Next Article in Journal
Phytochemical Investigation of Aquilaria agallocha and Identification of a Diarylheptanoid Exhibiting Anti-Tau Aggregation Activity
Previous Article in Journal
Exercise and Carnosine Modulate Microbiota-Derived Metabolites, Myokines, and Cardiometabolic Profiles in Rats: A Randomized Controlled Trial
Previous Article in Special Issue
Metabolic and Structural Consequences of GM3 Synthase Deficiency: Insights from an HEK293-T Knockout Model
error_outline You can access the new MDPI.com website here. Explore and share your feedback with us.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glycosphingolipids in Dementia: Insights from Mass Spectrometry and Systems Biology Approaches

1
National Institute for Research and Development in Electrochemistry and Condensed Matter, 300224 Timisoara, Romania
2
Department of Mathematics-Informatics, Aurel Vlaicu University of Arad, 310130 Arad, Romania
3
Department of Physics, West University of Timisoara, 300223 Timisoara, Romania
4
Neuroscience Department, Faculty of Medicine, Victor Babeş University of Medicine and Pharmacy, 300041 Timisoara, Romania
5
Department of Technical and Natural Sciences, Aurel Vlaicu University of Arad, 310130 Arad, Romania
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(12), 2854; https://doi.org/10.3390/biomedicines13122854
Submission received: 5 October 2025 / Revised: 13 November 2025 / Accepted: 20 November 2025 / Published: 22 November 2025

Abstract

This narrative literature review synthesizes recent evidence on glycosphingolipid (GSL) dysregulation in dementia, emphasizing discoveries enabled by mass spectrometry (MS) and systems biology. Focusing on the research published within the last decade, we selected studies that are relevant to GSL alterations in dementia and notable for their methodological advances. The findings were conceptually integrated to emphasize key molecular, analytical, and systems-level aspects across the major dementia types. The results from MS-based glycolipidomics in Alzheimer’s disease, dementia with Lewy bodies, frontotemporal dementia, Parkinson’s disease dementia, and Huntington’s disease consistently indicate altered GSL metabolism and shared molecular vulnerabilities in neuronal lipid regulation. At the same time, distinct GSL signatures differentiate individual dementias, reflecting the disease-specific mechanisms of neurodegeneration. The literature also reveals that recent advances in high-resolution MS and integrative analytical workflows have shifted GSL research from descriptive to mechanistic, facilitating the detailed mapping of species linked to neuroinflammation, protein aggregation, and synaptic dysfunction. Systems-level analyses combining MS data with other omics approaches increasingly depict GSLs as active regulators of neuronal function rather than inert membrane components. At the same time, emerging trends position GSLs as promising early biomarkers and potential therapeutic targets, while the growing use of artificial intelligence in MS data analysis is accelerating the detection of their subtle patterns, improving cross-disease comparisons. Together, these results reinforce the major role of MS-based platforms in discovering dementia-associated GSLs, identifying therapeutic targets, and influencing future strategies for diagnosis and treatment.

1. Introduction

Dementia encompasses a heterogeneous group of progressive neurodegenerative disorders that together represent one of the most critical global health challenges of the twenty-first century. Defined by cognitive decline severe enough to interfere with independent living, dementia is not a single disease but rather a syndrome with multiple etiologies and pathological substrates [1].
Dementia prevalence is rising inexorably. According to the most recent epidemiological estimates by the World Health Organization, more than 55 million people live at present with dementia worldwide, and projections suggest that this figure will exceed 135 million by 2050, due to increased life expectancy and population aging. The consequences extend beyond patients to caregivers, healthcare systems, and societies at large, with deep medical, social, and economic implications.
Despite the intense research efforts over the past few decades, the development of effective therapies has been hampered by the remarkable clinical and molecular heterogeneity of dementia syndromes [2,3], such as Alzheimer’s disease (AD), dementia with Lewy body (DLB), frontotemporal dementia (FTD), Parkinson’s disease dementia (PDD), Huntington’s disease (HD), and mixed dementia. Each exhibits distinct clinical and neuropathological features yet converges on shared mechanisms such as synaptic dysfunction, protein aggregation, mitochondrial impairment, oxidative stress, and neuroinflammation (Table 1) [4,5,6,7,8,9,10,11,12,13,14,15,16]. The molecular basis of the major dementia types, highlighting both shared pathways and disease-specific features, isillustrated in Figure 1. Progressively, the dysregulation of lipid metabolism has emerged as a critical contributor to these processes [17], pointing toward a dimension of dementia biology that has been neglected to some extent.
Lipids constitute nearly half of the brain’s dry weight and are essential for neuronal membrane structure, synaptic signaling, vesicular transport, and myelin stability [17,18]. Among them, glycolipids, particularly gangliosides (GGs) and sulfatides, are highly enriched in neuronal membranes and localized within lipid rafts, where they regulate receptor organization and signal transduction [19,20,21]. The disruption of glycolipid homeostasis is linked to both rare lysosomal storage disorders and common neurodegenerative diseases [22,23], influencing the aggregation and toxicity of proteins such as amyloid-β, tau, α-synuclein, and huntingtin [19].
Historically, research on glycolipids was limited by analytical challenges due to their chemical diversity and low abundance. Early techniques like thin-layer chromatography (TLC) and immunodetection lacked sensitivity and specificity [24,25].
In this context, the development of modern mass spectrometry (MS), including liquid chromatography coupled with MS (LC-MS), matrix-assisted laser desorption/ionization MS imaging (MALDI-MSI), and ion mobility spectrometry (IMS), has revolutionized the field of lipidomics, enabling the detailed profiling of glycolipids across brain tissue, cerebrospinal fluid (CSF), and plasma. Hence, for a better overview, Table 2 presents the strengths and limitations of different MS platforms for glycolipid biomarkers. These technologies have also facilitated the integration of lipidomics with systems biology approaches, enabling network-based models that place glycolipid dysregulation within the broader molecular landscape of dementia (Figure 2).
MS-based studies consistently report reductions in sulfatides and alterations in GG composition in AD [34], disruptions in glycosphingolipid (GSL) metabolism in FTD [35], shifts in specific glycolipid species in DLB [36], and impaired GG biosynthesis in HD [37]. Importantly, these findings are detectable not only in the postmortem brain but also in biofluids, supporting their potential as minimally invasive biomarkers for early diagnosis, subtype differentiation, and therapeutic monitoring. Furthermore, glycolipids are increasingly viewed as active drivers of neurodegenerative cascades and promising therapeutic targets, with ongoing efforts focused on modulating their metabolism and interactions [38,39].
Integrating MS-based lipidomics with systems biology enhances diagnostic capability, enabling the discovery of disease-specific glycolipid signatures, predicting disease progression, and informing targeted therapeutic interventions.
The aim of this review is to provide an integrative overview of current evidence on GSL dysregulation in major forms of dementia, with particular emphasis on the findings generated by MS and systems biology approaches. We first summarize the fundamental roles of GSLs in neuronal physiology, followed by an examination of their profiles in AD and LBD (Lewy body dementia), including Parkinson’s disease (PD), FTD, HD, and mixed dementia. Methodological advances in lipidomics and systems-level analyses are then discussed, highlighting how these have expanded our understanding of lipid-mediated mechanisms in neurodegeneration. Finally, we outline emerging links between GSL metabolism, protein aggregation, neuroinflammation, and synaptic dysfunction and consider their implications for biomarker discovery and therapeutic strategies.
To conduct this review, a comprehensive literature search across PubMed, Google Scholar, and Web of Science yielded 412 articles (PubMed: 96; Google Scholar: 157; Web of Science: 159). After eliminating duplicates and excluding studies outside the defined scope, 171 articles remained for full review, with 53.80% published post-2020. By placing glycolipid research within the broader landscape of dementia biology and by emphasizing the methodological advancements that have enabled its recent progress, overall, this review highlights the major importance of GSLs in the pathogenesis of neurodegenerative disease and their potential as targets for biomarker discovery and therapeutic innovation.

2. Alzheimer’s Disease (AD)

AD is the most common form of dementia, accounting for 60–70% of cases [26,38,40,41]. AD predominantly affects women [42] and typically develops after age 65 [26]. Its prevalence has risen from 4.08 million cases in 1992 to 9.84 million in 2021 [43]. Characterized by cognitive decline, AD impacts individuals’ daily functioning and independence, through symptoms like memory decline, impaired thinking, and change in behavior [44,45].
AD prevalence increases dramatically with age; family history; genetic mutations; and environmental, metabolic, energetic, and vascular factors [46]. Although a conclusive diagnosis requires postmortem histopathology, current clinical diagnosis relies on neuropsychological testing and neuroimaging. Macroscopically, the neuronal and synaptic loss in AD is marked by cerebral atrophy [26,47,48], while microscopically, it is defined by abnormal protein aggregation, such as Aβ-peptide forming senile plaques (SPs) and phosphorylated tau (p-tau) protein creating neurofibrillary tangles (NFTs) and neuropil threads [49,50,51,52].
Aβ derives from the sequential β- and γ-secretase cleavage of amyloid precursor protein (APP) [38,51], forming insoluble fibrillar deposits linked to gliosis, neuroinflammation, and oxidative stress [53,54]. NFT burden correlates strongly with disease severity [55].
Studies suggest that the clinical symptoms of AD appear long after neurodegeneration, which involves many molecular changes [47]. As cell membrane breakdown occurs [47], lipid metabolism become dysregulated, affecting lipid raft structure and function [19,56]. Changes occur across lipid classes, including glycerolipids, glycerophospholipids, sphingolipids, and cholesterol [47,56]. GGs are also altered: reduced levels of GM1, GD1a, GD1b, and GT1b GGs, alongside increased levels of simpler species such as GM2, GM3, and GD3 and cholinergic markers, like Chol-1α (GQ1bα) and GT1aα, are associated with AD [19,48,51,56,57,58,59,60]. Monoclonal antibody A2B5 selectively stain neurons undergoing neurofibrillary degeneration and neuritic processes within SP in AD, recognizing c-series GG, such as GQ1c, that reappear abnormally in AD brains [61].
Technologies based on MS, particularly MALDI-MSI and TOF-SIMS [51,59], have become popular for lipid analysis in AD. Studies in APP/PS1 transgenic mouse models (TMMs) have consistently demonstrated significant reductions in sulfatides (SHexCers) and glycerophosphoinositols (GroPIns) in the cerebral cortex, hippocampus, and cerebellum, indicating both aging- and AD-related neurodegeneration [59]. A multimodal MS approach in a TMM (tgAPPArcSwe) [51] revealed a distinct lipid signature within amyloid plaques, characterized by a global depletion of cortical sulfatides and accumulation of plaque-specific lipids, including ceramide (d18:1/18:0), GM2, and GM3, containing 18:0 fatty acid moieties, in plaque peripheries and a relative enrichment inGM1 in plaque cores. These findings underscore the central role of glycolipid dysregulation in plaque microenvironments and highlight MSI as a powerful tool in identifying lipid-based biomarkers and therapeutic targets in AD.
LC-MS/MS studies have extended these insights to human tissues. Mechref et al. [62] reported dysregulated lipids in AD brains, particularly the upregulation of phosphatidylcholine (PC), phosphatidylglycerol (PG), ganglioside GD2, phosphatidylinositol (PI), phosphatidylserine (PS), lysophosphatidic acid, lysophosphatidylcholine, and sphingomyelin (SM), along with the downregulation of GD1a in AD.Such findings implicate disrupted phospholipid and sphingolipid pathways in AD pathophysiology and suggest glycosylation changes linked to disease progression [62,63,64]. Furthermore, investigations of human brain tissues uncovered alterations in glycan modifications and coregulated glycoform networks in AD, suggesting that specific glycosylation changes are closely linked to disease progression [62]. Together, these findings [62,63,64] demonstrate that integrating lipidomics and glycomics through LC-MS/MS profiling can reveal critical molecular signatures for early detection and potential therapeutic targeting in dementia.
Quantitative ultra-high-performance LC (UHPLC)-MS/MS lipidomics in APP/PS1 mice identified 43 altered hippocampal lipid species, mainly glycerolipids, glycerophospholipids, and sphingolipids [56]. Elevated cholesteryl esters (CE 22:6, 22:4) promote Aβ aggregation and vascular dysfunction, while increases in PCs and phosphatidylethanolamines (PEs) indicate membrane remodeling. Declines in galactolipids and sphingolipids (hexosylceramide, HexCer, and ceramide, Cer) may impair membrane integrity and microglial function. The LC-MS profiling of GGs revealed 48 hippocampal species associated with AD, including potential biomarkers such as di-O-Ac-GT1a (d36:1) and O-Ac-GD1b (d36:1) [65]. MALDI-MSI studies further showed GM2 and GM3 accumulation near plaques correlating with HEXA gene expression, implicating impaired GG degradation [66].
Longitudinal MALDI-MSI in APP21 rats [67] and 5xFAD [68] mice highlighted age- and region-dependent shifts: simple GGs were elevated, complex GGs decreased, and the d20:1/d18:1 ratio increased due to the loss of d18:1 species. GM1 levels increased with age but decreased later in APP21 rats, while GM2 and GM3 accumulated, particularly GM3 (d18:1). High-resolution MSI demonstrated long-chain base-specific GG deposition within plaques: 18:1 species enriched subgranular zones; 20:1 localized to entorhinal pathways; and GM3 (d18:1/18:0), GM2 (d18:1/18:0), and GM1 (d18:1/18:0) colocalized with Aβ peptides [68].
Other MS studies demonstratedthe near-complete loss of ~20 GG species in the AD cerebellum and reductions in the right cerebral hemisphere, paralleling Aβ deposition and neuronal loss [69,70]. Human MSI confirmed region-specific GM1 alterations, notably reduced GM1 (d20:1)/(d18:1) ratios [45,58]. TLC/MALDI-TOF-MS corroborated decreases in GD1b and GT1b with the predominance of d18:1-containing GGs [71]. Serum LC-MS revealed CE (16:3) and GM3 (d18:1/18:1) as promising biomarkers for AD severity [72].
Autoantibodies against GGs also contribute to pathology. Elevated IgM and IgG antibodies against GM1, GD1b, GT1b, GQ1b, and cholinergic-specific GQ1bα were found in AD sera and in the cortex and hippocampus, potentially impairing cholinergic signaling and serving as early immunological biomarkers [42].
Several studies demonstrated that GM1 critically modulates AD pathogenesis by promoting Aβ aggregation and senile plaque formation. Elevated GM1 induces conformational alterations in γ-secretase, enhances Aβ production, and stabilizes GAβ complexes that insert into the lipid membrane [44,54]. Cholesterol-rich lipid rafts potentiate GM1-Aβ interactions via hydrogen bonding with GM1′s glycosidic linkage, accelerating peptide binding [73]. Structural studies further revealed that Aβ and α-synuclein recognize specific GGs via a shared motif; engineered peptides mimicking this motif can bind GM1 and block Aβ neuronal uptake, suggesting therapeutic potential [74]. A diagram illustrating the metabolism of GSLs and their interactions with key proteins, Aβ, α-synuclein, and tau, is presented in Figure 3.
Finally, detecting glycolipid-associated protein complexes, such as GM1-bound Aβ [75], apolipoprotein E isoforms [76], clusterin [77], complement C1q [78], and low-density lipoprotein receptor-related protein 1 [79], via immunoassays (ELISA, electrochemiluminescence, Luminex) bridges MS discoveries with clinical application [80]. GGs remain among the most valuable lipid biomarkers in AD, with region-dependent reductions: in early-onset AD, declines are prominent in gray and frontal white matter, whereas sporadic AD shows reductions mainly in the temporal cortex, hippocampus, and frontal regions [59].

