Skip to main content
Erschienen in: Alzheimer's Research & Therapy 1/2023

Open Access 01.12.2023 | Research

Body mass index and two-year change of in vivo Alzheimer’s disease pathologies in cognitively normal older adults

verfasst von: Seunghoon Lee, Min Soo Byun, Dahyun Yi, Min Jung Kim, Joon Hyung Jung, Nayeong Kong, Gijung Jung, Hyejin Ahn, Jun-Young Lee, Koung Mi Kang, Chul-Ho Sohn, Yun-Sang Lee, Yu Kyeong Kim, Dong Young Lee, for the KBASE Research Group

Erschienen in: Alzheimer's Research & Therapy | Ausgabe 1/2023

Abstract

Background

Low body mass index (BMI) or underweight status in late life is associated with an increased risk of dementia or Alzheimer’s disease (AD). However, the relationship between late-life BMI and prospective longitudinal changes of in-vivo AD pathology has not been investigated.

Methods

This prospective longitudinal study was conducted as part of the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer’s Disease (KBASE). A total of 194 cognitive normal older adults were included in the analysis. BMI at baseline was measured, and two-year changes in brain Aβ and tau deposition on PET imaging were used as the main outcomes. Linear mixed-effects (LME) models were used to examine the relationships between late-life BMI and longitudinal change in AD neuropathological biomarkers.

Results

A lower BMI at baseline was significantly associated with a greater increase in tau deposition in AD-signature region over 2 years (β, -0.018; 95% CI, -0.028 to -0.004; p = .008), In contrast, BMI was not related to two-year changes in global Aβ deposition (β, 0.0002; 95% CI, -0.003 to 0.002, p = .671). An additional exploratory analysis for each sex showed lower baseline BMI was associated with greater increases in tau deposition in males (β, -0.027; 95% CI, -0.046 to -0.009; p = 0.007), but not in females.

Discussion

The findings suggest that lower BMI in late-life may predict or contribute to the progression of tau pathology over the subsequent years in cognitively unimpaired older adults.
Hinweise

Publisher's Note

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

Introduction

A large amount of evidence indicates that body mass index (BMI) is related to the risk of Alzheimer’s disease (AD) dementia [13]. Several studies have shown that being overweight or obese in midlife increases the risk of AD dementia or cerebral beta-amyloid (Aβ) deposition [46]. However, multiple studies have also reported that low BMI or being underweight in late life was associated with an increased risk of dementia [1, 3, 7] and that higher BMI in late life was a protective factor for AD dementia [3, 8].
Several amyloid positron emission topography (PET) studies with cross-sectional design demonstrated that lower BMI in late life was associated with increased brain Aβ burden in cognitive normal(CN) elderly individuals [912]. Other cross-sectional studies also reported a correlation between lower late-life BMI and increased CSF total tau or phosphorylated-tau [9, 13, 14]. A study has reported that there is a correlation between frailty and brain atrophy as measured by MR imaging, with greater frailty being associated with greater brain atrophy in community dwelling older adults [15]. All these findings are consistent with the association between low BMI in late life and a higher risk of AD dementia. In regard of longitudinal approach, some prospective studies have reported that brain Aβ is associated with future decreased of BMI, suggesting that weight loss, as well as cognitive decline, may be a clinical manifestation of AD process [16, 17]. However, the relationship between late-life BMI and prospective longitudinal changes of in-vivo AD pathology has not yet been investigated. Understanding such relationship of current BMI and future prospective changes of AD pathological biomarkers in cognitively unimpaired older adults could make it clearer whether lower BMI can predict or contribute to the progression of AD pathology and subsequently to AD dementia risk.
In this context, we tested the hypothesis that a lower late-life BMI is related to a greater prospective increase in in-vivo AD pathology, including Aβ and tau deposition, in cognitively healthy individuals. Additionally, as several previous studies showed prominent sex-related differences for the relationship between BMI and AD dementia risk [18, 19] and brain Aβ deposition [11, 20], we explored the same relationship for each sex separately.

Methods

Participants

This study was performed as part of the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer’s Disease (KBASE), an ongoing prospective cohort study conducted from 2014 [21]. As of 2018, 297 CN adults between 55 and 90 years old were recruited and received a baseline evaluation, including a comprehensive clinical assessment and BMI measurement. Among them, 194 participants who had completed both baseline and two-year follow-up neuroimaging scans for brain Aβ deposition were included in the current study. The inclusion criteria were as follows: (a) age 55–90 years, (b) Clinical Dementia Rating score of 0, and (c) no diagnosis of mild cognitive impairment or dementia. The exclusion criteria were as follows: (a) any serious medical, psychiatric, or neurological disorder that could affect mental function; (b) any severe communication problem that would render clinical examination or brain scanning difficult; (c) contraindications to magnetic resonance imaging (MRI), such as a pacemaker or claustrophobia; (d) absence of a reliable informant; (e) illiteracy defined as a lack of the ability to read; and (f) participation in another clinical trial or treatment with an investigational product. Research clinicians determined the presence of any exclusion criteria by referring to the results of laboratory examinations and MRI scans. They also evaluated the clinical data collected by trained nurses during systematic interviews of participants and their reliable informants during the screening period. More detailed information on the recruitment of the KBASE cohort has been presented in a previous report [21]. The study was approved by the Institutional Review Board of the Seoul National University Hospital and SNU-SMG Boramae Medical Center, South Korea. All participants provided written informed consent.

Clinical assessment

The participants underwent comprehensive baseline clinical assessments based on the KBASE protocol [21] by trained psychiatrists. The assessments incorporated the Korean version of the Consortium to Establish a Registry for Alzheimer`s Disease Assessment (CERAD-K) [22, 23]. The presence of vascular risk factors (VRFs), including diabetes, hypertension, dyslipidemia, coronary heart disease, transient ischemic attack, and stroke, was assessed from data collected during systematic interviews by trained nurses with participants and their informants. Based on the number of VRFs, the vascular risk score (VRS) was calculated [24] and treated as a continuous variable for analyses. The Geriatric Depression Scale (GDS) [25] was used to measure the severity of depressive symptoms. Smoking status (never/former/smoker), alcohol intake status (never/former/drinker), and lifetime physical activity were evaluated through interviews with nurses. The Lifetime Total Physical Activity Questionnaire [26] was used to assess lifetime physical activity. A metabolic equivalent (MET) value was assigned to the intensity of activity based on the compendium of physical activities [27].

