Abstract
Objective:
To identify medications which may prevent psychosis in Alzheimer’s disease (AD) patients.
Methods:
We compared the frequency of medication usage among AD patients with or without psychosis symptoms (AD+P vs. AD-P). We also conducted survival analysis on time to psychosis for AD patients to identify drugs with beneficial effects. We further explored the potential molecular mechanisms of identified drugs by gene signature analysis. Specifically, the gene expression profiles induced by the identified drug(s) were collected to derive a list of most perturbed genes. These genes were further analyzed by the associations of their genetic variations with AD or psychosis-related phenotypes.
Results:
Vitamin D was used more often in AD-P patients than in AD+P patients. Vitamin D was also significantly associated with delayed time to psychosis. AD and/or psychosis-related genes were enriched in the list of genes most perturbed by Vitamin D, specifically genes involved in the regulation of calcium signaling downstream of the Vitamin D receptor.
Conclusion:
Vitamin D was associated with delayed onset of psychotic symptoms in AD patients. Its mechanisms of action provide a novel direction for development of drugs to prevent or treat psychosis in AD. In addition, genetic variations in Vitamin D-regulated genes may provide a biomarker signature to identify a subpopulation of patients who can benefit from Vitamin D treatment.
Keywords: Alzheimer’s Disease, Cognitive Decline, Vitamin D, Psychosis Symptoms, Systems Pharmacology, Clinical Data-Mining
1. Introduction
AD is the most common form of dementia (estimated to be 50−60% of all cases) and the sixth most common cause of death in the U.S. with the absolute numbers growing (1) though its prevalence is decreasing (2). Today, only five drugs approved by the U.S. Food and Drug Administration (FDA) are used to treat the cognitive dysfunction of AD, e.g. tacrine, donepezil, rivastigmine, galantamine, and memantine. These drugs are modestly efficacious for cognitive abilities in early to moderate stages of AD (3). However, none of them can stop or reverse the progression of this disease (3). As such, there remains a great demand for design and discovery of new medications or repurposing drugs that alter the course of AD. Our question is, can we find clues for AD treatment from the analysis of medication history of AD patients? The rationale for this approach arose from the fact that AD can co-occur with other diseases, and as such, multidrug therapy is common among AD patients. The substantial overlap between diseases in their risk factors points to the possibility of treating two relevant diseases with the same drugs.
In this study, we compared the medication history of two groups of AD populations, AD patients with or without psychotic symptoms (AD+P / AD-P), where psychotic symptoms include hallucinations and delusions. AD+P patients represent a subgroup with poor outcomes: more rapid cognitive and functional decline (4–7); higher rates of aggression (8); worse overall health (9); and shorter time to placement in a nursing home (7, 10), when compared to AD-P patients (11, 12). Our hypothesis is that a drug may have beneficial effects on AD patients if it is used more often in AD-P than in the AD+P population, because AD-P is a relatively positive outcome.
To explore the potential molecular mechanisms, other than the direct target(s) of an identified AD-beneficial drug, gene signature analysis can provide useful clues. Both disease and medications can alter gene expression patterns. It has been increasingly appreciated that such perturbations result in a specific transcriptome ‘signature’: i.e. a set of genes with a defined pattern of expression (13). Computational efforts have increasingly assembled and compared these signatures genome-wide, revealing unexpected drug–disease ‘connections’ when signatures correspond. If the transcriptome signature of a drug is negatively correlated with the disease signature, the drug might be able to reverse the disease transcriptome alterations, and hence, the disease phenotype itself. This approach has increasingly led to successful discoveries(13–16). For example, ursolic acid was identified as an inhibitor of fasting-induced muscle atrophy by gene-signature comparison(17). In the randomized, double-blinded, and placebo-controlled trial, right-handgrip strength of female subjects in the ursolic acid group was found to be significantly better than that of subjects in the control group, though no differences were found in the other outcomes(18). The application of this approach on repurposing drugs for AD treatment has also been reported (19). Thus, the objective of this study was to identify medication use associated with altered risk for psychosis in AD, and to perform gene signature analysis in an effort to identify potential pathways for modifying AD+P risk.
2. Methods
2.1. Subjects
Subject ascertainment and characterization were previously described in detail (11). In brief, all subjects were participants in the University of Pittsburgh ADRC, seen between May 2000 and August 2014. Subjects were included if they had an initial primary diagnosis of Mild Cognitive Impairment (MCI) (20, 21) or Probable or Possible AD (22) after a clinical diagnostic evaluation consisting of a baseline neurological, neuropsychological and psychiatric evaluation, laboratory studies, and brain imaging with annual re-evaluation of neurological presentation, behavioral symptoms, cognitive tests, and functioning (23–26). Initial and annual visits included the Mini-mental State Examination (MMSE)(27). Telephone assessments were conducted at approximately six-month intervals between annual in-person visits, and they were also conducted at the time of annual assessment for individuals unable to return to clinic. All subjects were required to have an age of onset of cognitive problems starting ≥60 years old and no current or prior psychosis, including no prior personal history of a primary psychotic disorder (e.g. schizophrenia).
Psychosis was assessed at visits and by telephone using the Consortium to Establish a Registry for Alzheimer Disease Behavioral Rating Scale (BRS) which was administered to an informant knowledgeable about the patients’ symptoms. The informant’s relationship to the participant (available for 97.8%) and frequency of contact (available for 90.9%) was recorded. Informants were predominantly a spouse (55%) or an adult child (38%). 83% had contact (5 or more days/week with the participant; 63% lived with the participant.
Patients were classified as psychotic if they had one or more of BRS items #33–45 rated as present at least three to eight days in the past month: delusional misidentification of people, self or objects; paranoia, beliefs of abandonment or infidelity; believing someone is an imposter, belief that characters on television are real; belief that there are people in or around house that aren’t there, belief that a dead person is still alive, belief that their house is not their home, auditory hallucinations, visual hallucinations. Symptoms were not rated if they occurred during an episode of delirium, were medication induced, or if the symptoms were hypnopompic or hypnogogic. AD-P was defined as the absence of all psychosis symptoms at all assessments.
All procedures were conducted under the research protocol approved by the University of Pittsburgh Institutional Review Board, and informed consent was obtained from subjects and/or their proxy.
2.2. Medication information
Information on all medications used by participants was collected at initial and annual visits, and during telephone evaluations. For AD+P patients, we only considered their medication usage from the date of the study entry to the first time of psychosis onset. For AD-P patients we considered all the available medication usage on and after the date of study entry. We selected the top 100 most used drugs for this analysis.
2.3. Statistical analysis
Chi-square test was performed to assess the association between psychosis and ever or never taking of each of the 100 candidate drugs during the follow-up. Log rank test was performed to test the difference in time to psychosis from time to entry of the study between patients who have ever or never taken each of the 100 candidate drugs during the follow-up. To adjust for multiple testing, Benjamini & Hochberg method (28) was used to control for the false discovery rate (FDR). For those drugs statistically significantly associated with time to psychosis with FDR ≤ 0.1, we also performed a multivariable Cox regression with baseline MMSE, baseline age, sex, race, and education, as covariates. We used the cox.zph function of R package survival to test the proportional hazard assumption (29).
The study data was maintained and managed using SPSS for Windows (v12–v15); the analyses were carried out using SAS (SAS Institute, Cary, North Carolina). p<0.05 was used as the threshold for statistical significance for all analyses except where stated otherwise.
2.4. Analysis of gene signatures induced by drugs
To understand the molecular mechanism behind the beneficial effects on AD, we analyzed the gene expression profiles induced by those drugs identified in the above analyses as significantly associated with occurrence and time to occurrence of psychosis. Differentially expressed genes (DEGs) induced by drugs were collected from commercial software Illumina BaseSpace (formerly Nextbio™) software (Santa Clara, CA, USA, http://www.nextbio.com). In BaseSpace, most of the raw gene expression datasets were from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/) and the Connectivity Map database (CMAP) (13). Only genes with p values <0.05 and absolute fold changes >1.2 were considered as DEGs. All the DEGs induced by a drug can be considered as a gene signature for this drug. The drug-induced gene expression datasets were selected by searching with drug names and were refined by only keeping datasets of one single drug treatment vs. control. All the qualified gene signatures for an individual drug were analyzed by the meta-analysis engine implemented in the BaseSpace database to generate a list of persistent genes from multiple instances of similar perturbations. All BaseSpace analyses were performed utilizing the default parameters.
2.5. Effect of drugs identified above on cognitive decline
To test for the effects of drugs identified above on the rate of decline in cognitive function, measured by MMSE, we applied a mixed-effect model with random intercept and random slope. We assessed the interaction term between treatment group and time controlling for the baseline MMSE, baseline age, race, sex, education and use of drugs identified above.
3. Results
3.1. Characteristics of subjects
The characteristics of the subjects are shown on Table 1. The subjects who developed psychosis symptoms had lower baseline MMSE, generally took fewer numbers of drugs and had lower education levels.
Table 1.
Variable | AD-P, N=367 | AD+P, N=256 | df | p value* |
---|---|---|---|---|
Follow-up, years | 2.5 (3.6) | 3.3 (3.1) | 595 | 0.003 |
Baseline MMSE | 21.8 (4.5) | 20.5 (5.3) | 490 | <0.001 |
Age at baseline | 76.9 (6.9) | 76.6 (6.4) | 576 | 0.54 |
Sex | ||||
female | 214 (58 %) | 164 (64 %) | 1 | 0.15 |
male | 153 (42 %) | 92 (36 %) | ||
Race | ||||
Asian | 2(1%) | 0(0%) | 2 | 0.48 |
African-American | 23 (6 %) | 20 (8 %) | ||
Caucasian | 342 (93 %) | 236 (92 %) | ||
Number of drugs taken | 9.7 (4.5) | 8.6 (4) | 587 | 0.002 |
Education, years | ||||
<12 | 162 (44 %) | 152 (59 %) | 1 | <0.001 |
>=12 | 205 (56 %) | 104 (41 %) |
AD-P: Alzheimer Disease without psychosis; AD+P: Alzheimer disease with psychosis; MMSE:
Mini-mental State Examination.
Results for AD-P and AD+P are mean (SD) for continuous variables and total (%) for categorical variables.
T-test for continuous variables (Follow-up, years; Baseline MMSE, Age at baseline), Chi-square test for sex and education, and Fisher exact test for race.
3.2. Vitamin D is identified as a drug with more frequent use in AD-P patients and which is associated with delayed onset of psychosis symptoms.
Of the 100 drugs analyzed (Table S1), only Vitamin D (Ergocalciferol) showed a significantly higher frequency of use among AD-P subjects compared to AD+P cases with q-value <0.001 (Table 2). Detailed Chi-square test results on the 100 drugs can be found in Table S2. We then analyzed the association between all 100 drugs and time to psychosis, identifying 6 drugs, including Vitamin D, with significant associations (Table S3; multivitamin, Vitamin D (ergocalciferol), memantine, warfarin, ferrous sulfate, and omeprazole). These six drugs were significantly associated with a delay to psychosis onset, as determined by Cox regression (Table 3).
Table 2.
Vitamin D (Ergocalciferol) | AD-P, N (%) | AD+P, N (%) | Chi-square | df | FDR Adjusted p value* |
---|---|---|---|---|---|
No | 237 (64.58%) | 215 (83.98%) | 28.5 | 1 | <0.001 |
Yes | 130 (35.42%) | 41 (16.02%) |
AD-P: Alzheimer Disease without psychosis; AD+P: Alzheimer disease with psychosis *Chi-square test adjusted by Benjamini & Hochberg method
Table 3.
Drug | HR (95% CI) | p value* |
---|---|---|
multivitamin | 0.6 (0.47,0.78) | 0.0001 |
memantine | 0.66 (0.51,0.86) | 0.002 |
Vitamin D(ergocalciferol) | 0.68 (0.48,0.96) | 0.03 |
omeprazole | 0.77 (0.49,1.19) | 0.24 |
warfarin | 0.53 (0.3,0.91) | 0.02 |
ferrous sulfate | 0.24 (0.08,0.77) | 0.02 |
Multivariable Cox regression controlled for baseline MMSE, baseline age, sex, race, and education. p values were obtained by Wald chi-square test and df is 1 for each of the variables.
3.3. Genes induced by Vitamin D are enriched in association with AD and psychosis diseases
We focused our studies of gene expression on Vitamin D, as it was the only common drug in the above analyses of association with presence of psychosis and time to psychosis occurrence. We identified 47 biosets of gene expression profiles from GEO and CMAP 2.0 database as shown in Table S4. These biosets were from single exposures of different cell types to Vitamin D, with different exposure durations. They were subjected to meta-analysis to derive a list of most perturbed genes (defined as having significant changes in 20 or more of the 47 biosets). As shown in Table 4, fifteen genes were found as the ones most perturbed by Vitamin D. These 15 genes are direct transcriptional targets of Vitamin D receptor (VDR) (30–38) except ACVRL1 and FOSL2. However, FOSL2 was reported to bind with the VDR promotor to regulate VDR gene expression (39). We then searched the GWAS CatLog (https://www.ebi.ac.uk/gwas/) to find associations of variations in these genes with AD- or psychosis-related phenotypes. Multiple variants in EFTUD1 have been associated with cognitive ability (40), intelligence (41), and educational attainment (42). Variation in EFTUD1 has also been associated with left superior temporal gyrus thickness in schizophrenia patients (43). SNPs in CLMN are associated with the cognitive decline in AD patients (44) and response to treatment of the antipsychotic drug perphenazine (45), an antagonist of dopamine D1 and D2 receptors. An SNP in SERPINB1 is associated with cerebrospinal fluid Aβ1–42 levels (46). Variations in HBEGF are associated with AD risk (47) and intelligence (41).
Table 4.
Gene ID | Gene Name | Specificity | Evidence of association with AD- or schizophrenia - related phenotypes | |||
---|---|---|---|---|---|---|
Phenotype | SNP | Reported p value* | Ref. | |||
CYP24A1 | cytochrome P450, family 24, subfamily A, polypeptide 1 | 33 (32 up/1 down)/47 | ||||
EFTUD1 | elongation factor Tu GTP binding domain containing 1 | 28(27 up/1 down)/47 | cognitive ability | rs867371 | 1 × 10−10 | (40) |
rs2134046 | 2 × 10−8 | (40) | ||||
Intelligence | rs28420834 | 1 × 10−10 | (41) | |||
rs1972460 | 2 × 10−10 | (41) | ||||
rs2665103 | 4 × 10−10 | (41) | ||||
rs12439619 | 1 × 10−8 | (41) | ||||
educational attainment | rs28420834 | 3 × 10−10 | (42) | |||
left superior temporal gyrus thickness in schizophrenia patients | rs1466921 | 9 × 10−7 | (43) | |||
CD14 | CD14 molecule | 27(26 up/1 down)/47 | ||||
CLMN | calmin (calponin-like, transmembrane) | 27(26 up/1 down)/47 | Cognitive decline in AD patients | rs115102486 | 2 × 10−8 | (44) |
response to perphenazine treatment | rs1187614 | 2 × 10−7 | (45) | |||
MAPK13 | mitogen-activated protein kinase 13 | 24(23 up/1 down)/47 | ||||
SERPINB1 | serpin peptidase inhibitor, clade B (ovalbumin), member 1 | 24(23 up/ 1 down)/47 | cerebrospinal fluid Aβ1–42 levels | rs316341 | 2 × 10−8 | (46) |
CEBPB | CCAAT/enhancer binding protein (C/EBP), beta | 23(22 up /1 down)/47 | ||||
THBD | Thrombomodulin | 22 (21 up /1 down)/47 | ||||
G0S2 | G0/G1switch 2 | 22(21 up /1 down)/47 | ||||
GEM | GTP binding protein overexpressed in skeletal muscle | 22(21 up / 1 down)/47 | ||||
CD97 | CD97 molecule | 22(22 up)/47 | ||||
ACVRL1 | activin A receptor type II-like 1 | 21(17 up/ 4 down)/47 | ||||
GRK5 | G protein-coupled receptor kinase 5 | 21(19 up /2 down)/47 | ||||
HBEGF | heparin-binding EGF-like growth factor | 20(18 up /2 down)/47 | AD risk | rs11168036 | 7 × 10−9 | (47) |
Intelligence | rs2282802 | 5 × 10−9 | (41) | |||
rs2074613 | 1 × 10−8 | (41) | ||||
FOSL2 | FOS-like antigen 2 | 20(18 up/2 down)/47 |
Reported p values were from literature (listed in Ref. column) reported genome-wide significance analysis (GWAS) studies.
3.4. Pathway analysis indicates an important role in regulation of mitochondrial functions by Vitamin D
According to the pathway enrichment analysis on DEGs after Vitamin D exposure, Vitamin D mainly enhances mitochondrial functions annotated by Gene Ontology (GO) database (48) or Broad MSigDB (49, 50), such as mitochondrial membrane (GO), mitochondrial inner membrane (GO), genes involved in the citric acid (TCA) cycle and respiratory electron transport (Broad MSigDB), cellular respiration (GO), mitochondrial matrix (GO), generation of precursor metabolites and energy (GO), mitochondrial membrane part (GO), genes involved in respiratory electron transport, ATP synthesis by chemiosmotic coupling and heat production by uncoupling proteins (Broad MSigDB), and energy derivation by oxidation of organic compounds (GO). Immune response-related pathways such as innate immune response (GO), regulation of defense response (GO), and inflammatory response (GO), are also on the top of the listed pathways (Table S5).
3.5. Association of Vitamin D usage with decline of cognitive function
The baseline characteristics of patients ever or not on Vitamin D during follow-up are shown on Table S6. The baseline MMSE is slightly higher for the subjects ever on Vitamin D (22.5 +/− 4.1 vs 10.8 +/− 5). More subjects in the group ever on Vitamin D had more than 12 years education than those in the group never on Vitamin D (62% vs 45%). The mixed-effect model estimates that the decline rates in MMSE in the Vitamin D group and the non-Vitamin D group are 1.84 (95% CI: 1.49–2.18) MMSE /year and 2.11 (95% CI: 1.89, 2.33) MMSE/year, respectively (F1,628=1.74, p=0.19).
4. Discussion
4.1. Vitamin D and psychosis
A link has been suggested between Vitamin D deficiency and the presence of schizophrenia-related symptoms by many studies. Adults and adolescents with higher dietary intakes of Vitamin D had a lower prevalence of psychotic-like symptoms in clinical observations. Patients with psychotic disorders were found to have lower Vitamin D levels than healthy controls, and this was mainly due to an observed association among schizophrenia patients (51). A similar conclusion was reached by a systematic review (52). Based on these reported studies, Vitamin D level has been significantly linked with psychotic symptoms and disorders, although our study is the first to extend this association to psychosis in AD. However, whether hypovitaminosis D may contribute mechanistically to psychosis, or is merely a correlate or confound, remains unknown.
4.2. Vitamin D and Alzheimer’s disease
Our findings are also congruent with prior evidence that Vitamin D deficiency is associated with cognitive impairment. Patients with AD have a high prevalence of Vitamin D deficiency, which is also associated with low mood and impaired cognitive performance in older people (53). A recent paper showed that the onset of hypovitaminosis D was accompanied by cognitive decline. Memantine was protective against hypovitaminosis D-related cognitive decline. This prompted a clinical trial of memantine together with Vitamin D for AD patients (NCT01409694), but no result has been released yet.
4.3. Role of Vitamin D receptor in AD and psychosis-related diseases
Vitamin D is recognized as playing several roles in the nervous system. The suggested protective effects of Vitamin D in AD were summarized (54) as the regulation of neurotrophic factor production, neurotransmitter levels, oxidative stress mechanisms, calcium (Ca2+) homeostasis and immune system functions, and induction of amyloid beta (Aβ) clearance (55–66). The molecular mechanism of Vitamin D on promoting Aβ clearance was reported to be through the recovery of Aβ phagocytosis by macrophages (65). In contrast, Vitamin D is not reported to affect accumulation of the other hallmark pathologic protein aggregate in AD, microtubule-associated protein tau (67).
Our pharmacogenomics analysis indicates that Vitamin D most likely regulates AD- and psychosis-related genes through the Vitamin D receptor (VDR), the bio-target of Vitamin D. In a small candidate gene study, polymorphism of the VDR gene was reported to be associated with AD (68), though this association has not been confirmed in GWAS analyses. In a functional study, both overexpression of VDR and Vitamin D treatment suppressed amyloid precursor protein (APP) transcription in vitro (69). Similarly, Vitamin D treatment in two AD mouse models reduced levels of soluble and insoluble Aβ and improved conditioned fear memory (70). Recently, our genetic association study of AD+P vs. AD-P patients found that polygenic risk for schizophrenia protects against the risk of psychosis in AD (71). Among the loci comprising this polygenic risk, we found that VDR can regulate the expression of TCF4 (indirect) (72) and CACNA1C (Calcium Voltage-Gated Channel Subunit Alpha1 C, down-regulate) (73). However, although these two genes are regulated by VDR, they were not among our most perturbed gene list. This may imply that their regulation is tissue-dependent as most of the biosets we used for the analysis were derived from non-neural cell types.
It would not be surprising if Vitamin D may modulate the AD progression to psychosis by regulating calcium (Ca2+) homeostasis. The most important role of Vitamin D in the periphery is to maintain skeletal calcium balance by promoting calcium absorption in the intestines, promoting bone resorption by increasing osteoclast number, maintaining calcium and phosphate levels for bone formation, and allowing proper functioning of parathyroid hormone to maintain serum calcium levels. Other possible effects of Vitamin D treatment may include fueling mitochondria through increasing the expression of related proteins (74) or modulating the gene expression of AD or psychosis-related genes as indicated in Table 3.
We would like to point out some limitations of our study: the dosage information for Vitamin D was not available, as such, we just roughly classified the subjects as ever or never taking the medication. We did a sensitivity analysis using the proportion of visits on Vitamin D and duration of time on Vitamin D, results consistent with our analyses of ever versus never using Vitamin D (Supplemental Methods and Results). Also, secular trends exist in Vitamin D usage. That is, the use of Vitamin D was rare before year 2008 in our dataset, but its usage became frequent in later years (Table S7). Data collected from 2008 to present revealed a more significant protective effect of Vitamin D (Supplemental Methods and Results). Finally, we note that the studies of perturbation of gene expression by Vitamin D are conducted in model systems that either cannot (e.g. cell culture), or have not (animal models), evaluated the age-dependent effects of Vitamin D on the transcriptome and have not specifically evaluated the effects in brain. Thus, it is possible that other gene perturbations relevant to any possible effects of Vitamin D on reduced risk of psychosis in AD went undetected by our gene signature analyses.
5. Conclusions
Our clinical outcome analyses supported the protective association of Vitamin D with delayed onset and reduced occurrence of psychotic symptoms in AD patients. We did not find an association of Vitamin D use with rate of cognitive decline, suggesting that any effects of Vitamin D on psychosis risk are independent of signaling pathways associated with cognitive decline. The AD- and psychosis-related genes that are enriched in the dataset of genes most perturbed after Vitamin D exposure imply that Vitamin D might modulate AD progression via these genes. These mechanisms of action of Vitamin D could therefore provide leads to novel approaches for the reduction of psychosis in AD. Finally, some patients did take Vitamin D without beneficial effects on either cognitive functions or psychosis symptoms. This may indicate that only patients bearing risk alleles for genes influenced by Vitamin D can benefit from the Vitamin D treatment. If this latter hypothesis can be validated by well-designed clinical studies, it would provide valuable guidance for personalized AD treatment.
Supplementary Material
Highlights.
Vitamin D was used more often in patients of Alzheimer’s disease without psychosis symptoms than in patients of Alzheimer’s disease with psychosis symptoms.
Vitamin D use was significantly associated with delayed time to psychosis in Alzheimer’s disease patients.
The molecular mechanism of the beneficial effect might be attributed to the facts that Vitamin D can regulate Alzheimer’s disease and/or psychosis-related genes.
Acknowledgment
This work was supported by Veterans Health Administration Grant BX000452 and NIH Grants AG027224, MH116046, and AG005133. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health, the Department of Veterans Affairs, or the United States Government.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. 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(3):263–9. doi: DOI 10.1016/j.jalz.2011.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Langa KM, Larson EB, Crimmins EM, Faul JD, Levine DA, Kabeto MU, Weir DR. A comparison of the prevalence of dementia in the United States in 2000 and 2012. JAMA Internal Medicine 2017;177(1):51–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Liu H, Wang L, Su W, Xie X-Q. Advances in recent patent and clinical trial drug development for Alzheimer’s disease. Pharmaceutical patent analyst 2014;3(4):429–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Emanuel JE, Lopez OL, Houck PR, Becker JT, Weamer EA, DeMichele-Sweet MAA, Kuller L, Sweet RA. Trajectory of cognitive decline as a predictor of psychosis in early Alzheimer disease in the cardiovascular health study. The American Journal of Geriatric Psychiatry 2011;19(2):160–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Seltman HJ, Mitchell S, Sweet RA. A Bayesian model of psychosis symptom trajectory in Alzheimer’s disease. International journal of geriatric psychiatry 2016;31(2):204–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Peters ME, Schwartz S, Han D, Rabins PV, Steinberg M, Tschanz JT, Lyketsos CG. Neuropsychiatric symptoms as predictors of progression to severe Alzheimer’s dementia and death: the Cache County Dementia Progression Study. American Journal of Psychiatry 2015;172(5):460–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Scarmeas N, Brandt J, Albert M, Hadjigeorgiou G, Papadimitriou A, Dubois B, Sarazin M, Devanand D, Honig L, Marder K. Delusions and hallucinations are associated with worse outcome in Alzheimer disease. Archives of neurology 2005;62(10):1601–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sweet RA, Pollock BG, Sukonick DL, Mulsant BH, Rosen J, Klunk WE, Kastango KB, DeKosky ST, Ferrell RE. The 5-HTTPR polymorphism confers liability to a combined phenotype of psychotic and aggressive behavior in Alzheimer disease. International Psychogeriatrics 2001;13(4):401–9. [DOI] [PubMed] [Google Scholar]
- 9.Bassiony MM, Steinberg MS, Warren A, Rosenblatt A, Baker AS, Lyketsos CG. Delusions and hallucinations in Alzheimer’s disease: prevalence and clinical correlates. International journal of geriatric psychiatry 2000;15(2):99–107. [DOI] [PubMed] [Google Scholar]
- 10.Steele C, Rovner B, Chase GA, Folstein M. Psychiatric symptoms and nursing home placement of patients with Alzheimer’s disease. The American journal of psychiatry 1990;147(8):1049. [DOI] [PubMed] [Google Scholar]
- 11.Weamer EA, DeMichele-Sweet MA, Cloonan YK, Lopez OL, Sweet RA. Incident Psychosis in Subjects With Mild Cognitive Impairment or Alzheimer’s Disease. J Clin Psychiatry 2016;77(12):e1564–e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gaugler JE, Yu F, Krichbaum K, Wyman JF. Predictors of nursing home admission for persons with dementia. Medical care 2009;47(2):191–8. [DOI] [PubMed] [Google Scholar]
- 13.Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M, Hieronymus H, Wei G, Armstrong SA, Haggarty SJ, Clemons PA, Wei R, Carr SA, Lander ES, Golub TR. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 2006;313(5795):1929–35. doi: 10.1126/science.1132939. [DOI] [PubMed] [Google Scholar]
- 14.Chang M, Smith S, Thorpe A, Barratt MJ, Karim F. Evaluation of phenoxybenzamine in the CFA model of pain following gene expression studies and connectivity mapping. Molecular pain 2010;6(1):56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dudley JT, Sirota M, Shenoy M, Pai RK, Roedder S, Chiang AP, Morgan AA, Sarwal MM, Pasricha PJ, Butte AJ. Computational repositioning of the anticonvulsant topiramate for inflammatory bowel disease. Science translational medicine 2011;3(96):96ra76–96ra76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Iorio F, Bosotti R, Scacheri E, Belcastro V, Mithbaokar P, Ferriero R, Murino L, Tagliaferri R, Brunetti-Pierri N, Isacchi A. Discovery of drug mode of action and drug repositioning from transcriptional responses. Proceedings of the National Academy of Sciences 2010;107(33):14621–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kunkel SD, Suneja M, Ebert SM, Bongers KS, Fox DK, Malmberg SE, Alipour F, Shields RK, Adams CM. mRNA expression signatures of human skeletal muscle atrophy identify a natural compound that increases muscle mass. Cell metabolism 2011;13(6):627–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cho YH, Lee SY, Kim CM, Kim ND, Choe S, Lee C-H, Shin J-H. Effect of loquat leaf extract on muscle strength, muscle mass, and muscle function in healthy adults: a randomized, double-blinded, and placebo-controlled trial. Evidence-Based Complementary and Alternative Medicine 2016;2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Siavelis JC, Bourdakou MM, Athanasiadis EI, Spyrou GM, Nikita KS. Bioinformatics methods in drug repurposing for Alzheimer’s disease. Briefings in bioinformatics 2015;17(2):322–35. [DOI] [PubMed] [Google Scholar]
- 20.Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, Snyder PJ, Carrillo MC, Thies B, Phelps CH. 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(3):270–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Petersen RC. Mild cognitive impairment as a diagnostic entity. J Intern Med 2004;256(3):183–94. [DOI] [PubMed] [Google Scholar]
- 22.McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA work group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s disease. Neurology 1984;34:939–44. [DOI] [PubMed] [Google Scholar]
- 23.Lopez OL, Becker JT, Chang YF, Sweet RA, Aizenstein H, Snitz B, Saxton J, McDade E, Kamboh MI, DeKosky ST, Reynolds CF III, Klunk WE. The long-term effects of conventional and atypical antipsychotics in patients with probable Alzheimer’s disease. Am J Psychiatry 2013;170(9):1051–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lopez OL, Becker JT, Klunk WE, Saxton J, Hamilton RL, Kaufer DI, Sweet RA, Meltzer C Cidis, Wisniewski SR, Kamboh MI, DeKosky ST. Research evaluation and diagnosis of probable Alzheimer’s disease over the last two decades. I. Neurology 2000;55:1854–62. [DOI] [PubMed] [Google Scholar]
- 25.Lopez OL, Becker JT, Klunk WE, Saxton J, Hamilton RL, Kaufer D, Sweet RA, Meltzer C Cidis, Wisniewski SR, Kamboh MI, DeKosky ST. Research evaluation and diagnosis of possible Alzheimer’s disease over the last two decades. II. Neurology 2000;55:1863–9. [DOI] [PubMed] [Google Scholar]
- 26.Wilkosz PA, Seltman HJ, Devlin B, Weamer EA, Lopez OL, DeKosky ST, Sweet RA. Trajectories of cognitive decline in Alzheimer’s disease. Int Psychogeriatr 2010;22(2):281–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975;12(3):189–98. [DOI] [PubMed] [Google Scholar]
- 28.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the royal statistical society Series B (Methodological) 1995:289–300. [Google Scholar]
- 29.Grambsch PM, Therneau TM. Proportional hazards tests and diagnostics based on weighted residuals. Biometrika 1994;81(3):515–26. [Google Scholar]
- 30.Goeman F, De Nicola F, De Meo PDO, Pallocca M, Elmi B, Castrignanò T, Pesole G, Strano S, Blandino G, Fanciulli M. VDR primary targets by genome-wide transcriptional profiling. The Journal of steroid biochemistry and molecular biology 2014;143:348–56. [DOI] [PubMed] [Google Scholar]
- 31.Milani C, Katayama MLH, de Lyra EC, Welsh J, Campos LT, Brentani MM, do Socorro Maciel M, Roela RA, Del Valle PR, Góes JCGS. Transcriptional effects of 1, 25 dihydroxyvitamin D 3 physiological and supra-physiological concentrations in breast cancer organotypic culture. BMC cancer 2013;13(1):119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.McMahon L, Schwartz K, Yilmaz O, Brown E, Ryan LK, Diamond G. Vitamin D-mediated induction of innate immunity in gingival epithelial cells. Infection and immunity 2011;79(6):2250–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wilfinger J, Seuter S, Tuomainen T-P, Virtanen JK, Voutilainen S, Nurmi T, de Mello VD, Uusitupa M, Carlberg C. Primary vitamin D receptor target genes as biomarkers for the vitamin D3 status in the hematopoietic system. The Journal of nutritional biochemistry 2014;25(8):875–84. [DOI] [PubMed] [Google Scholar]
- 34.Ryynänen J, Seuter S, Campbell MJ, Carlberg C. Gene regulatory scenarios of primary 1, 25-dihydroxyvitamin D3 target genes in a human myeloid leukemia cell line. Cancers 2013;5(4):1221–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kovalenko PL, Zhang Z, Cui M, Clinton SK, Fleet JC. 1, 25 dihydroxyvitamin D-mediated orchestration of anticancer, transcript-level effects in the immortalized, non-transformed prostate epithelial cell line, RWPE1. BMC genomics 2010;11(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wood RJ, Tchack L, Angelo G, Pratt RE, Sonna LA. DNA microarray analysis of vitamin D-induced gene expression in a human colon carcinoma cell line. Physiological genomics 2004;17(2):122–9. [DOI] [PubMed] [Google Scholar]
- 37.Wang T-T, Tavera-Mendoza LE, Laperriere D, Libby E, Burton MacLeod N, Nagai Y, Bourdeau V, Konstorum A, Lallemant B, Zhang R. Large-scale in silico and microarray-based identification of direct 1, 25-dihydroxyvitamin D3 target genes. Molecular endocrinology 2005;19(11):2685–95. [DOI] [PubMed] [Google Scholar]
- 38.Meyer MB, Benkusky NA, Sen B, Rubin J, Pike JW. Epigenetic plasticity drives adipogenic and osteogenic differentiation of marrow-derived mesenchymal stem cells. Journal of Biological Chemistry 2016;291(34):17829–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Halsall J, Osborne J, Hutchinson P, Pringle J. In silico analysis of the 5′ region of the Vitamin D receptor gene: functional implications of evolutionary conservation. The Journal of steroid biochemistry and molecular biology 2007;103(3–5):352–6. [DOI] [PubMed] [Google Scholar]
- 40.Lam M, Trampush JW, Yu J, Knowles E, Davies G, Liewald DC, Starr JM, Djurovic S, Melle I, Sundet K. Large-Scale Cognitive GWAS Meta-Analysis Reveals Tissue-Specific Neural Expression and Potential Nootropic Drug Targets. Cell reports 2017;21(9):2597–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hill W, Marioni R, Maghzian O, Ritchie S, Hagenaars S, McIntosh A, Gale C, Davies G, Deary I. A combined analysis of genetically correlated traits identifies 187 loci and a role for neurogenesis and myelination in intelligence. Molecular psychiatry 2018:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Okbay A, Beauchamp JP, Fontana MA, Lee JJ, Pers TH, Rietveld CA, Turley P, Chen G-B, Emilsson V, Meddens SFW. Genome-wide association study identifies 74 loci associated with educational attainment. Nature 2016;533(7604):539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wolthusen RP, Hass J, Walton E, Turner JA, Rössner V, Sponheim SR, Ho B-C, Holt DJ, Gollub RL, Calhoun V. Genetic underpinnings of left superior temporal gyrus thickness in patients with schizophrenia. The World Journal of Biological Psychiatry 2015;16(6):430–40. [PMC free article] [PubMed] [Google Scholar]
- 44.Sherva R, Tripodis Y, Bennett DA, Chibnik LB, Crane PK, De Jager PL, Farrer LA, Saykin AJ, Shulman JM, Naj A. Genome-wide association study of the rate of cognitive decline in Alzheimer’s disease. Alzheimer’s & dementia: the journal of the Alzheimer’s Association 2014;10(1):45–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Adkins DE, Åberg K, McClay JL, Bukszár J, Zhao Z, Jia P, Stroup TS, Perkins D, McEvoy JP, Lieberman JA. Genomewide pharmacogenomic study of metabolic side effects to antipsychotic drugs. Molecular psychiatry 2011;16(3):321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Deming Y, Li Z, Kapoor M, Harari O, Del-Aguila JL, Black K, Carrell D, Cai Y, Fernandez MV, Budde J. Genome-wide association study identifies four novel loci associated with Alzheimer’s endophenotypes and disease modifiers. Acta neuropathologica 2017;133(5):839–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jun GR, Chung J, Mez J, Barber R, Beecham GW, Bennett DA, Buxbaum JD, Byrd GS, Carrasquillo MM, Crane PK. Transethnic genome-wide scan identifies novel Alzheimer’s disease loci. Alzheimer’s & dementia: the journal of the Alzheimer’s Association 2017;13(7):727–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Consortium GO. The Gene Ontology (GO) database and informatics resource. Nucleic acids research 2004;32(suppl_1):D258–D61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences 2005;102(43):15545–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liberzon A, Subramanian A, Pinchback R, Thorvaldsdóttir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics 2011;27(12):1739–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Murri MB, Respino M, Masotti M, Innamorati M, Mondelli V, Pariante C, Amore M. Vitamin D and psychosis: mini meta-analysis. Schizophrenia research 2013;150(1):235–9. [DOI] [PubMed] [Google Scholar]
- 52.Adamson J, Lally J, Gaughran F, Krivoy A, Allen L, Stubbs B. Correlates of vitamin D in psychotic disorders: a comprehensive systematic review. Psychiatry research 2017;249:78–85. [DOI] [PubMed] [Google Scholar]
- 53.Lu’o’ng KVQ, Nguyễn LTH. The beneficial role of vitamin D in Alzheimer’s disease. American Journal of Alzheimer’s Disease & Other Dementias® 2011;26(7):511–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gezen-Ak D, Yılmazer S, Dursun E. Why vitamin D in Alzheimer’s disease? The hypothesis. Journal of Alzheimer’s Disease 2014;40(2):257–69. [DOI] [PubMed] [Google Scholar]
- 55.Feron F, Burne T, Brown J, Smith E, McGrath J, Mackay-Sim A, Eyles D. Developmental Vitamin D3 deficiency alters the adult rat brain. Brain research bulletin 2005;65(2):141–8. [DOI] [PubMed] [Google Scholar]
- 56.Cekic M, Sayeed I, Stein DG. Combination treatment with progesterone and vitamin D hormone may be more effective than monotherapy for nervous system injury and disease. Frontiers in neuroendocrinology 2009;30(2):158–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Garcion E, Wion-Barbot N, Montero-Menei CN, Berger F, Wion D. New clues about vitamin D functions in the nervous system. Trends in Endocrinology & Metabolism 2002;13(3):100–5. [DOI] [PubMed] [Google Scholar]
- 58.Dursun E, Gezen-Ak D, Yilmazer S. A new mechanism for amyloid-β induction of iNOS: vitamin D-VDR pathway disruption. Journal of Alzheimer’s Disease 2013;36(3):459–74. [DOI] [PubMed] [Google Scholar]
- 59.Bouillon R, Carmeliet G, Daci E, Segaert S, Verstuyf A. Vitamin D metabolism and action. Osteoporosis international 1998;8(8):S013–S9. [DOI] [PubMed] [Google Scholar]
- 60.Gezen-Ak D, Dursun E, Yilmazer S. The effects of vitamin D receptor silencing on the expression of LVSCC-A1C and LVSCC-A1D and the release of NGF in cortical neurons. PLoS One 2011;6(3):e17553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Dursun E, Gezen-Ak D, Yilmazer S. A novel perspective for Alzheimer’s disease: vitamin D receptor suppression by amyloid-β and preventing the amyloid-β induced alterations by vitamin D in cortical neurons. Journal of Alzheimer’s Disease 2011;23(2):207–19. [DOI] [PubMed] [Google Scholar]
- 62.Brown J, Bianco JI, McGrath JJ, Eyles DW. 1, 25-dihydroxyvitamin D3 induces nerve growth factor, promotes neurite outgrowth and inhibits mitosis in embryonic rat hippocampal neurons. Neuroscience letters 2003;343(2):139–43. [DOI] [PubMed] [Google Scholar]
- 63.Wang J-Y, Wu J-N, Cherng T-L, Hoffer BJ, Chen H-H, Borlongan CV, Wang Y. Vitamin D3 attenuates 6-hydroxydopamine-induced neurotoxicity in rats. Brain research 2001;904(1):67–75. [DOI] [PubMed] [Google Scholar]
- 64.Eyles DW, Burne TH, McGrath JJ. Vitamin D, effects on brain development, adult brain function and the links between low levels of vitamin D and neuropsychiatric disease. Frontiers in neuroendocrinology 2013;34(1):47–64. [DOI] [PubMed] [Google Scholar]
- 65.Mizwicki MT, Menegaz D, Zhang J, Barrientos-Durán A, Tse S, Cashman JR, Griffin PR, Fiala M. Genomic and nongenomic signaling induced by 1α, 25 (OH) 2-vitamin D 3 promotes the recovery of amyloid-β phagocytosis by Alzheimer’s disease macrophages. Journal of Alzheimer’s Disease 2012;29(1):51–62. [DOI] [PubMed] [Google Scholar]
- 66.Mizwicki MT, Liu G, Fiala M, Magpantay L, Sayre J, Siani A, Mahanian M, Weitzman R, Hayden EY, Rosenthal MJ. 1α, 25-Dihydroxyvitamin D 3 and Resolvin D1 Retune the Balance between Amyloid-β Phagocytosis and Inflammation in Alzheimer’s Disease Patients. Journal of Alzheimer’s Disease 2013;34(1):155–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Keeney JT, Butterfield DA. Vitamin D deficiency and Alzheimer disease: common links. Neurobiology of disease 2015;84:84–98. [DOI] [PubMed] [Google Scholar]
- 68.Gezen-Ak D, Dursun E, Ertan T, Hanagasi H, Gürvit H, Emre M, Eker E, Öztürk M, Engin F, Yilmazer S. Association between vitamin D receptor gene polymorphism and Alzheimer’s disease. The Tohoku journal of experimental medicine 2007;212(3):275–82. [DOI] [PubMed] [Google Scholar]
- 69.Wang L, Hara K, Van Baaren JM, Price JC, Beecham GW, Gallins PJ, Whitehead PL, Wang G, Lu C, Slifer MA. Vitamin D receptor and Alzheimer’s disease: a genetic and functional study. Neurobiology of Aging 2012;33(8):1844 e1-. e9. [DOI] [PubMed] [Google Scholar]
- 70.Durk MR, Han K, Chow EC, Ahrens R, Henderson JT, Fraser PE, Pang KS. 1α, 25-Dihydroxyvitamin D3 reduces cerebral amyloid-β accumulation and improves cognition in mouse models of Alzheimer’s disease. Journal of Neuroscience 2014;34(21):7091–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.DeMichele-Sweet MAA, Weamer EA, Klei L, Vrana DT, Hollingshead DJ, Seltman HJ, Sims R, Foroud T, Hernandez I, Moreno-Grau S. Genetic risk for schizophrenia and psychosis in Alzheimer disease. Molecular psychiatry 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Beildeck ME, Islam M, Shah S, Welsh J, Byers SW. Control of TCF-4 expression by VDR and vitamin D in the mouse mammary gland and colorectal cancer cell lines. PloS one 2009;4(11):e7872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dursun E, Gezen-Ak D, Yilmazer S. Beta amyloid suppresses the expression of the vitamin d receptor gene and induces the expression of the vitamin d catabolic enzyme gene in hippocampal neurons. Dementia and geriatric cognitive disorders 2013;36(1–2):76–86. [DOI] [PubMed] [Google Scholar]
- 74.Ferreira GB, Vanherwegen A-S, Eelen G, Gutiérrez ACF, Van Lommel L, Marchal K, Verlinden L, Verstuyf A, Nogueira T, Georgiadou M. Vitamin D3 induces tolerance in human dendritic cells by activation of intracellular metabolic pathways. Cell Reports 2015;10(5):711–25. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.