3. Lewy Body Dementia (LBD)

LBD represents an umbrella term that includes both dementia with Lewy bodies (DLB) and Parkinson’s disease dementia (PDD). LBD is recognized as the second most widespread form of degenerative dementia in older people after AD [81].

3.1. Dementia with Lewy Bodies (DLB)

DLB accounts for approximately 20% of cases [82] and is pathologically defined by the misfolding and aggregation of α-synuclein into Lewy bodies and neurites, leading to the disruption of synaptic integrity, neurotransmission failure, and progressive neuronal loss across cortical and subcortical regions [83]. Normally involved in synaptic vesicle cycling, α-synuclein aggregation disrupts network connectivity and cognition. Currently, DLB diagnosis is primarily clinical, relying on fluctuating cognition, REM sleep behavior disorder, parkinsonism, and visual hallucinations. While imaging modalities such as fluoro-deoxyglucose positron emission tomography (FDG-PET), revealing occipital hypometabolism or the cingulate island sign, and dopamine transporter–single-photon emission computed tomography (DAT-SPECT), showing reduced striatal dopamine transporter uptake, support diagnosis, biomarker interpretation is challenged by frequent copathology with AD, especially Aβ and tau deposition [84,85]. Despite these advances, definitive diagnosis remains postmortem, underscoring the need for molecular biomarkers capable of differentiating DLB from AD and PDD during one’s lifetime, which is essential for targeted management.
Initial biomarker studies targeting α-synuclein in CSF and plasma produced inconsistent results due to methodological variability [86,87,88]. Real-time quaking-induced conversion assays represent a major advancement, enabling the amplification and detection of misfolded α-synuclein aggregates with high diagnostic accuracy, including the clear differentiation of DLB from AD [89,90,91]. Additional candidates, including the Aβ42/40 ratio, p-tau, neurofilament light chain (NfL), and neurotransmitter metabolites, show supportive rather than definitive diagnostic value [92,93,94,95,96].
Machine learning (ML) integrated with MS-based lipidomics offers new diagnostic potential. Hence, by using UPLC-MS combined with ML, Shen et al. [36] identified distinct plasmalipidomic signatures discriminating DLB from healthy controls and AD, particularly alterations in sphingolipid (sphingoid bases, ceramides, and monohexosylceramides (Hex1Cers)) metabolism. A 13-lipid panel, including Hex1Cer (d18:1/24:0) and Hex1Cer (d18:1/23:0), showed high predictive accuracy. Across analyses, ceramides, sphingosines, PE, and PC emerged as the most consistently dysregulated lipid classes in DLB. These findings suggest that disruptions in sphingolipid signaling and membrane phospholipid remodeling may contribute to the pathophysiology of DLB [36].
Earlier plasma lipidomic studies also reported elevated plasma ceramides (16:0, 18:1, 20:0, 24:1) and Hex1Cer (18:1, 24:1) in both AD and DLB compared with controls, though they lacked disease specificity, indicating that systemic sphingolipid changes may reflect general neurodegenerative processes [97]. In contrast, CSF analyses revealed more distinct biochemical patterns. Lerche et al. [98] reported reduced galactosylsphingosine (GalSph) and ceramide levels in DLB compared with PD and controls, independent of GBA1 variant status, indicating more specific lipid alterations in DLB pathology.
Although the full potential of MS-based GSL profiling in DLB remains underexplored, current evidence highlights its value in uncovering subtle disease-specific lipid alterations and its promise as a non-invasive biomarker strategy. Plasma lipidomics reflects broader neurodegenerative changes, whereas CSF sphingolipid profiling appears to capture more specific biochemical signatures of DLB. The integration of MS-based lipidomics with ML approaches thus represents a powerful framework for enhancing differential diagnosis and deepening insight into the molecular mechanisms underlying DLB.

3.2. Parkinson’s Disease Dementia (PDD)

PD is the second most common neurodegenerative disorder after AD, pathologically characterized by dopaminergic neuron loss in the substantia nigra and the aggregation of misfolded α-synuclein into Lewy bodies, placing it among the α-synucleinopathies [99,100]. Because clinical diagnosis typically occurs after substantial neuronal loss, there is an urgent need for biomarkers that enable earlier and more accurate detection [101]. Cognitive decline represents a major non-motor complication of PD, while PDD affects most patients in later stages [102,103]. Although PDD and DLB share overlapping α-synuclein pathology, they differ mainly in the temporal sequence of cognitive and motor symptoms [104]. The onset of dementia in PD signals accelerated disease progression and increased mortality [105].
Following these clinical needs, high-throughput proteomics has provided important molecular insights into PD and PDD pathology. MS-based studies have revealed the post-translational modifications (PTMs) of key neurodegenerative proteins such as tau, α-synuclein, Aβ, and TDP-43 across various diseases [106,107,108,109].
In biofluids, phosphorylated and truncated tau and modified Aβ species can also be detected [106,107,110], demonstrating the ability of MS to resolve disease-specific proteoform diversity. However, most PTM-based biomarker research has concentrated mainly on AD and DLB, leaving PDD relatively underexplored. This gap has prompted growing interest in lipidomics and glycolipidomics as complementary molecular domains that may better capture the metabolic aspects of cognitive decline.
Building on this proteomic foundation, recent research has increasingly focused on lipidomic signatures, particularly GSL metabolism, as a promising field for PDD biomarker discovery. Targeted LC-MS/MS studies have revealed elevated plasma ceramide and glucosylceramide (GlcCer) levels in sporadic PD, correlating with poorer cognitive performance [111,112]. Using a validated HPLC-MS/MS platform, Xing et al. [112] identified specific ceramide species, namely 24:1 and 14:0, negatively associated with verbal memory, while 22:0 and 20:0 correlated with hallucinations and anxiety. These relationships remained significant after adjusting for confounders, suggesting that distinct ceramide glycoforms may serve as biomarkers of domain-specific cognitive and neuropsychiatric dysfunction in PDD.
Extending beyond these targeted analyses, high-resolution lipidomics has provided additional insights into PD progression and dementia risk. Zardini Buzatto et al. [105] applied a high-sensitivity LC-QTOF-MS lipidomics workflow to evaluate whether baseline serum lipid profiles could predict cognitive decline in PD. Over a three-year follow-up, they found that patients who later developed dementia exhibited elevated ceramides, diacylglycerols, and triacylglycerols, together with reduced PCs, bis(monoacyl)glycerophosphates (BMPs), and PSs, distinguishing them from patients who remained cognitively stable. Notably, multivariate models (PLS-DA, OPLS-DA, Random Forest) achieved excellent discrimination, with a five-lipid biomarker panel yielding an area under the curve of 0.993 with >95% accuracy in validation, demonstrating that serum lipidomic signatures can robustly predict dementia years before clinical onset.
Integrating MS-based sphingolipidomics with artificial intelligence (AI) and ML further enhances predictive precision. ML models combining LC-MS lipidomics with clinical data can forecast motor and non-motor trajectories up to two years in advance and identify lipid signatures associated with disease severity [113,114]. In summary, MS-based sphingolipidomics and AI-driven analyses provide compelling evidence that specific ceramide species and broader lipidomic signatures are tightly associated with cognitive and neuropsychiatric manifestations in PDD [105,112].
Overall, GSL metabolism represents a promising biomarker domain that complements protein-based markers such as α-synuclein and NfL, underscoring the translational potential of MS lipidomics for early stratification and therapeutic monitoring. Ultimately, continued research integrating lipidomics, proteomics, and advanced computational tools will deepen the understanding of PD and its progression to dementia. Future multi-omics studies correlated with clinical and imaging outcomes are essential to validate lipid-based biomarkers and develop practical diagnostic and prognostic tools, thereby supporting precision medicine in PD.

4. Frontotemporal Dementia (FTD)

FTD, historically referred to as Pick’s disease, comprises a heterogeneous group of non-Alzheimer’s neurodegenerative dementias, primarily affecting the frontal and temporal lobes, leading to progressive impairment in behavior, language, and executive function [115,116]. FTD represents the third most common neurodegenerative dementia after AD and DLB and the second leading cause of dementia in individuals under 65 years [117].
Clinical overlap with psychiatric disorders complicates diagnosis, underscoring the need for reliable fluid biomarkers to enable early detection and disease stratification. Genetic mutations, most commonly in GRN, C9orf72, and MAPT, account for 15–20% of FTD cases and highlight the role of lysosomal dysfunction and impaired lipid homeostasis in disease pathogenesis [118].
Beyond conventional biomarkers, such as NfL and progranulin (PGRN), glycolipid and sphingolipid profiling via MS provide mechanistic insight into membrane biology and neuronal–glial interactions and lipid-mediated signaling.The ability to detect these lipid alterations in peripheral biofluids through MS expands the potential for minimally invasive biomarkers and precision medicine approaches in FTD and related syndromes [22,119,120].
Recent lipidomic studies reveal both the shared and mutation-specific mechanisms of lipid dysregulation in FTD. In 2022, Boland et al. [121] demonstrated that GRN haploinsufficiency disrupts lysosomal homeostasis by reducing BMP levels, leading to GG (GM1, GD3, and GD1) accumulation in GRN-associated FTD-TDP. GT1 reduction was specific to GRN-FTD, and BMP supplementation normalized GM2 levels in PGRN-knockout cells, linking lysosomal dysfunction to lipid accumulation [121]. Similarly, increased GT1a and/or GD2, in Pick’s disease brains, indicates impaired GSL clearance in FTD [122].
Expanding this theme, Kim et al. [123] identified altered plasma lipid signatures in behavioral variant FTD (bvFTD), characterized by elevated triacylglycerol (TG) and reduced PS and PG, along with specific individual lipid species, such as TG (16:0/16:0/16:0), diglyceride DG (18:1/22:0), PC (32:0), PS (41:5), and SM (36:4), capable of distinguishing bvFTD from AD and healthy controls [123].
Given earlier evidence that GRN mutations interrupt lysosomal lipid catabolism, Marian et al. [124] compared lipid metabolism across GRN-FTD and C9orf72-FTD postmortem brains, revealing pronounced myelin lipid loss, cholesterol ester accumulation, and fatty acid metabolism disruption that was more pronounced in GRN-FTD, consistent with its more severe white matter pathology. Extending these tissue-based findings to a peripheral biomarker context, Marian et al. [120] found reduced plasma myelin-enriched glycolipids (HexCer), especially 22:0 GlcCer and GalCer that correlated inversely with disease duration and white matter damage suggesting their utility as peripheral biomarkers of FTD severity.
Mechanistically, PGRN deficiency also reduces β-glucocerebrosidase (GCase) activity and impairs enzyme maturation in FTD-GRN brains [125], linking enzyme-level lysosomal dysfunction with sphingolipid imbalance. Together, GG accumulation [121], myelin lipid loss [124], and plasma HexCer reductions [120] outline a coherent cascade in which GRN-related lysosomal defects drive both central and peripheral lipid abnormalities. Adding further complexity, He et al. [126] identified significant elevations of very-long-chain fatty acid (VLCFA)-containing phospholipids, particularly PC (30:5/18:1), PE (33:4/20:4), and PE (33:4/22:6), in the FTD cortex, correlated with ELOVL4 expression, implicating aberrant phospholipid elongation in disease mechanisms.
Taken together, these convergent findings delineate that lipid dysregulation in FTD is multifaceted, spanning GG accumulation, sphingolipid and myelin lipid loss, lysosomal enzyme dysfunction, and VLCFA-containing phospholipid elevations. Importantly, both central (brain tissue) and peripheral (plasma) lipid changes show potential as diagnostic and prognostic biomarkers while offering mechanistic insights into disease pathogenesis and therapeutic targets.

5. Huntington’s Disease (HD)

HD is a monogenic, autosomal dominant neurodegenerative disorder caused by the expansion of a Cytosine–Adenine–Guanine trinucleotide repeat in the HTT gene on chromosome 4, leading to an expanded polyglutamine tract in the HTT protein. Clinically, HD presents with motor, cognitive, and psychiatric symptoms that typically emerge in mid-adulthood after a prodromal phase [127]. Because the causal mutation is known, HD provides a unique model for investigating presymptomatic and early symptomatic stages, enabling the identification of biomarkers reflecting early molecular alterations and therapeutic response [128].
Cognitive impairment is an integral component of HD and progresses toward Huntington’s disease dementia (HDD) in advanced stages [129]. Subcortical cognitive deficits, especially in attention, executive function, and psychomotor speed, correlate with functional decline and dementia progression [130,131], underscoring the need for biomarkers that reflect early molecular changes, disease severity, or response to therapies.
MS-based lipidomics has revealed widespread metabolic dysregulation in HD. The untargeted UHPLC-MS/MS of plasma and CSF identified altered ceramides, HexCers, SMs, diacylglycerols, and PCs, which correlated with cognitive scores from the Stroop and Verbal Fluency tests [132]. Complementary MALDI-MSI studies of HD brain tissue revealed widespread sphingolipid and phospholipid dysregulation in the caudate and cortex, indicating disrupted neuronal membrane integrity, a shift from very-long-chain to long-chain species, and structural changes with dementia-related network dysfunction [133,134,135].
Within the GSL family, GM1 has emerged as a key molecule linking lipid metabolism to synaptic and cognitive vulnerability in HD. Multiple MS-based and cellular studies have shown markedly reduced GM1 levels in HD patient-derived fibroblasts and neural cultures [136], while GM1 supplementation restored synaptic function and improved motor and cognitive outcomes in HD mouse models [137,138]. These findings implicate GM1 deficiency as both a mechanistic biomarker and a potential therapeutic target for HD-related cognitive decline.
Mechanistically, lipids such as ceramides and GSLs function as bioactive signaling molecules that regulate apoptosis, synaptic transmission, and neuroinflammatory responses. Their dysregulation in HD links systemic metabolic stress to synaptic and cognitive dysfunction. While protein biomarkers such as NfL and mutant huntingtin (mHTT) remain robust indicators of axonal injury and genetic burden [133,139], they do not capture the lysosomal and membrane lipid abnormalities revealed by MS. Integrating proteomic and lipidomic markers may therefore enhance diagnostic precision and prognostic power for cognitive decline. Consistent with these findings, MALDI-IMS and LC-MS analyses of postmortem HD brain tissue revealed sphingolipid chainlength alterations and regional phospholipid depletion that impair neuronal connectivity in executive and memory networks [134].
A major limitation of current HD lipidomic research is that most studies remain cross-sectional, limiting the assessment of predictive value for conversion from prodromal to dementiastages. By contrast, longitudinal MS-based lipidomics in PD has successfully identified predictive biomarker panels for incipient dementia [105]. Implementing similar longitudinal MS-based approaches in HD could enable the early stratification of gene-positive individuals and guide preventive interventions. Recent advances in AI and ML offer new opportunities to enhance HD biomarker discovery and clinical prediction. AI-driven models integrating multimodal MS-based data have shown promise for improving prognostic accuracy and patient stratification in early-stage HD [140,141]. MS-based approaches, including LC-MS/MS, UHPLC-MS, and MALDI-IMS, consistently reveal alterations in ceramides, SMs, and GSLs that connect membrane lipid metabolism with cognitive decline [133,134,136]. In particular, GM1 depletion emerges as both a mechanistic biomarker of synaptic vulnerability and a potential therapeutic target [136,137,138]. Consequently, future longitudinal multi-omics studies integrating AI and ML-driven analytics will be crucial to validate GSLs and other lipidomic candidates as predictive and clinically actionable biomarkers of HDD.