BMI measurement

BMI was calculated as weight in kilograms divided by the square of the height in meters. It was measured at the baseline visit. Trained research nurses measured the participants’ height and body weight using standard anthropometric methods.

Measurement of Aβ biomarker

All participants underwent [11C] Pittsburgh compound B (PiB)-positron emission tomography (PET) scans using a 3.0 T Biograph mMR (PET-MR) scanner (Siemens, Washington DC, USA). These scans were conducted according to the manufacturer’s protocols at baseline and two-year follow-up visit. We described the details of PiB-PET image acquisition and preprocessing previously [28]. The automatic anatomic labeling algorithm and the region combination method [29] were used to determine regions of interest (ROIs) and to characterize PiB retention in the frontal, lateral parietal, posterior cingulate-precuneus, and lateral temporal regions. A global cortical ROI (consisting of the four smaller ROIs) was also defined. The global Aβ retention value, the standardized uptake value ratio (SUVR) for the global cortical ROI, was calculated by dividing the mean values for all voxels of the global cortical ROI by a mean reference region. For the analysis of baseline data, the inferior cerebellar gray matter in the spatially unbiased infratentorial template for the cerebellum (SUIT) atlas [30] was used as the reference region. A participant was classified as Aβ positive if the SUVR was > 1.21 [31]. For longitudinal analysis, the reference region included the inferior cerebellar grey matter, cerebellar white matter (thresholded at 50%), pons, and cerebrum white matter (thresholded at 95% and eroded by three voxels) [32, 33].

Measurement of cerebral tau deposition

A subset of subjects (n = 45) underwent two [18F] AV-1451 PET scans using a Biograph True Point 40 PET/CT platform (Siemens, USA) per the manufacturer’s guidelines at a two-year time interval. While the first PiB-PET imaging was performed during the baseline visit, the first AV-1451 PET imaging was performed at an average of 2.55 (standard deviation = 0.26) years after that visit. The details of AV-1451 PET imaging acquisition and preprocessing have been described previously [28]. We quantified the AV-1451 SUVR of a priori ROI of the “AD-signature region” of tau accumulation to estimate cerebral tau deposition. This was a size-weighted average of the partial volume-corrected uptake by the entorhinal, amygdala, parahippocampal, fusiform, inferior temporal, and middle temporal ROIs [34, 35]. It was done using the cerebral hemispheric white matter ROI from FreeSurfer in the partial volume code [36] as a reference region. The literature recommends using cerebral white matter as the reference region for intensity normalization in longitudinal AV-1451 PET data analysis [37].

Statistical analyses

We tested linear mixed-effects (LME) models with random intercepts to examine the relationships between late-life BMI and longitudinal change in AD neuropathological biomarkers. All models included Aβ or tau deposition values as dependent variables on the first and second PET scan. Model 1 included baseline BMI, age, sex, APOE4, baseline Aβ or Tau and their interactions with time. In Model 2, we additionally controlled for VRS and its interaction with time to adjust for the confounding effects of vascular risk factors, considering the well perceived role of vascular risk factors in AD development [38, 39]. A random intercept was included for each subject, and time was calculated as the number of years from baseline. For exploratory purposes, the LME model including baseline BMI, age, APOE4, baseline Aβ or tau and their interactions with time was analyzed for each sex. Statistical analyses were performed using R version 4.0, and jamovi version 2.2.1 (The jamovi project, www.​jamovi.​org). In all analyses, p < 0.05 was considered as statistical significance.

Availability of data and materials

The datasets generated and analyzed during the present study are not publicly available, owing to ethics considerations and privacy restrictions. Data might be obtained from the corresponding author after approval by the Institutional Review Board of the Seoul National University Hospital, South Korea.

Results

Participant characteristics

The demographic and clinical characteristics of all subjects are presented in Table 1.
Table 1
Participant characteristics
Variable
Total
Tau PET
No. of individuals
194
45
Age at baseline, year (mean ± SD)
68.4 ± 8.1
70.3 ± 7.3
Female, No. (%)
102 (53)
25 (55.6)
Education, year, median (IQR)
12 (7)
12(4)
APOE ε4 carriers, No. (%)
35 (18.0)
8 (17.8)
Baseline BMI, kg/m2 (mean ± SD)
24.20 ± 3.01
24.5 ± 2.55
Vascular risk factor, No. (%)
 Diabetes mellitus
35 (18.0)
10 (22.2)
 Hypertension
87 (44.8)
21 (46.7)
 Hyperlipidemia
69 (35.6)
15 (33.3)
 Coronary heart disease
11 (5.7)
3 (6.7)
 Stroke
0
0
 TIA
1 (0.5)
1 (2.2)
VRS, median (IQR)
1 (0–2)
1 (0–2)
Alcohol use, No. (%)
 Never
98 (50.5)
25(55.6)
 Former
23 (11.9)
7 (15.6)
 Drinker
73 (37.6)
13 (28.9)
Smoking status, No. (%)
 Never
125 (64.4)
30 (66.7)
 Former
57 (29.4)
13 (28.9)
 Drinker
12 (6.2)
2 (4.4)
Lifetime physical activity, MET, median (IQR)
68.7 (57.2)
64.5 (42.1)
Cerebral Aβ deposition, SUVR
 Baseline global Aβ retention, median (IQR)
1.12 (0.11)
1.13 (0.11)
 Baseline Aβ positive (> 1.20), No, (%)
43 (22)
13 (28.9)
Global Tau deposition, SUVR
 Baseline Tau retention, median (IQR)
1.02 (0.14)
1.00 (0.16)
Abbreviations: β-amyloid protein, IQR Interquartile range, MET metabolic equivalent, SD standard deviation, SUVR standardized uptake value ratio, VRS vascular risk score

Association of BMI at baseline with cerebral Aβ and tau deposition change over two years

Baseline BMI was not significantly associated with global Aβ deposition change during the two-year follow-up period for models 1 and 2. In contrast, a lower baseline BMI was significantly associated with a greater increase in tau deposition in the AD-signature region over two years (Table 2). When we conducted the same analyses including three BMI strata (below -1 SD, median BMI, above 1SD) instead of BMI as a continuous variable for the purpose of demonstration, the results were similar (Fig. 1 and Table 3). We also performed sensitivity analyses, including the GDS score, smoking status, alcohol intake status, and lifetime physical activity as additional covariates, which showed similar findings (Table 4).
Table 2
Association of the baseline BMI with neuroimaging biomarker changes for 2-year
 
Estimate
95% CI
t value
p value
Dependent variable: Aβ deposition
 Model 1a
  Baseline BMI x time
0.000
-0.003 to 0.002
-0.359
.720
 Model 2b
  Baseline BMI x time
0.000
-0.003 to 0.002
-0.426
.671
Dependent variable: Tau deposition
 Model 1a
  Baseline BMI x time
-0.018
-0.030 to -0.006
-3.027
.003
 Model 2b
  Baseline BMI x time
-0.016
-0.028 to -0.004
-2.703
.008
Abbreviations: β-amyloid protein, BMI body mass index, CI confidence interval
aAdjusted for age, sex, APOE4, baseline Aβ or Tau and their interactions with time
bAdjusted for age, sex, APOE4, baseline Aβ a or Tau, vascular risk score, and their interactions with time
Table 3
Association of the baseline BMI strata with neuroimaging biomarker changes for 2 years
 
Estimate
95% CI
t value
p value
Dependent variable: Aβ retention
 Baseline BMI strata x time
-0.004
-0.016 to 0.008
-0.653
.514
Dependent variable: Tau deposition
 Baseline BMI strata x time
-0.067
-0.118 to -0.017
-2.606
.011
Abbreviations: β-amyloid protein, APOE apolipoprotein e, BMI body mass index, CI confidence interval
Adjusted for age, sex, APOE e4, baseline Aβ or Tau and their interactions with time
Table 4
Results from sensitivity analyses for the association of the baseline BMI with neuroimaging biomarker changes for 2 years
 
Estimate
95% CI
t value
p value
Dependent variable: Aβ retention
 Baseline BMI x time
0.0003
-0.003 to 0.002
-0.232
.817
Dependent variable: Tau deposition
 Baseline BMI x time
-0.016
-0.028 to -0.004
-2.551
.012
Abbreviations: β-amyloid protein, APOE apolipoprotein e, BMI body mass index, CI confidence interval
Adjusted for smoking status, alcohol intake status, and lifetime physical activity as well as age, sex, apolipoprotein e4, vascular risk score, baseline Aβ or tau and their interactions with time

Association of BMI with cerebral Aβ and tau deposition change over two years stratified based on sex

A lower baseline BMI was associated with increased tau deposition over two years in men, but not in women (Table 5). As for Aβ changes, neither women nor men showed significant association between baseline BMI and cerebral Aβ changes over two years.
Table 5
Association of the baseline BMI with neuroimaging biomarker changes according to Sex
 
Female
Male
 
Estimate
95% CI
p value
Estimate
95% CI
p value
Dependent variable: global Aβ retention
 Baseline BMI x time
0.001
-0.002 to 0.004
.671
-0.002
-0.005 to 0.001
.250
Dependent variable: Global Tau deposition
 Baseline BMI x time
-0.013
-0.029 to 0.003
.115
-0.027
-0.046 to -0.009
.007
Abbreviations: β-amyloid protein, BMI body mass index, CI confidence interval
Adjusted for age, APOE4, baseline Aβ or tau and their interactions with time

Discussion

The present study found that a lower BMI was associated with greater increase of brain tau deposition over two years in cognitively healthy older adults. Further exploratory analyses showed that this association was significant in men, but not in women. In contrast, baseline BMI was not significantly associated with the change in cerebral Aβ deposition.
Our findings on the relationship between lower baseline BMI and greater increase in brain tau deposition are in agreement with previous reports of a cross-sectional association between lower BMI and higher CSF tau levels in older individuals [9, 13, 14, 40]. Although it is not easy to clearly explain the mechanisms underlying the relationship between lower BMI and greater increase in brain tau deposition, some possible explanations can be provided. First, the association between lower BMI and increased tau in the brain may be mediated by decreased leptin levels, a hormone synthesized from body fat that regulates appetite and energy metabolism [41]. Several laboratory studies have demonstrated that leptin reduces phosphorylated tau in in vivo and in vitro experiments [4244]. This possibility of leptin mediation may further explain why the association is more prominent in males than females. As leptin expression is higher in subcutaneous than visceral fat [41, 45], it is more likely to be lower in thin males than thin females. Even at the same BMI, males have less subcutaneous fat than females [41, 46]. Second, alterations in insulin regulation may influence brain tau pathology [47]. Insulin inhibits tau hyperphosphorylation [48, 49], and plasma insulin can be transported via the blood–brain barrier into the cerebrospinal fluid [50]. Given people with low BMI have lower plasma insulin levels than those with higher BMI [51], decreased insulin levels in thin individuals may accelerate the brain deposition of pathological tau protein by ameliorating the insulin function to inhibit tau phosphorylation.
Additional exploratory analyses demonstrated male-specific association between lower baseline BMI and increased tau deposition over two years. The finding is generally in line with our previous report which showed a male-specific association between mid-life lower BMI and reduced AD-signature region cortical thickness [11]. Both findings may explain the neuropathological links underlying sex-specific association between BMI and AD dementia risk repeatedly shown by epidemiological studies [18, 19, 52].
We did not find a significant relationship between baseline BMI and longitudinal brain Aβ changes for all participants. This disagrees with previous cross-sectional findings for the association between lower BMI and higher Aβ deposition in cognitively healthy older individuals [912]. Given very gradual accumulation of Aβ in the brain [53], the two-year follow-up period may be relatively short to assess changes in Aβ deposition. Such short-term observations may affect the null finding for the association between BMI and changes in Aβ deposition.
Our finding for the relationship between lower late-life BMI and prospective increase in in vivo tau pathology is a novel one. Nevertheless, the present study had several potential limitations that should be addressed. First, as the proportion of participants with obesity (BMI over 30 mg/kg2) and underweight (BMI below 18.5 mg/kg2) was very small in our sample [3.1% (n = 6) and 1% (n = 2) of overall participants, respectively], it might be difficult to investigate the influence of higher BMI, obesity or very low BMI on the change in AD pathologies. Second, the first tau PET was performed at an average of 2.55 years (standard deviation 0.26 years) after BMI measurement at baseline, whereas the first amyloid PET was performed at baseline. This temporal gap may have influenced the results. However, when we controlled for the temporal gap as an additional covariate, the results did not change. Third, only a subset of participants (n = 45) underwent two tau PET scans, whereas all participants underwent two amyloid PET scans. Despite the smaller sample size for tau, we found a statistically significant relationship between BMI and change in tau deposition. This indicates that a small sample size may not be a critical issue. Nevertheless, a study with a larger sample size is required to confirm the sex-specific association between BMI and pathological changes in AD patients. Finally, mood status and various lifestyle factors may confound the association between BMI and changes in AD biomarkers. To minimize this possibility, we performed additional sensitivity analyses including smoking status, alcohol status, lifetime physical activity, and GDS as additional covariates and still obtained similar results. However, we could not control for food intake or dietary quality due to the lack of information.

Conclusion

The present findings suggest that lower BMI in late life may predict or contribute to the progression of tau pathology over subsequent years in cognitively unimpaired older adults. Concerning the prevention of AD dementia or related cognitive decline, more attention needs to be paid to avoid being underweight in late life, particularly in men.

Acknowledgements

The authors are grateful to all participants in this study.
A complete list of KBASE research group members can be found at http://​kbase.​kr/​.

Declarations

All studies were conducted in accordance with the approved guidelines. The study was approved by the Institutional Review Board of the Seoul National University Hospital and SNU-SMG Boramae Medical Center, South Korea. All participants provided written informed consent.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This 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 Tolppanen A-M, Ngandu T, Kåreholt I, Laatikainen T, Rusanen M, Soininen H, et al. Midlife and late-life body mass index and late-life dementia: results from a prospective population-based cohort. J Alzheimers Dis. 2014;38:201–9.CrossRefPubMed Tolppanen A-M, Ngandu T, Kåreholt I, Laatikainen T, Rusanen M, Soininen H, et al. Midlife and late-life body mass index and late-life dementia: results from a prospective population-based cohort. J Alzheimers Dis. 2014;38:201–9.CrossRefPubMed
2.
Zurück zum Zitat Qizilbash N, Gregson J, Johnson ME, Pearce N, Douglas I, Wing K, et al. BMI and risk of dementia in two million people over two decades: a retrospective cohort study. Lancet Diabetes Endocrinol. 2015;3:431–6.CrossRefPubMed Qizilbash N, Gregson J, Johnson ME, Pearce N, Douglas I, Wing K, et al. BMI and risk of dementia in two million people over two decades: a retrospective cohort study. Lancet Diabetes Endocrinol. 2015;3:431–6.CrossRefPubMed
3.
Zurück zum Zitat Hughes T, Borenstein A, Schofield E, Wu Y, Larson E. Association between late-life body mass index and dementia The Kame Project. Neurology. 2009;72:1741–6.CrossRefPubMedPubMedCentral Hughes T, Borenstein A, Schofield E, Wu Y, Larson E. Association between late-life body mass index and dementia The Kame Project. Neurology. 2009;72:1741–6.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Xu W, Atti A, Gatz M, Pedersen N, Johansson B, Fratiglioni L. Midlife overweight and obesity increase late-life dementia risk A population-based twin study. Neurology. 2011;76:1568–74.CrossRefPubMedPubMedCentral Xu W, Atti A, Gatz M, Pedersen N, Johansson B, Fratiglioni L. Midlife overweight and obesity increase late-life dementia risk A population-based twin study. Neurology. 2011;76:1568–74.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Whitmer RA, Gunderson EP, Barrett-Connor E, Quesenberry CP, Yaffe K. Obesity in middle age and future risk of dementia: a 27 year longitudinal population based study. BMJ. 2005;330:1360.CrossRefPubMedPubMedCentral Whitmer RA, Gunderson EP, Barrett-Connor E, Quesenberry CP, Yaffe K. Obesity in middle age and future risk of dementia: a 27 year longitudinal population based study. BMJ. 2005;330:1360.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Gottesman RF, Schneider ALC, Zhou Y, Coresh J, Green E, Gupta N, et al. Association between midlife vascular risk factors and estimated brain amyloid deposition. JAMA. 2017;317:1443–50.CrossRefPubMedPubMedCentral Gottesman RF, Schneider ALC, Zhou Y, Coresh J, Green E, Gupta N, et al. Association between midlife vascular risk factors and estimated brain amyloid deposition. JAMA. 2017;317:1443–50.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Fitzpatrick AL, Kuller LH, Lopez OL, Diehr P, O’Meara ES, Longstreth W, et al. Midlife and late-life obesity and the risk of dementia: cardiovascular health study. Arch Neurol. 2009;66:336–42.CrossRefPubMedPubMedCentral Fitzpatrick AL, Kuller LH, Lopez OL, Diehr P, O’Meara ES, Longstreth W, et al. Midlife and late-life obesity and the risk of dementia: cardiovascular health study. Arch Neurol. 2009;66:336–42.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Atti AR, Palmer K, Volpato S, Winblad B, De Ronchi D, Fratiglioni L. Late-life body mass index and dementia incidence: nine-year follow-up data from the Kungsholmen Project. J Am Geriatr Soc. 2008;56:111–6.CrossRefPubMed Atti AR, Palmer K, Volpato S, Winblad B, De Ronchi D, Fratiglioni L. Late-life body mass index and dementia incidence: nine-year follow-up data from the Kungsholmen Project. J Am Geriatr Soc. 2008;56:111–6.CrossRefPubMed
9.
Zurück zum Zitat Vidoni ED, Townley RA, Honea RA, Burns JM, Alzheimer’s Disease Neuroimaging I. Alzheimer disease biomarkers are associated with body mass index. Neurology. 2011;77:1913–20. Vidoni ED, Townley RA, Honea RA, Burns JM, Alzheimer’s Disease Neuroimaging I. Alzheimer disease biomarkers are associated with body mass index. Neurology. 2011;77:1913–20.
10.
