ArticlePDF Available

Effect of vitamin D on cognitive decline: results from two ancillary studies of the VITAL randomized trial

Springer Nature
Scientific Reports
Authors:

Abstract and Figures

Low vitamin D levels have been associated with cognitive decline; however, few randomized trials have been conducted. In a trial, we evaluated vitamin D3 supplementation on cognitive decline. We included participants aged 60+ years (mean[SD] = 70.9[5.8] years) free of cardiovascular disease and cancer in two substudies in the VITAL 2 × 2 randomized trial of vitamin D3 (2000 IU/day of cholecalciferol) and fish oil supplements: 3424 had cognitive assessments by phone (eight neuropsychologic tests; 2.8 years follow-up) and 794 had in-person assessments (nine tests; 2.0 years follow-up). The primary, pre-specified outcome was decline over two assessments in global composite score (average z-scores of all tests); substudy-specific results were meta-analyzed. The pooled mean difference in annual rate of decline (MD) for vitamin D3 versus placebo was 0.01 (95% CI − 0.01, 0.02; p = 0.39). We observed no interaction with baseline 25-hydroxyvitamin-D levels (p-interaction = 0.84) and a significant interaction with self-reported race (p-interaction = 0.01). Among Black participants (19%), those assigned vitamin D3 versus placebo had better cognitive maintenance (MD = 0.04, 95% CI 0.01, 0.08, similar to that observed for Black participants 1.2 years apart in age). Thus, vitamin D3 (2000 IU/day cholecalciferol) supplementation was not associated with cognitive decline over 2–3 years among community-dwelling older participants but may provide modest cognitive benefits in older Black adults, although these results need confirmation. Trial registration ClinicalTrials.gov; VITAL (NCT01169259), VITAL-DEP (NCT01696435) and VITAL-Cog (NCT01669915); the date the registration for the parent trial (NCT01169259) was submitted to the registry: 7/26/2010 and the date of first patient enrollment in either of the ancillary studies for cognitive function in a subset of eligible VITAL participants: 9/14/2011.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports
Eect of vitamin D on cognitive
decline: results from two ancillary
studies of the VITAL randomized
trial
Jae H. Kang1*, Chirag M. Vyas2, Olivia I. Okereke1,2,3, Soshiro Ogata4, Michelle Albert5,
I.‑Min Lee3,6, Denise D’Agostino6, Julie E. Buring3,6, Nancy R. Cook3,6, Francine Grodstein1,7 &
JoAnn E. Manson3,6
Low vitamin D levels have been associated with cognitive decline; however, few randomized trials
have been conducted. In a trial, we evaluated vitamin D3 supplementation on cognitive decline.
We included participants aged 60+ years (mean[SD] = 70.9[5.8] years) free of cardiovascular
disease and cancer in two substudies in the VITAL 2 × 2 randomized trial of vitamin D3 (2000 IU/
day of cholecalciferol) and sh oil supplements: 3424 had cognitive assessments by phone (eight
neuropsychologic tests; 2.8 years follow‑up) and 794 had in‑person assessments (nine tests; 2.0 years
follow‑up). The primary, pre‑specied outcome was decline over two assessments in global composite
score (average z‑scores of all tests); substudy‑specic results were meta‑analyzed. The pooled mean
dierence in annual rate of decline (MD) for vitamin D3 versus placebo was 0.01 (95% CI − 0.01, 0.02;
p = 0.39). We observed no interaction with baseline 25‑hydroxyvitamin‑D levels (p‑interaction = 0.84)
and a signicant interaction with self‑reported race (p‑interaction = 0.01). Among Black participants
(19%), those assigned vitamin D3 versus placebo had better cognitive maintenance (MD = 0.04, 95%
CI 0.01, 0.08, similar to that observed for Black participants 1.2 years apart in age). Thus, vitamin
D3 (2000 IU/day cholecalciferol) supplementation was not associated with cognitive decline over
2–3 years among community‑dwelling older participants but may provide modest cognitive benets in
older Black adults, although these results need conrmation.
Trial registration ClinicalTrials.gov; VITAL (NCT01169259), VITAL‑DEP (NCT01696435) and VITAL‑
Cog (NCT01669915); the date the registration for the parent trial (NCT01169259) was submitted to
the registry: 7/26/2010 and the date of rst patient enrollment in either of the ancillary studies for
cognitive function in a subset of eligible VITAL participants: 9/14/2011.
Vitamin D is a fat-soluble steroid hormone essential for bone and muscle health. Yet, the discovery of vitamin
D’s autocrine pathways in multiple cell types has stimulated interest in its role in brain function114. Specically,
the vitamin D receptor is expressed in the cerebral cortex and hippocampus, critical for cognition and memory.
In animals, vitamin D deciency has been linked with decits in brain development and aging1517.
In humans, observational studies have implicated low vitamin D in cognitive impairment and dementia1820,
although the literature has been mixed21,22. Observational studies have used varying denitions of low vitamin
D and of cognitive impairment/dementia; further, the issue of reverse causation is important, as low vitamin D
concentrations may have resulted from lifestyle changes associated with incipient cognitive impairment/demen-
tia. ree major previous randomized clinical trials on cognitive change2325 have not shown benets of vitamin
OPEN
1Channing Division of Network Medicine, Brigham and Women’s Hospital/Harvard Medical School, 181 Longwood
Ave, Boston, MA 02115, USA. 2Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School,
Boston, MA, USA. 3Harvard T. H. Chan School of Public Health, Boston, MA, USA. 4Department of Preventive
Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan. 5University of
California at San Francisco School of Medicine, San Francisco, CA, USA. 6Division of Preventive Medicine, Brigham
and Women’s Hospital/Harvard Medical School, Boston, MA 02215, USA. 7Rush Alzheimer’s Disease Center, Rush
University Medical Center, Chicago, IL, USA. *email: nhjhk@channing.harvard.edu
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
D3. However, even a large trial (n = 4143) with 7.8years of follow-up that observed no eect of vitamin D3 and
calcium supplements on cognitive decline compared with placebo24 used a low dose of vitamin D3 (400IU/day).
us, we investigated whether vitamin D3 supplementation of 2000IU/day may delay cognitive decline over
2–3years compared to placebo among healthy participants aged 60+ years in VITAL (VITamin D and OmegA-3
TriaL; NCT01169259)2628, a randomized trial of vitamin D3 and omega-3 fatty acids in the prevention of major
chronic diseases. In addition, we conducted pre-specied subgroup analyses by race and baseline blood vitamin
D levels, given that supplementation may have stronger eects on subgroups with relatively lower blood vitamin
D levels, including Black adults who are also at higher risk of cognitive decline2931 and in whom we had observed
suggestively dierent eects of vitamin D supplementation in the parent VITAL trial32.
Results
e 3424 participants in VITAL-Cog were aged 60–91years (mean = 71.9; SD = 5.4) at the rst cognitive assess-
ment (Table1); 58.9% were women; 22.2% were Black participants and 49.8% had some years of post-graduate
studies. e mean change over an average of 2.8years of follow-up was − 0.25 (SD = 0.49) in those assigned to
placebo and − 0.24 (SD = 0.48) in those assigned to vitamin D. In CTSC-Cog (Table1), the 794 participants
were aged 60–87years (mean = 67.1; SD = 5.3) at the rst cognitive assessment; 50.4% were women; 5.7% were
Black participants and 55.5% had some post-graduate education. e mean change over a mean of 2.0years of
follow-up was 0.09 (SD = 0.39) in those assigned to placebo and 0.08 (SD = 0.40) in those assigned to vitamin D.