6. Mixed Dementia

Mixed dementia is a frequent and biologically heterogeneous form of cognitive impairment in which two or more neuropathological processes coexist often with synergistic effects on cognitive decline; most typically, these are AD proteinopathies [6]. Additional copathologies such as LB or TDP-43 inclusions are contributors that increase clinical complexity and disease progression [142].
Autopsy series and population studies indicate that mixed pathologies become prevalent with advancing age and that coexisting vascular and neurodegenerative lesions account for a substantial fraction of dementia cases in older adults. Consequently, a significant proportion of clinically diagnosed AD or vascular dementia (VD) actually represents mixed forms [6,143].
Clinically, mixed dementia often presents features of both component disorders: the episodic memory impairment characteristic of AD, as well asthe executive dysfunction, slowed processing speed, attention deficits, gait disturbance, and focal neurological signs associated with vascular contributions. When Lewy Body or TDP-43 pathology is also present, supplementary signs such as visual hallucinations, parkinsonism, or disproportionate temporal–hippocampal neuronal loss with hippocampal sclerosis occur, producing a phenotype that can shift over time as the different pathologies evolve [144].
Epidemiologically, mixed dementia incidence rises steeply with age and isassociated with vascular risk factors [145]; hence, creating strategies that target these factors at the population level isa major public health method for preventing this disease [146].
Modern diagnostic approaches attempt to deconvolute the contributions of degenerative and vascular processes in vivo by combining clinical assessment with (i) structural magnetic resonance imaging (MRI) to visualize infarcts, lacunes, white matter hyperintensities, microbleeds, and atrophy [147]; (ii) positron emission tomography (PET) for amyloid and tau [148]; (iii) CSF assays [149] of Aβ42 or Aβ42/40 or total and p-tau; and (iv) increasingly sensitive plasma biomarkers, such as p-tau217, p-tau181, Aβ ratios, and NfL, that provide scalable screening options [150].
These molecular tools increase specificity for AD and, in conjunction with MRI, are able to identify cases where vascular lesions and AD biomarkers coexist, although important limitations still remain. For instance, MRI underestimates minor cortical microinfarcts and diffuse small-vessel damage, PET is costly and not universally available, CSF sampling is invasive, plasma assays are influenced by peripheral factors and assay platform variability, and biomarker thresholds derived from pure disease cohorts may perform differently in mixed populations.
From a therapeutic perspective, treatment is focused on managing vascular risk factors [151,152] and the administration of AD drugs. While cholinesterase inhibitors and memantine may offer limited benefits [153], new anti-amyloid therapies are promising though remain unproven in mixed cohorts [154]. At the molecular level, mixed dementia arises from overlapping pathogenic cascades. In AD, abnormal APP processing generates toxic Aβ oligomers and plaques that induce oxidative stress and neuroinflammation, while hyperphosphorylated tau disrupts microtubules and neuronal integrity [155]. Vascular processes cause ischemia, blood–brain barrier (BBB) disruption, and inflammation, further amplifying neural injury.
Importantly, all these pathways interact: (i) vascular injury impairs the perivascular and endothelial clearance of Aβ, promoting its accumulation; (ii) Aβ deposition in vessels directly injures endothelial cells and pericytes; and (iii) apolipoprotein E ε4 modulates both amyloid accumulation and vascular integrity since ε4 carriers exhibit earlier BBB breakdown and pericyte dysfunction that can precede overt amyloid or tau pathology, creating an environment leading to mixed dementia.
Downstreamshared mechanisms across proteinopathies and ischemia include mitochondrial dysfunction, calcium dysregulation, chronic microglial and astrocytic activation, the failure of proteostatic systems, and metabolic shifts away from efficient oxidative phosphorylation toward impaired glycolysis, all of which converge to produce synaptic failure and neuronal death [156].
These molecular insights motivated an expanded biomarker agenda to include lipidomic and glycolipidomic signatures that reflect membrane, synaptic, and myelin integrity. Because GSLs participate in synapse formation and signal transduction, and modulate neuroinflammation and cell–cell interactions, their dysregulation reflects both degenerative and vascular/myelin damage and therefore holds promise as a biomarker set for mixed dementia [157].
In recent years, MS has been central to modern biomarker discovery and validation in neurodegenerative diseases because it enables the sensitive, high-resolution, and relatively unbiased profiling of biomolecules in brain tissue, CSF, and plasma without relying solely on antibody reagents [158,159,160,161]. Specific lipidomic studies of mixed dementia and related disorders illustrate these points. Hence, a comparative LC-MS lipidomic analysis of white and gray matter from the temporal cortex of subcortical ischemic vascular dementia patients, mixed dementia patients, and controls found pronounced alterations in sphingolipid classes and ceramides, as well as increases in some GG species, i.e., GM3 and markers of membrane breakdown, in mixed dementia white matter compared with controls. These features are consistent with the combined effects of neuronal degeneration and myelin/axonal injury in mixed pathology [162]. Such tissue-level findings provide mechanistic plausibility for glycolipid markers in biofluids and suggest candidate species for further study.
Additional MS-based investigations mapped GG distributions to Aβ plaques and periplaque regions and demonstrated the age- and disease-related dysregulation of GG degradation pathways with increases in GM2/GM3 in plaque-rich areas [66]. The results linked lipidomic profiles to imaging indices of vascular injury and white matter lesion burden, evidencing that lipid changes reflect the spatial interplay of degenerative and vascular lesions.
On the other hand, new developments in MALDI-MSI and related workflows, such as quantitative MSI combined with on-tissue extraction and LC-MS/MS, presently enablethenear-cellular mapping of lipids, permitting the direct visualization of how glycolipid perturbations colocalize with plaques, microinfarcts, or gliotic regions in human brain sections and model systems [163].
The importance of glycolipids as biomarkers derives from both biology and analytic feasibility: (i) GGs and GSLs directly report on membrane/synaptic composition and myelin health; (ii) they are sensitive to enzymatic shifts in lipid metabolism that occur with aging, hypoxia, and inflammation; and (iii) some species can be measured in CSF and, with advanced techniques, even in plasma. In AD specifically, Noel et al. [164] discovered altered GG patterns, including changes in GM1, GM2, GM3, GD1a/b, and GT1b and shifts in sulfatides and ceramides that correlate with amyloid and tau pathology or cognitive decline. In vascular and mixed dementias, white matter lipid degradation products and altered sphingolipid ratios reflect ischemic/myelin injury and overlap with degenerative signatures in mixed cases, suggesting that panels of glycolipids together with protein biomarkers could enhance sensitivity and specificity for mixed pathology [165].
In the past few years, modern and high-performance MS technologies have made several concrete achievements in glycolipid biomarker discovery for mixed dementia: (i) methodological refinements in extraction and chromatographic separation now permit the reliable detection and partial isomer resolution of GG species differing by sialylation state, ceramide backbone length, and unsaturation; (ii) high-resolution TOF instruments combined with MS/MS fragmentation and IMS allow for the structural assignment of glycan and lipid moieties at unparalleled sensitivity and reproducibility and with a wealth of compositional and structural data; (iii) MALDI-MSI and DESI imaging approaches enable spatially resolved lipidomics, linking molecular changes to histopathology in the same tissue section; and (iv) targeted LC-MS/MS assays developed for CSF and plasma enable quantitative panels suitable for larger cohort studies and longitudinal sampling, altogether moving glycolipid candidates from tissue discovery toward biofluid validation. Nevertheless, translation faces challenges such as inter-laboratory standardization; the low abundance and peripheral dilution of brain-derived lipids in plasma; confounding by diet/peripheral metabolism and renal function; and the need for large, well-phenotyped mixed dementia cohorts with paired imaging and neuropathology for validation [17].
Below, recent glycolipid biomarker candidates compiled in the concise Table 3 from MS-based tissue and biofluid investigation of species that have been implicated across studies in AD, VD, and mixed dementia are presented. Table 3 lists the representative glycolipid classes and specific molecular species that have appeared repeatedly as altered in disease cohorts; the table does not represent an exhaustive inventory but rather a prioritized set based on reproducible reports and biological plausibility in mixed pathology.
In glycolipidomics of mixed dementia, the emerging guidance from methodological investigations and discovery studies supports the following practical strategies: (i) the use of untargeted high-resolution IMS, LC-MS, or MSI with IMS for initial discovery in well-phenotyped brain tissue, paired with histology and proteomics to connect lipid changes to plaques, gliosis, and microinfarcts; (ii) the prioritization of candidate species that are abundant enough and biochemically plausible for measurement in CSF and the development of targeted LC-MS/MS selected reaction monitoring assays with isotopically labeled internal standards for validation in CSF/plasma cohorts; (iii) performing, whenever possible, matched imaging using MRI and fluid biomarker inventory, i.e., Aβ/tau/NfL plus a glycolipid panel, in order to determine whether glycolipid signals add diagnostic or prognostic value in mixed dementia beyond classical markers; and (iv) the standardization of sample collection, extraction, and instrument methods across centers to enable multi-site validation and regulatory qualification.
In conclusion, the advancements of MS and allied systems biology techniques such as high-resolution LC-MS/MS, IMS-MS, MALDI-MSI, and hybrid quantitative imaging workflows allow for the discovery of glycolipid biomarkers and targeted measurement.
The pathway forward requires large longitudinal cohorts with detailed clinical, imaging, and neuropathological characterization; standardized MS protocols; and multimodal MS which pairs MSI, high-resolution IMS, or LC-MS [160] and multi-omics integration to improve the diagnosis, prognostics, and monitoring of mixed dementia and enable trials of combinatorial therapies.

7. Cross-Dementia Comparison of GSL Profiles: Insights and Limitations

GSLs have emerged as critical modulators of neurodegenerative processes across multiple dementia subtypes, reflecting both the conserved and disease-specific mechanisms of pathology.
In AD, high-resolution MS studies consistently show reductions in GM1, GD1a, GD1b, and GT1b across key regions such as the hippocampus and frontal and temporal cortices. These losses are often accompanied by the accumulation of simpler structures, particularly GM2, GM3, and GD3, localized to the peripheries of Aβ plaques. These alterations facilitate the aggregation of both Aβ and tau, highlighting the role of GGs in both initiating and propagating AD pathology. Moreover, subtle changes in the ratio of long-chain base species, such as d18:1 versus d20:1, influence Aβ binding dynamics and plaque composition, suggesting that minor lipidomic variations can critically shape disease progression.
Despite the insights provided by these studies, several methodological and interpretative challenges exist. Many MS-based analyses rely on postmortem tissue, limiting the ability to assess longitudinal changes and early-stage disease processes. Animal models often exaggerate plaque pathology relative to humans, potentially inflating observed GSL alterations. Inter-study variability is common, reflecting differences in GSL extraction and purification methods, MS performances and parameters, and data normalization. Systems biology approaches suggest that GG loss in AD is coupled to the dysregulation of synaptic proteins, APP-processing complexes, and neuroinflammatory mediators. However, establishing causality between GG alterations and neurodegeneration remains a challenge, as lipid changes may represent both pathogenic drivers and compensatory responses.
FTD exhibits a distinct GSL signature relative to AD. GM1, GD1a, and GT1b are reduced in frontal and temporal white matter, whereas hippocampal regions are relatively spared. Notably, c-series GGs, including GQ1c and GT3, may accumulate in degenerating neurons, potentially reflecting compensatory mechanisms responding to tau-mediated cytoskeletal destabilization. Unlike AD, the accumulation of simple GGs such as GM2 and GM3 is less prominent, implying that the deposition of simple GGs is less central to FTD pathophysiology. This aspect highlights a critical distinction between tau-dominant dementias and amyloid-centered disorders: in FTD, GGs are linked to intracellular cytoskeletal disruptions rather than extracellular protein aggregation. Nevertheless, most insights derive from animal models or small patient cohorts; hence, regional heterogeneity may obscure broader systemic patterns.
DLB and PDD illustrate how disrupted GSL metabolism contributes to α-synuclein pathology. Simple GGs, particularly GM2 and GM3, promote Lewy body formation, while the depletion of GM1 and GD1a impairs lipid raft integrity, worsening cognitive and motor deficits. MSI shows region-specific GSL changes linked to symptom profiles, though distinguishing disease effects from aging remains challenging. Overall, alteredendocytosis, vesicular trafficking, and neuroinflammation appear to be related to the GSL-α-synuclein connection
HD shows early and severe deficits in GGs such as GM1, GM2, GD1a, and GD1b likely driven by mHTT-related disruptions in vesicular trafficking and signaling. Importantly, these changes may precede neurodegeneration and serve as early biomarkers, though most evidence comes from animal models. The causal link between GG loss and mHTT aggregation remains unclear, and human validation is limited. Furthermore, integrated analyses suggest that GG disturbances contribute to cytoskeletal, mitochondrial, and synaptic dysfunction underlying cognitive and motor decline.
Mixed dementia, a combination of AD and vascular pathology, exhibits synergistic GSL dysregulation. The early accumulation of GM3 and GM2 is observed in ischemic or hypoperfused regions, while the progressive depletion of GM1, GD1a, and GT1b mirrors amyloid-driven cortical and hippocampal degeneration. However, deciphering the contributions of vascular injury vs. amyloid pathology remains challenging, particularly due to the lack of standardized postmortem tissue protocols and longitudinal imaging approaches.
A comparative perspective across dementias highlights that complex GG reduction, especially GM1, GD1a, GD1b, and GT1b, is a common feature, likely contributing to lipid raft destabilization and synaptic vulnerability. Conversely, the accumulation of simple GGs such as GM2 and GM3 is prominent in AD, DLB, and mixed dementia but less evident in FTD, reflecting disease-specific proteinopathies and regional susceptibilities. C-series GGs, including GQ1c, appear selectively in FTD and AD, potentially marking tau-associated degeneration. Furthermore, shifts in long-chain base composition (d18:1 vs. d20:1) in AD affect plaque-associated GG distributions, revealing subtle lipidomic remodeling as a determinant of protein aggregation kinetics. These observations demonstrate the value of disease-specific glycolipidomic profiling for uncovering the mechanisms of neurodegeneration.
Overall, from amethodological point of view, MS has revolutionized the study of GSLs in dementia, providing unparalleled sensitivity, specificity, and resolution. MALDI-MSI, electrospray ionization (ESI)–MS and LC-MS/MS enable the detection of both quantitative and spatial changes in their composition, allowing for correlations with specific brain regions. However, limitations persist in all studies, and they are related to postmortem tissue degradation, inter-study variability, and the challenge of integrating MS data across platforms. Also, the cross-sectional nature of many studies limits insights into temporal dynamics, while animal models may not fully reproduce human disease, particularly for the cases of dementia. Moreover, systems biology is able to integrate glycolipid alterations with molecular networks to reveal potential therapeutic hubs, though translating these insights into clinical strategies remains challenging. Therapeutically, GSLs represent promising targets. In AD, GM1 supplementation modulated Aβ aggregation and enhanced neurotrophic signaling. Preclinical studies in HD and PD models suggest similar benefits, with GM1 restoring synaptic function and mitigating protein aggregation. Beyond direct supplementation, the modulation of GG metabolism via glycosyltransferase inhibitors, sphingolipid-targeted enzymes, or small molecules that stabilize specific GG species offers potential alternatives for disease-modifying interventions.
Finally, all findings indicate that, overall, GSLs act as both shared and disease-specific drivers of dementia, where common complex GG depletion contributes to synaptic vulnerability; however, GG patterns vary across diseases.

8. Conclusions and Perspectives

Research into the role of GSLs in dementia is beginning to redesign our perspectives on the molecular bases of neurodegenerative diseases. Data collected from multiple studies now clearly indicate that GSLs are not just inactive compositional building blocks of neuronal membranes but dynamic regulators of brain processes. As discussed above, the recurring patterns indicate core weaknesses in neuronal GSL regulation, whereas the distinct changes cause the diverse symptoms and progression seen across different dementias. In this context, Table 4 presents a synthetic view on the altered GSL expression specific to each dementia type discussed, along with possible biomarkers.
Technological refinements in MS have been crucial in enabling recent discoveries in glycolipid research, providing acritical basis for discovering novel lipid signatures in dementia, and reshaping our understanding of glycolipid biology. By offering unprecedented sensitivity, resolving power, and detailed structural information, modern MS has created opportunities to map complex lipid alterations with high accuracy and depth.
Up-to-date MS-based analytical platforms have the capability to detect and characterize a wide range of glycolipids with far greater precision than before, allowing us to build detailed maps of lipid changes in brains affected by dementia. When these lipidomic datasets are connected with systems biology approaches, they can be placed into larger networks of the processes involving other biomolecules. This kind of integrative view is crucial since dementia is not driven by a single factor but emerges from the intersection of many disrupted pathways.
Several promising pathways emerge when considering future directions. Hence, one key priority is to establish whether glycolipid changes occur early enough to serve as warning signals before clinical symptoms appear. This would provide the opportunity to use these glycoconjugates as biomarkers for risk assessment and early diagnosis. Equally critical is the alignment of methodologies and protocols across the laboratories involved in the study of dementia-associated glycolipids in order to generate results that are reliably compared, reproducible, and validated in larger patient cohorts.
Beyond description, there is also a pressing need to move toward mechanistic studies that clarify how specific glycolipid changes influence neuronal survival, immune responses, and protein aggregation.
Noteworthy also are the potential therapeutic implications and prospects. Hence, if certain glycolipids prove to be drivers rather than bystanders in dementia, they could become targets for interventions aimed at stabilizing neuronal membranes, modulating inflammation, or preventing toxic protein buildup. Even if they are not direct drivers, their measurable changes could complement other diagnostic tools, contributing to more precise personalized treatment strategies.
Moreover, the progress of AI presents opportunities for glycolipid research in dementia, facilitating the integration and analysis of complex datasets. ML algorithms can uncover subtle patterns in high-dimensional lipidomic data that might elude conventional analysis, linking glycolipid changes to specific disease stages or phenotypes. AI-driven models can integrate lipidomic profiles with other omics layers, such as genomics, proteomics, and transcriptomics, to construct systems-level maps of dementia pathophysiology.
When combined with MS, the potential of AI expands even further. MS produces vast, highly detailed datasets, while AI can process them at scale, revealing patterns and correlations that remain hidden to traditional approaches. This synergy enables a more precise mapping of glycolipid alterations across dementia types, enhancing biomarker discovery and deepening mechanistic insight. AI is also able to greatly improve MS workflows by automating data annotation, the interpretation of spectra, and structural identification while predicting the functional consequences of the detected changes in the glycolipid expression pattern.
In conclusion, MS and systems biology have developed to the point where detailed molecular insights can be translated into practical clinical advances. Based on ongoing research, which aims to connect novel findings at the molecular level with clinical outcomes, this research direction has the potential to significantly improve the methods of early dementia detection and its treatment.

Author Contributions

Conceptualization, A.D.Z., M.S. and L.D.; resources, M.S., R.I., M.-R.B. and L.D.; data curation, M.S., R.I., M.-R.B. and L.D.; writing—original draft preparation, M.S., R.I. and M.-R.B.; writing—review and editing, A.D.Z. and L.D.; supervision, A.D.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant of the Ministry of Research, Innovation and Digitization, CNCS-UEFISCDI, project number PN-IV-P2-2.1-TE-2023-0175, within PNCDI IV, and by Aurel Vlaicu University of Arad, Romania, grant number UAV-IRG-1-2025-8.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAlzheimer’s disease
AIartificial intelligence
APPamyloid precursor protein
amyloid-β peptide
BBBblood–brain barrier
BMPbis(monoacylglycero)phosphate
bvFTDbehavioral variant FTD
CEcholesteryl ester
CSFcerebrospinal fluid
DAT-SPECTdopamine transporter–single-photon emission computed tomography
DESIdesorption electrospray ionization
DLBdementia with Lewy body
ELISAenzyme-linked immunosorbent assay
ESIelectrospray ionization
FDG-PETfluoro-deoxyglucose positron emission tomography
FTDfrontotemporal dementia
GalCergalactosylceramide
GalSphgalactosylsphingosine
GCaseβ-glucocerebrosidase
GGganglioside
GlcCerglucosylceramide
GlcSphglucosylsphingosine
GRNprogranulin gene
GroPInglycerophosphoinositol
GSLglycosphingolipid
HDHuntington’s disease
HDDHuntington’s disease dementia
Hex1Cersmonohexosylceramides
HEXAhexosaminidase subunit alpha gene
HexCershexosylceramides
HTThuntingtin gene
IMSion mobility spectrometry
LacCerlactosylceramide
LBLewy body
LBDLewy body dementia
LC-MSliquid chromatography coupled with mass spectrometry
MALDImatrix-assisted laser desorption/ionization
mHTTmutant huntingtin
MRImagnetic resonance imaging
MLmachine learning
MSmass spectrometry
MS/MStandem mass spectrometry
MSImass spectrometry imaging
NfLneurofilament light chain
NFTneurofibrillary tangle
PCphosphatidylcholine
PDParkinson’s disease
PDDParkinson’s disease dementia
PEphosphatidylethanolamine
PETpositron emission tomography
PGphosphatidylglycerol
PGRNprogranulin
PIphosphatidylinositol
PSphosphatidylserine
p-tauphosphorylated tau
PTMpost-translational modification
QTOFquadrupole time of flight
SHexCersulfatide
SIMSsecondary ion mass spectrometry
SMsphingomyelin
SPsenile plaque
TGtriacylglycerol
TLCthin-layer chromatography
TMMtransgenic mouse model
UHPLC MS/MSultra-high-performance liquid chromatography coupled to tandem MS
VDvascular dementia
VLCFAvery-long-chain fatty acid

References

  1. Gale, S.A.; Acar, D.; Daffner, K.R. Dementia. Am. J. Med. 2018, 131, 1161–1169. [Google Scholar] [CrossRef]
  2. Duara, R.; Barker, W. Heterogeneity in Alzheimer’s Disease Diagnosis and Progression Rates: Implications for Therapeutic Trials. Neurotherapeutics 2022, 19, 8–25. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, J.; Zhang, Y.; Wang, J.; Xia, Y.; Zhang, J.; Chen, L. Recent Advances in Alzheimer’s Disease: Mechanisms, Clinical Trials and New Drug Development Strategies. Signal Transduct. Target. Ther. 2024, 9, 211. [Google Scholar] [CrossRef] [PubMed]
  4. Hobbs, N.Z.; Barnes, J.; Frost, C.; Henley, S.M.; Wild, E.J.; Macdonald, K.; Barker, R.A.; Scahill, R.I.; Fox, N.C.; Tabrizi, S.J. Onset and Progression of Pathologic Atrophy in Huntington Disease: A Longitudinal MR Imaging Study. AJNR Am. J. Neuroradiol. 2010, 31, 1036–1041. [Google Scholar] [CrossRef]
  5. Aarsland, D.; Kurz, M.W. The Epidemiology of Dementia Associated with Parkinson’s Disease. Brain Pathol. 2010, 20, 633–639. [Google Scholar] [CrossRef]
  6. Custodio, N.; Montesinos, R.; Lira, D.; Herrera-Pérez, E.; Bardales, Y.; Valeriano-Lorenzo, L. Mixed Dementia: A Review of the Evidence. Dement. Neuropsychol. 2017, 11, 364–370. [Google Scholar] [CrossRef]
  7. Attems, J.; Jellinger, K.A. The Overlap between Vascular Disease and Alzheimer’s Disease—Lessons from Pathology. BMC Med. 2014, 12, 206. [Google Scholar] [CrossRef]
  8. Vollhardt, A.; Frölich, L.; Stockbauer, A.C.; Danek, A.; Schmitz, C.; Wahl, A.-S. Towards a Better Diagnosis and Treatment of Dementia: Identifying Common and Distinct Neuropathological Mechanisms in Alzheimer’s and Vascular Dementia. Neurobiol. Dis. 2025, 208, 106638. [Google Scholar] [CrossRef]
  9. Mollah, S.A.; Nayak, A.; Barhai, S.; Maity, U. A Comprehensive Review on Frontotemporal Dementia: Its Impact on Language, Speech and Behavior. Dement. Neuropsychol. 2024, 18, e20230072. [Google Scholar] [CrossRef]
  10. Foxe, D.; Muggleton, J.; Cheung, S.C.; Mueller, N.; Ahmed, R.M.; Narasimhan, M.; Burrell, J.R.; Hwang, Y.T.; Cordato, N.J.; Piguet, O. Survival Rates in Frontotemporal Dementia and Alzheimer’s Disease. Neurodegener. Dis. Manag. 2025, 15, 191–197. [Google Scholar] [CrossRef]
  11. Manabe, T.; Fujikura, Y.; Mizukami, K.; Akatsu, H.; Kudo, K. Pneumonia-Associated Death in Patients with Dementia: A Systematic Review and Meta-Analysis. PLoS ONE 2019, 14, e0213825. [Google Scholar] [CrossRef] [PubMed]
  12. Amoatika, D.A.; Absher, J.R.; Khan, M.T.F.; Miller, M.C. Dementia Deaths Most Commonly Result from Heart and Lung Disease: Evidence from the South Carolina Alzheimer’s Disease Registry. Biomedicines 2025, 13, 1321. [Google Scholar] [CrossRef] [PubMed]
  13. Yao, J.; Liu, S.; Chen, Q. Mortality Rate of Pulmonary Infection in Senile Dementia Patients: A Systematic Review and Meta-Analysis. Medicine 2024, 103, e39816. [Google Scholar] [CrossRef] [PubMed]
  14. Walker, Z.; Possin, K.L.; Boeve, B.F.; Aarsland, D. Lewy Body Dementias. Lancet 2015, 386, 1683–1697. [Google Scholar] [CrossRef]
  15. Muangpaisan, W. Clinical Differences among Four Common Dementia Syndromes. Geriatr. Aging 2007, 10, 425–429. [Google Scholar]
  16. Harciarek, M.; Jodzio, K. Neuropsychological Differences between Frontotemporal Dementia and Alzheimer’s Disease: A Review. Neuropsychol. Rev. 2005, 15, 131–145. [Google Scholar] [CrossRef]
  17. He, S.; Xu, Z.; Han, X. Lipidome Disruption in Alzheimer’s Disease Brain: Detection, Pathological Mechanisms, and Therapeutic Implications. Mol. Neurodegener. 2025, 20, 11. [Google Scholar] [CrossRef]
  18. Osetrova, M.; Tkachev, A.; Mair, W.; Guijarro Larraz, P.; Efimova, O.; Kurochkin, I.; Stekolshchikova, E.; Anikanov, N.; Foo, J.C.; Cazenave-Gassiot, A.; et al. Lipidome Atlas of the Adult Human Brain. Nat. Commun. 2024, 15, 4455. [Google Scholar] [CrossRef]
  19. Grassi, S.; Giussani, P.; Mauri, L.; Prioni, S.; Sonnino, S.; Prinetti, A. Lipid Rafts and Neurodegeneration: Structural and Functional Roles in Physiologic Aging and Neurodegenerative Diseases. J. Lipid Res. 2020, 61, 636–654. [Google Scholar] [CrossRef]
  20. Ledeen, R.; Wu, G. Gangliosides of the Nervous System. Methods Mol. Biol. 2018, 1804, 19–55. [Google Scholar] [CrossRef]
  21. Blomqvist, M.; Zetterberg, H.; Blennow, K.; Månsson, J.E. Sulfatide in Health and Disease: The Evaluation of Sulfatide in Cerebrospinal Fluid as a Possible Biomarker for Neurodegeneration. Mol. Cell.Neurosci. 2021, 116, 103670. [Google Scholar] [CrossRef]
  22. Wei, J.; Wong, L.C.; Boland, S. Lipids as Emerging Biomarkers in Neurodegenerative Diseases. Int. J. Mol. Sci. 2023, 25, 131. [Google Scholar] [CrossRef]
  23. Ferreira, C.R.; Gahl, W.A. Lysosomal Storage Diseases. Transl. Sci. Rare Dis. 2017, 2, 1–71. [Google Scholar] [CrossRef]
  24. Müthing, J. High-Resolution Thin-Layer Chromatography of Gangliosides Methods. J. Chromatogr. A 1996, 720, 3–25. [Google Scholar] [CrossRef]
  25. Nishina, K.A.; Supattapone, S. Immunodetection of Glycophosphatidylinositol-Anchored Proteins Following Treatment with Phospholipase C. Anal. Biochem. 2007, 363, 318–320. [Google Scholar] [CrossRef] [PubMed]
  26. Dehelean, L.; Sarbu, M.; Petrut, A.; Zamfir, A.D. Trends in Glycolipid Biomarker Discovery in Neurodegenerative Disorders by Mass Spectrometry. Adv. Exp. Med. Biol. 2019, 1140, 703–729. [Google Scholar] [CrossRef] [PubMed]
  27. Touboul, D.; Gaudin, M. Lipidomics of Alzheimer’s Disease. Bioanalysis 2014, 6, 541–561. [Google Scholar] [CrossRef] [PubMed]
  28. Suzuki, A.; Suzuki, M.; Ito, E.; Nitta, T.; Inokuchi, J.I. Mass Spectrometry of Gangliosides. Methods Mol. Biol. 2018, 1804, 207–221. [Google Scholar] [CrossRef]
  29. Shon, H.K.; Son, J.G.; Lee, S.Y.; Moon, J.H.; Lee, G.S.; Kim, K.S.; Lee, T.G. Comparison Study of Mouse Brain Tissue by Using ToF-SIMS within Static Limits and Hybrid SIMS Beyond Static Limits (Dynamic Mode). Biointerphases 2023, 18, 031005. [Google Scholar] [CrossRef]
  30. Sarbu, M.; Fabris, D.; Vukelić, Ž.; Clemmer, D.E.; Zamfir, A.D. Ion Mobility Mass Spectrometry Reveals Rare Sialylated Glycosphingolipid Structures in Human Cerebrospinal Fluid. Molecules 2022, 27, 743. [Google Scholar] [CrossRef]
  31. Biricioiu, M.R.; Sarbu, M.; Ica, R.; Vukelić, Ž.; Clemmer, D.E.; Zamfir, A.D. Human Cerebellum Gangliosides: A Comprehensive Analysis by Ion Mobility Tandem Mass Spectrometry. J. Am. Soc. Mass Spectrom. 2024, 35, 683–695. [Google Scholar] [CrossRef]
  32. Ica, R.; Sarbu, M.; Biricioiu, R.; Fabris, D.; Vukelić, Ž.; Zamfir, A.D. Novel Application of Ion Mobility Mass Spectrometry Reveals Complex Ganglioside Landscape in Diffuse Astrocytoma Peritumoral Regions. Int. J. Mol. Sci. 2025, 26, 8433. [Google Scholar] [CrossRef]
  33. Xu, H.; Boucher, F.R.; Nguyen, T.T.; Taylor, G.P.; Tomlinson, J.J.; Ortega, R.A.; Simons, B.; Schlossmacher, M.G.; Saunders-Pullman, R.; Shaw, W.; et al. DMS as an Orthogonal Separation to LC/ESI/MS/MS for Quantifying Isomeric Cerebrosides in Plasma and Cerebrospinal Fluid. J. Lipid Res. 2019, 60, 200–211. [Google Scholar] [CrossRef] [PubMed]
  34. Michno, W.; Bowman, A.; Jha, D.; Minta, K.; Ge, J.; Koutarapu, S.; Zetterberg, H.; Blennow, K.; Lashley, T.; Heeren, R.M.A.; et al. Spatial Neurolipidomics at the Single Amyloid-β Plaque Level in Postmortem Human Alzheimer’s Disease Brain. ACS Chem. Neurosci. 2024, 15, 877–888. [Google Scholar] [CrossRef] [PubMed]
  35. Evers, B.M.; Rodriguez-Navas, C.; Tesla, R.J.; Prange-Kiel, J.; Wasser, C.R.; Yoo, K.S.; McDonald, J.; Cenik, B.; Ravenscroft, T.A.; Plattner, F.; et al. Lipidomic and Transcriptomic Basis of Lysosomal Dysfunction in Progranulin Deficiency. Cell Rep. 2017, 20, 2565–2574. [Google Scholar] [CrossRef]
  36. Shen, H.; Yu, Y.; Wang, J.; Nie, Y.; Tang, Y.; Qu, M. Plasma Lipidomic Signatures of Dementia with Lewy Bodies Revealed by Machine Learning, and Compared to Alzheimer’s Disease. Alzheimers Res. Ther. 2024, 16, 226. [Google Scholar] [CrossRef] [PubMed]
  37. Galleguillos, D.; Zhao, Y.; Pan, B.; Vandermeer, B.; Zaidi, A.; Al Hamarneh, Y.N.; Sarna, J.; Suchowersky, O.; Curtis, J.; Sipione, S. Plasma Gangliosides Correlate with Disease Stages and Symptom Severity in Huntington’s Disease Carriers. bioRxiv 2025. [Google Scholar] [CrossRef]
  38. Toprakcioglu, Z.; Jayaram, A.K.; Knowles, T.P.J. Ganglioside Lipids Inhibit the Aggregation of the Alzheimer’s Amyloid-β Peptide. RSC Chem. Biol. 2025, 6, 809–822. [Google Scholar] [CrossRef]
  39. Li, Q.; Jia, C.; Wu, H.; Liao, Y.; Yang, K.; Li, S.; Zhang, J.; Wang, J.; Li, G.; Guan, F.; et al. Nao Tan Qing Ameliorates Alzheimer’s Disease-like Pathology by Regulating Glycolipid Metabolism and Neuroinflammation: A Network Pharmacology Analysis and Biological Validation. Pharmacol. Res. 2022, 185, 106489. [Google Scholar] [CrossRef]
  40. Kamatham, P.T.; Shukla, R.; Khatri, D.K.; Vora, L.K. Pathogenesis, diagnostics, and therapeutics for Alzheimer’s disease: Breaking the memory barrier. Ageing Res. Rev. 2024, 101, 102481. [Google Scholar] [CrossRef]
  41. Zhang, H.; Tahami Monfared, A.A.; Zhang, Q.; Honig, L.S. Incidence and prevalence of Alzheimer’s disease in Medicare beneficiaries. Neurol. Ther. 2025, 14, 319–333. [Google Scholar] [CrossRef] [PubMed]
  42. Ariga, T.; Kubota, M.; Nakane, M.; Oguro, K.; Yu, R.K.; Ando, S. Anti-Chol-1 antigen, GQ1bα, antibodies are associated with Alzheimer’s disease. PLoS ONE 2013, 8, e63326. [Google Scholar] [CrossRef] [PubMed]
  43. Xu, L.; Wang, Z.; Li, M.; Li, Q. Global incidence trends and projections of Alzheimer disease and other dementias: An age-period-cohort analysis 2021. J. Glob. Health 2025, 15, 04156. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, X.; Zhou, R.; Sun, X.; Li, J.; Wang, J.; Yue, W.; Wang, L.; Liu, H.; Shi, Y.; Zhang, D. Preferential regulation of γ-secretase-mediated cleavage of APP by ganglioside GM1 reveals a potential therapeutic target for Alzheimer’s disease. Adv. Sci. 2023, 10, e2303411. [Google Scholar] [CrossRef]
  45. Hirano-Sakamaki, W.; Sugiyama, E.; Hayasaka, T.; Ravid, R.; Setou, M.; Taki, T. Alzheimer’s disease is associated with disordered localization of ganglioside GM1 molecular species in the human dentate gyrus. FEBS Lett. 2015, 589, 3611–3616. [Google Scholar] [CrossRef]
  46. Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
  47. Anand, S.; Barnes, J.M.; Young, S.A.; Garcia, D.M.; Tolley, H.D.; Kauwe, J.S.; Graves, S.W. Discovery and confirmation of diagnostic serum lipid biomarkers for Alzheimer’s disease using direct infusion mass spectrometry. J. Alzheimers Dis. 2017, 59, 277–290. [Google Scholar] [CrossRef]
  48. Han, X.; Holtzman, D.M.; McKeel, D.W., Jr.; Kelley, J.; Morris, J.C. Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer’s disease: Potential role in disease pathogenesis. J. Neurochem. 2002, 82, 809–818. [Google Scholar] [CrossRef]
  49. Yuyama, K.; Sun, H.; Sakai, S.; Mitsutake, S.; Okada, M.; Tahara, H.; Furukawa, J.-I.; Fujitani, N.; Shinohara, Y.; Igarashi, Y. Decreased amyloid-β pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in Alzheimer model mice. J. Biol. Chem. 2014, 289, 24488–24498. [Google Scholar] [CrossRef]
  50. González-Domínguez, R.; García-Barrera, T.; Gómez-Ariza, J.L. Metabolomic study of lipids in serum for biomarker discovery in Alzheimer’s disease using direct infusion mass spectrometry. J. Pharm. Biomed. Anal. 2014, 98, 321–326. [Google Scholar] [CrossRef]
  51. Michno, W.; Wehrli, P.M.; Zetterberg, H.; Blennow, K.; Hanrieder, J. GM1 locates to mature amyloid structures implicating a prominent role for glycolipid-protein interactions in Alzheimer pathology. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 458–467. [Google Scholar] [CrossRef]
  52. Sarbu, M.; Ica, R.; Zamfir, A.D. Gangliosides as biomarkers of human brain diseases: Trends in discovery and characterization by high-performance mass spectrometry. Int. J. Mol. Sci. 2022, 23, 693. [Google Scholar] [CrossRef]
  53. Matsuzaki, K. Aβ-Ganglioside Interactions in the Pathogenesis of Alzheimer’s Disease. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183233. [Google Scholar] [CrossRef] [PubMed]
  54. Chi, E.Y.; Frey, S.L.; Lee, K.Y. Ganglioside GM1-mediated amyloid-beta fibrillogenesis and membrane disruption. Biochemistry 2007, 46, 1913–1924. [Google Scholar] [CrossRef] [PubMed]
  55. Goux, W.J.; Liu, B.; Shumburo, A.M.; Parikh, S.; Sparkman, D.R. A quantitative assessment of glycolipid and protein associated with paired helical filament preparations from Alzheimer’s diseased brain. J. AlzheimersDis. 2001, 3, 455–466. [Google Scholar] [CrossRef] [PubMed]
  56. Xiao, S.; Wei, X.; Han, B.; Shi, X.; Wei, C.; Liang, R.; Sun, J.; Zhang, Z.; Han, Z.; Shen, L. Quantitative analysis of targeted lipidomics in the hippocampus of APP/PS1 mice employing the UHPLC-MS/MS method. Front. AgingNeurosci. 2025, 17, 1561831. [Google Scholar] [CrossRef]
  57. Chakraborty, A.; Praharaj, S.K.; Prabhu, R.K.; Prabhu, M.M. Lipidomics and cognitive dysfunction—A narrative review. Turk. J. Biochem. 2020, 45, 109–119. [Google Scholar] [CrossRef]
  58. Ollen-Bittle, N.; Pejhan, S.; Pasternak, S.H.; Keene, C.D.; Zhang, Q.; Whitehead, S.N. Co-registration of MALDI-MSI and histology demonstrates gangliosides co-localize with amyloid beta plaques in Alzheimer’s disease. Acta Neuropathol. 2024, 147, 105. [Google Scholar] [CrossRef]
  59. Zhang, Q.; Li, Y.; Sui, P.; Sun, X.H.; Gao, Y.; Wang, C.Y. MALDI mass spectrometry imaging discloses the decline of sulfoglycosphingolipid and glycerophosphoinositol species in the brain regions related to cognition in a mouse model of Alzheimer’s disease. Talanta 2024, 266, 125022. [Google Scholar] [CrossRef]
  60. Blank, M.; Hopf, C. Spatially resolved mass spectrometry analysis of amyloid plaque-associated lipids. J. Neurochem. 2021, 159, 330–342. [Google Scholar] [CrossRef]
  61. Ariga, T.; Jarvis, W.D.; Yu, R.K. Role of sphingolipid-mediated cell death in neurodegenerative diseases. J. Lipid Res. 1998, 39, 1–16. [Google Scholar] [CrossRef] [PubMed]
  62. Sanni, A.; Bennett, A.I.; Adeniyi, M.; Mechref, Y. Dysregulated Lipids in Alzheimer’s Disease: Insights into Biological Pathways through LC-MS/MS Analysis of Human Brain Tissues. ACS Chem. Neurosci. 2025, 16, 3694–3712. [Google Scholar] [CrossRef] [PubMed]
  63. Cho, B.G.; Veillon, L.; Mechref, Y. N-Glycan Profile of Cerebrospinal Fluids from Alzheimer’s Disease Patients Using Liquid Chromatography with Mass Spectrometry. J. Proteome Res. 2019, 18, 3770–3779. [Google Scholar] [CrossRef]
  64. Reyes, C.D.G.; Hakim, M.A.; Atashi, M.; Goli, M.; Gautam, S.; Wang, J.; Bennett, A.I.; Zhu, J.; Lubman, D.M.; Mechref, Y. LC-MS/MS Isomeric Profiling of N-Glycans Derived from Low-Abundant Serum Glycoproteins in Mild Cognitive Impairment Patients. Biomolecules 2022, 12, 1657. [Google Scholar] [CrossRef]
  65. Li, H.; Liu, Y.; Wang, Z.; Xie, Y.; Yang, L.; Zhao, Y.; Tian, R. Mass spectrometry-based ganglioside profiling provides potential insights into Alzheimer’s disease development. J. Chromatogr. A 2022, 1676, 463196. [Google Scholar] [CrossRef]
  66. Wang, W.; Myers, S.J.; Ollen-Bittle, N.; Whitehead, S.N. Elevation of ganglioside degradation pathway drives GM2 and GM3 within amyloid plaques in a transgenic mouse model of Alzheimer’s disease. Neurobiol. Dis. 2025, 205, 106798. [Google Scholar] [CrossRef]
  67. Caughlin, S.; Maheshwari, S.; Agca, Y.; Agca, C.; Harris, A.J.; Jurcic, K.; Yeung, K.K.-C.; Cechetto, D.F.; Whitehead, S.N. Membrane-lipid homeostasis in a prodromal rat model of Alzheimer’s disease: Characteristic profiles in ganglioside distributions during aging detected using MALDI imaging mass spectrometry. Biochim. Biophys. Acta Gen. Subj. 2018, 1862, 1327–1338. [Google Scholar] [CrossRef]
  68. Kaya, I.; Jennische, E.; Dunevall, J.; Lange, S.; Ewing, A.G.; Malmberg, P.; Baykal, A.T.; Fletcher, J.S. Spatial lipidomics reveals region and long chain base specific accumulations of monosialogangliosides in amyloid plaques in familial Alzheimer’s disease mice (5xFAD) brain. ACS Chem. Neurosci. 2020, 11, 14–24. [Google Scholar] [CrossRef]
  69. Zhang, Y.; Wang, J.; Liu, J.; Han, J.; Xiong, S.; Yong, W.; Zhao, Z. Combination of ESI and MALDI mass spectrometry for qualitative, semi-quantitative and in situanalysis of gangliosides in brain. Sci. Rep. 2016, 6, 25289. [Google Scholar] [CrossRef]
  70. Good, C.J.; Bowman, A.P.; Klein, C.; Awwad, K.; Buck, W.R.; Yang, J.; Wagner, D.S. Spatial mapping of gangliosides and proteins in amyloid beta plaques at cellular resolution using mass spectrometry imaging and MALDI-IHC. J. Mass Spectrom. 2025, 60, e5161. [Google Scholar] [CrossRef]
  71. Taki, T. An approach to glycobiology from glycolipidomics: Ganglioside molecular scanning in the brains of patients with Alzheimer’s disease by TLC-blot/matrix assisted laser desorption/ionization-time of flight MS. Biol. Pharm. Bull. 2012, 35, 1642–1647. [Google Scholar] [CrossRef]
  72. Zhang, L.; Li, L.; Meng, F.; Yu, J.; He, F.; Lin, Y.; Su, Y.; Hu, M.; Liu, X.; Liu, Y.; et al. Serum metabolites differentiate amnestic mild cognitive impairment from healthy controls and predict early Alzheimer’s disease via untargeted lipidomics analysis. Front. Neurol. 2021, 12, 704582. [Google Scholar] [CrossRef]
  73. Fantini, J.; Yahi, N.; Garmy, N. Cholesterol accelerates the binding of Alzheimer’s β-amyloid peptide to ganglioside GM1 through a universal hydrogen-bond-dependent sterol tuning of glycolipid conformation. Front. Physiol. 2013, 4, 120. [Google Scholar] [CrossRef] [PubMed]
  74. Yahi, N.; Fantini, J. Deciphering the glycolipid code of Alzheimer’s and Parkinson’s amyloid proteins allowed the creation of a universal ganglioside-binding peptide. PLoS ONE 2014, 9, e104751. [Google Scholar] [CrossRef] [PubMed]
  75. Kakio, A.; Nishimoto, S.I.; Yanagisawa, K.; Kozutsumi, Y.; Matsuzaki, K. Cholesterol-dependent formation of GM1 ganglioside-bound amyloid beta-protein, an endogenous seed for Alzheimer amyloid. J. Biol. Chem. 2001, 276, 24985–24990. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, C.C.; Kanekiyo, T.; Xu, H.; Bu, G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat. Rev. Neurol. 2013, 9, 106–118. [Google Scholar] [CrossRef]
  77. Thambisetty, M.; Simmons, A.; Velayudhan, L.; Hye, A.; Campbell, J.; Zhang, Y.; Wahlund, L.-O.; Westman, E.; Kinsey, A.; Güntert, A.; et al. Association of plasma clusterin concentration with severity, pathology, and progression in Alzheimer disease. Arch. Gen. Psychiatry 2010, 67, 739–748. [Google Scholar] [CrossRef]
  78. Fonseca, M.I.; Chu, S.H.; Hernandez, M.X.; Fang, M.J.; Modarresi, L.; Selvan, P.; MacGregor, G.R.; Tenner, A.J. Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain. J. Neuroinflammation 2017, 14, 48. [Google Scholar] [CrossRef]
  79. Kanekiyo, T.; Bu, G. The low-density lipoprotein receptor-related protein 1 and amyloid-β clearance in Alzheimer’s disease. Front. Aging Neurosci. 2014, 6, 93. [Google Scholar] [CrossRef]
  80. Peng, W.; Gutierrez Reyes, C.D.; Gautam, S.; Yu, A.; Cho, B.G.; Goli, M.; Donohoo, K.; Mondello, S.; Kobeissy, F.; Mechref, Y. MS-based glycomics and glycoproteomics methods enabling isomeric characterization. Mass Spectrom. Rev. 2023, 42, 577–616. [Google Scholar] [CrossRef]
  81. Prasad, S.; Katta, M.R.; Abhishek, S.; Sridhar, R.; Valisekka, S.S.; Hameed, M.; Kaur, J.; Walia, N. Recent advances in Lewy body dementia: A comprehensive review. Disease-a-Month 2023, 69, 101441. [Google Scholar] [CrossRef]
  82. McKeith, I.; Mintzer, J.; Aarsland, D.; Burn, D.; Chiu, H.; Cohen-Mansfield, J.; Dickson, D.; Dubois, B.; Duda, J.E.; Feldman, H.; et al. Dementia with Lewy bodies. Lancet Neurol. 2004, 3, 19–28. [Google Scholar] [CrossRef] [PubMed]
  83. Kasuga, K.; Nishizawa, M.; Ikeuchi, T. α-Synuclein as CSF and blood biomarker of dementia with Lewy bodies. Int. J. Alzheimers Dis. 2012, 2012, 437025. [Google Scholar] [CrossRef] [PubMed]
  84. Scamarcia, P.G.; Agosta, F.; Caso, F.; Filippi, M. Update on neuroimaging in non-Alzheimer’s disease dementia: A focus on the Lewy body disease spectrum. Curr. Opin. Neurol. 2021, 34, 532–538. [Google Scholar] [CrossRef] [PubMed]
  85. Kantarci, K.; Lowe, V.J.; Chen, Q.; Przybelski, S.A.; Lesnick, T.G.; Schwarz, C.G.; Senjem, M.L.; Gunter, J.L.; Jack, C.R., Jr.; Graff-Radford, J.; et al. β-Amyloid PET and neuropathology in dementia with Lewy bodies. Neurology 2020, 94, e282–e291. [Google Scholar] [CrossRef]
  86. Senanarong, V.; Wachirutmangur, L.; Rattanabunnakit, C.; Srivanitchapoom, P.; Udomphanthurak, S. Plasma alpha synuclein (α-syn) as a potential biomarker of diseases with synucleinopathy. Alzheimers Dement. 2020, 16, e044409. [Google Scholar] [CrossRef]
  87. Mukaetova-Ladinska, E.B.; Monteith, R.; Perry, E.K. Cerebrospinal fluid biomarkers for dementia with Lewy bodies. Int. J. Alzheimers Dis. 2010, 2010, 536538. [Google Scholar] [CrossRef]
  88. Mollenhauer, B.; Schlossmacher, M.G. CSF synuclein: Adding to the biomarker footprint of dementia with Lewy bodies. J. Neurol. Neurosurg. Psychiatry 2010, 81, 590–591. [Google Scholar] [CrossRef]
  89. Bongianni, M.; Ladogana, A.; Capaldi, S.; Klotz, S.; Baiardi, S.; Cagnin, A.; Perra, D.; Poleggi, A.; Antonelli, F.; Ciccocioppo, F.; et al. Alpha-synuclein RT-QuIC assay in cerebrospinal fluid of patients with dementia with Lewy bodies. Ann. Clin. Transl. Neurol. 2019, 6, 2120–2126. [Google Scholar] [CrossRef]
  90. Bargar, C.; Wang, W.; Gunzler, S.A.; LeFevre, A.; Wang, Z.; Siderowf, A.; Weintraub, D.; Lieberman, A.; Hurtig, H.I.; Espay, A.J.; et al. Streamlined alpha-synuclein RT-QuIC assay for various biospecimens in Parkinson’s disease and dementia with Lewy bodies. Acta Neuropathol. Commun. 2021, 9, 62. [Google Scholar] [CrossRef]
  91. Rossi, M.; Baiardi, S.; Teunissen, C.E.; Quadalti, C.; van de Beek, M.; Mammana, A.; Zenesini, C.; Bartoletti-Stella, A.; Baiardi, S.; Capellari, S.; et al. Diagnostic value of the CSF alpha-synuclein real-time quaking-induced conversion assay at the prodromal MCI stage of dementia with Lewy bodies. Neurology 2021, 97, e930–e940. [Google Scholar] [CrossRef]
  92. Mavroudis, I.; Petridis, F.; Kazis, D. Cerebrospinal fluid, imaging, and physiological biomarkers in dementia with Lewy bodies. Am. J. Alzheimers Dis. Other Dementias® 2019, 34, 421–432. [Google Scholar] [CrossRef]
  93. Vrillon, A.; Bousiges, O.; Götze, K.; Demuynck, C.; Muller, C.; Ravier, A.; Schorr, B.; Philippi, N.; Hourregue, C.; Cognat, E.; et al. Plasma biomarkers of amyloid, tau, axonal, and neuroinflammation pathologies in dementia with Lewy bodies. Alzheimers Res. Ther. 2024, 16, 146. [Google Scholar] [CrossRef]
  94. Peña-Bautista, C.; Bolsewig, K.; Gonzalez, M.C.; Ashton, N.J.; Aarsland, D.; Zetterberg, H.; Westman, E.; Bousiges, O.; Blanc, F.; E Teunissen, C.; et al. The association between plasma and MRI biomarkers in dementia with Lewy bodies. Alzheimers Res. Ther. 2025, 17, 197. [Google Scholar] [CrossRef]
  95. Wang, S.Y.; Chen, W.; Xu, W.; Li, J.-Q.; Hou, X.-H.; Ou, Y.-N.; Yu, J.-T.; Tan, L. Neurofilament light chain in cerebrospinal fluid and blood as a biomarker for neurodegenerative diseases: A systematic review and meta-analysis. J. Alzheimers Dis. 2019, 72, 1353–1361. [Google Scholar] [CrossRef]
  96. Lourenco, M.V.; Ribeiro, F.C.; Santos, L.E.; Beckman, D.; Melo, H.M.; Sudo, F.K.; Drummond, C.; Assunção, N.; Vanderborght, B.; Tovar-Moll, F.; et al. Cerebrospinal fluid neurotransmitters, cytokines, and chemokines in Alzheimer’s and Lewy body diseases. J. Alzheimers Dis. 2021, 82, 1067–1074. [Google Scholar] [CrossRef] [PubMed]
  97. Savica, R.; Murray, M.E.; Persson, X.M.; Kantarci, K.; Parisi, J.E.; Dickson, D.W.; Petersen, R.C.; Ferman, T.J.; Boeve, B.F.; Mielke, M.M. Plasma sphingolipid changes with autopsy-confirmed Lewy body or Alzheimer’s pathology. Alzheimers Dement. 2016, 3, 43–50. [Google Scholar] [CrossRef] [PubMed]
  98. Lerche, S.; Wurster, I.; Valente, E.M.; Avenali, M.; Samaniego, D.; Martínez-Vicente, M.; Hernández-Vara, J.; Laguna, A.; Sturchio, A.; Svenningsson, P.; et al. CSF d18:1 sphingolipid species in Parkinson disease and dementia with Lewy bodies with and without GBA1 variants. npjPark. Dis. 2024, 10, 198. [Google Scholar] [CrossRef]
  99. Miglis, M.G.; Adler, C.H.; Antelmi, E.; Arnaldi, D.; Baldelli, L.; Boeve, B.F.; Cesari, M.; Dall’ANtonia, I.; Diederich, N.J.; Doppler, K.; et al. Biomarkers of conversion to α-synucleinopathy in isolated rapid-eye-movement sleep behaviour disorder. Lancet Neurol. 2021, 20, 671–684. [Google Scholar] [CrossRef] [PubMed]
  100. Gomperts, S.N. Lewy body dementias: Dementia with Lewy bodies and Parkinson disease dementia. Continuum 2016, 22, 435–463. [Google Scholar] [CrossRef]
  101. Yamashita, K.Y.; Bhoopatiraju, S.; Silverglate, B.D.; Grossberg, G.T. Biomarkers in Parkinson’s disease: A state of the art review. Biomark. Neuropsychiatry 2023, 9, 100074. [Google Scholar] [CrossRef]
  102. Hely, M.A.; Reid, W.G.; Adena, M.A.; Halliday, G.M.; Morris, J.G. The Sydney multicenter study of Parkinson’s disease: The inevitability of dementia at 20 years. Mov. Disord. 2008, 23, 837–844. [Google Scholar] [CrossRef] [PubMed]
  103. Aarsland, D.; Andersen, K.; Larsen, J.P.; Lolk, A.; Kragh-Sørensen, P. Prevalence and characteristics of dementia in Parkinson disease: An 8-year prospective study. Arch. Neurol. 2003, 60, 387–392. [Google Scholar] [CrossRef] [PubMed]
  104. Savica, R.; Knopman, D.S. Dementia with Lewy bodies. In Neurodegeneration; Schapira, A., Wszolek, Z., Dawson, T.M., Wood, N., Eds.; Wiley-Blackwell: Hoboken, NJ, USA, 2017; pp. 83–92. [Google Scholar] [CrossRef]
  105. Zardini Buzatto, A.; Tatlay, J.; Bajwa, B.; Mung, D.; Camicioli, R.; Dixon, R.A.; Li, L. Comprehensive serum lipidomics for detecting incipient dementia in Parkinson’s disease. J. Proteome Res. 2021, 20, 4053–4067. [Google Scholar] [CrossRef] [PubMed]
  106. Azevedo, R.; Jacquemin, C.; Villain, N.; Fenaille, F.; Lamari, F.; Becher, F. Mass spectrometry for neurobiomarker discovery: The relevance of post-translational modifications. Cells 2022, 11, 1279. [Google Scholar] [CrossRef]
  107. Schmid, A.W.; Fauvet, B.; Moniatte, M.; Lashuel, H.A. Alpha-synuclein post-translational modifications as potential biomarkers for Parkinson disease and other synucleinopathies. Mol. Cell.Proteom. 2013, 12, 3543–3558. [Google Scholar] [CrossRef]
  108. Anderson, J.P.; Walker, D.E.; Goldstein, J.M.; de Laat, R.; Banducci, K.; Caccavello, R.J.; Barbour, R.; Huang, J.; Kling, K.; Lee, M.; et al. Phosphorylation of Ser-129 is the dominant pathological modification of alpha-synuclein in familial and sporadic Lewy body disease. J. Biol. Chem. 2006, 281, 29739–29752. [Google Scholar] [CrossRef]
  109. Neumann, M.; Sampathu, D.M.; Kwong, L.K.; Truax, A.C.; Micsenyi, M.C.; Chou, T.T.; Bruce, J.; Schuck, T.; Grossman, M.; Clark, C.M.; et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006, 314, 130–133. [Google Scholar] [CrossRef]