Zurück zum Zitat Hsu DC, Mormino EC, Schultz AP, Amariglio RE, Donovan NJ, Rentz DM, et al. Lower Late-Life Body-Mass Index is Associated with Higher Cortical Amyloid Burden in Clinically Normal Elderly. J Alzheimers Dis. 2016;1–9. Hsu DC, Mormino EC, Schultz AP, Amariglio RE, Donovan NJ, Rentz DM, et al. Lower Late-Life Body-Mass Index is Associated with Higher Cortical Amyloid Burden in Clinically Normal Elderly. J Alzheimers Dis. 2016;1–9.
11.
Zurück zum Zitat Lee SH, Byun MS, Lee JH, Yi D, Sohn BK, Lee JY, et al. Sex-Specific Association of Lifetime Body Mass Index with Alzheimer’s Disease Neuroimaging Biomarkers. J Alzheimers Dis. 2020;75:767–77. Lee SH, Byun MS, Lee JH, Yi D, Sohn BK, Lee JY, et al. Sex-Specific Association of Lifetime Body Mass Index with Alzheimer’s Disease Neuroimaging Biomarkers. J Alzheimers Dis. 2020;75:767–77.
12.
Zurück zum Zitat Thirunavu V, McCullough A, Su Y, Flores S, Dincer A, Morris JC, et al. Higher Body Mass Index Is Associated with Lower Cortical Amyloid-beta Burden in Cognitively Normal Individuals in Late-Life. J Alzheimers Dis. 2019;69:817–27. Thirunavu V, McCullough A, Su Y, Flores S, Dincer A, Morris JC, et al. Higher Body Mass Index Is Associated with Lower Cortical Amyloid-beta Burden in Cognitively Normal Individuals in Late-Life. J Alzheimers Dis. 2019;69:817–27.
13.
Zurück zum Zitat Ewers M, Schmitz S, Hansson O, Walsh C, Fitzpatrick A, Bennett D, et al. Body mass index is associated with biological CSF markers of core brain pathology of Alzheimer’s disease. Neurobiol Aging. 2012;33:1599–608.CrossRefPubMed Ewers M, Schmitz S, Hansson O, Walsh C, Fitzpatrick A, Bennett D, et al. Body mass index is associated with biological CSF markers of core brain pathology of Alzheimer’s disease. Neurobiol Aging. 2012;33:1599–608.CrossRefPubMed
14.
Zurück zum Zitat Sun Z, Wang ZT, Sun FR, Shen XN, Xu W, Ma YH, et al. Late-life obesity is a protective factor for prodromal Alzheimer’s disease: a longitudinal study. Aging. 2020;12:2005–17. Sun Z, Wang ZT, Sun FR, Shen XN, Xu W, Ma YH, et al. Late-life obesity is a protective factor for prodromal Alzheimer’s disease: a longitudinal study. Aging. 2020;12:2005–17.
15.
Zurück zum Zitat Del Brutto OH, Mera RM, Cagino K, Fanning KD, Milla-Martinez MF, Nieves JL, et al. Neuroimaging signatures of frailty: A population-based study in community-dwelling older adults (the Atahualpa Project). Geriatr Gerontol Int. 2017;17:270–6.CrossRefPubMed Del Brutto OH, Mera RM, Cagino K, Fanning KD, Milla-Martinez MF, Nieves JL, et al. Neuroimaging signatures of frailty: A population-based study in community-dwelling older adults (the Atahualpa Project). Geriatr Gerontol Int. 2017;17:270–6.CrossRefPubMed
16.
Zurück zum Zitat Buchman AS, Capuano AW, VanderHorst V, Wilson RS, Oveisgharan S, Schneider JA, et al. Brain β-amyloid links the association of change in BMI with cognitive decline in community-dwelling older adults. J Gerontol A Biol Sci Med Sci. 2021;glab320. Buchman AS, Capuano AW, VanderHorst V, Wilson RS, Oveisgharan S, Schneider JA, et al. Brain β-amyloid links the association of change in BMI with cognitive decline in community-dwelling older adults. J Gerontol A Biol Sci Med Sci. 2021;glab320.
17.
Zurück zum Zitat Xu W, Sun F-R, Tan C-C, Tan L, Alzheimer’s Disease Neuroimaging Initiative. Weight Loss is a Preclinical Signal of Cerebral Amyloid Deposition and Could Predict Cognitive Impairment in Elderly Adults. J Alzheimers Dis JAD. 2020;77:449–56. Xu W, Sun F-R, Tan C-C, Tan L, Alzheimer’s Disease Neuroimaging Initiative. Weight Loss is a Preclinical Signal of Cerebral Amyloid Deposition and Could Predict Cognitive Impairment in Elderly Adults. J Alzheimers Dis JAD. 2020;77:449–56.
18.
Zurück zum Zitat Beydoun MA, Lhotsky A, Wang Y, Dal Forno G, An Y, Metter EJ, et al. Association of adiposity status and changes in early to mid-adulthood with incidence of Alzheimer’s disease. Am J Epidemiol. 2008;168:1179–89.CrossRefPubMedPubMedCentral Beydoun MA, Lhotsky A, Wang Y, Dal Forno G, An Y, Metter EJ, et al. Association of adiposity status and changes in early to mid-adulthood with incidence of Alzheimer’s disease. Am J Epidemiol. 2008;168:1179–89.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Dahl AK, Löppönen M, Isoaho R, Berg S, Kivelä S. Overweight and obesity in old age are not associated with greater dementia risk. J Am Geriatr Soc. 2008;56:2261–6.CrossRefPubMed Dahl AK, Löppönen M, Isoaho R, Berg S, Kivelä S. Overweight and obesity in old age are not associated with greater dementia risk. J Am Geriatr Soc. 2008;56:2261–6.CrossRefPubMed
20.
Zurück zum Zitat Chuang Y, An Y, Bilgel M, Wong D, Troncoso J, O’Brien RJ, et al. Midlife adiposity predicts earlier onset of Alzheimer’s dementia, neuropathology and presymptomatic cerebral amyloid accumulation. Mol Psychiatry. 2015; Chuang Y, An Y, Bilgel M, Wong D, Troncoso J, O’Brien RJ, et al. Midlife adiposity predicts earlier onset of Alzheimer’s dementia, neuropathology and presymptomatic cerebral amyloid accumulation. Mol Psychiatry. 2015;
21.
Zurück zum Zitat Byun MS, Yi D, Lee JH, Choe YM, Sohn BK, Lee JY, et al. Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer’s Disease: Methodology and Baseline Sample Characteristics. Psychiatry Investig. 2017;14:851–63. Byun MS, Yi D, Lee JH, Choe YM, Sohn BK, Lee JY, et al. Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer’s Disease: Methodology and Baseline Sample Characteristics. Psychiatry Investig. 2017;14:851–63.
22.
Zurück zum Zitat Morris JC, Heyman A, Mohs RC, Hughes J, van Belle G, Fillenbaum G, et al. The consortium to establish a registry for Alzheimer’s disease (CERAD): I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology. 1989; Morris JC, Heyman A, Mohs RC, Hughes J, van Belle G, Fillenbaum G, et al. The consortium to establish a registry for Alzheimer’s disease (CERAD): I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology. 1989;
23.
Zurück zum Zitat Lee JH, Lee KU, Lee DY, Kim KW, Jhoo JH, Kim JH, et al. Development of the Korean Version of the Consortium to Establish a Registry for Alzheimer’s Disease Assessment Packet (CERAD-K): Clinical and Neuropsychological Assessment Batteries. J Gerontol B Psychol Sci Soc Sci. 2002;57:P47-53.CrossRefPubMed Lee JH, Lee KU, Lee DY, Kim KW, Jhoo JH, Kim JH, et al. Development of the Korean Version of the Consortium to Establish a Registry for Alzheimer’s Disease Assessment Packet (CERAD-K): Clinical and Neuropsychological Assessment Batteries. J Gerontol B Psychol Sci Soc Sci. 2002;57:P47-53.CrossRefPubMed
24.
Zurück zum Zitat DeCarli C, Mungas D, Harvey D, Reed B, Weiner M, Chui H, et al. Memory impairment, but not cerebrovascular disease, predicts progression of MCI to dementia. Neurology. 2004;63:220–7.CrossRefPubMed DeCarli C, Mungas D, Harvey D, Reed B, Weiner M, Chui H, et al. Memory impairment, but not cerebrovascular disease, predicts progression of MCI to dementia. Neurology. 2004;63:220–7.CrossRefPubMed
25.
Zurück zum Zitat Kim JY, Park JH, Lee JJ, Huh Y, Lee SB, Han SK, et al. Standardization of the Korean version of the geriatric depression scale: reliability, validity, and factor structure. Psychiatry Investig. 2008;5:232.CrossRefPubMedPubMedCentral Kim JY, Park JH, Lee JJ, Huh Y, Lee SB, Han SK, et al. Standardization of the Korean version of the geriatric depression scale: reliability, validity, and factor structure. Psychiatry Investig. 2008;5:232.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Friedenreich CM, Courneya KS, Bryant HE. The lifetime total physical activity questionnaire: development and reliability. Med Sci Sports Exerc. 1998;30:266–74.CrossRefPubMed Friedenreich CM, Courneya KS, Bryant HE. The lifetime total physical activity questionnaire: development and reliability. Med Sci Sports Exerc. 1998;30:266–74.CrossRefPubMed
27.
Zurück zum Zitat Ainsworth BE, Haskell WL, Herrmann SD, Meckes N, Bassett DR Jr, Tudor-Locke C, et al. 2011 Compendium of Physical Activities: a second update of codes and MET values. Med Sci Sports Exerc. 2011;43:1575–81.CrossRefPubMed Ainsworth BE, Haskell WL, Herrmann SD, Meckes N, Bassett DR Jr, Tudor-Locke C, et al. 2011 Compendium of Physical Activities: a second update of codes and MET values. Med Sci Sports Exerc. 2011;43:1575–81.CrossRefPubMed
28.
Zurück zum Zitat Park J-C, Han S-H, Yi D, Byun MS, Lee JH, Jang S, et al. Plasma tau/amyloid-β1–42 ratio predicts brain tau deposition and neurodegeneration in Alzheimer’s disease. Brain. 2019;142:771–86.CrossRefPubMed Park J-C, Han S-H, Yi D, Byun MS, Lee JH, Jang S, et al. Plasma tau/amyloid-β1–42 ratio predicts brain tau deposition and neurodegeneration in Alzheimer’s disease. Brain. 2019;142:771–86.CrossRefPubMed
29.
Zurück zum Zitat Reiman EM, Chen K, Liu X, Bandy D, Yu M, Lee W, et al. Fibrillar amyloid-β burden in cognitively normal people at 3 levels of genetic risk for Alzheimer’s disease. Proc Natl Acad Sci. 2009;106:6820–5.CrossRefPubMedPubMedCentral Reiman EM, Chen K, Liu X, Bandy D, Yu M, Lee W, et al. Fibrillar amyloid-β burden in cognitively normal people at 3 levels of genetic risk for Alzheimer’s disease. Proc Natl Acad Sci. 2009;106:6820–5.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Diedrichsen J, Balsters JH, Flavell J, Cussans E, Ramnani N. A probabilistic MR atlas of the human cerebellum. Neuroimage. 2009;46:39–46. Diedrichsen J, Balsters JH, Flavell J, Cussans E, Ramnani N. A probabilistic MR atlas of the human cerebellum. Neuroimage. 2009;46:39–46.
31.
Zurück zum Zitat Villeneuve S, Rabinovici GD, Cohn-Sheehy BI, Madison C, Ayakta N, Ghosh PM, et al. Existing Pittsburgh Compound-B positron emission tomography thresholds are too high: statistical and pathological evaluation. Brain. 2015;138:2020–33.CrossRefPubMedPubMedCentral Villeneuve S, Rabinovici GD, Cohn-Sheehy BI, Madison C, Ayakta N, Ghosh PM, et al. Existing Pittsburgh Compound-B positron emission tomography thresholds are too high: statistical and pathological evaluation. Brain. 2015;138:2020–33.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Lowe VJ, Lundt ES, Senjem ML, Schwarz CG, Min H-K, Przybelski SA, et al. White Matter Reference Region in PET Studies of 11C-Pittsburgh Compound B Uptake: Effects of Age and Amyloid-β Deposition. J Nucl Med. 2018;59:1583–9.CrossRefPubMedPubMedCentral Lowe VJ, Lundt ES, Senjem ML, Schwarz CG, Min H-K, Przybelski SA, et al. White Matter Reference Region in PET Studies of 11C-Pittsburgh Compound B Uptake: Effects of Age and Amyloid-β Deposition. J Nucl Med. 2018;59:1583–9.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Schwarz CG, Senjem ML, Gunter JL, Tosakulwong N, Weigand SD, Kemp BJ, et al. Optimizing PiB-PET SUVR change-over-time measurement by a large-scale analysis of longitudinal reliability, plausibility, separability, and correlation with MMSE. Neuroimage. 2017;144:113–27. Schwarz CG, Senjem ML, Gunter JL, Tosakulwong N, Weigand SD, Kemp BJ, et al. Optimizing PiB-PET SUVR change-over-time measurement by a large-scale analysis of longitudinal reliability, plausibility, separability, and correlation with MMSE. Neuroimage. 2017;144:113–27.
34.
Zurück zum Zitat Jack CR Jr, Wiste HJ, Weigand SD, Therneau TM, Lowe VJ, Knopman DS, et al. Defining imaging biomarker cut points for brain aging and Alzheimer’s disease. Alzheimers Dement. 2017;13:205–16.CrossRefPubMed Jack CR Jr, Wiste HJ, Weigand SD, Therneau TM, Lowe VJ, Knopman DS, et al. Defining imaging biomarker cut points for brain aging and Alzheimer’s disease. Alzheimers Dement. 2017;13:205–16.CrossRefPubMed
35.
Zurück zum Zitat Maass A, Landau S, Baker SL, Horng A, Lockhart SN, La Joie R, et al. Comparison of multiple tau-PET measures as biomarkers in aging and Alzheimer’s disease. Neuroimage. 2017;157:448–63. Maass A, Landau S, Baker SL, Horng A, Lockhart SN, La Joie R, et al. Comparison of multiple tau-PET measures as biomarkers in aging and Alzheimer’s disease. Neuroimage. 2017;157:448–63.
36.
37.
Zurück zum Zitat Harrison TM, La Joie R, Maass A, Baker SL, Swinnerton K, Fenton L, et al. Longitudinal tau accumulation and atrophy in aging and alzheimer disease. Ann Neurol. 2019;85:229–40.CrossRefPubMedPubMedCentral Harrison TM, La Joie R, Maass A, Baker SL, Swinnerton K, Fenton L, et al. Longitudinal tau accumulation and atrophy in aging and alzheimer disease. Ann Neurol. 2019;85:229–40.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Luchsinger JA, Reitz C, Honig LS, Tang MX, Shea S, Mayeux R. Aggregation of vascular risk factors and risk of incident Alzheimer disease. Neurology. Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology; 2005;65:545–51. Luchsinger JA, Reitz C, Honig LS, Tang MX, Shea S, Mayeux R. Aggregation of vascular risk factors and risk of incident Alzheimer disease. Neurology. Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology; 2005;65:545–51.
40.
Zurück zum Zitat Mathys J, Gholamrezaee M, Henry H, von Gunten A, Popp J. Decreasing body mass index is associated with cerebrospinal fluid markers of Alzheimer’s pathology in MCI and mild dementia. Exp Gerontol. 2017;100:45–53. Mathys J, Gholamrezaee M, Henry H, von Gunten A, Popp J. Decreasing body mass index is associated with cerebrospinal fluid markers of Alzheimer’s pathology in MCI and mild dementia. Exp Gerontol. 2017;100:45–53.
41.
Zurück zum Zitat Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends Endocrinol Metab. 2000;11:327–32.CrossRefPubMed Ahima RS, Flier JS. Adipose tissue as an endocrine organ. Trends Endocrinol Metab. 2000;11:327–32.CrossRefPubMed
42.
Zurück zum Zitat Marwarha G, Dasari B, Prasanthi JR, Schommer J, Ghribi O. Leptin reduces the accumulation of Abeta and phosphorylated tau induced by 27-hydroxycholesterol in rabbit organotypic slices. J Alzheimers Dis. 2010;19:1007–19. Marwarha G, Dasari B, Prasanthi JR, Schommer J, Ghribi O. Leptin reduces the accumulation of Abeta and phosphorylated tau induced by 27-hydroxycholesterol in rabbit organotypic slices. J Alzheimers Dis. 2010;19:1007–19.
43.
Zurück zum Zitat Greco SJ, Sarkar S, Johnston JM, Zhu X, Su B, Casadesus G, et al. Leptin reduces Alzheimer’s disease-related tau phosphorylation in neuronal cells. Biochem Biophys Res Commun. 2008;376:536–41.CrossRefPubMedPubMedCentral Greco SJ, Sarkar S, Johnston JM, Zhu X, Su B, Casadesus G, et al. Leptin reduces Alzheimer’s disease-related tau phosphorylation in neuronal cells. Biochem Biophys Res Commun. 2008;376:536–41.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Greco SJ, Bryan KJ, Sarkar S, Zhu X, Smith MA, Ashford JW, et al. Leptin reduces pathology and improves memory in a transgenic mouse model of Alzheimer’s disease. J Alzheimers Dis. 2010;19:1155–67.CrossRefPubMedPubMedCentral Greco SJ, Bryan KJ, Sarkar S, Zhu X, Smith MA, Ashford JW, et al. Leptin reduces pathology and improves memory in a transgenic mouse model of Alzheimer’s disease. J Alzheimers Dis. 2010;19:1155–67.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Van Harmelen V, Reynisdottir S, Eriksson P, Thörne A, Hoffstedt J, Lönnqvist F, et al. Leptin secretion from subcutaneous and visceral adipose tissue in women. Diabetes. 1998;47:913–7.CrossRefPubMed Van Harmelen V, Reynisdottir S, Eriksson P, Thörne A, Hoffstedt J, Lönnqvist F, et al. Leptin secretion from subcutaneous and visceral adipose tissue in women. Diabetes. 1998;47:913–7.CrossRefPubMed
46.
Zurück zum Zitat Pradhan AD. Sex differences in the metabolic syndrome: implications for cardiovascular health in women. Clin Chem. 2014;60:44–52.CrossRefPubMed Pradhan AD. Sex differences in the metabolic syndrome: implications for cardiovascular health in women. Clin Chem. 2014;60:44–52.CrossRefPubMed
47.
Zurück zum Zitat Craft S, Watson GS. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol. 2004;3:169–78.CrossRefPubMed Craft S, Watson GS. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol. 2004;3:169–78.CrossRefPubMed
48.
Zurück zum Zitat Hong M, Lee VMY. Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem. 1997;272:19547–53.CrossRefPubMed Hong M, Lee VMY. Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem. 1997;272:19547–53.CrossRefPubMed
49.
Zurück zum Zitat Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, et al. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci. 2003;23:7084–92.CrossRefPubMedPubMedCentral Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, et al. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci. 2003;23:7084–92.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Poduslo JF, Curran GL, Wengenack TM, Malester B, Duff K. Permeability of proteins at the blood–brain barrier in the normal adult mouse and double transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2001;8:555–67.CrossRefPubMed Poduslo JF, Curran GL, Wengenack TM, Malester B, Duff K. Permeability of proteins at the blood–brain barrier in the normal adult mouse and double transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2001;8:555–67.CrossRefPubMed
51.
Zurück zum Zitat Neeland IJ, Turer AT, Ayers CR, Powell-Wiley TM, Vega GL, Farzaneh-Far R, et al. Dysfunctional adiposity and the risk of prediabetes and type 2 diabetes in obese adults. JAMA. 2012;308:1150–9.CrossRefPubMedPubMedCentral Neeland IJ, Turer AT, Ayers CR, Powell-Wiley TM, Vega GL, Farzaneh-Far R, et al. Dysfunctional adiposity and the risk of prediabetes and type 2 diabetes in obese adults. JAMA. 2012;308:1150–9.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Rahmani F, Wang Q, McKay NS, Keefe S, Hantler N, Hornbeck R, et al. Sex-specific patterns of body mass index relationship with white matter connectivity. J Alzheimers Dis. 86:1831–48. Rahmani F, Wang Q, McKay NS, Keefe S, Hantler N, Hornbeck R, et al. Sex-specific patterns of body mass index relationship with white matter connectivity. J Alzheimers Dis. 86:1831–48.
53.
Zurück zum Zitat Jack CR, Wiste HJ, Lesnick TG, Weigand SD, Knopman DS, Vemuri P, et al. Brain β-amyloid load approaches a plateau. Neurology. Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology; 2013;80:890–6. Jack CR, Wiste HJ, Lesnick TG, Weigand SD, Knopman DS, Vemuri P, et al. Brain β-amyloid load approaches a plateau. Neurology. Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology; 2013;80:890–6.
Metadaten
Titel
Body mass index and two-year change of in vivo Alzheimer’s disease pathologies in cognitively normal older adults
verfasst von
Seunghoon Lee
Min Soo Byun
Dahyun Yi
Min Jung Kim
Joon Hyung Jung
Nayeong Kong
Gijung Jung
Hyejin Ahn
Jun-Young Lee
Koung Mi Kang
Chul-Ho Sohn
Yun-Sang Lee
Yu Kyeong Kim
Dong Young Lee
for the KBASE Research Group
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Alzheimer's Research & Therapy / Ausgabe 1/2023
Elektronische ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-023-01259-w

Weitere Artikel der Ausgabe 1/2023

Alzheimer's Research & Therapy 1/2023 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Bluttest erkennt Parkinson schon zehn Jahre vor der Diagnose

10.05.2024 Parkinson-Krankheit Nachrichten

Ein Bluttest kann abnorm aggregiertes Alpha-Synuclein bei einigen Menschen schon zehn Jahre vor Beginn der motorischen Parkinsonsymptome nachweisen. Mit einem solchen Test lassen sich möglicherweise Prodromalstadien erfassen und die Betroffenen früher behandeln.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Wartezeit nicht kürzer, aber Arbeit flexibler

Psychotherapie Medizin aktuell

Fünf Jahren nach der Neugestaltung der Psychotherapie-Richtlinie wurden jetzt die Effekte der vorgenommenen Änderungen ausgewertet. Das Hauptziel der Novellierung war eine kürzere Wartezeit auf Therapieplätze. Dieses Ziel wurde nicht erreicht, es gab jedoch positive Auswirkungen auf andere Bereiche.

Update Neurologie

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