal cortex. eLife 4, e06738 (2015).
    1. Coutrot A., Silva R., Manley E., de Cothi W., Sami S., Bohbot V. D., Wiener J. M., Hölscher C., Dalton R. C., Hornberger M., Spiers H. J., Global determinants of navigation ability. Curr. Biol. 28, 2861–2866.e4 (2018).
    1. Marchette S. A., Vass L. K., Ryan J., Epstein R. A., Anchoring the neural compass: Coding of local spatial reference frames in human medial parietal lobe. Nat. Neurosci. 17, 1598–1606 (2014).
    1. Thal D. R., Rüb U., Orantes M., Braak H., Phases of Aβ-deposition in the human brain and its relevance for the development of AD. Neurology 58, 1791–1800 (2002).
    1. Braak H., Alafuzoff I., Arzberger T., Kretzschmar H., Del Tredici K., Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 112, 389–404 (2006).
    1. Jagust W., Imaging the evolution and pathophysiology of Alzheimer disease. Nat. Rev. Neurosci. 19, 687–700 (2018).
    1. Huijbers W., Schultz A. P., Papp K. V., La Point M. R., Hanseeuw B., Chhatwal J. P., Hedden T., Johnson K. A., Sperling R. A., Tau accumulation in clinically normal older adults is associated with hippocampal hyperactivity. J. Neurosci. 39, 548–556 (2019).
    1. Bejanin A., Schonhaut D. R., La Joie R., Kramer J. H., Baker S. L., Sosa N., Ayakta N., Cantwell A., Janabi M., Lauriola M., O’Neil J. P., Gorno-Tempini M. L., Miller Z. A., Rosen H. J., Miller B. L., Jagust W. J., Rabinovici G. D., Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 140, 3286–3300 (2017).
    1. Howett D., Castegnaro A., Krzywicka K., Hagman J., Marchment D., Henson R., Rio M., King J. A., Burgess N., Chan D., Differentiation of mild cognitive impairment using an entorhinal cortex-based test of virtual reality navigation. Brain 142, 1751–1766 (2019).
    1. Ewers M., Sperling R. A., Klunk W. E., Weiner M. W., Hampel H., Neuroimaging markers for the prediction and early diagnosis of Alzheimer’s disease dementia. Trends Neurosci. 34, 430–442 (2011).
    1. Jagust W. J., Mormino E. C., Lifespan brain activity, β-amyloid, and Alzheimer’s disease. Trends Cogn. Sci. 15, 520–526 (2011).
    1. Prieto del Val L., Cantero J. L., Atienza M., APOE ε4 constrains engagement of encoding-related compensatory networks in amnestic mild cognitive impairment. Hippocampus 25, 993–1007 (2015).
    1. Bakker A., Krauss G. L., Albert M. S., Speck C. L., Jones L. R., Stark C. E., Yassa M. A., Bassett S. S., Shelton A. L., Gallagher M., Reduction of hippocampal hyperactivity improves cognition in amnestic mild cognitive impairment. Neuron 74, 467–474 (2012).
    1. Palop J. J., Mucke L., Network abnormalities and interneuron dysfunction in Alzheimer disease. Nat. Rev. Neurosci. 17, 777–792 (2016).
    1. Alzheimer’s Association , 2015 Alzheimer’s disease facts and figures. Alzheimers Dement. 11, 332–384 (2015).
    1. Hoenig M. C., Bischof G. N., Onur Ö. A., Kukolja J., Jessen F., Fliessbach K., Neumaier B., Fink G. R., Kalbe E., Drzezga A., van Eimeren T.; Alzheimer’s Disease Neuroimaging Initiative , Level of education mitigates the impact of tau pathology on neuronal function. Eur. J. Nucl. Med. Mol. Imaging 46, 1787–1795 (2019).
    1. Bécu M., Sheynikhovich D., Tatur G., Agathos C. P., Bologna L. L., Sahel J.-A., Arleo A., Age-related preference for geometric spatial cues during real-world navigation. Nat. Hum. Behav. 4, 88–99 (2020).
    1. Malkki H., Alzheimer disease: Effects of the APOE ε4 allele on brain development. Nat. Rev. Neurol. 10, 4 (2014).
    1. Heffernan A. L., Chidgey C., Peng P., Masters C. L., Roberts B. R., The neurobiology and age-related prevalence of the ε4 allele of apolipoprotein E in Alzheimer’s disease cohorts. J. Mol. Neurosci. 60, 316–324 (2016).
    1. Berens P., CircStat: A MATLAB toolbox for circular statistics. J. Stat. Softw. 31, 1–21 (2009).
    1. R Core Team, R: A Language and Environment for Statistical Computing (2018).
    1. Bates D., Mächler M., Bolker B. M., Walker S., Fitting linear mixed-effects models using lme4. J. Stat. Softw. 67, 1–48 (2014).
    1. R. Lenth, emmeans: Estimated Marginal Means, aka Least-Squares Means (2019).
    1. Hegarty M., Richardson A. E., Montello D. R., Lovelace K., Subbiah I., Development of a self-report measure of environmental spatial ability. Intelligence 30, 425–447 (2002).
    1. Langsrud Ø., ANOVA for unbalanced data: Use Type II instead of Type III sums of squares. Stat. Comput. 13, 163–167 (2003).
    1. Fischl B., van der Kouwe A., Destrieux C., Halgren E., Ségonne F., Salat D. H., Busa E., Seidman L. J., Goldstein J., Kennedy D., Caviness V., Makris N., Rosen B., Dale A. M., Automatically parcellating the human cerebral cortex. Cereb. Cortex 14, 11–22 (2004).
    1. Desikan R. S., Ségonne F., Fischl B., Quinn B. T., Dickerson B. C., Blacker D., Buckner R. L., Dale A. M., Maguire R. P., Hyman B. T., Albert M. S., Killiany R. J., An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest. Neuroimage 31, 968–980 (2006).
    1. Tombaugh T. N., McIntyre N. J., The mini-mental state examination: A comprehensive review. J. Am. Geriatr. Soc. 40, 922–935 (1992).
    1. Petersen R. C., Smith G. E., Waring S. C., Ivnik R. J., Tangalos E. G., Kokmen E., Mild cognitive impairment: Clinical characterization and outcome. Arch. Neurol. 56, 303–308 (1999).
    1. Albert M. S., DeKosky S. T., Dickson D., Dubois B., Feldman H. H., Fox N. C., Gamst A., Holtzman D. M., Jagust W. J., Petersen R. C., Snyder P. J., Carrillo M. C., Thies B., Phelps C. H., The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 7, 270–279 (2011).
    1. C. A. Helmstaedter, M. Lendth, S. Lux, Verbaler Lern- und Merkfähigkeitstest (Beltz Test, 2001).
    1. Morris J. C., The clinical dementia rating (CDR): Current version and scoring rules. Neurology 43, 2412–2414 (1993).
    1. Morris J. C., Heyman A., Mohs R. C., Hughes J. P., van Belle G., Fillenbaum G., Mellits E. D., Clark C., The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology 39, 1159–1165 (1989).
    1. A. Coutrot, E. Manley, D. Yesiltepe, R. C. Dalton, J. M. Wiener, C. Hölscher, M. Hornberger, H. J. Spiers, Cities have a negative impact on navigation ability: Evidence from 38 countries. bioRxiv 2020.01.23.917211 [Preprint]. 24 January 2020. .
    1. Bush D., Burgess N., Advantages and detection of phase coding in the absence of rhythmicity. Hippocampus 30, 745–762 (2020).
    1. Kraus B. J., Brandon M. P., Robinson R. J. II, Connerney M. A., Hasselmo M. E., Eichenbaum H., During running in place, grid cells integrate elapsed time and distance run. Neuron 88, 578–589 (2015).

Source: PubMed

3
Abonnieren