  110. Wesseling, H.; Mair, W.; Kumar, M.; Schlaffner, C.N.; Tang, S.; Beerepoot, P.; Fatou, B.; Guise, A.J.; Cheng, L.; Takeda, S.; et al. Tau PTM profiles identify patient heterogeneity and stages of Alzheimer’s disease. Cell 2020, 183, 1699–1713.e13. [Google Scholar] [CrossRef]
  111. Mielke, M.M.; Maetzler, W.; Haughey, N.J.; Bandaru, V.V.R.; Savica, R.; Deuschle, C.; Gasser, T.; Hauser, A.-K.; Gräber-Sultan, S.; Schleicher, E.; et al. Plasma ceramide and glucosylceramide metabolism is altered in sporadic Parkinson’s disease and associated with cognitive impairment: A pilot study. PLoS ONE 2013, 8, e73094. [Google Scholar] [CrossRef]
  112. Xing, Y.; Tang, Y.; Zhao, L.; Wang, Q.; Qin, W.; Ji, X.; Zhang, J.; Jia, J. Associations between plasma ceramides and cognitive and neuropsychiatric manifestations in Parkinson’s disease dementia. J. Neurol. Sci. 2016, 370, 82–87. [Google Scholar] [CrossRef]
  113. Galper, J.; Mori, G.; McDonald, G.; Rastegar, D.A.; Pickford, R.; Lewis, S.J.G.; Halliday, G.M.; Kim, W.S.; Dzamko, N. Prediction of motor and non-motor Parkinson’s disease symptoms using serum lipidomics and machine learning: A 2-year study. npjPark. Dis. 2024, 10, 123. [Google Scholar] [CrossRef] [PubMed]
  114. Avisar, H.; Guardia-Laguarta, C.; Area-Gomez, E.; Surface, M.; Chan, A.K.; Alcalay, R.N.; Lerner, B. Lipidomics prediction of Parkinson’s disease severity: A machine-learning analysis. J. Park. Dis. 2021, 11, 1141–1155. [Google Scholar] [CrossRef]
  115. Raz, L.; Knoefel, J.; Bhaskar, K. The neuropathology and cerebrovascular mechanisms of dementia. J. Cereb. Blood Flow Metab. 2016, 36, 172–186. [Google Scholar] [CrossRef] [PubMed]
  116. Peet, B.T.; Spina, S.; Mundada, N.; La Joie, R. Neuroimaging in frontotemporal dementia: Heterogeneity and relationships with underlying neuropathology. Neurotherapeutics 2021, 18, 728–752. [Google Scholar] [CrossRef] [PubMed]
  117. Erkkinen, M.G.; Kim, M.O.; Geschwind, M.D. Clinical neurology and epidemiology of the major neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2018, 10, a033118. [Google Scholar] [CrossRef]
  118. Lashley, T.; Rohrer, J.D.; Mead, S.; Revesz, T. An update on clinical, genetic and pathological aspects of frontotemporal lobar degenerations. Neuropathol. Appl. Neurobiol. 2015, 41, 858–881. [Google Scholar] [CrossRef]
  119. Ambaw, Y.A.; Ljubenkov, P.A.; Singh, S.; Hamed, A.; Boland, S.; Boxer, A.L.; Walther, T.C.; Farese, R.V. Plasma lipidome dysregulation in frontotemporal dementia reveals shared, genotype-specific, and severity-linked alterations. AlzheimersDement. 2025, 21, e70631. [Google Scholar] [CrossRef]
  120. Marian, O.C.; Matis, S.; Dobson-Stone, C.; Kim, W.S.; Kwok, J.B.; Piguet, O.; Halliday, G.M.; Landin-Romero, R.; Don, A.S. Reduced plasma hexosylceramides in frontotemporal dementia are a biomarker of white matter integrity. Alzheimers Dement. 2025, 17, e70131. [Google Scholar] [CrossRef]
  121. Boland, S.; Swarup, S.; Ambaw, Y.A.; Malia, P.C.; Richards, R.C.; Fischer, A.W.; Singh, S.; Aggarwal, G.; Spina, S.; Nana, A.L.; et al. Deficiency of the frontotemporal dementia gene GRN results in gangliosidosis. Nat. Commun. 2022, 13, 5924. [Google Scholar] [CrossRef]
  122. Kamp, P.E.; den Hartog Jager, W.A.; Maathuis, J.; de Groot, P.A.; de Jong, J.M.; Bolhuis, P.A. Brain gangliosides in the presenile dementia of Pick. J. Neurol. Neurosurg. Psychiatry 1986, 49, 881–885. [Google Scholar] [CrossRef]
  123. Kim, W.S.; Jary, E.; Pickford, R.; He, Y.; Ahmed, R.M.; Piguet, O.; Hodges, J.R.; Halliday, G.M. Lipidomics analysis of behavioral variant frontotemporal dementia: A scope for biomarker development. Front. Neurol. 2018, 9, 104. [Google Scholar] [CrossRef] [PubMed]
  124. Marian, O.C.; Teo, J.D.; Lee, J.Y.; Song, H.; Kwok, J.B.; Landin-Romero, R.; Halliday, G.; Don, A.S. Disrupted myelin lipid metabolism differentiates frontotemporal dementia caused by GRN and C9orf72 gene mutations. Acta Neuropathol. Commun. 2023, 11, 52. [Google Scholar] [CrossRef] [PubMed]
  125. Arrant, A.E.; Roth, J.R.; Boyle, N.R.; Kashyap, S.N.; Hoffmann, M.Q.; Murchison, C.F.; Ramos, E.M.; Nana, A.L.; Spina, S.; Grinberg, L.T.; et al. Impaired β-glucocerebrosidase activity and processing in frontotemporal dementia due to progranulin mutations. Acta Neuropathol. Commun. 2019, 7, 218. [Google Scholar] [CrossRef] [PubMed]
  126. He, Y.; Phan, K.; Bhatia, S.; Pickford, R.; Fu, Y.; Yang, Y.; Hodges, J.R.; Piguet, O.; Halliday, G.M.; Kim, W.S. Increased VLCFA-lipids and ELOVL4 underlie neurodegeneration in frontotemporal dementia. Sci. Rep. 2021, 11, 21348. [Google Scholar] [CrossRef]
  127. Aqel, S.; Ahmad, J.; Saleh, I.; Fathima, A.; Al Thani, A.A.; Mohamed, W.M.Y.; Shaito, A.A. Advances in Huntington’s disease biomarkers: A 10-year bibliometric analysis and a comprehensive review. Biology 2025, 14, 129. [Google Scholar] [CrossRef]
  128. Seeley, C.; Kegel-Gleason, K.B. Taming the Huntington’s disease proteome: What have we learned? J. Huntingt. Dis. 2021, 10, 239–257. [Google Scholar] [CrossRef]
  129. Peavy, G.M.; Jacobson, M.W.; Goldstein, J.L.; Hamilton, J.M.; Kane, A.; Gamst, A.C.; Lessig, S.L.; Lee, J.C.; Corey-Bloom, J. Cognitive and functional decline in Huntington’s disease: Dementia criteria revisited. Mov. Disord. 2010, 25, 1163–1169. [Google Scholar] [CrossRef]
  130. Snowden, J.S.; Craufurd, D.; Thompson, J.; Neary, D. Psychomotor, executive, and memory function in preclinical Huntington’s disease. J. Clin. Exp. Neuropsychol. 2002, 24, 133–145. [Google Scholar] [CrossRef]
  131. Hamilton, J.M.; Salmon, D.P.; Corey-Bloom, J.; Gamst, A.; Paulsen, J.S.; Jerkins, S.; Jacobson, M.W.; Peavy, G. Behavioural abnormalities contribute to functional decline in Huntington’s disease. J. Neurol. Neurosurg. Psychiatry 2003, 74, 120–122. [Google Scholar] [CrossRef]
  132. McGarry, A.; Gaughan, J.; Hackmyer, C.; Lovett, J.; Khadeer, M.; Shaikh, H.; Pradhan, B.; Ferraro, T.N.; Wainer, I.W.; Moaddel, R. Cross-sectional analysis of plasma and CSF metabolomic markers in Huntington’s disease for participants of varying functional disability: A pilot study. Sci. Rep. 2020, 10, 20490, Erratum in Sci. Rep. 2021, 11, 9947. https://doi.org/10.1038/s41598-021-89167-7. [Google Scholar] [CrossRef] [PubMed]
  133. Hunter, M.; Demarais, N.J.; Faull, R.L.M.; Grey, A.C.; Curtis, M.A. An imaging mass spectrometry atlas of lipids in the human neurologically normal and Huntington’s disease caudate nucleus. J. Neurochem. 2021, 157, 2158–2172. [Google Scholar] [CrossRef] [PubMed]
  134. Phillips, G.R.; Saville, J.T.; Hancock, S.E.; Brown, S.H.J.; Jenner, A.M.; McLean, C.; Fuller, M.; Newell, K.A.; Mitchell, T.W. The long and the short of Huntington’s disease: How the sphingolipid profile is shifted in the caudate of advanced clinical cases. BrainCommun. 2021, 4, fcab303. [Google Scholar] [CrossRef] [PubMed]
  135. Phillips, G.R.; Hancock, S.E.; Brown, S.H.J.; Jenner, A.M.; Kreilaus, F.; Newell, K.A.; Mitchell, T.W. Cholesteryl ester levels are elevated in the caudate and putamen of Huntington’s disease patients. Sci. Rep. 2020, 10, 20314. [Google Scholar] [CrossRef]
  136. Maglione, V.; Marchi, P.; Di Pardo, A.; Lingrell, S.; Horkey, M.; Tidmarsh, E.; Sipione, S. Impaired ganglioside metabolism in Huntington’s disease and neuroprotective role of GM1. J. Neurosci. 2010, 30, 4072–4080. [Google Scholar] [CrossRef]
  137. Di Pardo, A.; Maglione, V.; Alpaugh, M.; Horkey, M.; Atwal, R.S.; Sassone, J.; Ciammola, A.; Steffan, J.S.; Fouad, K.; Truant, R.; et al. Ganglioside GM1 Induces Phosphorylation of Mutant Huntingtin and Restores Normal Motor Behavior in Huntington Disease Mice. Proc. Natl. Acad. Sci. USA 2012, 109, 3528–3533. [Google Scholar] [CrossRef]
  138. Alpaugh, M.; Galleguillos, D.; Forero, J.; Morales, L.C.; Lackey, S.W.; Kar, P.; Di Pardo, A.; Holt, A.; Kerr, B.J.; Todd, K.G.; et al. Disease-modifying effects of ganglioside GM1 in Huntington’s disease models. EMBO Mol. Med. 2017, 9, 1537–1557. [Google Scholar] [CrossRef]
  139. Byrne, L.M.; Rodrigues, F.B.; Johnson, E.B.; Wijeratne, P.A.; De Vita, E.; Alexander, D.C.; Palermo, G.; Czech, C.; Schobel, S.; Scahill, R.I.; et al. Evaluation of mutant huntingtin and neurofilament proteins as potential markers in Huntington’s disease. Sci. Transl. Med. 2018, 10, eaat7108. [Google Scholar] [CrossRef]
  140. Ghofrani-Jahromi, M.; Poudel, G.R.; Razi, A.; Abeyasinghe, P.M.; Paulsen, J.S.; Tabrizi, S.J.; Saha, S.; Georgiou-Karistianis, N. Prognostic enrichment for early-stage Huntington’s disease: An explainable machine learning approach for clinical trial. Neuroimage Clin. 2024, 43, 103650. [Google Scholar] [CrossRef]
  141. Ganesh, S.; Chithambaram, T.; Krishnan, N.R.; Vincent, D.R.; Kaliappan, J.; Srinivasan, K. Exploring Huntington’s disease diagnosis via artificial intelligence models: A comprehensive review. Diagnostics 2023, 13, 3592. [Google Scholar] [CrossRef]
  142. Meneses, A.; Koga, S.; O’Leary, J.; Dickson, D.W.; Bu, G.; Zhao, N. TDP-43 pathology in Alzheimer’s disease. Mol. Neurodegener. 2021, 16, 84. [Google Scholar] [CrossRef] [PubMed]
  143. Knopman, D.S.; Parisi, J.E.; Boeve, B.F.; Cha, R.H.; Apaydin, H.; Salviati, A.; Edland, S.D.; Rocca, W.A. Vascular dementia in a population-based autopsy study. Arch. Neurol. 2003, 60, 569–575. [Google Scholar] [CrossRef] [PubMed]
  144. Buciuc, M.; Whitwell, J.L.; Boeve, B.F.; Ferman, T.J.; Graff-Radford, J.; Savica, R.; Kantarci, K.; Fields, J.A.; Knopman, D.S.; Petersen, R.C.; et al. TDP-43 is associated with a reduced likelihood of rendering a clinical diagnosis of dementia with Lewy bodies in autopsy-confirmed cases of transitional/diffuse Lewy body disease. J. Neurol. 2020, 267, 1444–1453. [Google Scholar] [CrossRef] [PubMed]
  145. Song, R.; Pan, K.Y.; Xu, H.; Qi, X.; Buchman, A.S.; Bennett, D.A.; Xu, W. Association of cardiovascular risk burden with risk of dementia and brain pathologies: A population-based cohort study. Alzheimers Dement. 2021, 17, 1914–1922. [Google Scholar] [CrossRef] [PubMed]
  146. Livingston, G.; Huntley, J.; Liu, K.Y.; Costafreda, S.G.; Selbæk, G.; Alladi, S.; Ames, D.; Banerjee, S.; Burns, A.; Brayne, C.; et al. Dementia prevention, intervention, and care: 2024 report of the Lancet standing Commission. Lancet 2024, 404, 572–628. [Google Scholar] [CrossRef]
  147. Leocadi, M.; Canu, E.; Paldino, A.; Agosta, F.; Filippi, M. Awareness impairment in Alzheimer’s disease and frontotemporal dementia: A systematic MRI review. J. Neurol. 2023, 270, 1880–1907. [Google Scholar] [CrossRef]
  148. Rowley, P.A.; Samsonov, A.A.; Betthauser, T.J.; Pirasteh, A.; Johnson, S.C.; Eisenmenger, L.B. Amyloid and Tau PET imaging of Alzheimer disease and other neurodegenerative conditions. Semin. Ultrasound CT MRI 2020, 41, 572–583. [Google Scholar] [CrossRef]
  149. Olsson, B.; Lautner, R.; Andreasson, U.; Öhrfelt, A.; Portelius, E.; Bjerke, M.; Hölttä, M.; Rosén, C.; Olsson, C.; Strobel, G.; et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: A systematic review and meta-analysis. Lancet Neurol. 2016, 15, 673–684. [Google Scholar] [CrossRef]
  150. Manjavong, M.; Kang, J.M.; Diaz, A.; Ashford, M.T.; Eichenbaum, J.; Aaronson, A.; Miller, M.J.; Mackin, S.; Tank, R.; Weiner, M.; et al. Performance of plasma biomarkers combined with structural MRI to identify candidate participants for Alzheimer’s disease-modifying therapy. J. Prev. Alzheimers Dis. 2024, 11, 1198–1205. [Google Scholar] [CrossRef]
  151. Mok, V.C.T.; Cai, Y.; Markus, H.S. Vascular cognitive impairment and dementia: Mechanisms, treatment, and future directions. Int. J. Stroke 2024, 19, 838–856. [Google Scholar] [CrossRef]
  152. Wong, E.C.; Chui, H.C. Vascular cognitive impairment and dementia. Continuum 2022, 28, 750–780. [Google Scholar] [CrossRef]
  153. Tisher, A.; Salardini, A. A comprehensive update on treatment of dementia. Semin. Neurol. 2019, 39, 167–178. [Google Scholar] [CrossRef] [PubMed]
  154. Kapasi, A.; James, B.D.; Yu, L.; Sood, A.; Arvanitakis, Z.; Bennett, D.A.; Boyle, P.; Schneider, J.A. Mixed pathologies and cognitive outcomes in persons considered for anti-amyloid treatment eligibility assessment: A community-based study. Neurology 2025, 105, e214004. [Google Scholar] [CrossRef] [PubMed]
  155. Alrouji, M.; Alshammari, M.S.; Tasqeeruddin, S.; Shamsi, A. Interplay between aging and tau pathology in Alzheimer’s disease: Mechanisms and translational perspectives. Antioxidants 2025, 14, 774. [Google Scholar] [CrossRef] [PubMed]
  156. Mani, S.; Wasnik, S.; Shandilya, C.; Srivastava, V.; Khan, S.; Singh, K.K. Pathogenic synergy: Dysfunctional mitochondria and neuroinflammation in neurodegenerative diseases associated with aging. Front. Aging 2025, 6, 1615764. [Google Scholar] [CrossRef]
  157. Yu, W.; Ying, J.; Wang, X.; Liu, X.; Zhao, T.; Yoon, S.; Zheng, Q.; Fang, Y.; Yang, D.; Hua, F. The involvement of lactosylceramide in central nervous system inflammation related to neurodegenerative disease. Front. Aging Neurosci. 2021, 13, 691230. [Google Scholar] [CrossRef]
  158. Teunissen, C.E.; Kimble, L.; Bayoumy, S.; Bolsewig, K.; Burtscher, F.; Coppens, S.; Das, S.; Gogishvili, D.; Fernandes Gomes, B.; Gómez de San José, N.; et al. Methods to discover and validate biofluid-based biomarkers in neurodegenerative dementias. Mol. Cell. Proteom. 2023, 22, 100629. [Google Scholar] [CrossRef]
  159. Yilmaz, A.; Ashrafi, N.; Ashrafi, R.; Akyol, S.; Saiyed, N.; Kerševičiūtė, I.; Gabrielaite, M.; Gordevicius, J.; Graham, S.F. Lipid profiling of Parkinson’s disease brain highlights disruption in lysophosphatidylcholines, and triacylglycerol metabolism. npjPark. Dis. 2025, 11, 159. [Google Scholar] [CrossRef]
  160. Kelley, A.R. Mass spectrometry-based analysis of lipid involvement in Alzheimer’s disease pathology—A review. Metabolites 2022, 12, 510. [Google Scholar] [CrossRef]
  161. Cilento, E.M.; Jin, L.; Stewart, T.; Shi, M.; Sheng, L.; Zhang, J. Mass spectrometry: A platform for biomarker discovery and validation for Alzheimer’s and Parkinson’s diseases. J. Neurochem. 2019, 151, 397–416. [Google Scholar] [CrossRef]
  162. Lam, S.M.; Wang, Y.; Duan, X.; Wenk, M.R.; Kalaria, R.N.; Chen, C.P.; Lai, M.K.; Shui, G. Brain lipidomes of subcortical ischemic vascular dementia and mixed dementia. Neurobiol. Aging 2014, 35, 2369–2381. [Google Scholar] [CrossRef]
  163. Hendriks, T.F.E.; Krestensen, K.K.; Mohren, R.; Vandenbosch, M.; De Vleeschouwer, S.; Heeren, R.M.A.; Cuypers, E. MALDI-MSI-LC-MS/MS workflow for single-section single step combined proteomics and quantitative lipidomics. Anal. Chem. 2024, 96, 4266–4274. [Google Scholar] [CrossRef] [PubMed]
  164. Noel, A.; Ingrand, S.; Barrier, L. Ganglioside and related-sphingolipid profiles are altered in a cellular model of Alzheimer’s disease. Biochimie 2017, 137, 158–164. [Google Scholar] [CrossRef] [PubMed]
  165. Chua, X.Y.; Torta, F.; Chong, J.R.; Venketasubramanian, N.; Hilal, S.; Wenk, M.R.; Chen, C.P.; Arumugam, T.V.; Herr, D.R.; Lai, M.K.P. Lipidomics profiling reveals distinct patterns of plasma sphingolipid alterations in Alzheimer’s disease and vascular dementia. Alzheimers Res. Ther. 2023, 15, 214. [Google Scholar] [CrossRef] [PubMed]
  166. Zimmer, V.C.; Lauer, A.A.; Haupenthal, V.; Stahlmann, C.P.; Mett, J.; Grösgen, S.; Hundsdörfer, B.; Rothhaar, T.; Endres, K.; Eckhardt, M.; et al. A bidirectional link between sulfatide and Alzheimer’s disease. Cell Chem. Biol. 2024, 31, 265–283.e7. [Google Scholar] [CrossRef]
  167. Reza, S.; Ugorski, M.; Suchański, J. Glucosylceramide and galactosylceramide, small glycosphingolipids with significant impact on health and disease. Glycobiology 2021, 31, 1416–1434. [Google Scholar] [CrossRef]
  168. Pujol-Lereis, L.M. Alteration of sphingolipids in biofluids: Implications for neurodegenerative diseases. Int. J. Mol. Sci. 2019, 20, 3564. [Google Scholar] [CrossRef]
  169. Koal, T.; Klavins, K.; Seppi, D.; Kemmler, G.; Humpel, C. Sphingomyelin SM(d18:1/18:0) is significantly enhanced in cerebrospinal fluid samples dichotomized by pathological amyloid-β42, tau, and phospho-tau-181 levels. J. Alzheimers Dis. 2015, 44, 1193–1201. [Google Scholar] [CrossRef]
  170. Montine, T.J.; Morrow, J.D. Fatty acid oxidation in the pathogenesis of Alzheimer’s disease. Am. J. Pathol. 2005, 166, 1283–1289. [Google Scholar] [CrossRef]
  171. Caughlin, S.; Hepburn, J.D.; Park, D.H.; Jurcic, K.; Yeung, K.K.-C.; Cechetto, D.F.; Whitehead, S.N. Increased expression of simple ganglioside species GM2 and GM3 detected by MALDI imaging mass spectrometry in a combined rat model of Aβ toxicity and stroke. PLoS ONE 2015, 10, e0130364. [Google Scholar] [CrossRef]
Figure 1. Molecular basis and hallmarks of major dementia types (schematic of neuron created in BioRender. Sarbu, M. (2025), https://BioRender.com/9gcz0t7, accessed on 5 October 2025).
Figure 1. Molecular basis and hallmarks of major dementia types (schematic of neuron created in BioRender. Sarbu, M. (2025), https://BioRender.com/9gcz0t7, accessed on 5 October 2025).
Biomedicines 13 02854 g001
Figure 2. A systems workflow illustrating the integration of MS-based glycolipid profiling with bioinformatic and systems biology approaches to characterize glycolipid dysregulation in dementia (Created in BioRender. Sarbu, M. (2025) https://BioRender.com/n5tserg, accessed on 3 November 2025).
Figure 2. A systems workflow illustrating the integration of MS-based glycolipid profiling with bioinformatic and systems biology approaches to characterize glycolipid dysregulation in dementia (Created in BioRender. Sarbu, M. (2025) https://BioRender.com/n5tserg, accessed on 3 November 2025).
Biomedicines 13 02854 g002
Figure 3. Metabolism of glycosphingolipids and their interactions with proteins.
Figure 3. Metabolism of glycosphingolipids and their interactions with proteins.
Biomedicines 13 02854 g003
Table 1. Key features of different dementia types [4,5,6,7,8,9,10,11,12,13,14,15,16].
Table 1. Key features of different dementia types [4,5,6,7,8,9,10,11,12,13,14,15,16].
DisorderDementia of Alzheimer’s Type (AD)Dementia with Lewy Body (DLB)Frontotemporal Dementia (FTD)Parkinson’s Disease Dementia (PDD)Huntington’s Disease (HD)Mixed Dementia
Features
OnsetPresenile or senileSenilePresenileLate onsetPresenileSenile onset
Age at diagnosis<65 s or >65 s50 s–80 s40 s and early 60 s>70 s30 s or 40 s>65
Patient profilePredominantly femaleSlight male predominanceMale predominanceMale predominanceNo gender preferenceNo gender preference
Brain abnormalitiesAccumulation of amyloid plaques and tau tangles α-synuclein aggregation in cortical and subcortical Lewy bodies (LBs); often coexists with ADAbnormal tau and TDP-43 proteins in the frontal and temporal lobesAccumulation of α-synuclein in LBsSpecific inherited gene mutation Accumulation of tau and amyloid plaques
Cerebral damageDiffuse cerebral atrophyWidespread LBpathology; variable cortical atrophySevere atrophy Atrophy in subcortical regions and cortical LB pathologyNeuronal loss in caudate nucleus and putamenCombination of AD and vascular lesions
Prominent symptomsMemory dysfunctionFluctuating cognition, visual hallucinations, and parkinsonism Personality and language disturbancesImpaired attention, executive dysfunction, memory issuesCognitive decline with behavioral disturbancesMemory loss, cognitive decline, executive dysfunction
Visuospatial abilitiesSeverely impairedMarkedly impairedPreservedModerately impairedImpairedOften impaired
Language problemsUnderstanding; speakingSpeakingThinking; understanding; readingThinking; speakingSpeakingVariable; may mirror AD difficulties
MoodDepression, anxiety, suspiciousnessDepression, anxiety, apathy, confusionMarked irritability, lack of guilt, alexithymia euphoria, apathyDepression, anxiety, apathyDepression, irritability, aggression, apathyDepression, anxiety, apathy
Intellectual deficitYesYesNoYesYesYes
Psychotic featuresDelusion of misidentification or prejudice secondary to memory impairment type Prominent visual hallucinations, delusionsRare persecutory delusionsand bizarre behaviorsVisual hallucinations, paranoid delusionsPsychosis Possible delusions and hallucinations
Appetite, dietary changeAnorexia and weight lossWeight loss Increased appetite, carbohydrate craving 80%, weight gainWeight loss Weight loss Variable
(weight loss or gain)
Progression to death11.8 ± 0.6 years5–8 years after diagnosis8.7 ± 1.2 years5–10 years after onset15–20 years after onsetVariable (faster than single dementia types)
Cause of deathAspiration pneumonia Aspiration pneumonia, complications of immobility, and infectionsPhysical changes that can cause skin, urinary tract, and/or lung infectionsComplications from immobility, aspiration pneumoniaComplications from immobility, infections, aspiration pneumoniaCardiovascular disease, pneumonia, infections
Table 2. MS platforms for glycolipid biomarkers. Strengths and limitations [26,27,28,29,30,31,32,33].
Table 2. MS platforms for glycolipid biomarkers. Strengths and limitations [26,27,28,29,30,31,32,33].
Platform/WorkflowKey FeaturesAdvantagesLimitationsTypical Use Cases
LC-MS/MS with Orbitrap/QuadrupoleTime of Flight (QTOF)Chromatographic separation
High-resolution tandem MS (MS/MS)
Quantitative, robust, reduces isobaric interference
Structural info on fatty acyl chains
Sensitive
Requires optimized chromatography
Need for derivatization or specialized columns
Discovery and validation
Shotgun Lipidomics (Direct Infusion, Orbitrap/TripleTOF)Rapid, high-throughput profiling
Direct infusion, no LC
Broad coverage
Quick surveys
Minimal preparation
Ion suppression
Poor isomer/isobar separation
Less quantitative
Initial screening before separation
MALDI-MSISpatial tissue mapping
Moderate to high resolution
Links molecular and anatomical data
Enables regional distribution analysis
High sensitivity with derivatization and high-resolution analyzers
Lower quantitation than LC-MS
Matrix/analyte suppression
Mapping
Correlation with plaques, vessels, microinfarcts
Desorption Electrospray Ionization (DESI)–MSI and Secondary Ion Mass Spectrometry (SIMS) ImagingAmbient ionization (DESI)
Utra-high-resolution imaging (SIMS)
Minimal sample preparation (DESI)
Sub-micron resolution (SIMS)
Limited mass range and fragmentation (SIMS)
Complex data analysis
Subcellular mapping
Complementary spatial lipidomics
IMS-MSSeparates isomers and isobars by shape/sizeResolves complex GGs
High confidence in structural identification
Boosts discovery
Requires specialized instrumentsBiomarker discovery
Detailed structural assignment
Targeted Derivatization and
Glycan-Specific Workflows
Chemical modifications
Specialized columns
Improves chromatographic behavior and MS sensitivity
Resolves isomers
Extra sample preparation complexityQuantifying disease-relevant isomers
Quantitative MSI and LC-MS HybridCombines MSI with microextraction and LC-MS/MSSpatial localization and quantitative data
Emerging gold standard
Complex workflowTissue-to-histopathology mapping
Anatomical and quantitative mapping
Table 3. Glycolipid classes relevant to mixed dementia discovered by MS-based approaches.
Table 3. Glycolipid classes relevant to mixed dementia discovered by MS-based approaches.
Glycolipid ClassRepresentative SpeciesRelevance to Mixed DementiaRepresentative Citation
GGs (mono-/di-/tri-sialo)GM1 (d18:1/18:0);
GM2; GM3 (d18:1/16:0; d18:1/18:0);
GD1a; GD1b; GT1b
Abundant in neuronal membranes and synapses
Altered sialylation indicates membrane degradation and inflammation
GM2/GM3 elevated near plaques and in white matter
[66]
Sulfatides (sulfated galactocerebrosides)ST (d18:1/24:0)Enriched in myelin
Early loss linked to AD and vascular myelin injury
Sensitive marker of demyelination
[166]
GalCer/GlcCerGalCer (d18:1/24:0);
GlcCer species
Core myelin lipids
Shifts indicate demyelination in ischemic regions
Reflect altered GSL metabolism
[167]
CeramidesCer (d18:1/16:0);
Cer (d18:1/24:1)
Products of SM breakdown
Elevated in neurodegeneration and vascular inflammation
Promote apoptosis and Aβ production
[168]
SphingomyelinsSM (d18:1/18:0);
SM (36:1)
Structural membrane lipids
SM/ceramide ratio changes reflect membrane injury
Altered in mixed dementia tissue
[169]
GG degradation intermediatesGM2; lactosylceramides (LacCer)Indicate increased glycosidase activity
Reflect lysosomal/autophagy dysfunction
Accumulate around plaques and ischemic zones
[66]
Glycolipid oxidized/
truncated forms
Oxidized ceramides; truncated GGsMarkers of oxidative stress and lipid peroxidation
Elevated near microinfarcts and plaques
[170]
Table 4. Overview of GSL expression in various types of dementia.
Table 4. Overview of GSL expression in various types of dementia.
Dementia TypeMost Promising GSL BiomarkersDirection of ChangeReferences
ADGM1, GM2, GM3, GD1a, GD1b, GD2, GD3, GT1a, GT1b, GQ1b, GQ1bα, di-O-Ac-GT1a, O-Ac-GD1b, ShexCer, GalNAc-GD1a, GGs (d18:1)↑ GM1 (d18:1/18:0), GM2, GM3, GD2, GD3, GT1a, GQ1b, GQ1bα, di-O-Ac-GT1a, O-Ac-GD1b, GGs (d18:1), TG, CE, PE
↓ GD1a, GD1b, GT1b, ShexCer, GalNAc-GD1a
[42,45,51,65,66,70,71,121,122,136,137,138,171]
DLBGM1 and GD1a, Cer (d18:1/18:0), GlcCer (d18:1/18:0), SphM (d18:1/18:0), GlcSph (d18:1), GalSph (d18:1)↑ GM2, GM3
↓ GM1 and GD1a, GalSph and Cer vs. controls/PD
[98]
PDDCer 24:1, 14:0, 18:0, 20:0↓ Cer 24:1, 14:0
↑ Cer 18:0, 20:0
[122]
FTDGM1, GM2, GM3, GD1, GD2, GD3, GT1a, GalNAc-GD1a, HexCers (22:0 GlcCer, GalCer)↑ GM1, GM2, GM3GD1, GD2, GD3, GT1a, GT3, GQ1c
↓GalNAc-GD1a, HexCers (22:0 GlcCer, GalCer)
[120,121,122]
HDCeramides (chain length shift), SM, GM1Loss of very-long-chain Cer/SM
Enrichment inlong-chain Cer/SM
↓ GM1, GM2, GD1a, and GD1b; improvement with exogenous GM1
[134,136,138]
Mixed
dementia
GM2, GM3, Cer (d16:1/24:0), Cer (d18:1/16:0), Hex2Cer (d16:1/16:0), HexCer (d18:1/18:0), SM (d16:1/16:0, 20:0), SM (d18:2/22:0), GlcCer, GalCer, sphingolipids (d16:1, d18:1)↑ GM2, GM3 (in/around Aβ plaques)
↑GM3 and membrane breakdown markers in mixed dementia
↓Sphingolipid d16:1 (VD), GM1, GD1a, and GT1b
↑Sphingolipid d18:1 (AD)
[66,162,165]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sarbu, M.; Ica, R.; Biricioiu, M.-R.; Dehelean, L.; Zamfir, A.D. Glycosphingolipids in Dementia: Insights from Mass Spectrometry and Systems Biology Approaches. Biomedicines 2025, 13, 2854. https://doi.org/10.3390/biomedicines13122854

AMA Style

Sarbu M, Ica R, Biricioiu M-R, Dehelean L, Zamfir AD. Glycosphingolipids in Dementia: Insights from Mass Spectrometry and Systems Biology Approaches. Biomedicines. 2025; 13(12):2854. https://doi.org/10.3390/biomedicines13122854

Chicago/Turabian Style

Sarbu, Mirela, Raluca Ica, Maria-Roxana Biricioiu, Liana Dehelean, and Alina D. Zamfir. 2025. "Glycosphingolipids in Dementia: Insights from Mass Spectrometry and Systems Biology Approaches" Biomedicines 13, no. 12: 2854. https://doi.org/10.3390/biomedicines13122854

APA Style

Sarbu, M., Ica, R., Biricioiu, M.-R., Dehelean, L., & Zamfir, A. D. (2025). Glycosphingolipids in Dementia: Insights from Mass Spectrometry and Systems Biology Approaches. Biomedicines, 13(12), 2854. https://doi.org/10.3390/biomedicines13122854

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop