Skip to main content
Erschienen in: Alzheimer's Research & Therapy 1/2022

Open Access 01.12.2022 | Research

The Aβ1–42/Aβ1–40 ratio in CSF is more strongly associated to tau markers and clinical progression than Aβ1–42 alone

verfasst von: Constance Delaby, Teresa Estellés, Nuole Zhu, Javier Arranz, Isabel Barroeta, María Carmona-Iragui, Ignacio Illán-Gala, Miguel Ángel Santos-Santos, Miren Altuna, Isabel Sala, M. Belén Sánchez-Saudinós, Laura Videla, Sílvia Valldeneu, Andrea Subirana, Mireia Tondo, Francisco Blanco-Vaca, Sylvain Lehmann, Olivia Belbin, Rafael Blesa, Juan Fortea, Alberto Lleó, Daniel Alcolea

Erschienen in: Alzheimer's Research & Therapy | Ausgabe 1/2022

Abstract

Background

Cerebrospinal fluid (CSF) Aβ1–42 levels and the Aβ1–42/Aβ1–40 ratio are markers of amyloid pathology, but previous studies suggest that their levels might be influenced by additional pathophysiological processes.

Aims

To compare Aβ1–42 and the Aβ1–42/Aβ1–40 ratio in CSF in different neurodegenerative disorders and study their association with other biomarkers (tTau, pTau181, and NfL) and with cognitive and functional progression.

Methods

We included all participants from the Sant Pau Initiative on Neurodegeneration (SPIN) with CSF Aβ1–42 and Aβ1–42/Aβ1–40. Participants had diagnoses of Alzheimer’s disease (AD), dementia with Lewy bodies, frontotemporal lobar degeneration-related syndromes, non-neurodegenerative conditions, or were cognitively normal. We classified participants as “positive” or “negative” according to each marker. We compared CSF levels of tTau, pTau181, and NfL between concordant and discordant groups through ANCOVA and assessed differences in cognitive (MMSE, FCSRT) and functional (GDS, CDR-SOB) progression using Cox regression and linear-mixed models.

Results

In the 1791 participants, the agreement between Aβ1–42 and Aβ1–42/Aβ1–40 was 78.3%. The Aβ1–42/Aβ1–40 ratio showed a stronger correlation with tTau and pTau181 than Aβ1–42 and an agreement with tTau and pTau181 of 73.1% and 77.1%, respectively. Participants with a low Aβ1–42/Aβ1–40 ratio showed higher tTau and pTau181 and worse cognitive and functional prognosis, regardless of whether they were positive or negative for Aβ1–42. The results were consistent across stages, diagnostic categories, and use of different cutoffs.

Conclusion

Although Aβ1–42 and Aβ1–42/Aβ1–40 are considered markers of the same pathophysiological pathway, our findings provide evidence favoring the use of the Aβ1–42/Aβ1–40 ratio in clinical laboratories in the context of AD.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13195-022-00967-z.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Cerebrospinal fluid (CSF) biomarkers of Alzheimer’s disease (AD) have changed the management of patients with cognitive impairment [1]. In particular, CSF levels of β-amyloid 1–42 (Aβ1–42), total tau (tTau), and its phosphorylated form on threonine 181 (pTau181) have shown very high accuracy for the diagnosis of AD [24]. They are consequently being implemented in clinical laboratories [59] both for diagnosis and research settings, as well as in clinical trials.
The role of Aβ1–42 in CSF as a marker of amyloid pathology is widely accepted [6, 9, 10]. However, a series of studies have shown that reduced levels of CSF Aβ1-42 can also be found in a variety of conditions different from AD, such as inflammatory diseases, prionopathies, amyloid angiopathy, or frontotemporal dementia [1015]. The Aβ1–42/Aβ1–40 ratio has proven to be of great value in detecting amyloid pathology both in CSF [16, 17] and plasma [18, 19] and has shown a better correlation with amyloid burden in PET than Aβ1–42 alone [2022]. However, Aβ1–40 levels are not systematically assessed in many clinical laboratories alleging that this marker alone has no diagnostic value. A large-scale head-to-head comparison between Aβ1–42 and the Aβ1–42/Aβ1–40 ratio would address the question of whether these two markers are equally tracking the same pathophysiological process. It would also inform laboratories on whether to implement the Aβ1–42/Aβ1–40 ratio into the clinical routine.
In the present work, we studied the CSF markers Aβ1–42 and Aβ1–42/Aβ1–40 ratio in a large cohort of participants with a variety of neurodegenerative disorders. We compared the agreement between both measures in different contexts and studied their association with other CSF biomarkers (tTau, pTau181, and NfL) and with cognitive and functional progression. This information is highly relevant in the implementation and interpretation of these markers in clinical routine.

Material and methods

Study participants and clinical classification

We included all participants in the Sant Pau Initiative on Neurodegeneration (SPIN) cohort [23] that underwent lumbar puncture for CSF biomarkers between November 2013 and August 2021. The diagnostic groups included patients with mild cognitive impairment (MCI) or dementia and with either pathophysiological evidence of Alzheimer’s disease (AD), frontotemporal lobar degeneration (FTLD)-related syndromes [24, 25], or probable dementia with Lewy bodies (DLB). Diagnoses were established following internationally accepted diagnostic criteria [6, 7, 9, 2628]. Clinical symptoms, neuroimaging and CSF biomarkers were considered for the diagnostic classification of patients. We also included participants with Down syndrome (DS) [29, 30] and cognitively normal controls (CN). All CN participants had normal cognitive scores in a formal neuropsychological evaluation [23, 31]. Patients with other diagnoses were grouped as “others” and included participants with prionopathy and other non-neurodegenerative conditions such as psychiatric etiology, vascular cognitive impairment, inflammatory, and those with uncertain etiology. Details about the SPIN cohort have been reported previously [23].

CSF collection and analysis

CSF was obtained by lumbar puncture, collected, and processed in polypropylene tubes following international recommendations [32, 33]. The same pre-analytical handling was followed in all samples [23]. Concentrations of Aβ1–42, Aβ1–40, total tau (tTau), and 181-phosphorylated tau (pTau181) in CSF were measured using commercially available kits in the Lumipulse fully automated platform (Fujirebio-Europe), as previously described [22, 34], and following provider’s instructions in line with Global Biomarker Standardization [35, 36]. For each sample, Aβ1–42, Aβ1–40, tTau, and pTau181 were quantified simultaneously in the same run immediately after the first freeze-thaw cycle of each sample using pristine aliquots containing 500 μL of CSF [22, 34]. The results of the Lumipulse G β-amyloid 1–42 have been standardized according to certified reference material developed by the International Federation of Clinical Chemistry and Laboratory Medicine as recommended by their working group for CSF proteins. Our laboratory participates in the Alzheimer’s Association Quality Control Program led by the University of Gothenburg [37]. Three levels of internal quality controls provided by the manufacturer were assessed for each analyte to assess the reproducibility of the assays. We included at least one level of quality control per analyte in each run. Inter-assay coefficients of variation (CV%) were between 1.7% and 6.8% for all levels and analytes.
Neurofilament light (NfL) levels in CSF were measured using a commercially available ELISA kit (NF-light, UMAN DIAGNOSTICS, Umea, Sweden) as previously described [24, 25]. The mean inter- and intra-assay coefficients of variation were 3.4% and 11.4%, respectively.

Definition of amyloid profile

To ensure that the cutoffs for Aβ1–42 and Aβ1–42/Aβ1–40 had comparable levels of sensitivity and specificity, we applied cutoffs corresponding to one-sided 95% quantile (Q95%) values in a middle-aged cognitively healthy population (age range 23–60 years, 67% female). This age range was selected to minimize the presence of preclinical AD in the reference population. We used these Q95% cutoffs for Aβ1–42 (637 pg/mL) and Aβ1–42/Aβ1–40 (0.070) to classify all participants in four different profiles: two concordant profiles in which both Aβ1–42 and Aβ1–42/Aβ1–40 ratio were above (Aβ1–42[−]Ratio[−]) or below (Aβ1–42[+]Ratio[+]) their respective cutoffs, and two discordant profiles, in which only one of the two amyloid parameters, Aβ1–42 or the Aβ1–42/Aβ1–40 ratio, was abnormal (Aβ1–42[+]Ratio[−] and Aβ1–42[−]Ratio[+], respectively). The objective of classifying participants in four amyloid profiles is to assess the particularities of those groups where Aβ1–42 and the Aβ1–42/Aβ1–40 ratio are discordant (Aβ1–42[−]Ratio[+] and Aβ1–42[+]Ratio[−]) and compare them to those that are clearly amyloid negative (Aβ1–42[–]Ratio[−]) or clearly amyloid positive (Aβ1–42[+]Ratio[+]) according to both markers. More details about the cognitively healthy reference population and results after applying other cutoffs can be found in Additional file 1.

Measures of cognitive and functional impairment

Cognition was assessed by the Mini-Mental State Examination (MMSE) and the free and cued selective reminding test (FCSRT). Global functional impairment was assessed by the Clinical Dementia Rating Scale Sum of Boxes (CDR-SOB) and the Global Deterioration Scale (GDS). Outcomes for cognitive and functional impairment were defined as MMSE < 24 and GDS ≥ 4, respectively [31].

APOE genotyping

DNA was extracted from whole blood using standard procedures and APOE was genotyped according to previously described methods [38].

Statistical analysis

Non-normally distributed variables were log-transformed. Differences in the frequency of categorical variables were assessed by the χ2 test, and we used age- and sex-adjusted analysis of covariance (ANCOVA) to compare CSF levels of tTau, pTau181, and NfL between concordant and discordant groups. We determined Spearman’s correlation coefficients between biomarkers in the whole sample and after stratification by diagnostic category, clinical stage, and amyloid profile. We assessed the association with cognitive and functional progression in patients with mild cognitive impairment through Kaplan-Meier survival curves and age- and sex-adjusted Cox regression analysis. We studied the association of amyloid profiles with a cognitive decline through linear-mixed models. The initial model included baseline MMSE score, baseline age, sex, years of education, pTau181 levels, diagnosis, time, APOE4 status, and amyloid profile together with its interaction with time and with APOE4 status as fixed factors. We defined random intercepts for diagnosis and at the participant level to account for repeated measures and modeled residual errors per diagnostic group. The alpha threshold was set at 0.05, and all analysis were performed using MEDCALC (MEDCALC software ver 15.2.2) and packages “survival” (v.3.1-12), “survminer” (v.0.4.6), “nlme” (v.3.1-147), “multcomp” (v.1.4-13), “ggplot2” (v.3.3.0), and “ggpubr” (v.0.3.0), as implemented in the R statistical software (v 3.6.2). The alpha threshold was set at 0.05 for all analyses.
All procedures in the study were approved by the Sant Pau Ethics Committee following the standards for medical research in humans recommended by the Declaration of Helsinki. All participants or their legally authorized representatives gave written informed consent.

Results

Demographics and core CSF biomarkers

We included a total of 1791 participants from the SPIN cohort. The demographic characteristics and biomarker results are summarized in Table 1. There were differences in age and male/female proportion between the groups. As expected, the APOEε4 allele was more frequent in AD patients (50%; p < 0.001), and no differences were observed among the other groups. MMSE scores were lower in all symptomatic groups compared to CN (p < 0.001).
Table 1
Demographics, clinical information, and biomarkers across diagnostic categories
  
CN
AD
DLB
FTLD
Down
Others
N
 
197
518
128
186
225
536
AGE, years
Mean (SD)
53.5 (12.5)
73.1 (6.88)
75.7 (5.49)
70.8 (8.58)
45.1 (10)
70 (9.18)
Median [IQR]
55 [46–62]
74 [69–78]
76 [71–80]
72 [66–77]
48 [40–52]
71 [65–77]
SEX, females/males (% females)
132/65 (67%)
311/207 (60%)
64/64 (50%)
77/109 (41.4%)
103/122 (45.8%)
310/226 (57.8%)
MMSE score
Mean (SD)
29.2 (0.889)
23.6 (4.52)
24 (4.09)
24 (5.08)
NAa
25.4 (4.12)
Median [IQR]
29 [29–30]
25 [22–27]
24.5 [22–27]
25 [21–28]
NAa
27 [24–28]
Education, years
Mean (SD)
15.6 (3.99)
10.7 (4.72)
9.18 (5.05)
12 (5.13)
15.3 (3.07)
10.7 (4.93)
Median [IQR]
16 [12–20]
10 [8–13]
8 [7–12]
12 [8–16]
NAa
9 [8–13]
APOEε4, APOEε4−/APOEε4+ (%APOEε4+)
46/151 (23.4%)
254/251 (50.3%)
33/93 (26.2%)
37/139 (21%)
44/177 (19.9%)
115/413 (21.8%)
Follow-up, years
Mean (SD)
2.02 (1.99)
1.09 (1.49)
3.33 (1.93)
1.71 (1.47)
1.76 (2.09)
0.59 (1.23)
Median [IQR]
1.71 [0–2.82]
0 [0–2.06]
3.46 [2.07–4.52]
1.52 [0.242–2.66]
NAa
0 [0–0.383]
Aβ1–42, pg/ml
Mean (SD)
1148 (397)
562 (165)
817 (399)
938 (446)
715 (417)
1000 (500)
Median [IQR]
1118 [849–1371]
556 [432–673]
703 [542–1009]
850 [569–1229]
583 [430–892]
896 [609–1323]
Aβ1–40, pg/ml
Mean (SD)
11,694 (3595)
12,790 (3781)
11,506 (4189)
10,806 (4357)
11,673 (4678)
11,399 (4390)
Median [IQR]
11,329 [9238–13,777]
12,541 [10,125–15,122]
10,885 [8882–14,234]
10,140 [7710–13,334]
11,035 [8346–14,594]
10,638 [8112–13,986]
Aβ1–42/Aβ1–40
Mean (SD)
0.0991 (0.0181)
0.0453 (0.0108)
0.0727 (0.0263)
0.0877 (0.0221)
0.0615 (0.0226)
0.0862 (0.0225)
Median [IQR]
0.104 [0.0986–0.109]
0.0445 [0.0376–0.0514]
0.0662 [0.0503–0.0996]
0.0964 [0.0731–0.103]
0.0562 [0.0422–0.078]
0.0943 [0.0665–0.103]
tTau, pg/ml
Mean (SD)
255 (152)
748 (358)
456 (334)
387 (260)
644 (520)
334 (231)
Median [IQR]
230 [174–291]
656 [488–915]
361 [253–525]
322 [222–456]
489 [262–870]
292 [213–378]
pTau181, pg/ml
Mean (SD)
37.3 (27.3)
122 (60.3)
70.5 (55.5)
49.9 (39.2)
100 (96.8)
45 (27.4)
Median [IQR]
31.6 [24.9–42]
105 [78.7–145]
51 [35.7–83]
39.7 [29.2–54.1]
63.6 [29.8–151]
41 [29.3–52.7]
NfL, pg/ml
Mean (SD)
475 (256)
1330 (1824)
1108 (570)
2079 (1836)
815 (773)
1488 (1340)
Median [IQR]
458 [320–533]
981 [791–1254]
918 [719–1297]
1412 [884–2767]
614 [357–1014]
1089 [595–1878]
Clinical stage, CN/MCI/dementia (% MCI)
177/0/0
0/296/208
2/60/64
5/90/80
NAa
27/348/125
Amyloid profile (Aβ1–42[−]Ratio[−]/Aβ1–42[−]Ratio[+]/Aβ1–42[+]Ratio[−]/Aβ1–42[+]Ratio[+])
165/13/11/8
4/154/2/358
50/29/7/42
120/8/23/35
61/37/13/114
345/45/47/100
MMSE Mini-Mental State Examination, CSF cerebrospinal fluid, CN cognitively normal, AD Alzheimer’s disease, DLB dementia with Lewy bodies; FTLD frontotemporal lobar degeneration-related syndrome, MCI mild cognitive impairment
aDue to specific particularities of the clinical and cognitive assessment in the context of intellectual disability, participants with Down syndrome were excluded from the prognostic analysis

Aβ1–42 and the Aβ1–42/Aβ1–40 ratio in CSF show high but not perfect agreement

Figure 1 shows the distribution of participants based on their CSF Aβ1–42 and Aβ1–42/Aβ1–40 ratio measures. The correlation between these two parameters was rho = 0.71, p < 0.001. Using Q95% cutoffs as described in the “Material and methods” section, Aβ1–42 and Aβ1–42/Aβ1–40 ratio had an overall agreement of 78.3% in the whole sample, as both measures were normal (Aβ1–42[−]Ratio[−]) in 41.6% and both were abnormal (Aβ1–42[+]Ratio[+]) in 36.7% of participants. Within each diagnostic category, the agreement ranged from 69.9% (AD group) to 87.9% (CN group). More details about the agreement after applying other cutoffs values and with other biomarkers can be found in Additional file 1.

Aβ1–42/Aβ1–40 ratio is more strongly associated with tau markers than Aβ1–42

We next studied the association of Aβ1–42 and the Aβ1–42/Aβ1–40 ratio with the other CSF biomarkers by assessing Spearman correlations in the whole sample and within diagnostic groups. In the whole sample, Aβ1–42 showed a significant correlation with tTau (rho = − 0.25, p < 0.001) and pTau181 (rho = − 0.32, p < 0.001). These correlations were stronger for the Aβ1–42/Aβ1–40 ratio (rho = − 0.69 and rho = − 0.75, respectively, both p < 0.001). Both Aβ1–42 and the Aβ1–42/Aβ1–40 ratio showed a similar correlation with NfL (rho = − 0.26 and rho = − 0.32, respectively, both p < 0.001). These stronger associations of Aβ1–42/Aβ1–40 ratio with tTau and pTau181 were observed within all symptomatic diagnostic categories (Additional file 1).
Next, we compared CSF levels of tTau, pTau181 and NfL between all four amyloid profiles. The objective of this analysis was to assess the particularities of those groups where the Aβ1–42 and the Aβ1–42/Aβ1–40 ratio are discordant. As expected, compared to the Aβ1–42[−]Ratio[−] group and after adjusting by age and sex, the Aβ1–42[+]Ratio[+] profile was associated with higher levels of tTau (Tukey post hoc p < 0.001), pTau181 (Tukey post hoc p < 0.001), and NfL (Tukey post hoc p = 0.001). But we also found differences in tTau and pTau181 levels between the two discordant profiles. As seen in Fig. 2, Aβ1–42[−]Ratio[+] participants showed higher levels of tTau (Tukey post hoc p < 0.001) and pTau181 (Tukey post hoc p < 0.001) compared to those with Aβ1–42[+]Ratio[−]. These differences were also observed within all diagnostic categories and in all clinical stages (Additional file 1). These results indicate that reduced levels of the Aβ1–42/Aβ1–40 ratio are associated with high levels of CSF tau markers, regardless of the status of Aβ1–42 alone.

Aβ1–42/Aβ1–40 ratio is more strongly associated with cognitive and functional progression than Aβ1–42

We studied the association of amyloid profiles with cognitive and functional progression in patients with MCI (n = 794) through Kaplan-Meier survival curves and age- and sex-adjusted Cox regression analysis. Supplementary Table 3 describes the characteristics of this subgroup. Due to specific particularities of the clinical and cognitive assessment in the context of intellectual disability, participants with Down syndrome were not included in this analysis. As displayed in Fig. 3, we found that patients with MCI with a low Aβ1–42/Aβ1–40 ratio had worse cognitive outcomes reflected by an earlier decline in MMSE scores. Compared to the Aβ1–42[−]Ratio[−] group, the adjusted risk of presenting a MMSE score lower than 24 during follow-up was 1.77 (1.25–2.49) times higher in the Aβ1–42[−]Ratio[+] group, 1.78 (1.33–2.39) times higher in the Aβ1–42[+]Ratio[+], but not different in the Aβ1–42[+]Ratio[−] group (p = 0.28). Similarly, the adjusted risk of progression to dementia was 1.55 (0.96–2.50) times higher in the Aβ1–42[−]Ratio[+] group and 2.07 (1.43–2.99) times higher in the Aβ1–42[+]Ratio[+] group compared to that of the Aβ1–42[−]Ratio[−]. The adjusted risk of progression to dementia in the Aβ1–42[+]Ratio[−] group was not significantly different from the Aβ1–42[−]Ratio[−] group (p = 0.26).
We also fitted linear-mixed models to assess the changes in longitudinal cognitive and functional measures by amyloid profiles. As shown in Fig. 4, after adjusting by baseline MMSE score, baseline age, sex, years of education, pTau181 levels, APOEε4 status and diagnosis, participants with low Aβ1–42/Aβ1–40 ratio presented larger decreases in MMSE scores. The model estimated an annual decrease of − 1.32 (− 1.55 to − 1.09) points when Aβ1–42 was low and of − 0.86 (− 1.18 to − 0.55) points when Aβ1–42 was in the normal range. However, the annual change in MMSE scores in the two groups with normal Aβ1–42/Aβ1–40 ratio was not significantly different from zero. As displayed in Fig. 4, we also found that both groups with low Aβ1–42/Aβ1–40 had larger annual decreases in the FCSRT total score, estimated in − 1.21 in Aβ1–42[−]Ratio[+] and − 1.5 in Aβ1–42[+]Ratio[+], compared to 0.05 in Aβ1–42[−]Ratio[−] and 0.1 in Aβ1–42[+]Ratio[−]. Positivity in the amyloid ratio was also associated to larger annual increases in the cognitive-functional scale CDR-SOB, of 0.46 (0.22–0.71) in Aβ1–42[−]Ratio[+] and 0.65 (0.46-0.85) in Aβ1–42[+]Ratio[+], compared to 0.27 (0.16–0.39) in Aβ1–42[−]Ratio[−] and no significant changes in Aβ1–42[+]Ratio[−].

Discussion

The results of our study indicate that, compared to Aβ1–42 alone, the Aβ1–42/Aβ1–40 CSF ratio is more strongly associated with tau markers and with cognitive and functional progression. Regardless of the Aβ1–42 status, participants with low CSF Aβ1–42/Aβ1–40 ratio showed a biochemical and clinical profile characterized by increased levels of CSF tTau and pTau181, worse functional prognosis and larger cognitive decline. Interestingly, when the Aβ1–42/Aβ1–40 ratio was normal, participants with low Aβ1–42 levels had a biochemical and clinical profile similar to that of participants with normal Aβ1–42. Our findings provide evidence suggesting that the use of the Aβ1–42/Aβ1–40 ratio is less confounded by other comorbidities or processes, and thus, this work favors the use of the Aβ1–42/Aβ1–40 ratio as a marker of AD in clinical laboratories and in clinical trials.
The use of CSF Aβ1–42 alone as a marker of amyloid plaques entails some limitations. Numerous studies have reported that low concentrations of Aβ1–42 can be found in some non-AD conditions such as prionopathies, bacterial meningitis, inflammatory diseases, amyloid angiopathy, or frontotemporal dementia [1014, 39], thus limiting its diagnostic accuracy. Different hypotheses have been suggested to explain these findings. Among other possibilities, low levels of Aβ1–42 in these contexts could be in relation with a decrease in Aβ generation due to neuronal or synaptic loss [10, 15] or the consequence of abnormal clearance through the blood-brain barrier [10]. As Aβ1–40 would be similarly affected by these processes, the use of the Aβ1-42/Aβ1-40 ratio could compensate the reduction in these situations to some extent, thus being less influenced by these processes and reflecting more accurately the presence of amyloid plaques. The stronger association of the Aβ1–42/Aβ1–40 ratio to other AD markers (tau markers) is in line with these hypotheses. Another limitation for the use of Aβ1–42 alone is that it is particularly sensitive to preanalytical and analytical variations, such as changes in the material of collection or storage tubes, number of freeze-thaw cycles, and volume of aliquoted CSF for storage [33, 40, 41]. In our study, we took advantage of a large cohort of subjects with various neurodegenerative and non-neurodegenerative conditions where CSF was collected using the same preanalytical protocol and analyzed under the same standard operating procedures. We found that the overall agreement between Aβ1–42 and the Aβ1–42/Aβ1–40 ratio did not exceed 85% in the whole sample, regardless of the cutoffs definition, suggesting that both markers might be tracking similar but not identical processes or that they are influenced differently by other factors.
We found that CSF concentrations of tTau and pTau181 were higher in the presence of low Aβ1–42/Aβ1–40 ratio, regardless of the Aβ1–42 status. This association was present in the whole sample but also within each diagnostic group. Thus, in the groups of CN, AD, and Down syndrome, less likely affected by non-AD pathology, low Aβ1–42/Aβ1–40 ratio values, but not low Aβ1–42 levels alone, were associated with high concentrations of markers of neurofibrillary pathology and neurodegeneration (pTau181, tTau, and NfL). In other contexts (DLB, FTLD, and other diagnoses), low levels of Aβ1–42 were only associated with markers of neurofibrillary pathology in the presence of reduced Aβ1–42/Aβ1–40 ratio. Our findings indicate that the ratio is more strongly associated to the AD pathophysiological process (both as main and comorbid pathology) and also support the idea that the isolated reduction of Aβ1–42 levels in CSF might reflect additional processes (such as neuronal or synaptic loss) or that the chrono pathology of their changes along the disease is not identical. These results are in line with our previous study showing that the Aβ1–42/Aβ1–40 ratio presents a stronger correlation with cerebral amyloid burden than Aβ1–42 alone [22].
Baseline levels of the Aβ1–42/Aβ1–40 ratio were also influential in the cognitive and functional outcomes of participants in our study. We found that participants with low Aβ1–42/Aβ1–40 ratio had faster cognitive and functional worsening, especially in the group with low Aβ1–42 but also when Aβ1–42 was in the normal range. The group with a low Aβ1–42/Aβ1–40 ratio also presented a more rapid decline in episodic memory measured by the free and cued selective reminding test. These findings support the use of the Aβ1–42/Aβ1–40 ratio over Aβ1–42 alone in the prognostic assessment of patients with cognitive decline.
The major strengths of our study are the large sample size and the inclusion of a variety of diagnoses. Another relevant strength is the fact that the same standard operating procedures were used in the processing and analysis of all samples. Aβ1–42 and Aβ1–40 were measured simultaneously, and we followed the same preanalytical and analytical protocol in all samples, thus minimizing the impact of confounders, which are critical in the case of amyloid-β peptides. Lastly, we replicated our results by using different levels of cutoffs, defined as percentiles from a cognitively normal population. This approach allowed us to match pairs of cutoffs for Aβ1–42 and Aβ1–42/Aβ1–40 ratio that had comparable levels of sensitivity and specificity. But we also acknowledge some limitations.

Limitations of the study

Despite the large sample size, extensive cognitive repeated measures were not available for all participants, thus limiting the statistical power in the longitudinal analysis. On the other hand, amyloid PET imaging was only available in a reduced group of participants previously reported [22].

Conclusions of the study

The present work highlights the importance of routinely measuring Aβ1–40 in the CSF in combination with Aβ1–42 to assess the Aβ1–42/Aβ1–40 ratio, as this measure reflects more accurately the presence of amyloid plaques and is a useful and robust tool for the diagnostic and prognostic evaluation of patients with cognitive decline. The fact that the Aβ1–42/Aβ1–40 ratio shows a stronger association than Aβ1–42 with markers of neurofibrillary pathology and with cognitive and functional decline strengths the utility of this ratio in the clinical context of symptomatic and preclinical AD but also to detect concomitant pathology in other neurodegenerative diseases.

Acknowledgements

We are grateful to all participants in the study and their families. We also thank all the clinical team members that were involved in the selection and assessment of participants in the SPIN cohort and the laboratory teams for sample handling, biomarker analyses, and structural support. We also thank Fujirebio for providing technical support and part of the reagents necessary to complete the study.

Declarations

The ethics committee of Hospital Sant Pau approved all procedures included in this study following the standards for medical research in humans recommended by the Declaration of Helsinki. All participants or their legally authorized representatives gave written informed consent before enrolment in the study.
All authors revised the manuscript for content and provided critical feedback.

Competing interests

Daniel Alcolea is employed by Hospital de la Santa Creu i Sant Pau and received research grants from Pla Estratègic de Recerca i Innovació en Salut (PERIS SLT006/17/125) and from Instituto de Salud Carlos III (PI18/00435 and INT19/00016). He participated in the advisory boards from Fujirebio-Europe and Roche Diagnostics and received speaker honoraria from Fujirebio-Europe, Roche Diagnostics, Nutricia, Zambon S.A.U., Esteve, and from Krka Farmacéutica S.L.
Constance Delaby is employed by Université de Montpellier and CHU de Montpellier. Declarations of interest: none
Teresa Estellés is employed by Biomedical Research Institute Sant Pau. Declarations of interest: none
Nuole Zhu is employed by Hospital de la Santa Creu i Sant Pau. Declarations of interest: none
Javier Arranz is employed by Biomedical Research Institute Sant Pau. Declarations of interest: none
Isabel Barroeta is employed by Hospital de la Santa Creu i Sant Pau. Declarations of interest: none
María Carmona-Iragui is employed by Hospital de la Santa Creu i Sant Pau. Declarations of interest: none
Ignacio Illán-Gala is supported by the Global Brain Health Institute (Atlantic Fellow for Equity in Brain Health and pilot award for global brain health leaders GBHI ALZ UK-21-720973) and the “Juan Rodés” grant from the Institute of Health Carlos III (JR20/00018).
Miguel Santos-Santos is employed by Hospital de la Santa Creu i Sant Pau. He is funded by a “Juan Rodés” research grant from the Institute of Health Carlos III.
Miren Altuna is employed by Biomedical Research Institute Sant Pau. Dr. Altuna is funded by a “Río Hortega” research grant from the Institute of Health Carlos III.
Isabel Sala is employed by Hospital de la Santa Creu i Sant Pau. Declarations of interest: none
M. Belén Sánchez-Saudinós is employed by Biomedical Research Institute Sant Pau. Declarations of interest: none
Laura Videla is employed by Fundació Catalana Síndrome de Down. Declarations of interest: none
Sílvia Valldeneu is employed by Biomedical Research Institute Sant Pau. Declarations of interest: none
Andrea Subirana is employed by Biomedical Research Institute Sant Pau. Declarations of interest: none
Mireia Tondo is employed by Hospital de la Santa Creu i Sant Pau. Declarations of interest: none
Francisco Blanco-Vaca is employed by Hospital de la Santa Creu i Sant Pau. Declarations of interest: none.
Sylvain Lehmann is employed by the University and the Hospital of Montpellier. He participated in advisory boards from Fujirebio-Europe and Roche.
Olivia Belbin is employed by Biomedical Research Institute Sant Pau. Dr. Belbin is funded by a “Miguel Servet” research grant from the Institute of Health Carlos III.
Rafael Blesa is employed by Hospital de la Santa Creu i Sant Pau and received research grants from Institute of Health Carlos III, Fundació Bancària Obra Social La Caixa, and Fundació La Marató de TV3. He participated in the advisory boards from Lilly and Nutricia, and he received speaker honoraria and travel funding from Novartis and Nutricia.
Juan Fortea is employed by Hospital de la Santa Creu i Sant Pau and received research grants from Institute of Health Carlos III, National Institutes of Health, Fundació La Marató de TV3, and Pla Estratègic de Recerca i Innovació en Salut (PERIS). Dr. Fortea has served as a consultant for Novartis and Lundbeck; has received honoraria for lectures from Roche, NovoNordisk, Esteve, and Biogen; and served at advisory boards for AC Immune, Zambon, and Lundbeck.
Alberto Lleó is employed by Hospital de la Santa Creu i Sant Pau and received research grants from CIBERNED, Institute of Health Carlos III, Generalitat de Catalunya (PERIS and AGAUR), and Fundación BBVA. He participated in the advisory boards from Fujirebio-Europe, Novartis, Roche Diagnostics, Otsuka Pharmaceutical, Nutricia, Zambón S.A.U., and Biogen and received speaker honoraria from Lilly, Biogen, KRKA, and Zambon.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, et al. Alzheimer’s disease. Lancet. 2021;397(10284):1577–90. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, et al. Alzheimer’s disease. Lancet. 2021;397(10284):1577–90.
2.
Zurück zum Zitat Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;4422:1–12. Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;4422:1–12.
3.
Zurück zum Zitat Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC, Holtzman DM. Cerebrospinal fluid tau/β-amyloid42 ratio as a prediction of cognitive decline in nondemented older adults. Arch Neurol. 2007;64:343–9.CrossRef Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC, Holtzman DM. Cerebrospinal fluid tau/β-amyloid42 ratio as a prediction of cognitive decline in nondemented older adults. Arch Neurol. 2007;64:343–9.CrossRef
4.
Zurück zum Zitat Gabelle A, Dumurgier J, Vercruysse O, Paquet C, Bombois S, Laplanche JL, et al. Impact of the 2008-2012 french Alzheimer plan on the use of cerebrospinal fluid biomarkers in research memory center: the PLM study. J Alzheimers Dis. 2013;34:297–305.CrossRef Gabelle A, Dumurgier J, Vercruysse O, Paquet C, Bombois S, Laplanche JL, et al. Impact of the 2008-2012 french Alzheimer plan on the use of cerebrospinal fluid biomarkers in research memory center: the PLM study. J Alzheimers Dis. 2013;34:297–305.CrossRef
5.
Zurück zum Zitat Dubois B, Feldman HH, Jacova C, Hampel H, Molinuevo JL, Blennow K, et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 2014;13:614–29.CrossRef Dubois B, Feldman HH, Jacova C, Hampel H, Molinuevo JL, Blennow K, et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 2014;13:614–29.CrossRef
6.
Zurück zum Zitat McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.CrossRef McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.CrossRef
7.
Zurück zum Zitat Dubois B, Villain N, Frisoni GB, Rabinovici GD, Sabbagh M, Cappa S, et al. Clinical diagnosis of Alzheimer’s disease: recommendations of the International Working Group. Lancet Neurol. 2021;20:484–96.CrossRef Dubois B, Villain N, Frisoni GB, Rabinovici GD, Sabbagh M, Cappa S, et al. Clinical diagnosis of Alzheimer’s disease: recommendations of the International Working Group. Lancet Neurol. 2021;20:484–96.CrossRef
8.
Zurück zum Zitat Lleó A, Cavedo E, Parnetti L, Vanderstichele H, Herukka SK, Andreasen N, et al. Cerebrospinal fluid biomarkers in trials for Alzheimer and Parkinson diseases. Nat Rev Neurol. 2015;11:41–55.CrossRef Lleó A, Cavedo E, Parnetti L, Vanderstichele H, Herukka SK, Andreasen N, et al. Cerebrospinal fluid biomarkers in trials for Alzheimer and Parkinson diseases. Nat Rev Neurol. 2015;11:41–55.CrossRef
9.
Zurück zum Zitat Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018;14:535–62.CrossRef Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018;14:535–62.CrossRef
10.
Zurück zum Zitat Lewczuk P, Riederer P, O’Bryant SE, Verbeek MM, Dubois B, Visser PJ, et al. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: an update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry. 2018;19:244–328.CrossRef Lewczuk P, Riederer P, O’Bryant SE, Verbeek MM, Dubois B, Visser PJ, et al. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: an update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry. 2018;19:244–328.CrossRef
11.
Zurück zum Zitat Iadanza MG, Jackson MP, Hewitt EW, Ranson NA, Radford SE. A new era for understanding amyloid structures and disease. Nat Rev Mol Cell Biol. 2018;19(12):755–73. Iadanza MG, Jackson MP, Hewitt EW, Ranson NA, Radford SE. A new era for understanding amyloid structures and disease. Nat Rev Mol Cell Biol. 2018;19(12):755–73.
12.
Zurück zum Zitat Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11:155–9.CrossRef Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11:155–9.CrossRef
13.
Zurück zum Zitat Spies PE, Slats D, Sjögren JMC, Kremer BPH, Verhey FRJ, Rikkert MGMO, et al. The cerebrospinal fluid amyloid beta42/40 ratio in the differentiation of Alzheimer’s disease from non-Alzheimer’s dementia. Curr Alzheimer Res. 2010;7:470–6.CrossRef Spies PE, Slats D, Sjögren JMC, Kremer BPH, Verhey FRJ, Rikkert MGMO, et al. The cerebrospinal fluid amyloid beta42/40 ratio in the differentiation of Alzheimer’s disease from non-Alzheimer’s dementia. Curr Alzheimer Res. 2010;7:470–6.CrossRef
14.
Zurück zum Zitat Llorens F, Schmitz M, Karch A, Cramm M, Lange P, Gherib K, et al. Comparative analysis of cerebrospinal fluid biomarkers in the differential diagnosis of neurodegenerative dementia. Alzheimers Dement. 2016;12:577–89.CrossRef Llorens F, Schmitz M, Karch A, Cramm M, Lange P, Gherib K, et al. Comparative analysis of cerebrospinal fluid biomarkers in the differential diagnosis of neurodegenerative dementia. Alzheimers Dement. 2016;12:577–89.CrossRef
15.
Zurück zum Zitat Spies PE, Verbeek MM, van Groen T, Claassen JAHR. Reviewing reasons for the decreased CSF Abeta42 concentration in Alzheimer disease. Front Biosci (Landmark Ed). 2012;17:2024–34.CrossRef Spies PE, Verbeek MM, van Groen T, Claassen JAHR. Reviewing reasons for the decreased CSF Abeta42 concentration in Alzheimer disease. Front Biosci (Landmark Ed). 2012;17:2024–34.CrossRef
16.
Zurück zum Zitat Lehmann S, Delaby C, Boursier G, Catteau C, Ginestet N, Tiers L, 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.CrossRef Lehmann S, Delaby C, Boursier G, Catteau C, Ginestet N, Tiers L, 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.CrossRef
17.
Zurück zum Zitat Hansson O, Lehmann S, Otto M, Zetterberg H, Lewczuk P. Advantages and disadvantages of the use of the CSF amyloid β (Aβ) 42/40 ratio in the diagnosis of Alzheimer’s disease. Alzheimers Res Ther. 2019;11:34.CrossRef Hansson O, Lehmann S, Otto M, Zetterberg H, Lewczuk P. Advantages and disadvantages of the use of the CSF amyloid β (Aβ) 42/40 ratio in the diagnosis of Alzheimer’s disease. Alzheimers Res Ther. 2019;11:34.CrossRef
18.
Zurück zum Zitat Cullen NC, Leuzy A, Janelidze S, Palmqvist S, Svenningsson AL, Stomrud E, et al. Plasma biomarkers of Alzheimer’s disease improve prediction of cognitive decline in cognitively unimpaired elderly populations. Nat Commun. 2021;12:3555.CrossRef Cullen NC, Leuzy A, Janelidze S, Palmqvist S, Svenningsson AL, Stomrud E, et al. Plasma biomarkers of Alzheimer’s disease improve prediction of cognitive decline in cognitively unimpaired elderly populations. Nat Commun. 2021;12:3555.CrossRef
19.
Zurück zum Zitat Alcolea D, Delaby C, Muñoz L, Torres S, Estellés T, Zhu N, et al. Use of plasma biomarkers for AT(N) classification of neurodegenerative dementias. J Neurol Neurosurg Psychiatry. 2021;92(11):1206–14. Alcolea D, Delaby C, Muñoz L, Torres S, Estellés T, Zhu N, et al. Use of plasma biomarkers for AT(N) classification of neurodegenerative dementias. J Neurol Neurosurg Psychiatry. 2021;92(11):1206–14.
20.
Zurück zum Zitat Doecke JD, Pérez-Grijalba V, Fandos N, Fowler C, Villemagne VL, Masters CL, et al. Total Aβ42/Aβ40 ratio in plasma predicts amyloid-PET status, independent of clinical AD diagnosis. Neurology. 2020;94:E1580–91.CrossRef Doecke JD, Pérez-Grijalba V, Fandos N, Fowler C, Villemagne VL, Masters CL, et al. Total Aβ42/Aβ40 ratio in plasma predicts amyloid-PET status, independent of clinical AD diagnosis. Neurology. 2020;94:E1580–91.CrossRef
21.
Zurück zum Zitat Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554:249–54.CrossRef Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554:249–54.CrossRef
22.
Zurück zum Zitat Alcolea D, Pegueroles J, Muñoz L, Camacho V, López-Mora D, Fernández-León A, et al. Agreement of amyloid PET and CSF biomarkers for Alzheimer’s disease on Lumipulse. Ann Clin Transl Neurol. 2019;6(9):1815–24. Alcolea D, Pegueroles J, Muñoz L, Camacho V, López-Mora D, Fernández-León A, et al. Agreement of amyloid PET and CSF biomarkers for Alzheimer’s disease on Lumipulse. Ann Clin Transl Neurol. 2019;6(9):1815–24.
23.
Zurück zum Zitat Alcolea D, Clarimón J, Carmona-Iragui M, Illán-Gala I, Morenas-Rodríguez E, Barroeta I, et al. The Sant Pau Initiative on Neurodegeneration (SPIN) cohort: a data set for biomarker discovery and validation in neurodegenerative disorders. Alzheimer’s Dement Transl Res Clin Interv. 2019;5:597–609.CrossRef Alcolea D, Clarimón J, Carmona-Iragui M, Illán-Gala I, Morenas-Rodríguez E, Barroeta I, et al. The Sant Pau Initiative on Neurodegeneration (SPIN) cohort: a data set for biomarker discovery and validation in neurodegenerative disorders. Alzheimer’s Dement Transl Res Clin Interv. 2019;5:597–609.CrossRef
24.
Zurück zum Zitat Alcolea D, Vilaplana E, Suárez-Calvet M, Illán-Gala I, Blesa R, Clarimón J, et al. CSF sAPPβ, YKL-40, and neurofilament light in frontotemporal lobar degeneration. Neurology. 2017;89:178–88.CrossRef Alcolea D, Vilaplana E, Suárez-Calvet M, Illán-Gala I, Blesa R, Clarimón J, et al. CSF sAPPβ, YKL-40, and neurofilament light in frontotemporal lobar degeneration. Neurology. 2017;89:178–88.CrossRef
25.
Zurück zum Zitat Illán-Gala I, Pegueroles J, Montal V, Alcolea D, Vilaplana E, Bejanin A, et al. APP-derived peptides reflect neurodegeneration in frontotemporal dementia. Ann Clin Transl Neurol. 2019;6:2518–30.CrossRef Illán-Gala I, Pegueroles J, Montal V, Alcolea D, Vilaplana E, Bejanin A, et al. APP-derived peptides reflect neurodegeneration in frontotemporal dementia. Ann Clin Transl Neurol. 2019;6:2518–30.CrossRef
26.
Zurück zum Zitat McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor J-PP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies. Neurology. 2017;89:88–100.CrossRef McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor J-PP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies. Neurology. 2017;89:88–100.CrossRef
27.
Zurück zum Zitat Strong MJ, Abrahams S, Goldstein LH, Woolley S, Mclaughlin P, Snowden J, et al. Amyotrophic lateral sclerosis - frontotemporal spectrum disorder (ALS-FTSD): revised diagnostic criteria. Amyotroph Lateral Scler Front Degener. 2017;18:153–74.CrossRef Strong MJ, Abrahams S, Goldstein LH, Woolley S, Mclaughlin P, Snowden J, et al. Amyotrophic lateral sclerosis - frontotemporal spectrum disorder (ALS-FTSD): revised diagnostic criteria. Amyotroph Lateral Scler Front Degener. 2017;18:153–74.CrossRef
28.
Zurück zum Zitat Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, et al. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:270–9.CrossRef Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, et al. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:270–9.CrossRef
29.
Zurück zum Zitat Fortea J, Carmona-Iragui M, Benejam B, Fernández S, Videla L, Barroeta I, et al. Plasma and CSF biomarkers for the diagnosis of Alzheimer’s disease in adults with Down syndrome: a cross-sectional study. Lancet Neurol. 2018;17:860–9.CrossRef Fortea J, Carmona-Iragui M, Benejam B, Fernández S, Videla L, Barroeta I, et al. Plasma and CSF biomarkers for the diagnosis of Alzheimer’s disease in adults with Down syndrome: a cross-sectional study. Lancet Neurol. 2018;17:860–9.CrossRef
30.
Zurück zum Zitat Fortea J, Vilaplana E, Carmona-Iragui M, Benejam B, Videla L, Barroeta I, et al. Clinical and biomarker changes of Alzheimer’s disease in adults with Down syndrome: a cross-sectional study. Lancet. 2020;395:1988–97.CrossRef Fortea J, Vilaplana E, Carmona-Iragui M, Benejam B, Videla L, Barroeta I, et al. Clinical and biomarker changes of Alzheimer’s disease in adults with Down syndrome: a cross-sectional study. Lancet. 2020;395:1988–97.CrossRef
31.
Zurück zum Zitat Sala I, Illán-Gala I, Alcolea D, Sánchez-Saudinós MBMB, Salgado SASA, Morenas-Rodríguez E, et al. Diagnostic and prognostic value of the combination of two measures of verbal memory in mild cognitive impairment due to Alzheimer’s disease. J Alzheimers Dis. 2017;58:909–18.CrossRef Sala I, Illán-Gala I, Alcolea D, Sánchez-Saudinós MBMB, Salgado SASA, Morenas-Rodríguez E, et al. Diagnostic and prognostic value of the combination of two measures of verbal memory in mild cognitive impairment due to Alzheimer’s disease. J Alzheimers Dis. 2017;58:909–18.CrossRef
32.
Zurück zum Zitat Alcolea D, Martínez-Lage P, Izagirre A, Clerigué M, Carmona-Iragui M, Alvarez RMRM, et al. Feasibility of lumbar puncture in the study of cerebrospinal fluid biomarkers for Alzheimer’s disease: a multicenter study in Spain. J Alzheimers Dis. 2014;39:719–26.CrossRef Alcolea D, Martínez-Lage P, Izagirre A, Clerigué M, Carmona-Iragui M, Alvarez RMRM, et al. Feasibility of lumbar puncture in the study of cerebrospinal fluid biomarkers for Alzheimer’s disease: a multicenter study in Spain. J Alzheimers Dis. 2014;39:719–26.CrossRef
33.
Zurück zum Zitat del Campo M, Mollenhauer B, Bertolotto A, Engelborghs S, Hampel H, Simonsen AH, et al. Recommendations to standardize preanalytical confounding factors in Alzheimer’s and Parkinson’s disease cerebrospinal fluid biomarkers: an update. Biomark Med. 2012;6:419–30.CrossRef del Campo M, Mollenhauer B, Bertolotto A, Engelborghs S, Hampel H, Simonsen AH, et al. Recommendations to standardize preanalytical confounding factors in Alzheimer’s and Parkinson’s disease cerebrospinal fluid biomarkers: an update. Biomark Med. 2012;6:419–30.CrossRef
34.
Zurück zum Zitat Delaby C, Muñoz L, Torres S, Nadal A, Le Bastard N, Lehmann S, et al. Impact of CSF storage volume on the analysis of Alzheimer’s disease biomarkers on an automated platform. Clin Chim Acta Elsevier. 2019;490:98–101.CrossRef Delaby C, Muñoz L, Torres S, Nadal A, Le Bastard N, Lehmann S, et al. Impact of CSF storage volume on the analysis of Alzheimer’s disease biomarkers on an automated platform. Clin Chim Acta Elsevier. 2019;490:98–101.CrossRef
36.
Zurück zum Zitat Carrillo MC, Blennow K, Soares H, Lewczuk P, Mattsson N, Oberoi P, et al. Global standardization measurement of cerebral spinal fluid for Alzheimer’s disease: an update from the Alzheimer’s Association Global Biomarkers Consortium. Alzheimers Dement. 2013;9:137–40.CrossRef Carrillo MC, Blennow K, Soares H, Lewczuk P, Mattsson N, Oberoi P, et al. Global standardization measurement of cerebral spinal fluid for Alzheimer’s disease: an update from the Alzheimer’s Association Global Biomarkers Consortium. Alzheimers Dement. 2013;9:137–40.CrossRef
37.
Zurück zum Zitat Mattsson N, Andreasson U, Persson S, Carrillo MC, Collins S, Chalbot S, et al. CSF biomarker variability in the Alzheimer’s Association quality control program. Alzheimers Dement. 2013;9:251–61.CrossRef Mattsson N, Andreasson U, Persson S, Carrillo MC, Collins S, Chalbot S, et al. CSF biomarker variability in the Alzheimer’s Association quality control program. Alzheimers Dement. 2013;9:251–61.CrossRef
38.
Zurück zum Zitat Carmona-Iragui M, Balasa M, Benejam B, Alcolea D, Fernández S, Videla L, et al. Cerebral amyloid angiopathy in Down syndrome and sporadic and autosomal-dominant Alzheimer’s disease. Alzheimers Dement. 2017;13:1251–60.CrossRef Carmona-Iragui M, Balasa M, Benejam B, Alcolea D, Fernández S, Videla L, et al. Cerebral amyloid angiopathy in Down syndrome and sporadic and autosomal-dominant Alzheimer’s disease. Alzheimers Dement. 2017;13:1251–60.CrossRef
39.
Zurück zum Zitat Strydom A, Coppus A, Blesa R, Danek A, Fortea J, Hardy J, et al. Alzheimer’s disease in Down syndrome: an overlooked population for prevention trials. Alzheimer’s Dement Transl Res Clin Interv. Elsevier Inc. 2018;4:703–13. Strydom A, Coppus A, Blesa R, Danek A, Fortea J, Hardy J, et al. Alzheimer’s disease in Down syndrome: an overlooked population for prevention trials. Alzheimer’s Dement Transl Res Clin Interv. Elsevier Inc. 2018;4:703–13.
40.
Zurück zum Zitat Perret-Liaudet A, Pelpel M, Tholance Y, Dumont B, Vanderstichele H, Zorzi W, et al. Risk of Alzheimer’s disease biological misdiagnosis linked to cerebrospinal collection tubes. J Alzheimers Dis. 2012;31:13–20.CrossRef Perret-Liaudet A, Pelpel M, Tholance Y, Dumont B, Vanderstichele H, Zorzi W, et al. Risk of Alzheimer’s disease biological misdiagnosis linked to cerebrospinal collection tubes. J Alzheimers Dis. 2012;31:13–20.CrossRef
41.
Zurück zum Zitat Lehmann S, Schraen S, Quadrio I, Paquet C, Bombois S, Delaby C, et al. Impact of harmonization o tubes on Alzheimer’s disease diagnosis. Alzheimers Dement. 2014;10:S390–S394.e2.PubMed Lehmann S, Schraen S, Quadrio I, Paquet C, Bombois S, Delaby C, et al. Impact of harmonization o tubes on Alzheimer’s disease diagnosis. Alzheimers Dement. 2014;10:S390–S394.e2.PubMed
Metadaten
Titel
The Aβ1–42/Aβ1–40 ratio in CSF is more strongly associated to tau markers and clinical progression than Aβ1–42 alone
verfasst von
Constance Delaby
Teresa Estellés
Nuole Zhu
Javier Arranz
Isabel Barroeta
María Carmona-Iragui
Ignacio Illán-Gala
Miguel Ángel Santos-Santos
Miren Altuna
Isabel Sala
M. Belén Sánchez-Saudinós
Laura Videla
Sílvia Valldeneu
Andrea Subirana
Mireia Tondo
Francisco Blanco-Vaca
Sylvain Lehmann
Olivia Belbin
Rafael Blesa
Juan Fortea
Alberto Lleó
Daniel Alcolea
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Alzheimer's Research & Therapy / Ausgabe 1/2022
Elektronische ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-022-00967-z

Weitere Artikel der Ausgabe 1/2022

Alzheimer's Research & Therapy 1/2022 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Obstipation Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.