Cerebrospinal Fluid Biomarkers of Alzheimer Disease
- Authors: Nevzorova K.V.1, Shpilyukova Y.A.1, Shabalina А.A.1, Fedotova E.Y.1, Illarioshkin S.N.1
-
Affiliations:
- Russian Center of Neurology and Neurosciences
- Issue: Vol 19, No 2 (2025)
- Pages: 82-91
- Section: Technologies
- Submitted: 05.08.2024
- Accepted: 24.09.2024
- Published: 26.06.2025
- URL: https://annaly-nevrologii.com/pathID/article/view/1185
- DOI: https://doi.org/10.17816/ACEN.1185
- EDN: https://elibrary.ru/YTDDIC
- ID: 1185
Cite item
Abstract
Alzheimer disease (AD) is a chronic neurodegenerative disorder and the most common cause of dementia in the elderly. Current international guidelines for the clinical diagnosis of AD consider the diagnosis to be both clinical and biological. It requires a specific clinical phenotype and a confirmed biological origin based on biomarkers of amyloid and tau pathology. In Russia, only a few research centers perform laboratory diagnosis of AD using cerebrospinal fluid (CSF) biomarkers. Better access to laboratory diagnosis of AD and wider use of CSF biomarkers in clinical practice will help to assess the true prevalence of AD in the Russian population and to select patients for targeted pathogenic therapies based on the use of monoclonal antibodies against abnormal brain proteins, which have been actively developed in recent years. This review summarizes information on the main CSF biomarkers of AD and their diagnostic and prognostic value.
Full Text
Introduction
Alzheimer disease (AD) is a chronic neurodegenerative disorder and the most common cause of dementia in the elderly [1]. Neuropathologically, AD is characterized by the deposition of beta-amyloid (Aβ) in the brain as extracellular plaques and the formation of intracellular neurofibrillary tangles of phosphorylated tau protein [2].
In Russia, there are approximately 9,000 registered patients with AD [3]. However, it is estimated that more than 90% of AD cases in Russia remain undiagnosed [4]. This is mainly due to a lack of awareness among primary care physicians about the early signs of AD (when symptoms are interpreted as part of normal aging or cerebrovascular disease), a reluctance to make the diagnosis at more advanced stages because of the potential social consequences, or the presence of an atypical clinical phenotype that makes it difficult to identify the nature of the neurodegenerative process without additional diagnostic tools.
Until recently, the diagnosis of AD was based primarily on clinical data, namely on the development of a typical cognitive deficit [5]. However, according to the latest guidelines from the International Working Group for the Clinical Diagnosis of Alzheimer Disease (2021), diagnosis of AD should be based on both clinical and biological features and requires a specific clinical phenotype and confirmation of biological origin based on amyloid and tau pathology biomarkers [6]. Amyloid pathology can be confirmed by low levels of Aβ1-42 in the cerebrospinal fluid (CSF) or the detection of abnormal amyloid deposition in the brain using positron emission tomography (PET). Tau pathology can be diagnosed by high CSF levels of phosphorylated tau protein or abnormal deposition of tau protein identified by brain PET with an appropriate ligand.
In Russia, only a few research centers are able to perform laboratory diagnosis of AD based on CSF biomarkers, whereas it remains inaccessible for most Russian clinics [7–10]. PET scans with ligands for Aβ and tau proteins are not available in every clinic in Russia. However, the need to verify the diagnosis for targeted therapy will soon require a sharp increase in the availability of laboratory diagnostic tools for AD as well as a wider clinical use of CSF biomarkers (as a more accessible method compared to PET).
The aim of this review was to summarize data on the key CSF biomarkers of AD and their diagnostic and predictive value.
Main Pathogenic Mechanisms of Alzheimer Disease
In 1906, Alois Alzheimer first described a clinical case of dementia in a young woman with progressive memory loss, speech, movement and behavioral disorders, and hallucinations. Postmortem brain pathomorphology revealed macroscopic signs of extensive brain atrophy. Using a novel silver impregnation technique for brain histology, Alzheimer identified typical neuropathologic changes namely extracellular amyloid plaques and intracellular neurofibrillary tangles [11]. n 1987, the gene APP (amyloid precursor protein), which encodes the amyloid precursor protein located on chromosome 21, was identified [12]. In 1992, the official amyloid hypothesis of AD was proposed [13].
APP is a transmembrane protein found in many tissues of the body. However, its physiological functions are not fully understood. This protein may be involved in learning, memory and neuroplasticity, including synaptogenesis, which may be a key element of neuroprotection [14]. Proteolytic cleavage of APP can involve two pathways: a non-amyloidogenic pathway that results in the production of soluble α-amyloid and an amyloidogenic pathway that results in the formation of insoluble and aggregation-prone Aβ fragments [15]. According to the amyloid theory, a critical role in AD is attributed to the altered cleavage pattern of the APP protein, leading to excessive production of Aβ peptides. Aβ is formed by the sequential cleavage of APP by specific enzymes such as β-secretase and γ-secretase (identified as a presenilin complex) [16]. γ-Secretase-mediated APP cleavage results in the production of amyloid peptides consisting of 36–43 amino acids [17]: a peptide consisting of 40 amino acids (Aβ40) is produced in larger quantities and a peptide consisting of 42 amino acids (Aβ42) is produced in smaller quantities [18]. Although different isoforms of Aβ can be detected in AD patients, the levels of Aβ42 and Aβ40 and their ratio are considered the most reliable biomarkers for AD [19].
Historically, hyperproduction of Aβ was thought to be the main cause of AD [20, 21]. In recent years, however, a defect in Aβ clearance mechanisms has been suggested to play a major role [22, 23]. Yoon et al. identified four main mechanisms of Aβ clearance, divided into non-enzymatic and enzymatic pathways [24]. The non-enzymatic pathway includes three mechanisms:
1) Drainage of interstitial fluid into the blood through periarterial Virchow–Robin spaces [22];
2) Phagocytosis by microglia or astrocytes [25];
3) Transport across blood vessel walls, mediated by some clearance receptors (low density lipoprotein receptor-related protein 1 (LRP1); very-low-density-low-density lipoprotein receptor (VLDLR); P-glycoprotein) [26].
The enzymatic pathway involves the cleavage of Aβ by proteases, including neprilysin [27], insulin-degrading enzyme [28], matrix metalloproteinase-9 [29], and glutamyl carboxypeptidase II [30]. The imbalance between Aβ peptide production and clearance initiates a cascade of pathological reactions that are the main cause of AD development [15].
