Skip to main content
Erschienen in: European Journal of Medical Research 1/2023

Open Access 01.12.2023 | Research

Causal relationships between delirium and Alzheimer's disease: a bidirectional two-sample Mendelian randomization study

verfasst von: Jiang Zheng, Xiaohui Du, Liu Yang, Hong Fu

Erschienen in: European Journal of Medical Research | Ausgabe 1/2023

Abstract

Background

Previous observational studies have reported that delirium has an association with an increased risk of Alzheimer's disease (AD), and that patients with AD have a higher risk of developing delirium. However, due to the limitations of observational study, it is challenging to confirm whether delirium has a causal effect on AD or reverse causation exists.

Methods

A bidirectional two-sample Mendelian randomization (MR) was performed to investigate the relationship between delirium and AD. Summary statistics from genome-wide association studies of delirium and AD phenotypes were utilized. Inverse-variance weighted (IVW) method was used as the main analysis approach, and additional analyses were performed using MR Egger, weighted median, simple mode and weighted mode to ensure result accuracy. Heterogeneity and horizontal pleiotropy were assessed using Cochran's Q statistics and MR Egger intercept, separately.

Results

The MR analyses showed that genetically predicted delirium was not associated with AD (IVW: odds ratio [OR] 0.98, 95% CI 0.91–1.05, P = 0.544; MR Egger: OR 0.98, 95% CI 0.83–1.15, P = 0.780; weighted median: OR 0.96, 95% CI 0.88–1.05, P = 0.323; simple mode: OR 0.91, 95% CI 0.80–1.04, P = 0.212; weighted mode: OR 0.93, 95% CI 0.83–1.05, P = 0.277). However, in the reverse direction, AD was associated with delirium (IVW: OR 1.32, 95% CI 1.13–1.54, P = 3.91E-04; MR Egger: OR 1.42, 95% CI 1.02–1.98, P = 5.60E-02; Weighted median: OR 1.39, 95% CI 1.18–1.63, P = 8.22E-05; Simple mode: OR 1.41, 95% CI 1.10–1.80, P = 1.41E−02; Weighted mode: OR 1.39, 95% CI 1.16–1.67, P = 3.23E-03).

Conclusion

Based on the results of our MR study, there is no bidirectional causality between delirium and AD, delirium is not associated with an increased risk of AD, while genetically predicted AD is a potential causal risk factor for delirium.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40001-023-01245-w.
Jiang Zheng and Xiaohui Du contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AD
Alzheimer’s disease
MR
Mendelian randomization
IVW
Inverse-variance weighted
SNPs
Single nucleotide polymorphisms
GWAS
Genome-wide association study
IGAP
International Genomics of Alzheimer’s Project
LD
Linkage disequilibrium
OR
Odds ratio

Introduction

Delirium is an acute cognitive function statues changing in a short time among hospitalized older adults, which can lead to serious outcomes and increase the medical costs [1, 2]. Delirium is often associated with longer hospital stays, serious complications, and mortality [35]. In addition, there is growing evidence suggesting that delirium may also increase the risk of developing dementia. Delirium has been reported to be a strong risk factor for dementia in the patients aged over 85 years and can increase the rate of cognitive deterioration in Alzheimer’s disease (AD) patients [68]. However, dementia itself is a significant predisposing factor for delirium, and nearly half of dementia cases go undiagnosed, whether delirium is related to new-onset dementia remains controversial [911]. AD is the most common cause of dementia, which accounts of 60–80% of all cases [12]. Although previous observational studies have identified a connection between dementia and delirium, the limitations of those observational studies (i.e., the inability to completely eliminate potential confounding factors, selection bias and small sample size) and the expensive and time-consuming nature of randomized controlled trails, make it challenging to determine a causal association between delirium and AD.
Mendelian randomization (MR) is a valuable tool for exploring causality between exposures and outcomes. By using single nucleotide polymorphisms (SNPs) that linked with exposures, MR can avoid the distribution of confounding bias and reverse causality that are present in observational studies [13, 14]. Therefore, we aimed to investigate the relationship between delirium and AD using a bidirectional two-sample MR in this study.

Materials and methods

Study design and MR assumptions

Figure 1 presents an overview of the design of our study. The MR analysis rests on following three critical assumptions: (a) the association hypothesis: genetic variant is associated with the exposure; (b) the independence hypothesis: genetic variant is not associated with any confounding factors; and (c) the exclusivity hypothesis: genetic variables only influence the outcome through the exposure.

Data source

We obtained genome-wide association study (GWAS) summary data related to delirium were sourced from the FinnGen Consortium, based on 2,612 cases and 325,306 controls of Finnish ancestry, and 21,168,109 SNPs were identified. The definition of delirium was based on the International Classification of Diseases, 10th Revision (ICD-10).
Summary data for the GWAS of AD from the International Genomics of Alzheimer’s Project (IGAP) consortium [15]. The GWAS study involved 21,982 patients with AD and 41,944 controls who were non-Hispanic White individuals, and 10,528,610 SNPs were identified.

Mendelian randomization

SNPs strongly (P < 5 × 10−8) associated with AD were used as instrumental variables (IVs), and a relaxed threshold (P < 5 × 10−6) was used to acquire more IVs when exploring the effect of delirium on AD, as a threshold has been used in previous study [16]. For the selection of independent instruments, SNPs that were not in linkage disequilibrium (LD) with each other (r2 < 0.001) and a distance of 10,000 kb apart were chosen. We then extracted the same SNPs from the outcome GWAS dataset. For removing the ambiguous SNPs, we used the “harmonise_data” function to coordinate exposure and outcome datasets. Each SNP was queried on the website PhenoScanner (http://​www.​phenoscanner.​medschl.​cam.​ac.​uk/​), in order to identify and remove potential confounding factors. F-statistic is a valuable tool to assess the strength of the IVs. We calculated the F-statistic for each SNP (β2/se2) and retained SNPs with an F-statistic greater than 10. This threshold was chosen to ensure that the correlation between the IVs and exposure is sufficiently strong to minimize instrumental bias in the MR analysis [17, 18].
We used the random-effect inverse-variance weighted (IVW) method as primary analysis. Multiplicative random-effect IVW model was used when heterogeneity existed, otherwise, a fixed-effect IVW was used [19]. To ensure the accuracy of the results, we used additional methods such ad MR Egger, simple mode, weighted median and weighted mode. As sensitivity analysis, we performed the Cochran Q-test and MR Egger intercept separately to detect any heterogeneity and directional pleiotropy in the IVW model. A significance level of p < 0.05 was considered indicative of possible heterogeneity or horizontal pleiotropy. MR Egger can evaluate the pleiotropy using the intercept term, when the intercept term equals zero, the result of MR Egger is consistent with IVW, proving that there is no horizontal pleiotropy [20]. Moreover, a leave-one-out method was used to determine if the combined IVW estimate was driven by any individual SNP.
A bilateral P-value < 0.05 was considered statistical significance. We conducted the analysis by the package TwoSampleMR using the R software (Version 4.2.1), and GraphPad Prism for Windows (Version 7.00) was used for visualization.

Results

Effect of delirium on Alzheimer’s disease

In the MR analysis, we chose a relaxed threshold (P < 5 × 10−6) to get more SNPs associated with delirium. Nine SNPs were obtained as IVs after excluding one palindromic SNP (rs963891), one AD-related SNP (rs429558) and two SNPs related to confounding factors (Type II diabetes: rs1353361; major depressive disorder male: rs1669794) [21]. All IVs had F statistics above 10 (rang 21–35), indicating that the strength of the instruments was robust (Additional file 1: Table S1).
Our MR analysis showed that delirium was not associated with AD (IVW: odds ratio [OR] 0.98, 95% CI 0.91–1.05, P = 0.544), other MR methods showed a consistent result with IVW (MR Egger: OR 0.98, 95% CI 0.83–1.15, P = 0.780; weighted median: OR 0.96, 95% CI 0.88–1.05, P = 0.323; simple mode: OR 0.91, 95% CI 0.80–1.04, P = 0.212; weighted mode: OR 0.93, 95% CI 0.83–1.05, P = 0.277; Figs. 2, 3). There was no evidence of heterogeneity (Q = 24.67, p = 0.244) and horizontal pleiotropy (p = 0.973) (Additional file 1: Table S2). Additionally, the result of leave-one-out sensitivity analysis, also indicated that the causal effect was not driven by a single SNP (Fig. 4).

Effect of Alzheimer’s disease on delirium

We identified SNPs associated with AD at genome-wide significance level (P < 5 × 10−8) that were independent from each other (r2 < 0.001 and kb > 10,000). After excluding palindromic SNPs (rs114812713) and delirium-related SNPs (rs12972156), we obtained 15 SNPs as IVs for our analysis. The F-statistics of all IVs were above 10 (range 30–204), indicating there was no weak instrument (Additional file 1: Table S3).
Figure 2 illustrates the results of the MR analysis. All four methods except MR Egger showed a statistical significance (IVW: OR 1.32, 95% CI 1.13–1.54, P = 3.91E−04). Weighted median, simple mode and weighted mode methods all show a consistent result with IVW (weighted median: OR 1.39, 95% CI 01.18–1.63, P = 8.22E−05; simple mode: OR 1.41, 95% CI 1.10–1.80, P = 1.41E−02; weighted mode: OR 1.39, 95% CI 1.16–1.67, P = 3.23E−03). Although MR Egger did not show an association in AD and delirium, it was close to statistical significance (OR 1.42, 95% CI 1.02–1.98, P = 5.70E-02) (Figs. 2, 3). Heterogeneity was observed in the Cochran’s Q test (Q = 24.67, p = 0.029). No horizontal pleiotropy was observed in the MR Egger intercept test (p = 0.630), indicating that the associations were not confounded by pleiotropic effects (Additional file 1: Table S2). MR leave-one-out sensitivity analysis suggested that no single SNP changed the causal effect (Fig. 4).

Discussion

We conducted a bidirectional two-sample MR approach to evaluate the causal association between delirium and AD. Results from IVW and other MR methods, showed that there was no bidirectional causality between delirium and AD. Specifically, we found that patients with AD may have a higher risk of delirium, but delirium was not significant in relation to AD. To the best of our knowledge, the present study firstly explored a causal relationship between delirium and AD using MR. Our findings provide valuable insights into the potential causal relationship between these two conditions.
Previous studies have found a correlation between delirium and AD. Delirium has been identified as a risk factor that increases the likelihood of developing or exacerbating dementia, and it can accelerate cognitive decline in AD patients [6, 22, 23]. However, it is important to note that in the present study, we only included patients with or without AD, and our research focused on assessing the causal relationship between delirium and AD. The process and development of AD go beyond the scope of this specific study. Although we did not find that delirium can increase the risk for developing AD, it is not contradictory to the conclusion that delirium can accelerate cognitive decline in AD patients. Inflammatory response is believed to be a primary driver of acute brain dysfunction or delirium, in addition, critical illness with acute inflammatory insult was also a risk factor for AD [24, 25]. A meta-analysis including 28 observational studies found that biomarkers associated with dementia, such as elevated blood-based levels of IL-6, C-reactive protein, S100B, and NfL, were also associated with delirium [26]. The fact that delirium and AD share similar biomarkers suggests that they may share similar pathologies in the development of disease. These biomarkers associated delirium may also cause harm to the process of AD and speed up cognitive decline instead of delirium itself.
There are common risk factors, such as low vitamin D concentrations, that may contribute to both delirium and AD. Two MR analysis studies have found an association between low vitamin D concentrations and the development of AD and delirium [27, 28]. Witlox J et al. concluded that patients with delirium have a higher incidence of developing AD in a meta-analysis. This meta-analysis included two prospective follow-up studies that were conducted over a period of 5 and 3 years [5, 29, 30]. However, causality cannot be established because of the small sample size and the limitation of observational studies. In this study, we used five MR analysis methods and found consistent results showing that delirium is not an independent risk factor for AD. The results were robust, as we did not observe any pleiotropy or heterogeneity in the sensitivity analysis. For all that, we cannot completely deny the possibility that delirium may influence the process of AD through similar pathological mechanisms, such as abnormal nerve transmission and neuroinflammation. Future studies including lager sample sizes should be done, to confirm whether delirium has an effect on the process of AD and to explore the true underlying factors that contribute to accelerating the process of AD.
It is well known that AD is the risk factors for delirium. In our study, the effect estimate of IVW (OR 1.32, 95% CI 1.13–1.54) demonstrates that AD is indeed a risk factor for delirium, which aligns with previous research demonstrating that dementia and recognition decline are independent risk factors for delirium [31, 32]. Although the exact mechanisms of delirium are not completely understood, it is believed to be a multifactorial process that can result in brain dysfunction, often involving abnormal nerve transmission and neuroinflammation [10, 33, 34]. Patients with dementia and pre-existing neurodegeneration may be more prone to neuroinflammatory cytokines, and trigger greater oxidative stress during surgery, pain or other stressful situations, potentially increasing the risk of delirium. Furthermore, patients with delirium superimposed on dementia have poorer outcomes, such as higher mortality rates, greater dependence on walking and increased healthcare expenses [3537]. Therefore, recognition and prevention of the occurrence of delirium at an early stage is crucial for improving patient outcomes.
Delirium is currently classified into major subtypes: hyperactive type (which is more apparent), hypoactive type (which is more prevalent but often overlooked) and a combined hyperactive and hypoactive types [38]. Research has found that staff and volunteers tend to focus more on treating hyperactive symptoms, while may miss the prevention and early recognition of hypoactive delirium [39]. Clinicians and nurses may overlook patients with hypoactive delirium because they appear to be performing normally, even though they actually unwell. Prevention is always better than treatment, and there are two types of risk factors for delirium: predisposing and precipitating factors. The predisposing factors are associated with the patients’ internal state, such as advanced age, functional deficiency, and comorbidities. Precipitating factors, on the other hand, refer to external disturbances, such a trauma, sedative drugs, sepsis, surgery and anesthesia. Patients with more predisposing factors more likely to develop delirium with fewer precipitating factors [2, 40]. Post-operative delirium is a common post-surgical complication, particularly in older adults [41]. Although it may be difficult to make changes in predisposing factors, it possible to mitigate the risk of precipitating factors. Clinicians can avoid the use of benzodiazepines or anticholinergic drugs, manage pain, encourage early post-operative mobilization and regular ambulation to reduce the risk of post-operative delirium. Implementation nonpharmacologic prevention strategies can also significantly improve outcomes for older patients [42]. Therefore, the clinicians and nurses should pay close attention to patients with AD, and the presence of predisposing risk factors for delirium. By taking early preventative measures, clinicians can improve the chances of early recognition and effective prevention of delirium.
Several limitations of our study should be noted. Firstly, our findings may not be applicable to populations with different ethnicities or geographic locations as our study only included individuals of European ancestry. Further data collection and analysis is needed to confirm the generalizability of our findings. Secondly, we only included the delirium phenotype as a binary variable, and did not consider the three subtypes or delirium or the severity of delirium. Therefore, we could not assess the association between AD and delirium subtypes or severity. It is worth noting that previous study has shown that patients with persistent delirium have worse outcomes than those with transient delirium [43]. Thirdly, the Cochran’s Q test revealed heterogeneity in the effect of AD on delirium. Various GWAS cohort studies might have differences in gene annotation analysis platforms, inclusion and exclusion criteria for cases, which could contribute to the heterogeneity in our study [44]. Additionally, the GWAS data for AD are obtained from several consortia, may contribute to the heterogeneity. However, this heterogeneity did not impact the main analysis, as the consistent results from other MR methods, such as MR Egger and weighted median model, are robust to genetic heterogeneity and can provide valid estimates [4547]. Moreover, the other three MR methods (simple mode, weighted median and weighted mode) showed similar result (p < 0.05), and the p value was also close to statistical significance using MR Egger method (P = 5.70E−02). In addition, no directional pleiotropy was observed in the MR Egger intercept analysis. The leave-one-out sensitivity analysis was also performed, and it showed that even when individual SNPs were removed one by one, the overall results remained stable (the overall error line did not fluctuate significantly). These findings strongly suggest that the results of our MR analysis are robust.

Conclusions

Our bidirectional two-sample MR analysis showed that although AD increase the risk for delirium, while delirium is not associated with AD. These findings may suggest that clinicians should identify and prevent the occurrence of delirium superimposed on dementia in an early stage. Future study should investigate the mechanisms underlying the effect of AD on delirium, and researchers could include larger sample sizes to confirm the effect of delirium on AD.

Acknowledgements

We want to acknowledge the participants and investigators of the FinnGen study and IGAP (International Genomics of Alzheimer’s Project) consortium.

Declarations

All these GWAS summary data are publicly available, and all studies included were approved by relevant ethics committee.
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 Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK. One-year health care costs associated with delirium in the elderly population. Arch Intern Med. 2008;168(1):27–32.PubMedPubMedCentral Leslie DL, Marcantonio ER, Zhang Y, Leo-Summers L, Inouye SK. One-year health care costs associated with delirium in the elderly population. Arch Intern Med. 2008;168(1):27–32.PubMedPubMedCentral
3.
Zurück zum Zitat Omuojine JP, Bello T, Wemakor S, Mante PK, Amponsah GS, Kusi-Mensah K, et al. Risk factors and outcomes of delirium in hospitalized older Ghanaians. Int J Geriatr Psychiatry. 2023;38(4): e5912.PubMed Omuojine JP, Bello T, Wemakor S, Mante PK, Amponsah GS, Kusi-Mensah K, et al. Risk factors and outcomes of delirium in hospitalized older Ghanaians. Int J Geriatr Psychiatry. 2023;38(4): e5912.PubMed
4.
Zurück zum Zitat Kinchin I, Mitchell E, Agar M, Trépel D. The economic cost of delirium: a systematic review and quality assessment. Alzheimers Dement. 2021;17(6):1026–41.PubMed Kinchin I, Mitchell E, Agar M, Trépel D. The economic cost of delirium: a systematic review and quality assessment. Alzheimers Dement. 2021;17(6):1026–41.PubMed
5.
Zurück zum Zitat Witlox J, Eurelings LS, de Jonghe JF, Kalisvaart KJ, Eikelenboom P, van Gool WA. Delirium in elderly patients and the risk of postdischarge mortality, institutionalization, and dementia: a meta-analysis. JAMA. 2010;304(4):443–51.PubMed Witlox J, Eurelings LS, de Jonghe JF, Kalisvaart KJ, Eikelenboom P, van Gool WA. Delirium in elderly patients and the risk of postdischarge mortality, institutionalization, and dementia: a meta-analysis. JAMA. 2010;304(4):443–51.PubMed
6.
Zurück zum Zitat Davis DH, Muniz Terrera G, Keage H, Rahkonen T, Oinas M, Matthews FE, et al. Delirium is a strong risk factor for dementia in the oldest-old: a population-based cohort study. Brain. 2012;135(Pt 9):2809–16.PubMedPubMedCentral Davis DH, Muniz Terrera G, Keage H, Rahkonen T, Oinas M, Matthews FE, et al. Delirium is a strong risk factor for dementia in the oldest-old: a population-based cohort study. Brain. 2012;135(Pt 9):2809–16.PubMedPubMedCentral
7.
Zurück zum Zitat Weiner MF. Impact of delirium on the course of Alzheimer disease. Arch Neurol. 2012;69(12):1639–40.PubMed Weiner MF. Impact of delirium on the course of Alzheimer disease. Arch Neurol. 2012;69(12):1639–40.PubMed
8.
Zurück zum Zitat Davis DH, Muniz-Terrera G, Keage HA, Stephan BC, Fleming J, Ince PG, et al. Association of delirium with cognitive decline in late life: a neuropathologic study of 3 population-based cohort studies. JAMA Psychiat. 2017;74(3):244–51. Davis DH, Muniz-Terrera G, Keage HA, Stephan BC, Fleming J, Ince PG, et al. Association of delirium with cognitive decline in late life: a neuropathologic study of 3 population-based cohort studies. JAMA Psychiat. 2017;74(3):244–51.
9.
Zurück zum Zitat Sampson EL, Blanchard MR, Jones L, Tookman A, King M. Dementia in the acute hospital: prospective cohort study of prevalence and mortality. Br J Psychiatry. 2009;195(1):61–6.PubMed Sampson EL, Blanchard MR, Jones L, Tookman A, King M. Dementia in the acute hospital: prospective cohort study of prevalence and mortality. Br J Psychiatry. 2009;195(1):61–6.PubMed
10.
Zurück zum Zitat Inouye SK, Westendorp RG, Saczynski JS. Delirium in elderly people. Lancet. 2014;383(9920):911–22.PubMed Inouye SK, Westendorp RG, Saczynski JS. Delirium in elderly people. Lancet. 2014;383(9920):911–22.PubMed
11.
Zurück zum Zitat Khachaturian AS, Hayden KM, Devlin JW, Fleisher LA, Lock SL, Cunningham C, et al. International drive to illuminate delirium: a developing public health blueprint for action. Alzheimers Dement. 2020;16(5):711–25.PubMed Khachaturian AS, Hayden KM, Devlin JW, Fleisher LA, Lock SL, Cunningham C, et al. International drive to illuminate delirium: a developing public health blueprint for action. Alzheimers Dement. 2020;16(5):711–25.PubMed
12.
Zurück zum Zitat 2021 Alzheimer's disease facts and figures. Alzheimers Dement. 2021;17(3):327-406. 2021 Alzheimer's disease facts and figures. Alzheimers Dement. 2021;17(3):327-406.
13.
Zurück zum Zitat Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet Epidemiol. 2013;37(7):658–65.PubMedPubMedCentral Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet Epidemiol. 2013;37(7):658–65.PubMedPubMedCentral
14.
Zurück zum Zitat Holmes MV, Ala-Korpela M, Smith GD. Mendelian randomization in cardiometabolic disease: challenges in evaluating causality. Nat Rev Cardiol. 2017;14(10):577–90.PubMedPubMedCentral Holmes MV, Ala-Korpela M, Smith GD. Mendelian randomization in cardiometabolic disease: challenges in evaluating causality. Nat Rev Cardiol. 2017;14(10):577–90.PubMedPubMedCentral
15.
Zurück zum Zitat Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, Naj AC, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet. 2019;51(3):414–30.PubMedPubMedCentral Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, Naj AC, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet. 2019;51(3):414–30.PubMedPubMedCentral
16.
Zurück zum Zitat Kwok MK, Schooling CM. Herpes simplex virus and Alzheimer’s disease: a Mendelian randomization study. Neurobiol Aging. 2021;99:101e11-e13. Kwok MK, Schooling CM. Herpes simplex virus and Alzheimer’s disease: a Mendelian randomization study. Neurobiol Aging. 2021;99:101e11-e13.
17.
Zurück zum Zitat Burgess S, Thompson SG. Avoiding bias from weak instruments in Mendelian randomization studies. Int J Epidemiol. 2011;40(3):755–64.PubMed Burgess S, Thompson SG. Avoiding bias from weak instruments in Mendelian randomization studies. Int J Epidemiol. 2011;40(3):755–64.PubMed
18.
Zurück zum Zitat Feng R, Lu M, Xu J, Zhang F, Yang M, Luo P, et al. Pulmonary embolism and 529 human blood metabolites: genetic correlation and two-sample Mendelian randomization study. BMC Genom Data. 2022;23(1):69.PubMedPubMedCentral Feng R, Lu M, Xu J, Zhang F, Yang M, Luo P, et al. Pulmonary embolism and 529 human blood metabolites: genetic correlation and two-sample Mendelian randomization study. BMC Genom Data. 2022;23(1):69.PubMedPubMedCentral
19.
Zurück zum Zitat Papadimitriou N, Dimou N, Tsilidis KK, Banbury B, Martin RM, Lewis SJ, et al. Physical activity and risks of breast and colorectal cancer: a Mendelian randomisation analysis. Nat Commun. 2020;11(1):597.PubMedPubMedCentral Papadimitriou N, Dimou N, Tsilidis KK, Banbury B, Martin RM, Lewis SJ, et al. Physical activity and risks of breast and colorectal cancer: a Mendelian randomisation analysis. Nat Commun. 2020;11(1):597.PubMedPubMedCentral
20.
Zurück zum Zitat Burgess S, Thompson SG. Interpreting findings from Mendelian randomization using the MR-Egger method. Eur J Epidemiol. 2017;32(5):377–89.PubMedPubMedCentral Burgess S, Thompson SG. Interpreting findings from Mendelian randomization using the MR-Egger method. Eur J Epidemiol. 2017;32(5):377–89.PubMedPubMedCentral
21.
Zurück zum Zitat Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396(10248):413–46.PubMedPubMedCentral Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396(10248):413–46.PubMedPubMedCentral
22.
Zurück zum Zitat Fong TG, Jones RN, Shi P, Marcantonio ER, Yap L, Rudolph JL, et al. Delirium accelerates cognitive decline in Alzheimer disease. Neurology. 2009;72(18):1570–5.PubMedPubMedCentral Fong TG, Jones RN, Shi P, Marcantonio ER, Yap L, Rudolph JL, et al. Delirium accelerates cognitive decline in Alzheimer disease. Neurology. 2009;72(18):1570–5.PubMedPubMedCentral
23.
Zurück zum Zitat Kunicki ZJ, Ngo LH, Marcantonio ER, Tommet D, Feng Y, Fong TG, et al. Six-year cognitive trajectory in older adults following major surgery and delirium. JAMA Intern Med. 2023;183(5):442–50.PubMed Kunicki ZJ, Ngo LH, Marcantonio ER, Tommet D, Feng Y, Fong TG, et al. Six-year cognitive trajectory in older adults following major surgery and delirium. JAMA Intern Med. 2023;183(5):442–50.PubMed
24.
Zurück zum Zitat Cunningham C. Systemic inflammation and delirium: important co-factors in the progression of dementia. Biochem Soc Trans. 2011;39(4):945–53.PubMedPubMedCentral Cunningham C. Systemic inflammation and delirium: important co-factors in the progression of dementia. Biochem Soc Trans. 2011;39(4):945–53.PubMedPubMedCentral
25.
Zurück zum Zitat Walker KA, Le Page LM, Terrando N, Duggan MR, Heneka MT, Bettcher BM. The role of peripheral inflammatory insults in Alzheimer’s disease: a review and research roadmap. Mol Neurodegener. 2023;18(1):37.PubMedPubMedCentral Walker KA, Le Page LM, Terrando N, Duggan MR, Heneka MT, Bettcher BM. The role of peripheral inflammatory insults in Alzheimer’s disease: a review and research roadmap. Mol Neurodegener. 2023;18(1):37.PubMedPubMedCentral
26.
Zurück zum Zitat Wang S, Greene R, Song Y, Chan C, Lindroth H, Khan S, et al. Postoperative delirium and its relationship with biomarkers for dementia: a meta-analysis. Int Psychogeriatr. 2022;34(4):377–90. Wang S, Greene R, Song Y, Chan C, Lindroth H, Khan S, et al. Postoperative delirium and its relationship with biomarkers for dementia: a meta-analysis. Int Psychogeriatr. 2022;34(4):377–90.
27.
Zurück zum Zitat Bowman K, Jones L, Pilling LC, Delgado J, Kuchel GA, Ferrucci L, et al. Vitamin D levels and risk of delirium: a mendelian randomization study in the UK Biobank. Neurology. 2019;92(12):e1387–94.PubMedPubMedCentral Bowman K, Jones L, Pilling LC, Delgado J, Kuchel GA, Ferrucci L, et al. Vitamin D levels and risk of delirium: a mendelian randomization study in the UK Biobank. Neurology. 2019;92(12):e1387–94.PubMedPubMedCentral
28.
Zurück zum Zitat Meng L, Wang Z, Ming YC, Shen L, Ji HF. Are micronutrient levels and supplements causally associated with the risk of Alzheimer’s disease? A two-sample Mendelian randomization analysis. Food Funct. 2022;13(12):6665–73.PubMed Meng L, Wang Z, Ming YC, Shen L, Ji HF. Are micronutrient levels and supplements causally associated with the risk of Alzheimer’s disease? A two-sample Mendelian randomization analysis. Food Funct. 2022;13(12):6665–73.PubMed
29.
Zurück zum Zitat Lundström M, Edlund A, Bucht G, Karlsson S, Gustafson Y. Dementia after delirium in patients with femoral neck fractures. J Am Geriatr Soc. 2003;51(7):1002–6.PubMed Lundström M, Edlund A, Bucht G, Karlsson S, Gustafson Y. Dementia after delirium in patients with femoral neck fractures. J Am Geriatr Soc. 2003;51(7):1002–6.PubMed
30.
Zurück zum Zitat Bickel H, Gradinger R, Kochs E, Förstl H. High risk of cognitive and functional decline after postoperative delirium. A three-year prospective study. Dement Geriatr Cogn Disord. 2008;26(1):26–31.PubMed Bickel H, Gradinger R, Kochs E, Förstl H. High risk of cognitive and functional decline after postoperative delirium. A three-year prospective study. Dement Geriatr Cogn Disord. 2008;26(1):26–31.PubMed
31.
Zurück zum Zitat Koster S, Hensens AG, Schuurmans MJ, van der Palen J. Risk factors of delirium after cardiac surgery: a systematic review. Eur J Cardiovasc Nurs. 2011;10(4):197–204.PubMed Koster S, Hensens AG, Schuurmans MJ, van der Palen J. Risk factors of delirium after cardiac surgery: a systematic review. Eur J Cardiovasc Nurs. 2011;10(4):197–204.PubMed
32.
Zurück zum Zitat Zaal IJ, Devlin JW, Peelen LM, Slooter AJ. A systematic review of risk factors for delirium in the ICU. Crit Care Med. 2015;43(1):40–7.PubMed Zaal IJ, Devlin JW, Peelen LM, Slooter AJ. A systematic review of risk factors for delirium in the ICU. Crit Care Med. 2015;43(1):40–7.PubMed
33.
Zurück zum Zitat Jahangir S, Allala M, Khan AS, Muyolema Arce VE, Patel A, Soni K, et al. A review of biomarkers in Delirium Superimposed on Dementia (DSD) and their clinical application to personalized treatment and management. Cureus. 2023;15(5):e38627.PubMedPubMedCentral Jahangir S, Allala M, Khan AS, Muyolema Arce VE, Patel A, Soni K, et al. A review of biomarkers in Delirium Superimposed on Dementia (DSD) and their clinical application to personalized treatment and management. Cureus. 2023;15(5):e38627.PubMedPubMedCentral
34.
Zurück zum Zitat Oldham MA, Holloway RG. Delirium disorder: Integrating delirium and acute encephalopathy. Neurology. 2020;95(4):173–8.PubMed Oldham MA, Holloway RG. Delirium disorder: Integrating delirium and acute encephalopathy. Neurology. 2020;95(4):173–8.PubMed
35.
Zurück zum Zitat Vo QT, Koethe B, Holmes S, Simoni-Wastila L, Briesacher BA. Patient outcomes after delirium screening and incident Alzheimer’s disease or related dementias in skilled nursing facilities. J Gen Intern Med. 2023;38(2):414–20.PubMed Vo QT, Koethe B, Holmes S, Simoni-Wastila L, Briesacher BA. Patient outcomes after delirium screening and incident Alzheimer’s disease or related dementias in skilled nursing facilities. J Gen Intern Med. 2023;38(2):414–20.PubMed
36.
Zurück zum Zitat Morandi A, Davis D, Fick DM, Turco R, Boustani M, Lucchi E, et al. Delirium superimposed on dementia strongly predicts worse outcomes in older rehabilitation inpatients. J Am Med Dir Assoc. 2014;15(5):349–54.PubMedPubMedCentral Morandi A, Davis D, Fick DM, Turco R, Boustani M, Lucchi E, et al. Delirium superimposed on dementia strongly predicts worse outcomes in older rehabilitation inpatients. J Am Med Dir Assoc. 2014;15(5):349–54.PubMedPubMedCentral
37.
Zurück zum Zitat Fick DM, Agostini JV, Inouye SK. Delirium superimposed on dementia: a systematic review. J Am Geriatr Soc. 2002;50(10):1723–32.PubMed Fick DM, Agostini JV, Inouye SK. Delirium superimposed on dementia: a systematic review. J Am Geriatr Soc. 2002;50(10):1723–32.PubMed
38.
Zurück zum Zitat Breitbart W, Rosenfeld B, Roth A, Smith MJ, Cohen K, Passik S. The memorial delirium assessment scale. J Pain Symptom Manage. 1997;13(3):128–37.PubMed Breitbart W, Rosenfeld B, Roth A, Smith MJ, Cohen K, Passik S. The memorial delirium assessment scale. J Pain Symptom Manage. 1997;13(3):128–37.PubMed
39.
Zurück zum Zitat Featherstone I, Siddiqi N, Jones L, Coppo E, Sheldon T, Hosie A, et al. It’s tough It is hard’: A qualitative interview study of staff and volunteers caring for hospice in-patients with delirium. Palliat Med. 2023;37(7):993–1005.PubMedPubMedCentral Featherstone I, Siddiqi N, Jones L, Coppo E, Sheldon T, Hosie A, et al. It’s tough It is hard’: A qualitative interview study of staff and volunteers caring for hospice in-patients with delirium. Palliat Med.  2023;37(7):993–1005.PubMedPubMedCentral
40.
Zurück zum Zitat Inouye SK, Charpentier PA. Precipitating factors for delirium in hospitalized elderly persons. Predictive model and interrelationship with baseline vulnerability. JAMA. 1996;275(11):852–7.PubMed Inouye SK, Charpentier PA. Precipitating factors for delirium in hospitalized elderly persons. Predictive model and interrelationship with baseline vulnerability. JAMA. 1996;275(11):852–7.PubMed
41.
Zurück zum Zitat Cascella M, Muzio MR, Bimonte S, Cuomo A, Jakobsson JG. Postoperative delirium and postoperative cognitive dysfunction: updates in pathophysiology, potential translational approaches to clinical practice and further research perspectives. Minerva Anestesiol. 2018;84(2):246–60.PubMed Cascella M, Muzio MR, Bimonte S, Cuomo A, Jakobsson JG. Postoperative delirium and postoperative cognitive dysfunction: updates in pathophysiology, potential translational approaches to clinical practice and further research perspectives. Minerva Anestesiol. 2018;84(2):246–60.PubMed
42.
Zurück zum Zitat Oh ES, Fong TG, Hshieh TT, Inouye SK. Delirium in older persons: advances in diagnosis and treatment. JAMA. 2017;318(12):1161–74.PubMedPubMedCentral Oh ES, Fong TG, Hshieh TT, Inouye SK. Delirium in older persons: advances in diagnosis and treatment. JAMA. 2017;318(12):1161–74.PubMedPubMedCentral
43.
Zurück zum Zitat McCusker J, Cole M, Dendukuri N, Han L, Belzile E. The course of delirium in older medical inpatients: a prospective study. J Gen Intern Med. 2003;18(9):696–704.PubMedPubMedCentral McCusker J, Cole M, Dendukuri N, Han L, Belzile E. The course of delirium in older medical inpatients: a prospective study. J Gen Intern Med. 2003;18(9):696–704.PubMedPubMedCentral
44.
Zurück zum Zitat Guo Z, Du H, Guo Y, Jin Q, Liu R, Yun Z, et al. Association between leptin and NAFLD: a two-sample Mendelian randomization study. Eur J Med Res. 2023;28(1):215.PubMedPubMedCentral Guo Z, Du H, Guo Y, Jin Q, Liu R, Yun Z, et al. Association between leptin and NAFLD: a two-sample Mendelian randomization study. Eur J Med Res. 2023;28(1):215.PubMedPubMedCentral
45.
Zurück zum Zitat Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–25.PubMedPubMedCentral Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–25.PubMedPubMedCentral
46.
Zurück zum Zitat Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40(4):304–14.PubMedPubMedCentral Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40(4):304–14.PubMedPubMedCentral
47.
Zurück zum Zitat Adewuyi EO, O’Brien EK, Porter T, Laws SM. Relationship of cognition and Alzheimer’s Disease with gastrointestinal tract disorders: a large-scale genetic overlap and mendelian randomisation analysis. Int J Mol Sci. 2022;23(24):16199.PubMedPubMedCentral Adewuyi EO, O’Brien EK, Porter T, Laws SM. Relationship of cognition and Alzheimer’s Disease with gastrointestinal tract disorders: a large-scale genetic overlap and mendelian randomisation analysis. Int J Mol Sci. 2022;23(24):16199.PubMedPubMedCentral
Metadaten
Titel
Causal relationships between delirium and Alzheimer's disease: a bidirectional two-sample Mendelian randomization study
verfasst von
Jiang Zheng
Xiaohui Du
Liu Yang
Hong Fu
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01245-w

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe