Journal Home > Volume 7 , Issue 1

Alzheimer’s disease (AD) is the most common neurodegenerative disorder. However, it still has no available disease-modifying therapies. Its pathology cascade begins decades before symptomatic presentation. For these reasons, highly sensitive and highly specific fluid biomarkers should be developed for the early diagnosis of AD. In this study, the well-established and emerging fluid biomarkers of AD are summarized, and recent advances on their role in early diagnosis and progression monitoring as well as their correlations with AD pathology are highlighted. Future prospects and related research directions are also discussed.


menu
Abstract
Full text
Outline
About this article

The fluid biomarkers of Alzheimer's disease

Show Author's information Qinyu PengZhentao Zhang( )
Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China

Abstract

Alzheimer’s disease (AD) is the most common neurodegenerative disorder. However, it still has no available disease-modifying therapies. Its pathology cascade begins decades before symptomatic presentation. For these reasons, highly sensitive and highly specific fluid biomarkers should be developed for the early diagnosis of AD. In this study, the well-established and emerging fluid biomarkers of AD are summarized, and recent advances on their role in early diagnosis and progression monitoring as well as their correlations with AD pathology are highlighted. Future prospects and related research directions are also discussed.

Keywords: dementia, cerebrospinal fluid, amyloid-β, tau, blood

References(112)

[1]
World Alzheimer Report 2019 Attitudes to dementia. https://www.alz.co.uk/research/WorldAlzhei mer Report2019-Summary.pdf (accessed 1 Sep, 2020)
[2]
Musiek ES, Holtzman DM. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat Neurosci 2015, 18(6): 800-806.
[3]
Blennow K, Leon MJ de L, Zetterberg H. Alzheimer's disease. Lancet 2006. 368(9533): 387-403.
[4]
Hansson O, Seibyl J, Stomrud E, et al. CSF biomarkers of Alzheimer's disease concord with amyloid-β PET and predict clinical progression: a study of fully automated immunoassays in BioFINDER and ADNI cohorts. Alzheimer's Dement 2018, 14(11): 1470-1481.
[5]
Rizzi L, Missiaggia L, Roriz-Cruz M. CSF Aβ1-42, but not p-Tau181, predicted progression from amnestic MCI to Alzheimer's disease dementia. NeuroMolecular Med 2018, 20(4): 491-497.
[6]
Rizzi L, Maria PM, Batista CEA, et al. CSF Aβ1-42, but not p-Tau181, differentiates aMCI from SCI. Brain Res 2018, 1678: 27-31.
[7]
Ye LQ, Li XY, Zhang YB, et al. The discriminative capacity of CSF β-amyloid 42 and Tau in neurodegenerative diseases in the Chinese population. J Neurol Sci 2020, 412: 116756.
[8]
Baiardi S, Abu-Rumeileh S, Rossi M, et al. Antemortem CSF Aβ42/Aβ40 ratio predicts Alzheimer's disease pathology better than Aβ42 in rapidly progressive dementias. Ann Clin Transl Neurol 2018, 6(2): 263-273.
[9]
Lehmann S, Delaby C, Boursier G, et al. Relevance of Aβ42/40 Ratio for detection of Alzheimer disease pathology in clinical routine: The PLMR Scale. Front Aging Neurosci 2018, 10: 138.
[10]
Vogelgsang J, Wedekind D, Bouter C, et al. Reproducibility of Alzheimer's disease cerebrospinal fluid-biomarker measurements under clinical routine conditions. J Alzheimers Dis 2018, 62(1): 203-212.
[11]
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. Alzheimers Res Ther 2018, 10(1): 33.
[12]
Schauer SP, Mylott WR, Jr., Yuan M, et al. Preanalytical approaches to improve recovery of amyloid-β peptides from CSF as measured by immunological or mass spectrometry-based assays. Alzheimers Res Ther 2018, 10(1): 118.
[13]
Meyer PF, Binette AP, Gonneaud J, et al. Characterization of Alzheimer disease biomarker discrepancies using cerebrospinal fluid phosphorylated Tau and AV1451 positron emission tomography. JAMA Neurol 2020, 77(4): 508-516.
[14]
Llibre-Guerra JJ, Li Y, Schindler SE, et al. Association of longitudinal changes in cerebrospinal fluid total tau and phosphorylated tau 181 and brain atrophy with disease progression in patients with alzheimer disease. JAMA Netw Open 2019, 2(12): e1917126.
[15]
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.
[16]
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.
[17]
Blennow K, Chen C, Cicognola C, et al. Cerebrospinal fluid tau fragment correlates with tau PET: a candidate biomarker for tangle pathology. Brain 2020, 143(2): 650-660.
[18]
Blennow K, Shaw LM, Stomrud E, et al. Predicting clinical decline and conversion to Alzheimer's disease or dementia using novel Elecsys Aβ(1-42), pTau and tTau CSF immunoassays. Alzheimers Res Ther 2019, 9(1): 19024.
[19]
Bos I, Vos SJB, Schindler SE, et al. Vascular risk factors are associated with longitudinal changes in cerebrospinal fluid tau markers and cognition in preclinical Alzheimer's disease. Alzheimers Dement 2019, 15(9): 1149-1159.
[20]
García-Chamé M, Gutiérrez-Sanz Ó, Ercan-Herbst E, et al. A transistor-based label-free immunosensor for rapid detection of tau protein. Biosens Bioelectron 2020, 159: 112129.
[21]
Zhu KC, Xiang XY, Filser S, et al. Beta-site amyloid precursor protein cleaving enzyme 1 inhibition impairs synaptic plasticity via seizure protein 6. Biol Psychiatry 2018, 83(5): 428-437.
[22]
Zhao J, Fu Y, Yasvoina M, et al. Beta-site amyloid precursor protein cleaving enzyme 1 levels become elevated in neurons around amyloid plaques: implications for Alzheimer's disease pathogenesis. J Neurosci 2007, 27(14): 3639-3649.
[23]
Vassar R, Bennett BD, Babu-Khan S, et al. Beta-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Science 1999, 286(5440): 735-741.
[24]
Vassar R. BACE1: the beta-secretase enzyme in Alzheimer's disease. J Mol Neurosci 2004, 23(1/2): 105-114.
[25]
Alexopoulos P, Thierjung N, Grimmer T, et al. Cerebrospinal fluid BACE1 activity and sAβPPβ as biomarker candidates of Alzheimer's disease. Dement Geriatr Cogn Disord 2018, 45(3/4): 152-161.
[26]
Kirsebom BE, Nordengen K, Selnes P, et al. Cerebrospinal fluid neurogranin/β-site APP-cleaving enzyme 1 predicts cognitive decline in preclinical Alzheimer's disease. Alzheimers Dement (N. Y.) 2018, 4: 617-627.
[27]
Schipke CG, De Vos A, Fuentes M, et al. Neurogranin and BACE1 in CSF as potential biomarkers differentiating depression with cognitive deficits from early Alzheimer's disease: A pilot study. Dement Geriatr Cogn Dis Extra 2018, 8(2): 277-289.
[28]
Bridel C, Van Wieringen WN, Zetterberg H, et al. Diagnostic value of cerebrospinal fluid neurofilament light protein in neurology: a systematic review and meta-analysis. JAMA Neurol 2019, 76(9): 1035-1048.
[29]
Khalil M, Teunissen CE, Otto M, et al. Neurofilaments as biomarkers in neurological disorders. Nat Rev Neurol 2018, 14(10): 577-589.
[30]
Lleó A, Alcolea D, Martínez-Lage P, et al. Longitudinal cerebrospinal fluid biomarker trajectories along the Alzheimer's disease continuum in the BIOMARKAPD study. Alzheimers Dement 2019, 15(6): 742-753.
[31]
Bos I, Vos S, Verhey F, et al. Cerebrospinal fluid biomarkers of neurodegeneration, synaptic integrity, and astroglial activation across the clinical Alzheimer's disease spectrum. Alzheimers Dement 2019, 15(5): 644-654.
[32]
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.
[33]
Andersson E, Janelidze S, Lampinen B, et al. Blood and cerebrospinal fluid neurofilament light differentially detect neurodegeneration in early Alzheimer's disease. Neurobiol Aging 2020, 95: 143-153.
[34]
Moore EE, Hohman TJ, Badami FS, et al. Neurofilament relates to white matter microstructure in older adults. Neurobiol Aging 2018, 70: 233-241.
[35]
Rojas JC, Bang J, Lobach IV, et al. CSF neurofilament light chain and phosphorylated tau 181 predict disease progression in PSP. Neurology 2018, 90(4): e273-e281.
[36]
Scherling CS, Hall T, Berisha F, et al. Cerebrospinal fluid neurofilament concentration reflects disease severity in frontotemporal degeneration. Ann Neurol 2014, 75(1): 116-126.
[37]
Skillbäck T, Farahmand B, Bartlett JW, et al. CSF neurofilament light differs in neurodegenerative diseases and predicts severity and survival. Neurology 2014, 83(21): 1945-1953.
[38]
Ulland TK, Song WM, Huang SC, et al. TREM2 maintains microglial metabolic fitness in Alzheimer's disease. Cell 2017, 170(4): 649–663.e13.
[39]
Rauchmann BS, Schneider-Axmann T, Alexopoulos P, et al. CSF soluble TREM2 as a measure of immune response along the Alzheimer's disease continuum. Neurobiol Aging 2019, 74: 182-190.
[40]
Ma LZ, Tan L, Bi YL, et al. Dynamic changes of CSF sTREM2 in preclinical Alzheimer's disease: the CABLE study. Mol Neurodegener 2020, 15(1): 25.
[41]
Suárez-Calvet M, Morenas-Rodríguez E, Kleinberger G, et al. Early increase of CSF sTREM2 in Alzheimer's disease is associated with tau related-neurodegeneration but not with amyloid-β pathology. Mol Neurodegener 2019, 14(1): 1.
[42]
Ewers M, Biechele G, Suárez-Calvet M, et al. Higher CSF sTREM2 and microglia activation are associated with slower rates of beta-amyloid accumulation. EMBO Mol Med 2020, 12(9): e12308.
[43]
Casati M, Ferri E, Gussago C, et al. Increased expression of TREM2 in peripheral cells from mild cognitive impairment patients who progress into Alzheimer's disease. Eur J Neurol 2018, 25(6): 805-810.
[44]
Falcon C, Monté-Rubio GC, Grau-Rivera O, et al. CSF glial biomarkers YKL40 and sTREM2 are associated with longitudinal volume and diffusivity changes in cognitively unimpaired individuals. NeuroImage Clin 2019, 23: 101801.
[45]
Cantó E, Tintoré M, Villar LM, et al. Chitinase 3-like 1: prognostic biomarker in clinically isolated syndromes. Brain 2015, 138(Pt 4): 918-931.
[46]
Querol-Vilaseca M, Colom-Cadena M, Pegueroles J, et al. YKL-40 (Chitinase 3-like I) is expressed in a subset of astrocytes in Alzheimer's disease and other tauopathies. J Neuroinflammation 2017, 14(1): 118.
[47]
Zhang H, Ng KP, Therriault J, et al. Cerebrospinal fluid phosphorylated tau, visinin-like protein-1, and chitinase-3-like protein 1 in mild cognitive impairment and Alzheimer's disease. Transl Neurodegener 2018, 7: 23.
[48]
Nordengen K, Kirsebom BE, Henjum K, et al. Glial activation and inflammation along the Alzheimer's disease continuum. J Neuroinflammation 2019, 16(1): 46.
[49]
Janelidze S, Mattsson N, Stomrud E, et al. CSF biomarkers of neuroinflammation and cerebrovascular dysfunction in early Alzheimer disease. Neurology 2018, 91(9): e867-e877.
[50]
Thordardottir S, Almkvist O, Johansson C, et al. Cerebrospinal fluid YKL-40 and neurogranin in familial Alzheimer's disease: a pilot study. J Alzheimers Dis 2020, 76(3): 941-953.
[51]
Gerendasy DD, Sutcliffe JG. RC3/neurogranin, a postsynaptic calpacitin for setting the response threshold to calcium influxes. Mol Neurobiol 1997, 15(2): 131-163.
[52]
Milà-Alomà M, Salvadó G, Gispert JD, et al. Amyloid beta, tau, synaptic, neurodegeneration, and glial biomarkers in the preclinical stage of the Alzheimer's continuum. Alzheimers Dement 2020, 16(10): 1358-1371.
[53]
Kvartsberg H, Lashley T, Murray CE, et al. The intact postsynaptic protein neurogranin is reduced in brain tissue from patients with familial and sporadic Alzheimer's disease. Acta Neuropathol 2019, 137(1): 89-102.
[54]
Xue M, Sun FR, Ou YN, et al. Association of cerebrospinal fluid neurogranin levels with cognition and neurodegeneration in Alzheimer's disease. Aging (Albany NY) 2020, 12(10): 9365-9379.
[55]
Headley A, De Leon-Benedetti A, Dong CH, et al. Neurogranin as a predictor of memory and executive function decline in MCI patients. Neurology 2018, 90(10): e887-e895.
[56]
Liu WL, Lin HW, He XJ, 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.
[57]
Nakamura A, Kaneko N, Villemagne VL, et al. High performance plasma amyloid-β biomarkers for Alzheimer's disease. Nature 2018, 554(7691): 249-254.
[58]
Vergallo A, Mégret L, Lista S, et al. Plasma amyloid β 40/42 ratio predicts cerebral amyloidosis in cognitively normal individuals at risk for Alzheimer's disease. Alzheimers Dement 2019, 15(6): 764-775.
[59]
Vogelgsang J, Shahpasand-Kroner H, Vogelgsang R, et al. Multiplex immunoassay measurement of amyloid-β42 to amyloid-β40 ratio in plasma discriminates between dementia due to Alzheimer's disease and dementia not due to Alzheimer's disease. Exp Brain Res 2018, 236(5): 1241-1250.
[60]
Verberk IMW, Slot RE, Verfaillie SCJ, et al. Plasma amyloid as prescreener for the earliest alzheimer pathological changes. Ann Neurol 2018, 84(5): 648-658.
[61]
Pérez-Grijalba V, Arbizu J, Romero J, et al. Plasma Aβ42/40 ratio alone or combined with FDG-PET can accurately predict amyloid-PET positivity: a cross-sectional analysis from the AB255 Study. Alzheimers Res Ther 2019, 11(1): 96.
[62]
Palmqvist S, Janelidze S, Stomrud E, et al. Performance of fully automated plasma assays as screening tests for Alzheimer disease-related β-amyloid status. JAMA neurol 2019, 76(9): 1060-1069.
[63]
Pérez-Grijalba V, Romero J, Pesini P, et al. Plasma Aβ42/40 ratio detects early stages of Alzheimer's disease and correlates with CSF and neuroimaging biomarkers in the AB255 study. J Prev Alzheimers Dis 2019, 6(1): 34-41.
[64]
Amouzadeh Tabrizi M, Ferré-Borrull J, Marsal LF. Highly sensitive aptasensor based on interferometric reflectance spectroscopy for the determination of amyloid β as an Alzheimer's disease biomarkers using nanoporous anodic alumina. Biosens Bioelectron 2019, 137: 279-286.
[65]
Chen WL, Gao G, Jin Y, et al. A facile biosensor for Aβ40O based on fluorescence quenching of Prussian blue nanoparticles. Talanta 2020, 216: 120930.
[66]
Yin YM, Chen GF, Gong L, et al. DNAzyme-powered three-dimensional DNA walker nanoprobe for detection amyloid β-peptide oligomer in living cells and in vivo. Anal Chem 2020, 92(13): 9247-9256.
[67]
Zhao YN, Li X, Yang Y, et al. A simple aptasensor for Aβ40 oligomers based on tunable mismatched base pairs of dsDNA and graphene oxide. Biosens Bioelectron 2020, 149: 111840.
[68]
Youn YC, Lee BS, Kim GJ, et al. Blood amyloid-β oligomerization as a biomarker of Alzheimer's disease: a blinded validation study. J Alzheimers Dis 2020, 75(2): 493-499.
[69]
Meng X, Li T, Wang X, et al. Association between increased levels of amyloid-β oligomers in plasma and episodic memory loss in Alzheimer's disease. Alzheimers Res Ther 2019, 11(1): 89.
[70]
Pase MP, Beiser AS, Himali JJ, et al. Assessment of plasma total tau level as a predictive biomarker for dementia and related endophenotypes. JAMA Neurol 2019, 76(5): 598-606.
[71]
De Wolf F, Ghanbari M, Licher S, et al. Plasma tau, neurofilament light chain and amyloid-β levels and risk of dementia; a population-based cohort study. Brain 2020, 143(4): 1220-1232.
[72]
Park JC, Han SH, Yi D, et al. Plasma tau/amyloid-β1-42 ratio predicts brain tau deposition and neurodegeneration in Alzheimer's disease. Brain 2019, 142(3): 771-786.
[73]
Karikari TK, Pascoal TA, Ashton NJ, et al. Blood phosphorylated tau 181 as a biomarker for Alzheimer's disease: a diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol 2020, 19(5): 422-433.
[74]
Mielke MM, Hagen CE, Xu J, et al. Plasma phospho-tau181 increases with Alzheimer's disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement 2018, 14(8): 989-997.
[75]
Lantero Rodriguez J, Karikari TK, Suárez-Calvet M, et al. Plasma p-tau181 accurately predicts Alzheimer's disease pathology at least 8 years prior to post-mortem and improves the clinical characterisation of cognitive decline. Acta Neuropathol 2020, 140(3): 267-278.10.1007/s00401-020-02195-x
[76]
Barthélemy NR, Horie K, Sato C, et al. Blood plasma phosphorylated-tau isoforms track CNS change in Alzheimer's disease. J Exp Med 2020, 217(11): e20200861.
[77]
Palmqvist S, Janelidze S, Quiroz YT, et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 2020, 324(8): 772.
[78]
Vergallo A, Houot M, Cavedo E, et al. Brain Aβ load association and sexual dimorphism of plasma BACE1 concentrations in cognitively normal individuals at risk for AD. Alzheimers Dement 2019, 15(10): 1274-1285.
[79]
Shen Y, Wang HB, Sun QY, et al. Increased plasma beta-secretase 1 may predict conversion to Alzheimer's disease dementia in individuals with mild cognitive impairment. Biol Psychiatry 2018, 83(5): 447-455.
[80]
Cervellati C, Trentini A, Rosta V, et al. Serum beta-secretase 1 (BACE1) activity as candidate biomarker for late-onset Alzheimer's disease. Geroscience 2020, 42(1): 159-167.
[81]
Lewczuk P, Ermann N, Andreasson U, et al. Plasma neurofilament light as a potential biomarker of neurodegeneration in Alzheimer's disease. Alzheimers Res Ther 2018, 10(1):71.
[82]
Nyberg L, Lundquist A, Nordin Adolfsson A, et al. Elevated plasma neurofilament light in aging reflects brain white-matter alterations but does not predict cognitive decline or Alzheimer's disease. Molecular Psychiatry 2020, 12(1):e12050.
[83]
Schultz SA, Strain JF, Adedokun A, et al. Serum neurofilament light chain levels are associated with white matter integrity in autosomal dominant Alzheimer's disease. Neurobiol Dis 2020, 142: 104960.
[84]
Quiroz YT, Zetterberg H, Reiman EM, et al. Plasma neurofilament light chain in the presenilin 1 E280A autosomal dominant Alzheimer's disease kindred: a cross-sectional and longitudinal cohort study. Lancet Neurol 2020, 19(6): 513-521.
[85]
Kapogiannis D, Mustapic M, Shardell MD, et al. Association of extracellular vesicle biomarkers with Alzheimer disease in the Baltimore Longitudinal Study of Aging. JAMA Neurol 2019, 76(11): 1340-1351.
[86]
Eren E, Hunt JFV, Shardell M, et al. Extracellular vesicle biomarkers of Alzheimer's disease associated with sub-clinical cognitive decline in late middle age. Alzheimers Dement 2020, 16(9): 1293-1304.
[87]
Jia LF, Qiu QQ, Zhang H, et al. Concordance between the assessment of Aβ42, T-tau, and P-T181-tau in peripheral blood neuronal-derived exosomes and cerebrospinal fluid. Alzheimers Dement 2019, 15(8): 1071-1080.
[88]
Perrotte M, Haddad M, Le Page A, et al. Profile of pathogenic proteins in total circulating extracellular vesicles in mild cognitive impairment and during the progression of Alzheimer's disease. Neurobiol Aging 2020, 86: 102-111.
[89]
Jia LF, Zhu M, Kong CJ, et al. Blood neuro-exosomal synaptic proteins predict Alzheimer's disease at the asymptomatic stage. Alzheimers Dement 2021, 17(1): 49-60.
[90]
Goetzl EJ, Abner EL, Jicha GA, et al. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer's disease. FASEB J 2018, 32(2): 888-893.
[91]
Agliardi C, Guerini FR, Zanzottera M, et al. SNAP-25 in serum is carried by exosomes of neuronal origin and is a potential biomarker of Alzheimer's disease. Mol Neurobiol 2019, 56(8): 5792-5798.
[92]
Gámez-Valero A, Campdelacreu J, Vilas D, et al. Exploratory study on microRNA profiles from plasma-derived extracellular vesicles in Alzheimer's disease and dementia with Lewy bodies. Transl Neurodegener 2019, 8: 31.
[93]
Takousis P, Sadlon A, Schulz J, et al. Differential expression of microRNAs in Alzheimer's disease brain, blood, and cerebrospinal fluid. Alzheimers Dement 2019, 15(11): 1468-1477.
[94]
Nagaraj S, Zoltowska KM, Laskowska-Kaszub K, et al. MicroRNA diagnostic panel for Alzheimer's disease and epigenetic trade-off between neurodegeneration and cancer. Ageing Res Rev 2019, 49: 125-143.
[95]
Wu HZY, Thalamuthu A, Cheng L, et al. Differential blood miRNA expression in brain amyloid imaging-defined Alzheimer's disease and controls. Alzheimers Rresearch Ther 2020, 12(1): 59.
[96]
Siedlecki-Wullich D, Català-Solsona J, Fábregas C, et al. Altered microRNAs related to synaptic function as potential plasma biomarkers for Alzheimer's disease. Alzheimers Res Ther 2019, 11(1): 1-11.
[97]
de Felice B, Montanino C, Oliva M, et al. MicroRNA expression signature in mild cognitive impairment due to Alzheimer's disease. Mol Neurobiol 2020, 57(11): 4408-4416.
[98]
Jain G, Stuendl A, Rao P, et al. A combined miRNA-PiRNA signature to detect Alzheimer's disease. Transl Psychiatry 2019, 9(1): 1-12.
[99]
González-Sánchez M, Bartolome F, Antequera D, et al. Decreased salivary lactoferrin levels are specific to Alzheimer's disease. EBioMedicine 2020, 57: 102834.
[100]
Carro E, Bartolomé F, Bermejo-Pareja F, et al. Early diagnosis of mild cognitive impairment and Alzheimer's disease based on salivary lactoferrin. Alzheimers Dementia (Amst) 2017, 8: 131-138.
[101]
Bermejo-Pareja F, del Ser T, Valentí M, et al. Salivary lactoferrin as biomarker for Alzheimer's disease: Brain-immunity interactions. Alzheimers Dement 2020, 16(8): 1196-1204.
[102]
Yilmaz A, Ugur Z, Bisgin H, et al. Targeted metabolic profiling of urine highlights a potential biomarker panel for the diagnosis of Alzheimer's disease and mild cognitive impairment: a pilot study. Metabolites 2020, 10(9): E357.
[103]
Li YX, Guan SC, Jin H, et al. The relationship between urinary Alzheimer-associated neuronal thread protein and blood biochemical indicators in the general population. Aging (Albany NY) 2020, 12(15): 15260-15280.
[104]
Ku BD, Kim H, Kim YK, et al. Comparison of urinary alzheimer-associated neural thread protein (AD7c-NTP) levels between patients with amnestic and nonamnestic mild cognitive impairment. Am J Alzheimers Dis Other Demen 2020, 35: 1533317519 880369.
[105]
Li YX, Kang MM, Wang HX, et al. Urinary Alzheimer-associated neuronal thread protein is not elevated in patients with subjective cognitive decline and patients with depressive state. J Alzheimers Dis 2019, 71(4): 1115-1123.
[106]
Ma LN, Chen J, Wang R, et al. The level of Alzheimer-associated neuronal thread protein in urine may be an important biomarker of mild cognitive impairment. J Clin Neurosci 2015, 22(4): 649-652.
[107]
Xu Y, Shen YY, Zhang XP, et al. Diagnostic potential of urinary monocyte chemoattractant protein-1 for Alzheimer's disease and amnestic mild cognitive impairment. Eur J Neurol 2020, 27(8): 1429-1435.
[108]
Zhang YQ, Tang YB, Dammer E, et al. Dysregulated urinary arginine metabolism in older adults with amnestic mild cognitive impairment. Front Aging Neurosci 2019, 11: 90.
[109]
Li WW, Shen YY, Tian DY, et al. Brain amyloid-β deposition and blood biomarkers in patients with clinically diagnosed Alzheimer's disease. J Alzheimers Dis 2019, 69(1): 169-178.
[110]
Rózga M, Bittner T, Batrla R, et al. Preanalytical sample handling recommendations for Alzheimer's disease plasma biomarkers. Alzheimers Dement (Amst) 2019, 11: 291-300.
[111]
Lewczuk P, Gaignaux A, Kofanova O, et al. Interlaboratory proficiency processing scheme in CSF aliquoting: implementation and assessment based on biomarkers of Alzheimer's disease. Alzheimers Res Ther 2018, 10(1): 87.
[112]
Willemse EAJ, van Maurik IS, Tijms BM, et al. Diagnostic performance of Elecsys immunoassays for cerebrospinal fluid Alzheimer's disease biomarkers in a nonacademic, multicenter memory clinic cohort: The ABIDE project. Alzheimers Dement (Amst) 2018, 10: 563-572.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 01 October 2020
Revised: 02 December 2020
Accepted: 24 December 2020
Published: 05 March 2021
Issue date: March 2021

Copyright

© The authors 2021

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (No. 81822016, No. 81771382) to Zhentao Zhang.

Rights and permissions

This article is published with open access at journals.sagepub.com/home/BSA

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/ en-us/nam/open-access-at-sage).

Return