Intracellular accumulation of soluble amyloidogenic Aβ oligomers has a neurotoxic effect even before the formation of extracellular plaques, leading to synaptic dysfunction, postsynaptic hyperexcitability, disruption of homeostasis, and increased production of reactive oxygen species in neuronal mitochondria [31, 32]. The extracellular aggregates of insoluble fibrils containing Aβ peptides (amyloid plaques) also have a neurotoxic effect. Simultaneous dysfunction of astrocytes and microglia, the brain’s immune cells, develops. Overproduction of inflammatory cytokines occurs and phagocytosis of Aβ is impaired. These processes activate cell signaling pathways associated with apoptosis and neuronal death [33].
Tau protein is associated with microtubules, is expressed primarily in neurons and is encoded by the MAPT (microtubule-associated protein tau) gene located on chromosome 17. Neuroimaging studies show that the onset and location of tau pathology correspond to both the onset and type of cognitive deficit [34, 35]. The main functions of this protein include stimulation of tubulin polymerization, stabilization of microtubules, and transport of intracellular organelles [36]. Tau aggregation is a multi-step process that likely begins with the hyperphosphorylation of tau protein and its detachment from microtubules. During aggregation, tau protein moves into the somatodendritic regions of neurons where further phosphorylation and structural changes occur. Misfolded proteins begin to aggregate, forming freely spreading pathogenic oligomers, which leads to further disease development, affecting healthy cells and causing neuronal death [37].
Several mechanisms leading to tau hyperphosphorylation and conformational changes and formation of neurofibrillary tangles are described:
1) activation by Aβ proteins of specific enzymes that catalyze hyperphosphorylation;
2) neuroinflammation triggered by Aβ deposition and promoting the activation of pro-inflammatory cytokines;
3) decreased ability to degrade hyperphosphorylated tau proteins;
4) axonal transport defect [38].
Aβ oligomers first induce phosphorylation of tau protein at specific epitopes and then cause cytoskeleton collapse and neuronal degeneration [39].
Cerebrospinal Fluid Biomarkers of Alzheimer Disease
Lumbar puncture is a routine medical procedure used for diagnostic and therapeutic purposes. The CSF is in direct contact with the extracellular space of the brain and spinal cord, and its biochemical changes may reflect the characteristics of neurodegenerative diseases. CSF is the main biological fluid used for the diagnosis of AD [40]. Aβ1-42 and Aβ1-40, total tau (t-tau), and phosphorylated tau (p-tau) are the best-known CSF biomarkers for AD [41].
Aβ1-42
The Aβ1-42 protein in CSF is recognized as a key biomarker for AD. Reduced levels of Aβ1-42 have been shown in several international studies to be highly accurate in diagnosing dementia and mild cognitive impairment in AD. This biomarker has high sensitivity and specificity in diagnosing AD at all stages [42–45]. Reduced levels of Aβ1-42 in CSF are found to be the earliest pathological change in AD, preceding Aβ ligand PET imaging [46]. The concentration of Aβ1-42 declines long before the onset of clinical symptoms [47], making this biomarker particularly suitable for early diagnosis [48].
The mechanisms leading to decreased CSF levels of Aβ1-42 in patients with AD are still unclear. Some authors suggest that this may be due to excessive deposition of Aβ42 in amyloid plaques, as the aggregated state impedes transport of Aβ1-42 from the interstitial fluid into the CSF [49]. Other hypotheses include decreased production rates of Aβ42 [23], increased Aβ42 degradation due to proteolytic breakdown [50] or microglial phagocytosis [51], as well as increased clearance of Aβ1-42 into the blood [52], although these are considered less likely [53].
One limitation of isolated Aβ1-42 studies in CSF is the frequent finding of decreased levels of this biomarker in other neurodegenerative diseases, such as cerebral microangiopathy [54], dementia with Lewy bodies [55], Creutzfeldt–Jakob disease [56], and frontotemporal dementia (FTD) [57]. Although the levels of Aβ1-42 in AD are usually significantly lower than in these diseases, this overlap limits the differential diagnosis.
Aβ1-40
While Aβ1-42 constitutes approximately 10% of the total Aβ peptide population, the protein Aβ1-40 is the dominant form in the brain, CSF, and plasma [58]. The total concentration of Aβ varies little between diseases, and the concentration of Aβ1-40 does not differ significantly between patients with AD, healthy individuals, and patients with dementia of other origin [59]. Therefore, CSF levels of Aβ1-40 may be considered the most accurate reflection of total brain Aβ burden, although the value of this test in isolation remains controversial. Aβ1-40 levels are primarily used to evaluate the Aβ1-42/Aβ1-40 ratio.
The Aβ1-42/Aβ1-40 Ratio
The Aβ1-42/Aβ1-40 ratio was proposed in the late 1990s to improve the differential diagnosis of AD [60]. This ratio is important and accounts for constitutive interindividual differences in total CSF burden of Aβ between high and low amyloid production [61]. Studies have found a high correlation between a lower Aβ1-42/Aβ1-40 ratio and higher levels of total and phosphorylated tau protein [62]. Patients with a lower Aβ1-42/Aβ1-40 ratio show a faster cognitive and functional decline and a more rapid decline in episodic memory [63]. These data demonstrate the advantage of using the Aβ1-42/Aβ1-40 ratio over isolated CSF levels of Aβ1-42 for predicting the progression of cognitive impairment.
Total Tau Protein
The first study that successfully evaluated total t-tau in CSF was published in 1995 and showed that t-tau levels were significantly higher in patients with AD compared to patients with other neurodegenerative diseases and controls [64]. Similar results have been found in hundreds of other studies [65]. However, elevated CSF levels of t-tau were later found in some acute conditions (such as stroke [66], traumatic brain injury [67], Wernicke encephalopathy [68]), as well as in rapidly progressive neurodegenerative diseases (such as Creutzfeldt–Jakob disease [69]). Based on the data obtained, the level of t-tau is proposed to be used as a marker of the activity of the neurodegenerative process or the severity of acute neuronal damage in the brain [70]. In patients with AD, higher levels of t-tau may predict faster clinical progression of the disease [71].
Phosphorylated Tau Protein
Tau protein undergoes multiple post-translational modifications, such as glycosylation [72], glycation (non-enzymatic glycosylation) [73], phosphorylation, etc. Phosphorylation is the most important modification and level phosphorylation regulates the biological activity of the tau protein [74]. Normally, more than 30 different sites of the protein at serine, threonine, or proline positions are phosphorylated [75]. These modifications can control normal biological functions of tau, such as regulating the stability of microtubules, and lead to the development of pathological processes associated with the protein’s ability to self-assemble into neuronal filaments found in neurodegenerative diseases [76].
Tau protein phosphorylated at threonine 181 (p-tau181) in CSF is the best understood form of p-tau as an AD biomarker used in current disease diagnosis [77]. This biomarker (in combination with Aβ42) accurately discriminates between patients with AD and healthy individuals and can also predict cognitive decline in preclinical and prodromal stages of the disease [78]. Levels of p-tau181 are significantly higher in AD than in other tauopathies, including FTD, progressive supranuclear palsy, and corticobasal degeneration. Therefore, this parameter can be used in the differential diagnosis of dementia in these conditions [57, 79, 80].
The levels of tau phosphorylated at positions 217 (p-tau217) and 231 (p-tau231) have received considerable attention in recent years. For example, elevated levels of p-tau217 in CSF have been shown to be the most specific parameter for detecting both preclinical and advanced stages of AD [81]. CSF p-tau217 levels in patients with prodromal stage and AD dementia were several times higher than p-tau181 levels in the same patients [82]. The superiority of p-tau217 over p-tau181 has also been demonstrated in studies showing stronger correlations of p-tau217 with amyloid PET results [83].
For p-tau231, there is evidence that it is most sensitive to the earliest manifestations of amyloid pathology in the medial orbitofrontal cortex, precuneus, and posterior cingulate cortex, before the threshold of pathological amyloid ligand accumulation is reached on PET scans [84]. This biomarker is thought to reach diagnostically significant abnormal levels only at disease onset [85] and may be key to identifying the recently described pre-amyloid phase of AD [86], which occurs before the abnormal accumulation of Aβ is detectable by PET. Stronger correlations between CSF p-tau231 levels and amyloid PET burden in individuals without clinically evident cognitive impairment suggest that increases in CSF p-tau231 levels occur during the lag phase of Aβ protein aggregation in the brain [84].
Markers of Neurodegeneration and Microglial Activation
Although a hallmark of AD is the formation of Aβ and tau protein aggregates in the brain, there are also typical neuroinflammatory responses that occur in the affected brain areas, leading to neuronal dysfunction, neuronal death, and synapse loss [87]. Further research into the diverse pathogenetic mechanisms of AD is needed to identify alternative therapeutic approaches.
Accumulating data in recent years suggest an association between synaptic loss in AD and neurogranin (Ng). Neurogranin is a neuron-specific postsynaptic protein that is abundantly expressed in the brain, particularly in the dendrites of hippocampal and cortical neurons [88]. It binds to calmodulin at low calcium ion concentrations and regulates synaptic plasticity of neurons by modulating Ca2+/calmodulin-dependent pathways. It is also involved in long-term potentiation, which is important for learning and memory processes [89]. Another hallmark of AD is an increase in CSF levels of Ng, which gradually increases with cognitive decline and negatively correlates with Mini Mental State Examination scores, likely reflecting synaptic damage due to Aβ aggregation with plaque accumulation [90, 91]. Some authors report a significant increase in CSF Ng levels in AD compared to Lewy body dementia, FTD, and amyotrophic lateral sclerosis [92], while others report only a high correlation between CSF Ng levels and CSF t-tau and p-tau181 levels [93]. Therefore, the value of CSF Ng level assessment is still controversial.
Neurofilament light chain (NfL) is a scaffold protein of the neuronal cytoskeleton that plays an important role in axon and dendrite branching and growth. Following axonal injury, CSF NfL levels increase and serve as a biomarker for axonal injury and neurodegeneration [94]. In recent years, the use of this biomarker to assess the progression of various neurological diseases, including AD, has increased significantly [95]. Higher CSF levels of NfL are also found in cognitively healthy individuals with hippocampal atrophy on neuroimaging [96] and in preclinical stages of AD [97, 98]. Longitudinal studies in patients with AD have shown that an increase in CSF NfL level is associated with a more intense progression of brain atrophy and cognitive decline. Therefore, higher NfL levels in early clinical stages of AD appear to predict faster conversion to dementia [99]. However, the specificity of this biomarker for AD is low since the highest levels are found in other neurodegenerative disorders such as amyotrophic lateral sclerosis, FTD, corticobasal degeneration, and progressive supranuclear palsy [100].
The pathogenesis of AD is also accompanied by reactive astrogliosis, which is characterized by morphological, molecular, and functional remodeling of astrocytes [101]. Glial fibrillary acidic protein (GFAP) is a type III intermediate filament protein that is predominantly expressed by astrocytes in the CNS [102]. In animal models, high levels of GFAP expression are found in astrocytes of the hippocampus, the corpus callosum and the cerebral peduncles [103]. Its expression is significantly increased in neurodegenerative diseases, including AD, reflecting neuroinflammatory processes and astrocyte activation [104]. In AD, elevated CSF levels of GFAP are a potential marker of progressive cognitive impairment; these levels have been shown to increase as cognitive deficits progress [105]. However, these changes are not specific to AD, because an increase in GFAP levels with progressive cognitive impairment has also been described in patients with Parkinson’s disease, FTD, multiple sclerosis, and other neurological disorders [106].
These biomarkers are being studied for research purposes, but are not yet used in clinical practice due to insufficient specificity for diagnosing AD.
Clinical Use of CSF Biomarkers for AD in Neurology
As mentioned above, according to the International Working Group Recommendations for the Clinical Diagnosis of Alzheimer’s Disease (2021), a diagnosis of AD requires the presence of a specific clinical phenotype and confirmation of the biological origin of the disease based on biomarker testing [6]. These guidelines distinguish between common and rare AD phenotypes. The main clinical phenotypes of AD include the classic amnestic (hippocampal) variant, posterior cortical atrophy, and the logopenic variant primary progressive aphasia. Rare phenotypes include frontal (behavioral/dysregulation) variant, corticobasal syndrome, and semantic and agrammatic variants of primary progressive aphasia. It is proposed to establish probability levels for AD as the primary diagnosis based on the combination of the clinical phenotype and the results of key biomarker testing (in CSF or by PET). The diagnosis of AD is categorized as definite, probable, and possible, with additional categories of unlikely and excluded. For controversial cases, recommendations for further patient evaluation are provided (Table 1).
Table 1. International Working Group Recommendations for the Clinical Diagnosis of Alzheimer's Disease (2021)
Phenotype | Likelihood of AD as a primary diagnosis | Further investigation |
Common clinical phenotypes in AD (amnestic variant, posterior cortical atrophy, logopenic variant primary progressive aphasia) | ||
Amyloid positive; tau positive | Highly probable – established | None required |
Amyloid positive; tau unknown | Probable | Consider a tau measure (PET, CSF) |
Amyloid positive; tau negative | Probable | Consider an additional tau measure (PET, CSF) |
Тau positive; amyloid unknown | Possible | Consider an amyloid measure (PET, CSF) |
Тau positive; amyloid negative | Possible | Consider an additional amyloid measure (PET, CSF) |
Аmyloid negative; tau unknown | Unlikely | Full investigation of cause and consider |
Аmyloid unknown; tau negative | Unlikely | Full investigation of cause and consider |
Аmyloid unknown; tau negative | Highly unlikely – excluded | Full investigation of cause* |
Аmyloid unknown; tau unknown | Non-assessable | Consider tau and amyloid and measure (PET, CSF) |
Uncommon clinical phenotypes in AD (frontal variant, corticobasal syndrome, semantic and agrammatic variants of primary progressive aphasia) | ||
Amyloid positive; tau positive | Probable | None required; careful follow-up needed: |
Amyloid positive; tau unknown | Possible | Consider a tau measure (PET, CSF) |
Amyloid positive; tau negative | Possible | Consider an additional tau measure (PET, CSF) |
Tau positive; amyloid unknown | Unlikely | Full investigation of cause and consider |
Tau positive; amyloid negative | Unlikely | Full investigation of cause*⛛ |
Amyloid negative; tau unknown | Highly unlikely – excluded | Full investigation of cause*⛛ |
Amyloid negative; tau negative | Highly unlikely – excluded | Full investigation of cause*⛛ |
Amyloid unknown; tau negative | Highly unlikely – excluded | Full investigation of cause*⛛ |
Amyloid unknown; tau unknown | Non-assessable | Full investigation of cause and |
Note. *Full investigation of cause depends on the specific clinical phenotype and can imply, for example, 18F-fluorodeoxyglucose PET (FDG-PET), dopamine transporter imaging with single-photon emission computed tomography (DaT-SPECT), serum progranulin assay, genetic analysis, oculomotor recordings, or electromyoneurography. ⛛Consider a new Alzheimer’s disease biomarker investigation only if there is a reasonable doubt about the validity of the biomarker results.
Conclusion
According to international guidelines, the key CSF biomarkers for the clinical diagnosis of AD (gold standard) include Аβ1-42, Аβ1-42/Аβ1-40 ratio, and p-tau181. CSF t-tau levels can be used to assess the activity of the neurodegenerative process and predict the clinical progression of the disease. Novel biomarkers of tau pathology (including CSF p-tau217 and p-tau231 levels) could also be used to diagnose AD, because they are both highly sensitive and specific even in the preclinical stages of the disease. The clinical applicability of markers of neurodegeneration and astrocyte activation (Ng, NfL, GFAP) requires further discussion, so their use is currently warranted only in research settings.
The wider availability of CSF biomarkers in Russian clinical practice will allow for the assessment of the true prevalence of AD in the Russian population, as well as the selection of patients for targeted therapy, which has been actively developed in recent years.
About the authors
Kseniya V. Nevzorova
Russian Center of Neurology and Neurosciences
Author for correspondence.
Email: nevzorova.k.v@neurology.ru
ORCID iD: 0009-0000-9148-0203
postgraduate student, neurologist, 5th Neurological department with a molecular genetic laboratory, Institute of Clinical and Preventive Neurology
Russian Federation, 80, Volokolamskoye shosse, Moscow, 125367Yuliya A. Shpilyukova
Russian Center of Neurology and Neurosciences
Email: nevzorova.k.v@neurology.ru
ORCID iD: 0000-0001-7214-583X
Cand. Sci. (Med.), researcher, 5th Neurological department with a molecular genetic laboratory, Institute of Clinical and Preventive Neurology
Russian Federation, 80, Volokolamskoye shosse, Moscow, 125367Аlla A. Shabalina
Russian Center of Neurology and Neurosciences
Email: nevzorova.k.v@neurology.ru
ORCID iD: 0000-0001-7393-0979
Dr. Sci. (Med.), leading researcher, Head, Laboratory diagnostics department
Russian Federation, 80, Volokolamskoye shosse, Moscow, 125367Ekaterina Yu. Fedotova
Russian Center of Neurology and Neurosciences
Email: nevzorova.k.v@neurology.ru
ORCID iD: 0000-0001-8070-7644
Dr. Sci. (Med.), leading researcher, Head, 5th Neurological department with a molecular genetic laboratory, Institute of Clinical and Preventive Neurology
Russian Federation, 80, Volokolamskoye shosse, Moscow, 125367Sergey N. Illarioshkin
Russian Center of Neurology and Neurosciences
Email: nevzorova.k.v@neurology.ru
ORCID iD: 0000-0002-2704-6282
Dr. Sci. (Med.), Prof., Full member of the RAS, Deputy director, Director, Brain Institute
Russian Federation, 80, Volokolamskoye shosse, Moscow, 125367References
- Weller J, Budson A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Res. 2018;7:F1000 Faculty Rev-1161. doi: 10.12688/f1000research.14506.1
- Duyckaerts C, Delatour B, Potier MC. Classification and basic pathology of Alzheimer disease. Acta Neuropathol. 2009;118(1):5–36. doi: 10.1007/s00401-009-0532-1
- Васенина Е.Е., Левин О.С., Сонин А.Г. Современные тенденции в эпидемиологии деменции и ведении пациентов с когнитивными нарушениями. Журнал неврологии и психиатрии им. С.С. Корсакова. Спецвыпуски. 2017;117(6-2):87–95. Vasenina EE, Levin OS, Sonin AG. Modern trends in epidemiology of dementia and management of patients with cognitive impairment. S.S. Korsakov Journal of Neurology and Psychiatry. 2017;117(6-2):87–95. doi: 10.17116/jnevro20171176287-95
- Коберская Н.Н. Болезнь Альцгеймера. Неврология, нейропсихиатрия, психосоматика. 2019;11(3S):52–60. Koberskaya NN. Alzheimer’s disease. Neurology, Neuropsychiatry, Psychosomatics. 2019;11(3S):52–60. doi: 10.14412/2074-2711-2019-3S-52-60
- Преображенская И.С. Современные подходы к диагностике и лечению болезни Альцгеймера. Медицинский cовет. 2017;(10):26–31. Preobrazhenskaya IS. Modern approaches to diagnostics and therapy of Alzheimer disease. Medical Council. 2017;(10):26–31. doi: 10.21518/2079-701X-2017-10-26-31
- Dubois B., Villain N, Frisoni GB, et al. Clinical diagnosis of Alzheimer’s disease: recommendations of the Intenational Working Group. Lancet Neurol. 2021;20(6):484–496. doi: 10.1016/S1474-4422(21)00066-1
- Таппахов А.А., Николаева Т.Я., Попова Т.Е., Шнайдер Н.А. Трудности диагностики атипичных вариантов болезни Альцгеймера. Российский неврологический журнал. 2021;26(5):16–23. Tappakhov AA, Nikolaeva TYa, Popova TE, Shnayder NA. Difficulties in diagnosing atypical variants of Alzheimer’s disease. Russian neurological journal. 2021;26(5):16–23. doi: 10.30629/2658-7947-2021-26-5-16-23
- Шпилюкова Ю.А., Шабалина А.А., Ахмадуллина Д.Р., Федотова Е.Ю. Опыт использования лабораторных биомаркеров в диагностике нейродегенеративных деменций. Бюллетень Национального общества по изучению болезни Паркинсона и расстройств движений. 2022;(2):227–230. Shpilyukova YuA, Shabalina AA, Akhmadullina DR, Fedotova EYu. The experience of using laboratory biomarkers in the diagnosis of neurodegenerative dementia. Bulletin of the National Society for the Study of Parkinson’s Disease and Movement Disorders. 2022;(2):227–230. doi: 10.24412/2226-079X-2022-12474
- Nevzorova K, Shpilyukova Y, Shabalina A, et al. Biomarkers of Alzheimer’s disease pathology in atypical non-amnestic clinical phenotypes. Alzheimers Dement., 2023;19(S15):e076010. doi: 10.1002/alz.076010
- Гришина Д.А., Хаялиева Н.А., Гринюк В.В., Тюрина А.Ю. Диагностика болезни Альцгеймера с использованием биологических маркеров при синдроме задней корковой атрофии. Неврология, нейропсихиатрия, психосоматика. 2024;16(2):47–55. Grishina DA, Khayalieva NA, Grinyuk VV, Tyurina AYu. Diagnosis of Alzheimer’s disease by using biological markers in posterior cortical atrophy. Neurology, Neuropsychiatry, Psychosomatics. 2024;16(2):47–53. doi: 10.14412/2074-2711-2024-2-47-53
- Alzheimer A. Uber eigenartige Erkrankung der Hirnrinde. Über eine eigenartige Erkrankung der Hirnrinde. Allgemeine Zeitschrift fur Psychiatrie und Psychisch-gerichtliche Medizin. 1907;64:146–148.
- Kang J, Lemaire HG, Unterbeck A, et al. The precursor of Alzheimer’s disease amyloid A4 protein resembles a cell-surface receptor. Nature. 1987;325(6106):733–736. doi: 10.1038/325733a0
- Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–185. doi: 10.1126/science.1566067
- Nalivaeva NN, Turner AJ. The amyloid precursor protein: a biochemical enigma in brain development, function and disease. FEBS Lett. 2013;587(13):2046–2054. doi: 10.1016/j.febslet.2013.05.010
- Литвиненко И.В., Емелин А.Ю., Лобзин В.Ю. и др. Амилоидная гипотеза болезни Альцгеймера: прошлое и настоящее, надежды и разочарования. Неврология, нейропсихиатрия, психосоматика. 2019;11(3):4–10. Litvinenko IV, Emelin AYu, Lobzin VYu, et al. The amyloid hypothesis of Alzheimer’s disease: past and present, hopes and disappointments. Neurology, Neuropsychiatry, Psychosomatics. 2019;11(3):4–10. doi: 10.14412/2074-2711-2019-3-4-10
- Molinuevo JL, Ayton S, Batrla R, et al. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 2018;136(6):821–853. doi: 10.1007/s00401-018-1932-x
- Стефанова Н.А., Колосова Н.Г. Эволюция представлений о патогенезе болезни Альцгеймера. Вестник Московского университета. Серия 16. Биология. 2016;(1):6–13. Stefanova NA, Kolosova NG. Evolution of understanding of Alzheimer’s disease pathogenesis. Vestnik Moskovskogo universiteta. Seriya 16. Biologiya. 2016;(1):6–13.
- Кухарский М.С., Овчинников Р.К., Бачурин С.О. Молекулярные аспекты патогенеза и современные подходы к фармакологической коррекции болезни Альцгеймера. Журнал неврологии и психиатрии им. С.С. Корсакова. 2015;115(6):103–114. Kukharskiĭ MS, Ovchinnikov RK, Bachurin SO. Molecular aspects of the pathogenesis and current approaches to pharmacological correction of Alzheimer’s disease. S.S. Korsakov Journal of Neurology and Psychiatry. 2015;115(6):103–114. doi: 10.17116/jnevro20151156103-114
- Lee JC, Kim SJ, Hong S, Kim Y. Diagnosis of Alzheimer’s disease utilizing amyloid and tau as fluid biomarkers. Exp Mol Med. 2019;51(5):1–10. doi: 10.1038/s12276-019-0250-2
- Glenner GG, Wong CW. Alzheimer’s disease and Down’s syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem Biophys Res Commun. 1984;122(3):1131–1135. doi: 10.1016/0006-291x(84)91209-9
- Scheuner D, Eckman C, Jensen M, et al. Secreted amyloid β-protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the Presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nat Med. 1996;2(8):864–870. doi: 10.1038/nm0896-864
- Weller RO, Massey A, Kuo YM, Roher AE. Cerebral amyloid angiopathy: accumulation of A-beta in interstitial fluid drainage pathways in Alzheimer’s disease. Ann N Y Acad Sci. 2000;903:110–117. doi: 10.1111/j.1749-6632.2000.tb06356.x
- Mawuenyega KG, Sigurdson W, Ovod V, et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science. 2010;330(6012):1774. doi: 10.1126/science.1197623
- Yoon SS, Jo SA. Mechanisms of amyloid-β peptide clearance: potential therapeutic targets for Alzheimer’s disease. Biomol Ther (Seoul). 2012;20(3):245–255. doi: 10.4062/biomolther.2012.20.3.245
- Wyss-Coray T, Loike JD, Brionne TC, et al. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med. 2003;9(4):453–457. doi: 10.1038/nm838
- Shibata M, Yamada S, Kumar SR, et al. Clearance of Alzheimer’s amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest. 2000;106(12):1489–1499. doi: 10.1172/JCI10498
- Shirotani K, Tsubuki S, Iwata N, et al. Neprilysin degrades both amyloid peptides 1-40 and 1-42 most rapidly and efficiently among thiorphan- and phosphoramidon-sensitive endopeptidases. J Biol Chem. 2001;276(24):21895–21901. doi: 10.1074/jbc.M008511200
- Chesneau V, Vekrellis K, Rosner MR, Selkoe DJ. Purified recombinant insulin-degrading enzyme degrades amyloid beta-protein but does not promote its oligomerization. Biochem J. 2000;351(Pt 2):509–516.
- Yin KJ, Cirrito JR, Yan P, et al. Matrix metalloproteinases expressed by astrocytes mediate extracellular amyloid-beta peptide catabolism. J Neurosci. 2006;26(43):10939–10948. doi: 10.1523/JNEUROSCI.2085-06.2006
- Kim MJ, Chae SS, Koh YH, et al. Glutamate carboxypeptidase II: an amyloid peptide-degrading enzyme with physiological function in the brain. FASEB J. 2010;24(11):4491–4502. doi: 10.1096/fj.09-148825
- Hensley K, Hall N, Subramaniam R, et al. Brain regional correspondence between Alzheimer’s disease histopathology and biomarkers of protein oxidation. J Neurochem. 1995;65(5):2146–2156. doi: 10.1046/j.1471-4159.1995.65052146.x
- Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2006;27(10):1372–1384. doi: 10.1016/j.neurobiolaging.2005.09.012
- Григорьева В.Н., Машкович К.А. Ликворологические биомаркеры болезни Альцгеймера (обзор). Медицинский альманах. 2021;2(67):22–32. Grigoreva VN, Mashkovich KA. Cerebrospinal fluid biomarkers of Alzheimer’s disease (review). Medicinskij alʹmanah. 2021;2(67):22–32.
- Schöll M, Lockhart SN, Schonhaut DR, et al. PET imaging of Tau deposition in the aging human brain. Neuron. 2016;89(5):971–982. doi: 10.1016/j.neuron.2016.01.028
- Ossenkoppele R, Schonhaut DR, Schöll M, et al. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain. 2016;139(Pt 5):1551–1567. doi: 10.1093/brain/aww027
- Zhang CC, Xing A, Tan MS, et al. The role of MAPT in neurodegenerative diseases: genetics, mechanisms and therapy. Mol Neurobiol. 2016;53(7):4893–4904. doi: 10.1007/s12035-015-9415-8
- Абдуллаева Н., Алиева Г. Взаимосвязь тау-белка с патологией болезни Альцгеймера. Norwegian Journal of Development of the International Science. 2021;63-1:9–12. Abdullayeva N, Aliyeva G. The relationship of tau-protein with the pathology of Alzheimer’s disease. Norwegian Journal of Development of the International Science. 2021;63-1:9–12. doi: 10.24412/3453-9875-2021-63-1-9-12
- Alonso A, Zaidi T, Novak M, et al. Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments. Proc Natl Acad Sci U S A. 2001;98(12):6923–6928. doi: 10.1073/pnas.121119298
- Jin M, Shepardson N, Yang T, et al. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108(14):5819–5824. doi: 10.1073/pnas.1017033108
- Counts SE, Ikonomovic MD, Mercado N, et al. Biomarkers for the early detection and progression of Alzheimer’s disease. Neurotherapeutics. 2017;14(1):35–53. doi: 10.1007/s13311-016-0481-z
- Van Harten AC, Wiste HJ, Weigand SD, et al. Detection of Alzheimer’s disease amyloid beta 1-42, p-tau, and t-tau assays. Alzheimers Dement. 2022;18(4):635–644. doi: 10.1002/alz.12406
- Frederiksen KS, Nielsen TR, Appollonio I, et al. Biomarker counseling, disclosure of diagnosis and follow-up in patients with mild cognitive impairment: a European Alzheimer’s disease consortium survey. Int J Geriatr Psychiatry. 2021;36(2):324–333. doi: 10.1002/gps.5427
- Vlassenko AG, McCue L, Jasielec MS, et al. Imaging and cerebrospinal fluid biomarkers in early preclinical Alzheimer disease. Ann Neurol. 2016;80(3):379–387. doi: 10.1002/ana.24719
- Stomrud E, Minthon L, Zetterberg H, et al. Longitudinal cerebrospinal fluid biomarker measurements in preclinical sporadic Alzheimer’s disease: a prospective 9-year study. Alzheimers Dement (Amst). 2015;1(4):403–411. doi: 10.1016/j.dadm.2015.09.002
- Shaw LM, Vanderstichele H, Knapik-Czajka M, et al. Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol. 2009;65(4):403–413. doi: 10.1002/ana.21610
- Palmqvist S, Mattsson N, Hansson O. Cerebrospinal fluid analysis detects cerebral amyloid-β accumulation earlier than positron emission tomography. Brain. 2016;139(Pt 4):1226–1236. doi: 10.1093/brain/aww015
- Skoog I, Davidsson P, Aevarsson O. et al. Cerebrospinal fluid beta-amyloid 42 is reduced before the onset of sporadic dementia: a population-based study in 85-year-olds. Dement Geriatr Cogn Disord. 2003;15(3):169–176. doi: 10.1159/000068478
- Kuhlmann J, Andreasson U, Pannee J, et al. CSF Aβ1-42 — an excellent but complicated Alzheimer’s biomarker — a route to standardisation. Clin Chim Acta. 2017;467:27–33. doi: 10.1016/j.cca.2016.05.014
- Gravina SA, Ho L, Eckman CB, et al. Amyloid beta protein (A beta) in Alzheimer’s disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at A beta 40 or A beta 42(43). J Biol Chem. 1995;270(13):7013–7016. doi: 10.1074/jbc.270.13.7013
- Leissring MA, Farris W, Chang AY, et al. Enhanced proteolysis of beta-amyloid in APP transgenic mice prevents plaque formation, secondary pathology, and premature death. Neuron. 2003;40(6):1087–1093. doi: 10.1016/s0896-6273(03)00787-6
- Wegiel J, Wang KC, Imaki H, et al. The role of microglial cells and astrocytes in fibrillar plaque evolution in transgenic APP(SW) mice. Neurobiol Aging. 2001;22(1):49–61. doi: 10.1016/s0197-4580(00)00181-0
- Wilhelmus MM, Otte-Höller I, van Triel JJ, et al. Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells. Am J Pathol. 2007;171(6):1989–1999. doi: 10.2353/ajpath.2007.070050
- Spies PE, Verbeek MM, Van Groen T, Claassen J. Reviewing reasons for the decreased CSF Abeta42 concentration in Alzheimer disease. Front Biosci (Landmark Ed). 2012;17(6), 2024–2034. doi: 10.2741/4035
- Bjerke M, Andreasson U, Rolstad S, et al. Subcortical vascular dementia biomarker pattern in mild cognitive impairment. Dement Geriar Cogn Disord. 2009;28(4):348–356. doi: 10.1159/000252773
- Slaets S, Le Bastard N, Theuns J, et al. Amyloid pathology influences aβ1-42 cerebrospinal fluid levels in dementia with Lewy bodies. J Alzheimers Dis. 2013;35(1):137–146. doi: 10.3233/JAD-122176
- Dorey A, Tholance Y, Vighetto A, et al. Association of cerebrospinal fluid prion protein levels and the distinction between Alzheimer disease and Creutzfeldt–Jakob disease. JAMA Neurol. 2015;72(3):267–275. doi: 10.1001/jamaneurol.2014.4068
- Koopman K, Le Bastard N, Martin JJ, et al. Improved discrimination of autopsy-confirmed Alzheimer’s disease (AD) from non-AD dementias using CSF P-tau (181P). Neurochem Int. 2009;55(4):214–218. doi: 10.1016/j.neuint.2009.02.017
- Sehlin D, Englund H, Simu B, et al. Large aggregates are the major soluble Aβ species in AD brain fractionated with density gradient ultracentrifugation. PLoS One. 2012;7(2):e32014. doi: 10.1371/jurnal.pone.0032014
- Dorey A, Perret-Liaudet A, Tholance Y, et al. Cerebrospinal fluid Aβ40 improves the interpretation of Aβ42 concentration for diagnosing Alzheimer’s disease. Front Neurol. 2015;6:247. doi: 10.3389/fneur.2015.00247
- Kanai M, Matsubara E, Isoe K, et al. Longitudinal study of cerebrospinal fluid levels of tau, A beta1-40, and A beta1-42(43) in Alzheimer’s disease: a study in Japan. Ann Neurol. 1998;44(1):17–26. doi: 10.1002/ana.410440108
- Dumurgier J, Schraen S, Gabelle A, et al. Cerebrospinal fluid amyloid-β 42/40 ratio in clinical setting of memory centers: a multicentric study. Alzheimers Res Ther. 2015;7(1):30. doi: 10.1186/s13195-015-0114-5
- Delaby C, Estellés T, Zhu N, et al. The Aβ1-42/Aβ1-40 ratio in CSF is more strongly associated to tau markers and clinical progression than Aβ1-42 alone. Alzheimers Res Ther. 2022;14(1):20. doi: 10.1186/s13195-022-00967-z
- Baldeiras I, Santana I, Leitão MJ, et al. Addition of the Aβ42/40 ratio to the cerebrospinal fluid biomarker profile increases the predictive value for underlying Alzheimer’s disease dementia in mild cognitive impairment. Alz Res Therapy. 2018;10(1):33. doi: 10.1186/s13195-018-0362-2
- Arai H, Terajima M, Miura M, et al. Tau in cerebrospinal fluid: a potential diagnostic marker in Alzheimer’s disease. Ann Neurol. 1995;38(4):649–652. doi: 10.1002/ana.410380414
- Olsson B, Lautner R, Andreasson U, et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;15(7):673–684. doi: 10.1016/S1474-4422(16)00070-3
- Hesse C, Rosengren L, Andreasen N, et al. Transient increase in total tau but not phospho-tau in human cerebrospinal fluid after acute stroke. Neurosci Lett. 2001;297(3):187–190. doi: 10.1016/s0304-3940(00)01697-9
- Zetterberg H, Hietala MA, Jonsson M, et al. Neurochemical aftermath of amateur boxing. Arch Neurol. 2006;63(9):1277–1280. doi: 10.1001/archneur.63.9.1277
- Matsushita S, Miyakawa T, Maesato H, et al. Elevated cerebrospinal fluid tau protein levels in Wernicke’s encephalopathy. Alcohol Clin Exp Res. 2008;32(6):1091–1095. doi: 10.1111/j.1530-0277.2008.00671.x
- Skillbäck T, Rosén C, Asztely F, et al. Diagnostic performance of cerebrospinal fluid total tau and phosphorylated tau in Creutzfeldt–Jakob disease: results from the Swedish Mortality Registry. JAMA Neurol. 2014;71(4):476–483. doi: 10.1001/jamaneurol.2013.6455
- Blennow K, Wallin A, Agren H, et al. Tau protein in cerebrospinal fluid: a biochemical marker for axonal degeneration in Alzheimer disease? Mol Chem Neuropathol. 1995;26(3):231–245. doi: 10.1007/BF02815140
- Wallin AK, Blennow K, Zetterberg H, et al. CSF biomarkers predict a more malignant outcome in Alzheimer disease. Neurology. 2010;74(19):1531–1537. doi: 10.1212/WNL.0b013e3181dd4dd8
- Wang JZ, Grundke-Iqbal I, Iqbal K. Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer’s disease. Nat Med. 1996;2(8):871–875. doi: 10.1038/nm0896-871
- Liu F, Iqbal K, Grundke-Iqbal I, et al. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2004;101(29):10804–10809. doi: 10.1073/pnas.0400348101
- Chung SH. Aberrant phosphorylation in the pathogenesis of Alzheimer’s disease. BMB Rep. 2009;42(8):467–474. doi: 10.5483/bmbrep.2009.42.8.467
- Billingsley ML, Kincaid RL. Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. Biochem J. 1997;323(Pt 3):577–591. doi: 10.1042/bj3230577
- Pîrşcoveanu DFV, Pirici I, Tudorică V, et al. Tau protein in neurodegenerative diseases — a review. Rom J Morphol Embryol. 2017;58(4):1141–1150.
- Holper S, Watson R, Yassi N. Tau as a biomarker of neurodegeneration. Int J Mol Sci. 2022;23(13):7307. doi: 10.3390/ijms23137307
- Vos SJ, Xiong C, Visser PJ, et al. Preclinical Alzheimer’s disease and its outcome: a longitudinal cohort study. Lancet Neurol. 2013;12(10):957–965. doi: 10.1016/S1474-4422(13)70194-7
- Schoonenboom NS, Reesink FE, Verwey NA, et al. Cerebrospinal fluid markers for differential dementia diagnosis in a large memory clinic cohort. Neurology. 2012;78(1):47–54. doi: 10.1212/WNL.0b013e31823ed0f0
- Lleó A, Irwin DJ, Illán-Gala I, et al. A 2-Step cerebrospinal algorithm for the selection of frontotemporal lobar degeneration subtypes. JAMA Neurol. 2018;75(6):738–745. doi: 10.1001/jamaneurol.2018.0118
- Barthélemy NR, Bateman RJ, Hirtz C, et al. Cerebrospinal fluid phospho-tau T217 outperforms T181 as a biomarker for the differential diagnosis of Alzheimer’s disease and PET amyloid-positive patient identification. Alzheimers Res Ther. 2020;12(1):26. doi: 10.1186/s13195-020-00596-4
- Leuzy A, Janelidze S, Mattsson-Carlgren N, et al. Comparing the clinical utility and diagnostic performance of CSF P-Tau181, P-Tau217, and P-Tau231 assays. Neurology. 2021;97(17):e1681–e1694. doi: 10.1212/WNL.0000000000012727
- Janelidze S, Stomrud E, Smith R, et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat Commun. 2020;11(1):1683. doi: 10.1038/s41467-020-15436-0
- Ashton NJ, Benedet AL, Pascoal TA, et al. Cerebrospinal fluid p-tau231 as an early indicator of emerging pathology in Alzheimer’s disease. EBioMedicine. 2022;76:103836. doi: 10.1016/j.ebiom.2022.103836
- Ercan-Herbst E, Ehrig J, Schöndorf DC, et al. A post-translational modification signature defines changes in soluble tau correlating with oligomerization in early stage Alzheimer’s disease brain. Acta Neuropathol Commun. 2019;7(1):192. doi: 10.1186/s40478-019-0823-2
- Uhlmann RE, Rother C, Rasmussen J, et al. Acute targeting of pre-amyloid seeds in transgenic mice reduces Alzheimer-like pathology later in life. Nat Neurosci. 2020;23(12):1580–1588. doi: 10.1038/s41593-020-00737-w
- Kinney JW, Bemiller SM, Murtishaw AS, et al. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. (N Y). 2018;4:575–590. doi: 10.1016/j.trci.2018.06.014
- Pak JH, Huang FL, Li J, et al. Involvement of neurogranin in the modulation of calcium/calmodulin-dependent protein kinase II, synaptic plasticity, and spatial learning: a study with knockout mice. Proc Natl Acad Sci U S A. 2000;97(21):11232–11237. doi: 10.1073/pnas.210184697
- Huang KP, Huang FL, Jäger T, et al. Neurogranin/RC3 enhances long-term potentiation and learning by promoting calcium-mediated signaling. J Neurosci. 2004;24(47):10660–10669. doi: 10.1523/JNEUROSCI.2213-04.2004
- Liu W, Lin H, He X, et al. Neurogranin as a cognitive biomarker in cerebrospinal fluid and blood exosomes for Alzheimer’s disease and mild cognitive impairment. Transl Psychiatry. 2020;10(1):125. doi: 10.1038/s41398-020-0801-2
- Portelius E, Zetterberg H, Skillbäck T, et al. Cerebrospinal fluid neurogranin: relation to cognition and neurodegeneration in Alzheimer’s disease. Brain. 2015;138(Pt 11):3373–3385. doi: 10.1093/brain/awv267
- Portelius E, Olsson B, Höglund K, et al. Cerebrospinal fluid neurogranin concentration in neurodegeneration: relation to clinical phenotypes and neuropathology. Acta Neuropathol. 2018;136(3):363–376. doi: 10.1007/s00401-018-1851-x
- Willemse EAJ, Sieben A, Somers C, et al. Neurogranin as biomarker in CSF is non-specific to Alzheimer’s disease dementia. Neurobiol Aging. 2021;108:99–109. doi: 10.1016/j.neurobiolaging.2021.08.002
- Yuan A, Rao MV, Veeranna RP, Nixon RA. Neurofilaments and neurofilament proteins in health and disease. Cold Spring Harb Perspect. Biol. 2017;9(4):a018309. doi: 10.1101/cshperspect.a018309
- Meeker KL, Butt OH, Gordon BA, et al. Cerebrospinal fluid neurofilament light chain is a marker of aging and white matter damage. Neurobiol Dis. 2022;166:105662. doi: 10.1016/j.nbd.2022.105662
- Idland AV, Sala-Llonch R, Borza T, et al. CSF neurofilament light levels predict hippocampal atrophy in cognitively healthy older adults. Neurobiol Aging. 2017;49:138–144. doi: 10.1016/j.neurobiolaging.2016.09.012
- Dhiman K, Gupta VB, Villemagne VL, et al. Cerebrospinal fluid neurofilament light concentration predicts brain atrophy and cognition in Alzheimer’s disease. Alzheimers Dement (Amst). 2020;12(1):e12005. doi: 10.1002/dad2.12005
- Dhiman K, Villemagne VL, Fowler C, et al. Cerebrospinal fluid neurofilament light predicts risk of dementia onset in cognitively healthy individuals and rate of cognitive decline in mild cognitive impairment: a prospective longitudinal study. Biomedicines. 2022;10(5):1045. doi: 10.3390/biomedicines10051045
- Lim B, Grøntvedt GR, Bathala P, et al. CSF neurofilament light may predict progression from amnestic mild cognitive impairment to Alzheimer’s disease dementia. Neurobiol Aging. 2021;107:78–85. doi: 10.1016/j.neurobiolaging.2021.07.013
- Delaby C, Alcolea D, Carmona-Iragui M, et al. Differential levels of neurofilament light protein in cerebrospinal fluid in patients with a wide range of neurodegenerative disorders. Sci Rep. 2020;10(1):9161. doi: 10.1038/s41598-020-66090-x
- Benedet AL, Milà-Alomà M, Vrillon A, et al. Differences between plasma and cerebrospinal fluid glial fibrillary acidic protein levels across the Alzheimer disease continuum. JAMA Neurol. 2021;78(12):1471–1483. doi: 10.1001/jamaneurol.2021.3671
- Li D, Liu X, Liu T, et al. Neurochemical regulation of the expression and function of glial fibrillary acidic protein in astrocytes. Glia. 2020;68(5):878–897. doi: 10.1002/glia.23734
- Zhang Z, Ma Z, Zou W, et al. The appropriate marker for astrocytes: comparing the distribution and expression of three astrocytic markers in different mouse cerebral regions. Biomed Res. Int. 2019;2019:9605265. doi: 10.1155/2019/9605265
- Van Hulle C, Jonaitis EM, Betthauser TJ, et al. An examination of a novel multipanel of CSF biomarkers in the Alzheimer’s disease clinical and pathological continuum. Alzheimers Dement. 2021;17(3):431–445. doi: 10.1002/alz.12204
- Fukuyama R, Izumoto T, Fushiki S. The cerebrospinal fluid level of glial fibrillary acidic protein is increased in cerebrospinal fluid from Alzheimer’s disease patients and correlates with severity of dementia. Eur Neurol. 2001;46(1):35–38. doi: 10.1159/000050753
- Heimfarth L, Passos FRS, Monteiro BS, et al. Serum glial fibrillary acidic protein is a body fluid biomarker: a valuable prognostic for neurological disease — a systematic review. Int Immunopharmacol. 2022;107:108624. doi: 10.1016/j.intimp.2022.108624