Skip to main content
Brain logoLink to Brain
. 2024 Nov 22;148(5):1577–1587. doi: 10.1093/brain/awae352

Hypometabolic mismatch with atrophy and tau pathology in mixed Alzheimer’s and Lewy body disease

Michael Tran Duong 1,2, Sandhitsu R Das 3,4, Pulkit Khandelwal 5,6, Xueying Lyu 7,8, Long Xie 9,10, Emily McGrew 11, Nadia Dehghani 12, Corey T McMillan 13,14, Edward B Lee 15,16, Leslie M Shaw 17,18, Paul A Yushkevich, Alzheimer’s Disease Neuroimaging Initiative19,20,21, David A Wolk 22,23,24,, Ilya M Nasrallah 25,26,27,
PMCID: PMC12073973  PMID: 39573823

Abstract

Polypathology is a major driver of heterogeneity in the clinical presentation and extent of neurodegeneration (N) in patients with Alzheimer’s disease (AD). Beyond amyloid (A) and tau (T) pathologies, over half of patients with AD have concomitant pathology such as α-synuclein (S) in mixed AD with Lewy body disease (LBD). Patients with multiple aetiology dementia such as AD + LBD have faster progression and potentially differential responses to targeted treatments, although the diagnosis of AD + LBD can be challenging given the overlapping clinical and imaging features. Development and validation of improved in vivo biomarkers are required to study relationships between N and S and identify imaging patterns reflecting mixed AD + LBD pathologies.

We hypothesized that individual proteinopathies, such as T and S, are associated with commensurate levels of N. Thus, we assessed biomarkers of A, T, N and S with PET, MRI and CSF seeding amplification assay (SAA) data to determine molecular presentations of mixed A+S+ versus A+S– cognitively impaired patients from the Alzheimer’s Disease Neuroimaging Initiative (ADNI).

Strikingly, A+S+ patients had parieto-occipital 18F-fluorodeoxyglucose hypometabolism (a measure of N) disproportionate to the degree of regional atrophy or T burden, highlighting worse hypometabolism associated with S+ status on SAA. Following up on this hypometabolic mismatch with CSF metabolite and proteome analyses, we found that A+S+ patients exhibited lower CSF levels of dopamine metabolites and synaptic markers like neuronal pentraxin-2 (NPTX2), suggesting that altered neurotransmission and neuron integrity contribute to this dissociation between metabolic PET and MRI. Potential confounders exist when studying relations between N, AD and LBD pathologies, including neuroinflammation and other non-Alzheimer’s pathologies in mixed dementia, although our findings imply posterior hypometabolic mismatch is related more to S than vascular or TDP-43 pathology.

Overall, A+S+ patients had posterior mismatch with excessive 18F-fluorodeoxyglucose hypometabolism relative to atrophy or T load, possibly reflecting impaired neuronal integrity. Further research must disentangle the impact of multiple proteinopathies and clinicopathologic factors on hypometabolism and atrophy. Cumulatively, patients with mixed AD + LBD aetiologies harbour a unique metabolic PET mismatch signature.

Keywords: multiple aetiology dementia, Alzheimer’s disease, Lewy body, α-synuclein, metabolism, biomarkers


By assessing imaging and CSF biomarkers of amyloid, tau, α-synuclein and neurodegeneration, Duong et al. demonstrate that patients with mixed Alzheimer’s and Lewy body disease show a signature pattern of posterior hypometabolism that is disproportionate to the degree of atrophy on MRI and tau pathology as measured by PET.

Introduction

Over half of patients with Alzheimer’s disease (AD) have concomitant non-Alzheimer’s pathologies at autopsy,1-6 including α-synuclein (S) and TDP-43. Such patients with mixed pathologies are classified as multiple aetiology dementia. AD is often an amnestic disorder characterized by amyloid-β plaques and tau-based neurofibrillary tangles (NFTs) that can be detected in vivo by amyloid (A) and tau (T) biomarkers. NFTs are more highly associated with neuronal and synaptic loss or neurodegeneration (N).7,8 Conversely, Lewy body disease (LBD) is a disorder with movement and neuropsychiatric symptoms linked to α-synuclein Lewy bodies.9,10 LBD encompasses a spectrum of neuronal α-synucleinopathies, including dementia with Lewy bodies (DLB) and Parkinson’s disease (PD) based on recent biological frameworks.11,12 Furthermore, patients with multiple aetiology dementia tend to have faster progression of cognitive presentation than patients with fewer pathologies.7 While some treatments, such as anti-cholinesterase medications, may overlap between AD and DLB,9 it is uncertain the degree to which copathology influences the efficacy of emerging anti-amyloid treatment or other therapies in clinical trials. Therefore, systematic characterization of neural biomarkers in multiple aetiology dementia is paramount to determining accurate diagnosis and treatment response.

Promising α-synuclein biomarkers are now available, including the seeding amplification assay (SAA) used to detect α-synuclein aggregates from CSF13 and peripheral skin and salivary glands.14 α-synuclein SAA has been validated in patients with PD and parkinsonian disorders,13,15,16 cognitively impaired patients (including mixed AD + LBD)17 and cognitively unimpaired older adults.16-18 SAA determines binary α-synuclein status, but does not currently provide information on regional distribution or relative load of α-synuclein pathology throughout the brain.

From the Alzheimer’s Disease Neuroimaging Initiative (ADNI) study, we assessed the neurodegeneration reflecting underlying α-synuclein SAA status to determine the systematic patterns of neurodegeneration attributable to latent α-synuclein pathology. Non-Alzheimer copathologies can be inferred by measuring cellular responses as seen by changes in neurodegeneration not adequately accounted for by AD pathology alone, particularly in multiple aetiology dementia. While tau burden is used to stage AD and is tightly linked to neurodegeneration, measured as structural atrophy (NS) or lowered metabolism (NM) in AD,19-23 we hypothesized that coexisting non-Alzheimer pathologies may independently drive neurodegeneration and thus distort the relationship between neurodegeneration and tau burden, creating a mismatch.10,24-27 Hence, comparing the observed atrophy on MRI or hypometabolism on 18F-fluorodeoxyglucose (18F-FDG) PET with the expected neurodegeneration based on the degree of AD pathology (tau PET) can highlight the impact of α-synuclein SAA status on neurodegeneration. Particularly, we aim to identify the regional hypometabolism profile attributable to SAA-based S+ status, where hypometabolism is disproportionate to the extent of tau pathology in A+S+ patients compared to A+S– patients. To this point, our group has shown in a series of studies that a mismatch of neurodegeneration relative to tau load (‘TN mismatch’) may predict the presence of non-Alzheimer’s pathologies, such as TDP-43-related markers.24,26,27 Now, we extend this work for α-synuclein SAA in mixed AD + LBD.

In addition to TNM mismatch, we explore the ‘NSNM mismatch’ between MRI and 18F-FDG PET that may occur in the setting of mixed AD + LBD. Cortical thickness and glucose metabolism on MRI and 18F-FDG PET typically decrease in tandem in AD23 but not in LBD.28,29 In fact, compared to AD, patients with LBD may not exhibit as many overall structural changes.28-30 Posterior parieto-occipital hypometabolic measures, such as the cingulate island sign on 18F-FDG PET are prototypical molecular signatures associated with LBD31-35 that are not matched by a corresponding degree of atrophy.28-30,33 Indeed, deficits in glucose metabolism seemingly precede reductions in cortical thickness and neurotransmission changes as detected by MRI28 and dopamine transporter molecular imaging,36 respectively. This convergence of evidence pointing to mismatch between MRI and 18F-FDG PET led us to explore this discordance in A+S+ patients.

Here, we analysed data from 246 cognitively impaired older adults from ADNI with amyloid, tau and SAA α-synuclein status, of which 185 had both MRI and 18F-FDG PET measures. We compared regional levels of TNM and NSNM mismatches to study the hypometabolic patterns linked with α-synuclein that are not explained by tau or structural atrophy, respectively. Initially, we found that α-synuclein markers were associated with patients with amyloid pathology. Based on the literature, we hypothesized that A+S+ patients have posterior hypometabolism disproportionate to the degree of tau or atrophy. Finally, we predicted CSF metabolomic and proteomic studies would point to potential mechanisms connecting α-synuclein to altered neurotransmission and ‘neuronal integrity,’ a term encompassing neuronal dysfunction, as well as death, and therefore representing visible and occult changes on MRI. These hypotheses could account for how hypometabolism on 18F-FDG PET may potentially precede atrophy on MRI and support our goal to identify a unique hypometabolic signature in AD + LBD.

Materials and methods

Patient cohort for biomarkers

From the ADNI cohort database (http://adni.loni.usc.edu), we included 246 cognitively impaired patients with a measure of amyloid (A), tau (T) and α-synuclein (S) status (last accessed 12/2023). We found 180 participants with a diagnosis of mild cognitive impairment (MCI), and 66 patients with dementia (Table 1). Evaluation of A status utilized amyloid PET (18F-florbetapir or 18F-florbetaben by ADNI AMYLOID_STATUS, n = 242, UCBERKELEY_AMY_6MM, accessed 1/2024) or Elecsys CSF assay37 (CSF amyloid-β42 < 980 pg/ml, n = 4, UPENNBIOMK_MASTER_FINAL_21Dec2023, accessed 1/2024). T status was determined by 18F-flortaucipir PET images, obtained within 1 year from A testing. S status was obtained via Amprion α-synuclein SAA13,38 obtained within 2 years of imaging (AMPRION_ASYN_SAA, accessed 1/2024). This range was extended to include additional patients with S– SAA results >2 years after imaging or S+ SAA results >2 years before imaging, since CSF SAA rarely converts from S+ to S– status.14,16,39 From the whole cohort, 185 patients were found to have MRI and 18F-FDG PET within 1 year of tau PET. Median time between tau versus amyloid PET was 14 days (84% of cases within 3 months). Median time between tau PET versus α-synuclein CSF SAA was 34 days (63% within 3 months). Median time between tau versus 18F-FDG PET was 13 days (91% within 3 months). For cognitive data, we selected ADNI clinical testing sessions closest to imaging for the AD Assessment Scale-Cognition 13 item (ADAS, where higher score is worse, accessed 1/2024) and Neuropsychiatric Inventory (NPI, accessed 1/2024) for patients who presently had or developed hallucinations (NPI-B).

Table 1.

Clinical characteristics of the Alzheimer's disease neuroimaging initiative cohort with amyloid, tau and α-synuclein markers

AS Group Disease (AD/LBD) MCI/Dementia Sex (F/M) Age (years) Education (years) ADAS-Cog Hallucinations (%)
A–S– (n = 81) Neither AD nor LBD 74/7 32/49 71.3 (8.4) 16.3 (2.6) 14.0 (6.3) 0%
A–S+ (n = 19) LBD without AD 16/3 4/15 78.3 (6.4) 16.3 (3.0) 16.2 (8.5) 5.3%
A+S– (n = 99) AD without LBD 70/29 44/55 75.0 (7.9) 15.8 (2.6) 20.7 (9.7) 7.1%
A+S+ (n = 47) Mixed AD + LBD 20/27 24/23 75.0 (7.6) 15.8 (2.4) 27.4 (11.8)** 10.6%

Frequencies are shown for disease severity [mild cognitive impairment (MCI)/dementia] and sex [female/male (F/M)]. Mean (and standard deviations) are shown for age, education and Alzheimer’s Disease Assessment Scale-Cognitive (ADAS-Cog). Percentages are shown for the proportion of patients with/developing hallucinations over time. Significant differences were calculated using likelihood ratio tests with the amyloid+ α-synuclein− (A+S–) group as the reference, covariates of age, sex and tau (T) burden in the inferior temporal gyrus. *P < 0.05 and **P < 0.005. LBD = Lewy body disease.

Imaging data

Post-processed PET images from the ADNI data archive (http://adni.loni.usc.edu/data-samples/access-data/) were obtained. Tau PET imaging was originally performed using the ADNI protocol with 30-min brain scans (six 5-min frames) starting 75 min after intravenous administration of ∼10.0 mCi 18F-flortaucipir. 18F-FDG PET imaging consisted of a 30-min scan (six 5-min frames) at 30 min after 5.0 mCi 18F-FDG injection. For amyloid PET, a 20-min brain scan (four 5-min frames) was performed 50 min after ∼10.0 mCi 18F-florbetapir or 90 min following ∼8.1 mCi 18F-florbetaben injection. Processed tau and 18F-FDG PET images with uniform isotropic resolution (8 mm full-width-at-half-maximum) were obtained with the ADNI archive description ‘Coreg, Avg, Std Img and Vox Size, Uniform Resolution.’ ADNI MRI included a T1-weighted structural scan (resolution 1.0 × 1.0 × 1.2 mm3) and fluid-attenuated inversion recovery (FLAIR) sequence scan acquired in the same session.

Image processing and PET regional analysis

MRI studies were processed using the ANTs pipeline40 for inhomogeneity correction, brain extraction, template registration and cortical thickness measurement.41,42 MRI scans were divided into cortical, subcortical, brainstem and cerebellar regions of interest (ROIs) with multi-atlas segmentation43,44 (http://neuromorphometrics.com/ParcellationProtocol_2010-04-05.PDF). PET images were co-registered to T1-weighted MRI with ANTs for rigid-body transformation.40 Standardized uptake value ratio (SUVR) maps were generated with reference regions specific for each tracer: inferior cerebellar cortex for 18F-flortaucipir45 and cerebellar cortex for 18F-FDG.46 Mean regional T, NM and NS measures were extracted from tau SUVR, 18F-FDG SUVR and cortical thickness maps, respectively. The cingulate island sign represents metabolic sparing of posterior cingulate cortex relative to precuneus and cuneus and has been associated with S+ status. It was quantified as the ratio of posterior cingulate/precuneus/cuneus 18F-FDG SUVR; higher cingulate island ratio is linked to LBD.32,33 Likewise, the I/MTL/FSO ratio represents sparing of inferior temporal gyrus relative to medial temporal lobe and frontal supraorbital gyrus in 18F-FDG SUVR; higher I/MTL/FSO ratio is associated with TDP-43 disease.47

Regional TNM and NSNM mismatch

Spatial patterns of TNM mismatch were investigated using a previously published residual method.25,26 Briefly, robust linear regressions were performed comparing individual 18F-FDG SUVR versus a log transform of tau SUVR or cortical thickness (in mm) across all cohort patients in each of the grey matter ROIs. A bi-square weighting function minimized the influence of outliers in robust regression. Regional mean residuals were calculated from regressions and visualized in two-dimensional maps and three-dimensional renderings by ITK-SNAP48 and MRIcroGL.49 For general 3D rendering maps, the white colour reflected a mismatch residual value close to about 0.6 standard deviations (where about half of the patients reside) from the regression line, and blue colour denoted negative mismatch residual with NM worse than expected. For significant difference renderings, white colour represented no significant difference between A+S+ and A+S– groups, and blue colour depicted a negative mismatch residual that significantly differs between groups.

CSF metabolite and proteomics analysis

Processed CSF proteomics and metabolomics results50,51 for each protein and metabolite were available from ADNI in 544 participants with A and CSF SAA-based S markers within 2 years of each other (58% within 3 months) (CruchagaLab_CSF_SOMAscan7k_Protein_matrix_postQC, CruchagaLab_CSF_metabolomic_matrix_20230620, accessed 1/2024). This cohort included A–S– (n = 161), A–S+ (n = 35), A+S– (n = 246) and A+S+ (n = 102). Metabolites were assessed by targeted hypothesis-driven analysis of dopamine metabolites (homovanillic acid and 3-methoxytyramine sulfate) per literature.52,53 We next investigated proteome differences between patients with mixed AD + LBD versus AD without LBD or LBD without AD. Groups were compared by ANOVA to establish group-wise differences (P < 0.05) followed by two-sample t-tests to assess hypothesis-driven pairwise differences between A+S+ versus A+S– and A+S+ versus A–S+ groups, with significant hits identified by a stringent false discovery rate (FDR) <0.005 threshold after Benjamini-Hochberg adjustment for multiple comparisons. Based on comparisons, there were 276 protein hits for A+S+ versus A+S– and 210 proteins for A+S+ versus A–S+ analyses, which were then input into the ontology functional database of the WebGestalt program (https://www.webgestalt.org/). Over-representation analysis was conducted for biological process and cellular compartment in the Homo sapiens protein-coding genome with FDR <0.05 after Benjamini-Hochberg adjustment and redundancy reduction by weighted set cover. Data were visualized by graphs of fold enrichment, protein count and –log10(FDR) per ontology class.

Statistical analysis

Statistical analysis was performed in R (v4.1.2). All statistical tests were two-sided. Comparisons for variables such as tau and 18F-FDG SUVRs were performed with likelihood ratio tests by linear regression. Covariates included sex, age and tau SUVR in the inferior temporal gyrus, a region where T correlates with disease severity.8,54 Multiple test adjustment by Benjamini-Hochberg correction with FDR <0.05 was conducted for pairwise comparisons of proteomics data. Relative frequency comparisons were performed with χ2 tests.

Results

Amyloid, tau and α-synuclein markers can be compared in vivo

First, we evaluated in vivo SAA-based S status and its associations with A and T pathology from an ADNI cohort of 246 cognitively impaired participants with A, T and S status. A+S+ patients had significantly worse impairment compared to A+S– patients on the ADAS-Cog after adjusting for covariates of age, sex and T burden (P = 0.003), in keeping with more severe cognitive presentation in mixed AD + LBD (Table 1). A+S+ patients trended towards a higher frequency of hallucinations.

Consistent with autopsy cohorts,3 cognitively impaired patients with SAA-based S+ status were more likely to have AD pathology on amyloid PET, although this could further be affected by the recruitment criteria for ADNI (see the ‘Discussion’ section). In our ADNI cohort, 32% of A+ patients were S+, compared to just 19% of A– patients (P = 0.022).

Combining Alzheimer’s and Lewy body disease markers highlights a metabolic mismatch signature

From our ADNI cohort of cognitively impaired participants with known A, T and S status, we assessed the dissociation between NM versus T (TNM mismatch) and NM versus NS (NSNM mismatch) in the 185 patients with available MRI and 18F-FDG PET. TNM mismatch is based on the hypothesis that the degree of molecular pathologies is linked to a commensurate level of cellular injury (Fig. 1A). Evaluating correlations across the entire sample together, greater T is associated with worse NM (Fig. 1B), while structural and metabolic measures of N are positively correlated (Fig. 1C and D). However, the degree of variability in these TNM and NSNM relationships, as measured by regression model residuals, can reflect the impact of non-Alzheimer pathology on N that is not accounted for by AD pathology alone. Here, S+ status is associated with TNM mismatch where worse NM is disproportionate to the extent of T burden, as illustrated in two example cases (Fig. 1E and F) and quantitative group analysis (Fig. 2). Moreover, different pathologies (T, S, TDP-43) may potentially impact individual neurodegenerative measures differently. For instance, imaging evidence implies that S+ status is linked more to hypometabolism on 18F-FDG PET than atrophy on MRI,28,29 therefore leading to an NSNM mismatch that we study further.

Figure 1.

Figure 1

Imaging examples highlight mismatch residuals between tau and neuronal 18F-FDG PET hypometabolism, and MRI/18F-FDG PET mismatch relationships.

(A) Mismatch between tau pathology (T) and neuronal 18F-fluorodeoxyglucose (18F-FDG) hypometabolism (NM) occurs in mixed Alzheimer’s and Lewy body disease (AD + LBD) since NM may reflect a balance of both T and α-synuclein (S) pathologies. (B) Associations between worse 18F-FDG hypometabolism and greater T burden show outliers with worse NM than expected based on T. (C) Mismatch between atrophy on MRI (NS) and NM occurs in mixed AD + LBD since S appears to impact NM on PET moreso than NS on MRI. (D) Associations between worse 18F-FDG hypometabolism and lower cortical thickness (mm) in the inferior temporal gyrus highlight outliers with worse NM than expected based on NS. Amyloid (A), T, S status, 18F-FDG PET and MRI are shown for two cognitively impaired older adults, whose residuals are highlighted in B and C. (D) A 70-year-old male with A+T+S+ markers has AD Assessment Scale-Cognitive (ADAS-Cog) score of 29 (higher is worse) and cingulate island sign, while a (F) 75-year-old female with A+T+S– markers has an ADAS-Cog score of 24 and higher metabolism, despite both having similar A and T burdens. Regression lines of best fit and standard deviation based-lines are shown.

Figure 2.

Figure 2

Imaging reveals a metabolic mismatch signature in patients with concomitant amyloid+synuclein pathology. Mean regional residual z-score maps show posterior mismatch (arrows) in amyloid+ α-synuclein+ (A+S+) patients compared with A+S– patients in [A(i)] mismatch residuals between tau and 18F-fluorodeoxyglucose (18F-FDG) PET (TNM) and [B(i)] mismatch residuals between MRI and 18F-FDG PET (NSNM). Here, the white colour represents a mismatch residual value close to about 0.6 standard deviations from the regression line, and the blue colour denotes a negative mismatch residual with an NM worse than expected. Mismatch maps of average residual z-scores in regions that significantly differ between A+S+ and A+S– groups are seen for [A(ii)] TNM and [B(ii)] NSNM mismatch, after adjusting for age, sex and tau load. Here, white colour represents no significant difference between A+S+ and A+S– groups, and blue colours depict a negative mismatch residual that significantly differs between groups, where residual values (and hence the colour) in A(i) and B(i) are similar to those shown in A(ii) and B(ii), respectively.

Regional TNM and NSNM mismatch residual maps in A+S+ patients reveal a posterior hypometabolism signature, where hypometabolism is greater than expected given the level of tau tracer uptake [Fig. 2A(i)] or atrophy [Fig. 2B(i)]. Conversely, regions outside the parieto-occipital cortex generally did not have significant deviations between observed and expected NM. Maps visualizing only the regions where residuals significantly differ between A+S+ and A+S– groups (adjusting for age, sex and tau burden) also accentuate posterior metabolic mismatch for TNM [Fig. 2A(ii)] and NSNM [Fig. 2B(ii)]. Significant differences in mismatch residuals are localized primarily in medial retrosplenial and posterolateral parieto-occipital areas, such as precuneus and occipital pole. Hence, compared to A+S– status, comorbid A+S+ pathology is associated with a posterior hypometabolic phenotype that is disproportionately worse than expected given the degree of either T or NS.

Occipital hypometabolic mismatch does not correlate with vascular or TDP-43 markers

We investigated the role of non-Alzheimer’s, non-LBD aetiologies that may contribute to TNM and NSNM mismatch. Posterior mismatch does not correlate significantly with in vivo white matter hyperintensity volumes on FLAIR MRI or a hypometabolic I/MTL/FSO ratio on 18F-FDG PET suggestive of TDP-43 disease47 in A+S+ or A+S– patients (Supplementary Fig. 1). While additional autopsy validation in a large cohort is needed, this may support our hypothesis that posterior hypometabolic mismatch is related to α-synuclein more than cerebrovascular or TDP-43 pathology.

CSF markers reflect altered neurotransmission and neuronal integrity with S+ status

To further evaluate why there may be a more prominent phenotype on 18F-FDG PET than MRI in S+ pathology, we next performed exploratory analyses of CSF metabolite and proteomics data50,51 from 544 ADNI participants with A and S biomarkers. The A+S+ group shows lower metabolism of dopamine, a neurotransmitter reduced in LBD due to hallmark dopaminergic neuron loss. Significant depletion was observed in A+S+ relative to A+S– groups for dopamine catabolites (Fig. 3A) such as homovanillic acid (P < 0.001) and 3-methoxytyramine sulfate (P = 0.004) after correcting for age and sex covariates (Fig. 3B and C). These findings are concordant with the literature on disrupted dopamine metabolism in LBD.52,53

Figure 3.

Figure 3

CSF metabolite and proteome analysis reveal altered dopamine neurotransmission and neuron integrity in patients with concomitant amyloid+synuclein pathology. (A) Dopamine is metabolized by enzymes, including catechol-O-methyltransferase (COMT), monoamine oxidase (MAO), aldehyde dehydrogenase (ALDH) and sulfotransferase (SULT). The CSF metabolite study demonstrated decreased dopamine metabolites (B) homovanillic acid and (C) 3-methoxytyramine sulfate (normalized concentration) with amyloid+ α-synuclein+ (A+S+) status. (D) The synaptic protein neuronal pentraxin-2 (NPTX2) is significantly decreased in A+S+. RFU = relative fluorescence unit. Box plots: filled circles = data points; × = mean; mid-line = median; lower and upper edges of each filled box = first (Q1) and third (Q3) quartiles (interquartile range, IQR); whiskers = minimum/maximum points and outliers based on thresholds <Q1 – 1.5(IQR) or >Q3 + 1.5(IQR). (E) Ontology of CSF proteomics comparing A+S+ versus A+S– indicates that S+ status is associated with neuron signalling, synapse organization and vesicles. (F) Comparing A+S+ versus A–S+ highlights that A+ status is associated with immune responses, protein degradation and lipoparticles. False discovery rate correction, Benjamini–Hochberg adjustment and weighted set cover were performed. Comparisons were made using likelihood ratio tests with covariates of age and sex, where *P < 0.05 and **P < 0.005.

CSF proteome changes related to S+ status may involve altered neuronal integrity. Prior biomarker studies revealed that CSF levels of synaptic neuronal pentraxin-2 (NPTX2) are reduced in neurodegenerative diseases such as AD55 and PD, where NPTX2 correlates with dopaminergic pre-synaptic integrity characterized by dopamine transporter scan.56 Here, we find that A+S+ patients have significantly attenuated NPTX2 CSF concentrations than patients with A+S– (P = 0.002) and A–S+ (P = 0.005) status after adjusting for age and sex covariates (Fig. 3D), suggesting that AD + LBD polypathology may lead to worse neuronal integrity than in AD or LBD alone.

From a proteomic screen of over 7000 proteins, significant differences in protein expression were enriched in A+S+ for certain pathway ontologies. To better isolate the effect of α-synuclein in mixed AD + LBD pathology versus AD alone, we identified the protein ontologies by over-representation analysis reflecting differential CSF protein levels in A+S+ versus A+S– groups after multiple comparison adjustment (Fig. 3E). Compared to A+S–, the A+S+ group had CSF protein changes related to biological processes of synapse organization, cell-cell signaling and stimulus response. Dysregulated CSF proteins generally localized to the apparatus of neurotransmission and transport (receptor complex, cell surface/junction) and endolysosomal pathway (secretory/cytoplasmic vesicles, endoplasmic reticulum and Golgi complex).

We next evaluated the role of amyloid-β in the A+S+ group by contrasting proteome expression in mixed AD + LBD versus LBD alone (Fig. 3F). Compared to A–S+, A+S+ status had CSF protein changes related to biological processes of proteolysis, inflammation and immune mechanisms (wound and defence responses). Proteins with aberrant CSF expression were enriched for biochemical compartments including serum lipoparticles, myelin sheath and extracellular matrix. Together, these exploratory CSF studies suggest that the hypometabolic mismatch phenotype on imaging in A+S+ status may reflect selective injury of dopamine pathways and impaired neuron integrity seen with α-synuclein rather than the neuroimmune changes that may perhaps be more widespread with amyloid-β.

Discussion

Polypathology is increasingly recognized as a cause of clinical and biological variability in AD and multiple aetiology dementia. The confluence of amyloid, tau and α-synuclein pathologies in mixed AD + LBD can lead to accelerated neurodegeneration and worse outcomes,2,3,7 raising questions on whether targeted therapies have similar efficacy in ‘pure’ versus mixed disease. While various SAA-based tests are available13,38 and specific PET markers of α-synuclein are being developed,57,58 much can be learned from the cellular responses to α-synuclein pathology by comparing MRI, 18F-FDG PET and measures of AD and LBD pathology. From a cohort of cognitively impaired ADNI participants with markers of amyloid, tau, α-synuclein, cortical thickness and glucose metabolism, we found that S+ status was associated with A+ status, similar to autopsy studies.3 Next, we uncovered that A+S+ patients have a posterior parieto-occipital pattern of reduced glucose metabolism that is disproportionate to both the degree of AD pathology (NFTs) and the degree of atrophy in these patients. This mismatch could be used as a ‘virtual biomarker’ for AD + LBD that does not correlate with cerebrovascular or TDP-43 biomarkers. In probing why this dissociation between 18F-FDG PET and MRI exists in AD + LBD, exploratory CSF analysis suggested this hypometabolic mismatch may be related to aberrant neuronal integrity with disrupted metabolism and death of critical neurons in dopamine-mediated pathways51,52 (altering levels of dopamine catabolites and synaptic marker NPTX2), which may be better detected by 18F-FDG PET than by MRI.

Posterior hypometabolic mismatch is consistent with but not entirely explained by the cingulate island sign on 18F-FDG PET, the prototypical hypometabolic signature in LBD.31-35 Cingulate island sign emphasizes the lack of hypometabolism in the posterior cingulate relative to the extensive hypometabolism in the cuneus. With the cingulate island of LBD, neither the spared posterior cingulate cortex nor the hypometabolic cuneus have corresponding atrophy on MRI.28-30,33 Conversely, our TNM and NSNM mismatch measures highlight greater parieto-occipital hypometabolism relative to local tau load and atrophy. These additional measures of tau and atrophy are not explicitly incorporated into the cingulate island sign.

Our findings are concordant with the literature exemplifying hypometabolism over atrophy as a feature of LBD, possibly reflecting a pathobiological basis of α-synuclein. Patients with LBD, even those with severe neocortical α-synuclein load, have fairly preserved cortical volume on ante-mortem and post-mortem assessment.28,29,33,59 While LBD may be associated with some temporal atrophy,29,30 this is much less than the degree of parieto-occipital hypometabolism.28,31 In fact, occipital cortex thickness has even been reported to increase in patients with AD and hallucinations (who may be enriched for mixed AD + LBD).60 Glucose hypometabolism on 18F-FDG PET also precedes neurotransmission changes seen on dopamine transporter molecular imaging.36 From a network perspective, LBD displays broad metabolic abnormalities in the brain, which could reflect damage at key pathway nodes in dopaminergic neuronal ensembles that are susceptible to α-synuclein pathology,52,53,59-64 and/or regional alterations in neuronal integrity that could appear on 18F-FDG PET despite the lack of change on structural MRI.28-30 This selective neurodegeneration of vulnerable dopamine pathway neurons in LBD9,56 may be associated with wider metabolic network changes36,60-64 and disproportionate posterior 18F-FDG hypometabolism relative to atrophy when compared to a more generalized neurodegeneration and glial activation in AD.7,65 At the cellular/molecular level, neuronal activity modulates the phosphorylation and aggregation kinetics of α-synuclein,66 further supporting that pathophysiological manifestations of α-synuclein may be better detected by changes in neuronal integrity more associated with 18F-FDG PET than by cortical thickness on MRI. Intriguingly, NPTX2 colocalizes with α-synuclein aggregates in Lewy bodies and neurites and is connected to dopamine neuron dysfunction56,67 and AMPA-mediated excitotoxicity.67,68 Misregulated NPTX2 expression has been reported among patients with various neurodegenerative proteinopathies55,56,68 and we found NPTX2 CSF levels were lower with A+S+ status than A+S– or A–S+ status, perhaps reflecting a greater impact on neuron integrity in mixed AD + LBD. Overall, we posit that converging findings implicate α-synuclein pathology in disrupted neuronal integrity, especially in the dopaminergic pathway, that is associated more with hypometabolism on 18F-FDG PET than changes seen on MRI.

In the absence of α-synuclein pathology, A+S– patients demonstrated a relative lack of dissociation between MRI and 18F-FDG PET. This consilience between neurodegenerative measures may stem from differences in metabolic activity in neurons versus glia in AD versus LBD. CSF proteome analyses comparing to A+S+ patients suggested that the presence of amyloid pathology favor an inflammatory or innate immune response phenotype, more so than the selective dopaminergic injury and neuronal integrity changes observed with the presence of α-synuclein. Considering AD pathology, numerous studies support the hypothesis that amyloid-β incites a strong neuroinflammatory response by astrocytes, microglia and immune cells64 while tau hyperphosphorylation and NFTs are more closely associated with neuronal loss.7,19 Because 18F-FDG PET detects both neuronal ‘hypo’metabolism in neurodegeneration and glial ‘hyper’metabolism in neuroinflammation, the extent of net hypometabolism visualized in AD is likely dampened in magnitude. This is supported by dual tracer studies with 18F-FDG PET and the microglia-related Translocator protein (TSPO) PET,69 demonstrating that microglial inflammation may be present in AD and potentially contribute to measurable 18F-FDG PET uptake. Thus, neuronal hypometabolism on 18F-FDG PET could be masked by microglial activation in some regions. Differing degrees of neuroinflammation in AD versus LBD may also contribute to this mismatch between MRI and 18F-FDG PET. While α-synuclein pathology provokes a neuroimmune reaction,70 including adaptive immune CD4+ T cells,71,72 LBD may recruit a less robust or more confined neuroinflammatory response of glia and innate immunity than in AD,73-75 especially at early stages of selective dopaminergic neuron injury. Indeed, microglial and astrocyte activation at autopsy is more closely associated with AD rather than LBD pathologies, even in patients with mixed AD + LBD.73 For these reasons, 18F-FDG PET may be better suited to identify neuronal responses to LBD and mixed AD + LBD.

Limitations and future directions

Our study has several limitations and next steps. Examining the relationships between A, T, N and S is based on hypotheses of associations between proteinopathies and cellular responses (atrophy and/or hypometabolism), and therefore not causal in nature. Extended in vitro, in vivo and ex vivo investigations are required to pinpoint precise mechanisms of neuronal and glial injury from multiple pathologies. The clinical cohort available was based on patients with primarily amnestic phenotype in ADNI, such that the sample is more biased towards predominantly an AD-like presentation and underlying AD neuropathology, which would most likely be enriched for A+T+ status. Concomitant non-Alzheimer pathologies may be present but underrepresented in ADNI relative to the general population. ADNI largely excludes patients with substantial neurological diseases besides AD, systemic illnesses, substance use and primary psychiatric disorders and is thus not representative of a typical clinical population with comorbidities that could influence 18F-FDG PET and MRI. Additional cohorts such as patients with predominantly LBD-like presentation or community-based samples with more patients with typical presentations of multiple aetiology dementia should be examined to corroborate our signatures of hypometabolic mismatch. There are limitations to the SAA measure. Several SAA tests exhibit different sensitivities for α-synuclein pathology based on different stages, such as the lower detection rates in patients with lower α-synuclein load or olfactory involvement.16,76 Hence, it may be more difficult to assess the role of hypometabolic mismatch in patients with low α-synuclein levels wherein the SAA test is insensitive. Future work may relate the relative degree of α-synuclein load based on quantitative SAA biofluid measures. Development of α-synuclein PET tracers,57,58 specifically those sensitive to LBD, would likely allow for greater spatiotemporal resolution of α-synuclein spread, its relationship with neurodegeneration and the mismatch metrics examined here. Time differences between imaging and CSF SAA studies could contribute to more variability, although this was addressed by choosing scans and CSF collection times that were close together (median time was 34 days, with 63% within 3 months). Based on the goal to maximize power and increase sample size, the time interval of 2 years was selected within reason from the literature, similar to the time between diagnostic imaging and CSF collection at enrollment seen in some SAA studies16,39 such as the Parkinson’s Progression Marker Initiative16 (though some studies of A and S markers in the ADNI cohort have applied a 6-month timeframe38). The conversion rate from negative to positive result is slower for α-synuclein CSF SAA than for amyloid markers in cognitively unimpaired and impaired older adults,18,38 which supports our selected intervals. Nevertheless, more studies may highlight ideal intervals between biomarker studies for cross-comparison. Additional temporal considerations include the limitation of cross-sectional studies to detect atrophy and hypometabolism due to individual variability, which could potentially be over- or under-called based on one set of scans. Future analyses of longitudinal change on MRI and PET can improve the ability to evaluate neurodegeneration robustly. Certain technical factors may saturate radiotracer retention, creating a ceiling effect for tau burden. Moreover, since each participant may be at a different disease stage, there may be patients with earlier-stage pathological burden where TNM and NSNM mismatch may not have yet manifested despite the presence of early α-synuclein inclusions. Therefore, analysis with alternate measures of neurodegeneration and α-synuclein SAA, pathological classification and autopsy validation can strengthen results. Further inquiries into non-Alzheimer’s, non-LBD pathologies such as TDP-43 and cerebrovascular disease can be ascertained with exploratory biomarkers and neuropathology. Mismatch based on regressions performed within different subgroups of patients based on markers of amyloid-β, tau, α-synuclein and other pathologies can also be evaluated further, although we did not find major between-group differences in regressions across regions. The relationship and mismatch between tau PET, 18F-FDG PET and thickness on MRI may vary across disease stage or at higher levels of cognitive impairment. Non-Alzheimer’s copathologies may also increase at later stages of AD and LBD. Future studies with additional markers and longitudinal data can clarify such relations. Exploratory CSF proteome ontologies primarily found extracellular and secreted protein hits, so novel studies may aim to elucidate intracellular expression levels of protein/RNA from specific cell types. Proteome findings related to neuronal integrity could represent changes in both neuroglial structure and function, so additional investigations are required to obtain a more granular view linking microscopic and macroscopic features of α-synucleinopathy. This work may help contextualize emerging biological frameworks for AD7,77 and LBD.11,12

Overall, we found evidence supporting the hypothesis that 18F-FDG metabolism is lower than expected based on the degree of tau pathology or cortical thinning in the setting of mixed AD + LBD aetiologies. This posterior hypometabolism was consonant with CSF proteomics and metabolite studies highlighting altered dopamine transmission and neuronal integrity related to α-synuclein pathology and cognition but not vascular disease or TDP-43 pathology. While further validation is required, hypometabolic mismatch may represent a unique feature in the pathobiology of mixed AD + LBD that may assist in precise diagnosis and treatment.

Supplementary Material

awae352_Supplementary_Data

Acknowledgements

The authors thank our lab members for helpful and constructive discussions and the Alzheimer's Disease Neuroimaging Initiative (ADNI) investigators, staff, participants and families. This manuscript is dedicated to the families of the authors, as well as the patients and families from ADNI. Data collection and sharing for ADNI was funded by the National Institutes of Health (NIH U01 AG024904) and the Department of Defense (DOD ADNI 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 generous 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 Pharmaceuticals, 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 Pharmaceutical 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 Pharmaceutical 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). The ADNI grantee organization is the Northern California Institute for Research and Education and the study is coordinated by the Alzheimer’s Therapeutic Research Institute at the University of Southern California. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California. Data used in preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf

Contributor Information

Michael Tran Duong, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.

Sandhitsu R Das, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Pulkit Khandelwal, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.

Xueying Lyu, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.

Long Xie, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.

Emily McGrew, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Nadia Dehghani, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Corey T McMillan, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Edward B Lee, Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Leslie M Shaw, Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Paul A Yushkevich, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

David A Wolk, Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Ilya M Nasrallah, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.

Data availability

Raw and processed data, including scans and spreadsheets, are available on the Alzheimer’s Disease Neuroimaging Initiative (ADNI) data archive (http://adni.loni.usc.edu). Code for image processing is available at http://stnava.github.io/ANTs/ and code for analyses can be provided upon reasonable request.

Funding

Funding was provided by the National Institute on Aging (NIA) via a Ruth L. Kirschstein National Research Service Award (NIA F30 AG074524), research project grants (NIA R01 AG072796 and R01 AG069474) and a University of Pennsylvania Alzheimer’s Disease Core Center grant (NIA P30 AG072979).

Competing interests

L.X. received personal consulting fees from Galileo CDS, Inc. L.X. has become an employee of Siemens Healthineers since May 2022 but the current study was conducted during his employment at the University of Pennsylvania. D.A.W. reports grants from Merck, Biogen, Eli Lilly/Avid and additional fees from GE Healthcare, Functional Neuromodulation and Neuronix, all outside this work. I.M.N. reports fees from Biogen outside this work. The remaining authors report no competing interests.

Supplementary material

Supplementary material is available at Brain online.

References

  • 1. Nelson PT, Brayne C, Flanagan ME, et al. Frequency of LATE neuropathologic change across the spectrum of Alzheimer’s disease neuropathology: Combined data from 13 community based or population-based autopsy cohorts. Acta Neuropathol. 2022;144:27–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Robinson JL, Richardson H, Xie SX, et al. The development and convergence of copathologies in Alzheimer's disease. Brain. 2021;144:953–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Colom-Cadena M, Gelpi E, Charif S, et al. Confluence of α-synuclein, tau, and β-amyloid pathologies in dementia with Lewy bodies. J Neuropathol Exp Neurol. 2013;72:1203–1212. [DOI] [PubMed] [Google Scholar]
  • 4. Thomas AJ, Mahin-Babaei F, Saidi M, et al. Improving the identification of dementia with Lewy bodies in the context of an Alzheimer's-type dementia. Alzheimers Res Ther. 2018;10:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Irwin DJ, Grossman M, Weintraub D, et al. Neuropathological and genetic correlates of survival and dementia onset in synucleinopathies: A retrospective analysis. Lancet Neurol. 2017;16:55–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Merdes AR, Hansen LA, Jeste DV, et al. Influence of Alzheimer pathology on clinical diagnostic accuracy in dementia with Lewy bodies. Neurology. 2003;60:1586–1590. [DOI] [PubMed] [Google Scholar]
  • 7. Jack CR, Bennett DA, Blennow K, et al. NIA-AA research framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Jack CR, Wiste HJ, Therneau TM, et al. Associations of amyloid, tau, and neurodegeneration biomarker profiles with rates of memory decline among individuals without dementia. JAMA Neurol. 2019;321:2316–2325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. McKeith IG, Boeve BF, Dickson DW, et al. Diagnosis and management of dementia with Lewy bodies fourth consensus report of the DLB consortium. Neurology. 2017;89:88–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Landau SM, Lee J, Murphy A, et al. Individuals with Alzheimer's disease and low tau burden: Characteristics and implications. Alzheimers Dement. 2024;20:2113–2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Simuni T, Chahine LM, Poston K, et al. A biological definition of neuronal α-synuclein disease: Towards an integrated staging system for research. Lancet Neurol. 2024;23:178–190. [DOI] [PubMed] [Google Scholar]
  • 12. Höglinger GU, Adler CH, Berg D, et al. A biological classification of Parkinson's disease: The SynNeurGe research diagnostic criteria. Lancet Neurol. 2024;23:191–204. [DOI] [PubMed] [Google Scholar]
  • 13. Concha-Marambio L, Pritzkow S, Shahnawaz M, Farris CM, Soto C. Seed amplification assay for the detection of pathologic alpha-synuclein aggregates in cerebrospinal fluid. Nat Protoc. 2023;18:1179–1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Chahine LM, Beach TG, Adler CH, et al. Central and peripheral α-synuclein in Parkinson disease detected by seed amplification assay. Ann Clin Transl Neurol. 2023;10:696–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Grossauer A, Hemicker G, Krismer F, et al. α-synuclein seed amplification assays in the diagnosis of synucleinopathies using cerebrospinal fluid-A systematic review and meta-analysis. Mov Disord Clin Pract. 2023;10:737–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Siderowf A, Concha-Marambio L, Lafontant DE, et al. Assessment of heterogeneity among participants in the Parkinson's progression markers initiative cohort using α-synuclein seed amplification: A cross-sectional study. Lancet Neurol. 2023;22:407–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Arnold MR, Coughlin DG, Brumbach BH, et al. α-synuclein seed amplification in CSF and brain from patients with different brain distributions of pathological α-synuclein in the context of co-pathology and non-LBD diagnoses. Ann Neurol. 2022;92:650–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Palmqvist S, Rossi M, Hall S, et al. Cognitive effects of Lewy body pathology in clinically unimpaired individuals. Nat Med. 2023;29:1971–1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Gómez-Isla T, Hollister R, West H, et al. Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer's disease. Ann Neurol. 1997;41:17–24. [DOI] [PubMed] [Google Scholar]
  • 20. Hanseeuw B, Betensky RA, Schultz AP, et al. Fluorodeoxyglucose metabolism associated with tau-amyloid interaction predicts memory decline. Ann Neurol. 2017;81:583–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Das S, Xie L, Wisse LEM, et al. Longitudinal and cross-sectional structural magnetic resonance imaging correlates of AV-1451 uptake. Neurobiol Aging. 2018;66:49–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Iaccarino L, Tammewar G, Ayakta N, et al. Local and distant relationships between amyloid, tau and neurodegeneration in Alzheimer's disease. Neuroimage Clin. 2018;17:452–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Strom A, Iaccarino L, Edwards L, et al. Cortical hypometabolism reflects local atrophy and tau pathology in symptomatic Alzheimer's disease. Brain. 2022;145:713–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. de Flores R, Wisse LEM, Das SR, et al. Contribution of mixed pathology to medial temporal lobe atrophy in Alzheimer's disease. Alzheimers Dement. 2020;16:843–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Das SR, Lyu X, Duong MT, et al. Tau-Atrophy variability reveals phenotypic heterogeneity in Alzheimer’s disease. Ann Neurol. 2021;90:751–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Duong MT, Das SR, Lyu X, et al. Dissociation of tau pathology and neuronal hypometabolism within the ATN framework of Alzheimer’s disease. Nat Commun. 2022;13:1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Lyu X, Duong MT, Xie L, et al. Tau-neurodegeneration mismatch reveals vulnerability and resilience to comorbidities in Alzheimer's continuum. Alzheimers Dement. 2023;20:1586–1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Ishii K, Soma T, Kono AK, et al. Comparison of regional brain volume and glucose metabolism between patients with mild dementia with Lewy bodies and those with mild Alzheimer's disease. J Nucl Med. 2007;48:704–711. [DOI] [PubMed] [Google Scholar]
  • 29. Harper L, Bouwman F, Burton EJ, et al. Patterns of atrophy in pathologically confirmed dementias: A voxelwise analysis. J Neurol Neurosurg Psychiatry. 2017;88:908–916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kang SW, Jeon S, Yoo HS, et al. Effects of Lewy body disease and Alzheimer disease on brain atrophy and cognitive dysfunction. Neurology. 2019;92:e2015–e2026. [DOI] [PubMed] [Google Scholar]
  • 31. Lim SM, Katsifis A, Villemagne VL, et al. The 18F-FDG PET cingulate island sign and comparison to 123I-β-CIT SPECT for diagnosis of dementia with Lewy bodies. J Nucl Med. 2009;50:1638–1645. [DOI] [PubMed] [Google Scholar]
  • 32. Imabayashi E, Yokoyama K, Tsukamoto T, et al. The cingulate island sign within early Alzheimer’s disease-specific hypoperfusion volumes of interest is useful for differentiating Alzheimer’s disease from dementia with Lewy bodies. EJNMMI Res. 2016;6:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Patterson L, Firbank MJ, Colloby SJ, et al. Neuropathological changes in dementia with Lewy bodies and the cingulate island sign. J Neuropathol Exp Neurol. 2019;78:714–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Silva-Rodríguez J, Labrador-Espinosa MA, Moscoso A, Schöll M, Mir P, Grothe MJ. Differential effects of tau stage, Lewy body pathology, and substantia Nigra degeneration on 18F-FDG PET patterns in clinical Alzheimer disease. J Nucl Med. 2023a;64:274–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Silva-Rodríguez J, Labrador-Espinosa MA, Moscoso A, et al. Characteristics of amnestic patients with hypometabolism patterns suggestive of Lewy body pathology. Brain. 2023b;146:4520–4531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Stockbauer A, Beyer L, Huber M, et al. Metabolic network alterations as a supportive biomarker in dementia with Lewy bodies with preserved dopamine transmission. Eur J Nucl Med Mol Imaging. 2023;51:1023–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Hansson O, Seibyl J, Stomrud E, et al. CSF biomarkers of Alzheimer's disease concord with amyloid-β PET and predict clinical progression: A study of fully automated immunoassays in BioFINDER and ADNI cohorts. Alzheimers Dement. 2018;14:1470–1481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Tosun D, Hausle Z, Iwaki H, et al. A cross-sectional study of α-synuclein seed amplification assay in Alzheimer's disease neuroimaging initiative: Prevalence and associations with Alzheimer's disease biomarkers and cognitive function. Alzheimers Dement. 2024;20:5114–5131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Poggiolini I, Gupta V, Lawton M, et al. Diagnostic value of cerebrospinal fluid alpha-synuclein seed quantification in synucleinopathies. Brain. 2022;145:584–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Avants BB, Epstein CL, Grossman M, Gee JC. Symmetric diffeomorphic image registration with cross correlation: Evaluating automated labeling of elderly and neurodegenerative brain. Med Image Anal. 2008;12:26–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Das SR, Avants BB, Grossman M, Gee JC. Registration based cortical thickness measurement. Neuroimage. 2009;45:867–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Tustison NJ, Cook PA, Klein A, et al. Large-scale evaluation of ANTs and FreeSurfer cortical thickness measurements. Neuroimage. 2014;99:166–179. [DOI] [PubMed] [Google Scholar]
  • 43. Wang H, Suh JW, Das S, et al. Regression-based label fusion for multi-atlas segmentation. IEEE Conf Comput Vis Pattern Recognit Work. 2011;20:1113–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Landman B, Warfield S. MICCAI 2012 Workshop on multi-atlas labeling. In: Medical Image Computing and Computer-Assisted Intervention, 15th International Conference, Nice, France, October 1-5, 2012, Proceedings. Springer; 2012.
  • 45. Sepulcre J, Grothe MJ, Sabuncu M, et al. Hierarchical organization of tau and amyloid deposits in the cerebral cortex. JAMA Neurol. 2017;74:813–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Kimura N, Aso Y, Yabuuchi K, et al. Association of modifiable lifestyle factors with cortical amyloid burden and cerebral glucose metabolism in older adults with mild cognitive impairment. JAMA Netw Open. 2020;3:e205719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Buciuc M, Botha H, Murray ME, et al. Utility of FDG-PET in diagnosis of Alzheimer-related TDP-43 proteinopathy. Neurology. 2020;95:e23–e34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Yushkevich PA, Piven J, Hazlett HC. User-guided 3D active contour segmentation of anatomical structures: Significantly improved efficiency and reliability. Neuroimage. 2006;31:1116–1128. [DOI] [PubMed] [Google Scholar]
  • 49. Rorden C, Brett M. Stereotaxic display of brain lesions. Behav Neurol. 2000;12:191–200. [DOI] [PubMed] [Google Scholar]
  • 50. Panyard DJ, McKetney J, Deming YK, et al. Large-scale proteome and metabolome analysis of CSF implicates altered glucose and carbon metabolism and succinylcarnitine in Alzheimer's disease. Alzheimers Dement. 2023;19:5447–5470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Sung YJ, Yang C, Norton J, et al. Proteomics of brain, CSF, and plasma identifies molecular signatures for distinguishing sporadic and genetic Alzheimer's disease. Sci Transl Med. 2023;15:eabq5923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Olsson R, Roos BE. Concentrations of 5-hydroxyindoleacetic acid and homovanillic acid in the cerebrospinal fluid after treatment with probenecid in patients with Parkinson's disease. Nature. 1968;219:502–503. [DOI] [PubMed] [Google Scholar]
  • 53. Goto R, Kurihara M, Kameyama M, et al. Correlations between cerebrospinal fluid homovanillic acid and dopamine transporter SPECT in degenerative parkinsonian syndromes. J Neural Transm (Vienna). 2023;130:513–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Maass A, Landau S, Baker SL, et al. Comparison of multiple tau-PET measures as biomarkers in aging and Alzheimer’s disease. Neuroimage. 2017;157:448–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Libiger O, Shaw LM, Watson MH, et al. Longitudinal CSF proteomics identifies NPTX2 as a prognostic biomarker of Alzheimer's disease. Alzheimers Dement. 2021;17:1976–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Nilsson J, Constantinescu J, Nellgård B, et al. Cerebrospinal fluid biomarkers of synaptic dysfunction are altered in Parkinson's disease and related disorders. Mov Disord. 2023;38:267–277. [DOI] [PubMed] [Google Scholar]
  • 57. Smith R, Capotosti F, Schain M, et al. The α-synuclein PET tracer [18F] ACI-12589 distinguishes multiple system atrophy from other neurodegenerative diseases. Nat Commun. 2023;14:6750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Kim HY, Chia WK, Hsieh C-J, et al. A novel brain PET radiotracer for imaging alpha synuclein fibrils in multiple system atrophy. J Med Chem. 2023;66:12185–12202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Whitwell JL, Weigand SD, Shiung MM, et al. Focal atrophy in dementia with Lewy bodies on MRI: A distinct pattern from Alzheimer's disease. Brain. 2007;130(Pt 3):708–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Phan TX, Baratono S, Drew W, et al. Increased cortical thickness in Alzheimer's disease. Ann Neurol. 2024;95:929–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Schumacher J, Peraza LR, Firbank M, et al. Functional connectivity in dementia with Lewy bodies: A within- and between-network analysis. Hum Brain Mapp. 2018;39:1118–1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Caminiti SP, Tettamanti M, Sala A, et al. Metabolic connectomics targeting brain pathology in dementia with Lewy bodies. J Cereb Blood Flow Metab. 2017;37:1311–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Han L, Lu J, Tang Y, et al. Dopaminergic and metabolic correlations with cognitive domains in non-demented Parkinson's disease. Front Aging Neurosci. 2021;13:627356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Liu FT, Ge JJ, Wu JJ, et al. Clinical, dopaminergic, and metabolic correlations in Parkinson disease: A dual-tracer PET study. Clin Nucl Med. 2018;43:562–571. [DOI] [PubMed] [Google Scholar]
  • 65. De Strooper B, Karran E. The cellular phase of Alzheimer’s disease. Cell. 2006;164:603–615. [DOI] [PubMed] [Google Scholar]
  • 66. Parra-Rivas LA, Madhivanan K, Aulston BD, et al. Serine-129 phosphorylation of α-synuclein is an activity-dependent trigger for physiologic protein-protein interactions and synaptic function. Neuron. 2023;111:4006–4023.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Moran LB, Hickey L, Michael GJ, et al. Neuronal pentraxin II is highly upregulated in Parkinson's disease and a novel component of Lewy bodies. Acta Neuropathol. 2008;115:471–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Hruska-Plochan M, Wiersma VI, Betz KM, et al. A model of human neural networks reveals NPTX2 pathology in ALS and FTLD. Nature. 2024;626:1073–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Xiang X, Wind K, Wiedemann T, et al. Microglial activation states drive glucose uptake and FDG-PET alterations in neurodegenerative diseases. Sci Transl Med. 2021;13:eabe5640. [DOI] [PubMed] [Google Scholar]
  • 70. Kwon HS, Koh SH. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl Neurodegener. 2020;9:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Iba M, Kim C, Sallin M, et al. Neuroinflammation is associated with infiltration of T cells in Lewy body disease and α-synuclein transgenic models. J Neuroinflammation. 2020;17:214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Gate D, Tapp E, Leventhal O, et al. CD4+ T cells contribute to neurodegeneration in Lewy body dementia. Science. 2021;374:868–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wetering JV, Geut H, Bol JJ, et al. Neuroinflammation is associated with Alzheimer's disease co-pathology in dementia with Lewy bodies. Acta Neuropathol Commun. 2024;12:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Zhao N, Attrebi ON, Ren Y, et al. APOE4 exacerbates α-synuclein pathology and related toxicity independent of amyloid. Sci Transl Med. 2020;12:eaay1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Amin J, Holmes C, Dorey RB, et al. Neuroinflammation in dementia with Lewy bodies: A human post-mortem study. Transl Psychiatry. 2020;10:267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Mastenbroek SE, Vogel JW, Collij LE, et al. Disease progression modelling reveals heterogeneity in trajectories of Lewy-type α-synuclein pathology. bioRxiv. [Preprint] 10.1101/2023.12.05.569878 [DOI] [PMC free article] [PubMed]
  • 77. Jack CR, Andrews JS, Beach TG, et al. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement. 2024;20(8):5143–5169. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

awae352_Supplementary_Data

Data Availability Statement

Raw and processed data, including scans and spreadsheets, are available on the Alzheimer’s Disease Neuroimaging Initiative (ADNI) data archive (http://adni.loni.usc.edu). Code for image processing is available at http://stnava.github.io/ANTs/ and code for analyses can be provided upon reasonable request.


Articles from Brain are provided here courtesy of Oxford University Press

RESOURCES