In VITAL-Cog, we did not observe an eect of vitamin D3 supplementation on cognitive function at the end
of follow-up (mean = 2.8years (range = 1.4–4.3years); Table2): the least squares mean for the global score was
0.28 standard units (SE = 0.01) for the vitamin D3 group and − 0.26 (SE = 0.02) for the placebo group (mean
dierence = − 0.02, 95% CI − 0.06, 0.02). We observed no multivariable-adjusted dierences in the global score
annual rate of decline by assignment (Table3; model 2, multivariable-adjusted mean dierence = 0.01, 95% CI
0.01, 0.02). Similarly, multivariable-adjusted dierences in annual rates of decline were not signicant for the
secondary outcomes: 0.01 (95% CI − 0.01, 0.03), verbal memory composite score; 0.01 (95% CI − 0.01, 0.02),
executive function/attention score and 0.03 (95% CI − 0.04, 0.09), TICS.
In CTSC-Cog (Table2), we did not observe an eect of vitamin D3 supplementation on cognition at the end of
follow-up (mean 2.0years (range = 1.0–3.1years)): the least squares mean for the global score was 0.11 (SE = 0.03)
for the vitamin D3 group and 0.06 (SE = 0.03) for the placebo group (mean dierence = 0.05, 95% CI − 0.04,
0.14). e multivariable-adjusted mean dierence in the global score annual rate of decline was − 0.004 (95% CI
− 0.04, 0.03; p = 0.83; Table3). Similarly, multivariable-adjusted mean dierences in annual rates of decline for
secondary cognitive systems were not signicant: 0.01 (95% CI − 0.05, 0.06) for the verbal memory composite
score; − 0.01 (95% CI − 0.05, 0.02) for the executive function/attention score and 0.04 (95% CI − 0.08, 0.17) for
the 3MS score (re-scaled to have range 0–41 points like the TICS). Results for individual tests are in TableS1.
We observed no heterogeneity in the results by substudy (p for heterogeneity ≥ 0.28); thus, the multivariable-
adjusted results were meta-analyzed (Table3). e pooled eect of vitamin D3 supplementation was a mean
dierence in the annual rate of decline of 0.01 (95% CI − 0.01, 0.02; p = 0.39) for the global score; 0.01 (95% CI
0.01, 0.02), verbal composite score; 0.01 (95% CI − 0.01, 0.02), executive function/attention composite score;
and 0.03 (95% CI − 0.03, 0.09), for general cognition (TICS/3MS).
For pre-specied interaction analysis (Table4) by race for the global composite score, we observed a signi-
cant interaction (pooled p-interaction = 0.01), where among Black participants, the vitamin D3 group showed
a signicantly slower rate of decline than placebo (pooled multivariable-adjusted mean dierence in annual
rate of decline = 0.04, 95% CI 0.01, 0.08), but not in other races (pooled multivariable-adjusted mean dier-
ence = − 0.001, 95% CI − 0.01, 0.01). To help interpret these results, among Black participants, at the 2nd assess-
ment in VITAL-Cog, those on vitamin D had a 0.03 standard units higher global score performance than those
on placebo; this dierence was equivalent to that observed with Black participants who were 1.2years apart in
age, indicating an overall modest eect. e benecial vitamin D3 eect among Black participants was stronger
for the executive function/attention score, where the eect was equivalent to the dierence observed between
Black participants who were 3.8years apart in age (when the more conservative xed eects summary was used
for estimation (Table5 footnote)). We observed no signicant interaction with the other pre-specied modi-
er of baseline blood 25(OH)D concentrations for the global score (pooled p-interaction = 0.84; Table4) or the
secondary outcomes (pooled p-interaction ≥ 0.12; Table5). Finally, we observed no signicant interactions for
the other 13 eect modiers evaluated for the global score (pooled p-interactions ≥ 0.18; Table4; for secondary
outcomes, see Table6).
In sensitivity analyses where we restricted the analyses in both substudies to those who reported no hearing
impairment (68% in VITAL-Cog; 86% in CTSC-Cog; pooled mean dierence in the annual rate of decline in
the global score was 0.01 (95% CI − 0.01, 0.02; p = 0.37)) or restricted the analyses to those enrolled from the 1st
assessment in VITAL-Cog (pooled mean dierence was 0.01 (95% CI − 0.01, 0.02; p = 0.39)) or restricted the
analyses in CTSC-Cog to those who did not have neuropsychiatric disorders or possible dementia at baseline
(72%; pooled mean dierence was 0.01 (95% CI − 0.01, 0.02; p = 0.40)) or restricted the analyses in both sub-
studies to those who were in the top 90% of performance in each outcome (to avoid oor eects and to remove
those with possible dementia, especially in VITAL-Cog; pooled mean dierence was − 0.001 (95% CI − 0.01,
0.01; p = 0.93)), results were similar to the main results. Results also did not dier when we additionally adjusted
for practice eects (pooled mean dierence was 0.01 (95% CI − 0.01, 0.02; p = 0.31)) or when we additionally
adjusted for season of cognitive assessment (pooled mean dierence was 0.01 (95% CI − 0.01, 0.02; p = 0.38)).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
Table 1. Baseline characteristics of participants aged 60+ years in the VITAL cognitive substudy by vitamin
D supplement assignment for VITAL-Cog (n = 3424) and CTSC-Cog (n = 794). 3MS Modied Mini-
Mental Status exam (range = 0–100)33; CTSC Clinical and Translational Science Collaborative Center for
VITAL in Boston, MA; EBMT East Boston Memory Test (range = 0–12)34; OTMT Oral Trail Making Test
(range = 0–120s)35,36; SD standard deviation; TICS Telephone Interview for Cognitive Status (range = 0–41)37;
TMT Trail Making Test (range = 0–150s for part A and range = 0–300s for part B)38,39. a Characteristics as
of randomization unless noted otherwise; for categorical variables, the percentages do not add to 100%
due to rounding errors and numbers do not add to the total due to missing values, which were taken out of
descriptive statistical analyses. In the VITAL-Cog, 501 completed only the baseline, 440 completed only the
2nd assessment and 2483 completed both assessments. In the CTSC-Cog, 497 completed both assessments,
279 completed only the baseline and 18 completed only the 2nd assessment. b“ Other race/ethnicity” includes
“Non-Black/African-American Hispanic”, “Asian”, “Native Hawaiian or other Pacic Islander” or “American
Indian/Alaska Native. c Depression is dened as a lifetime history of a depression diagnosis or of treatment for
depression; current use of antidepressants; experiencing two or more weeks of depression in the past 2years or
scoring 10 points or higher on the Patient Health Questionnaire-8.
VITAL-Cog (n = 3424) CTSC-Cog (n = 794)
Vitamin D Group (n = 1710) Placebo Group (n = 1714) Vitamin D3 Group (n = 396) Placebo Group (n = 398)
Mean (SD)
Age at 1st interview, yearsa (n = 2984 in VITAL-Cog;
n = 776 in CTSC-Cog) 71.9 (5.4)
(n = 1480) 71.8 (5.4)
(n = 1504) 66.9 ± 5.2
(n = 385) 67.3 ± 5.4
(n = 391)
Age at 2nd interview, yearsa (n = 2923 in VITAL-Cog;
n = 515 in CTSC-Cog) 73.3 (5.7)
(n = 1466) 73.4 (5.7)
(n = 1457) 69.2 ± 5.1
(n = 254) 69.8 ± 5.6
(n = 261)
Cognitive test scores at 1st interview
VITAL-Cog only tests
TICS 33.9 (2.8) 34.0 (2.8)
OTMT-Part A (s) 10.7 (3.9) 10.3 (3.3)
OTMT-Part B (s) 38.0 (24.2) 37.9 (24.1)
Digit span backwards 6.7 (2.3) 6.8 (2.4)
CTSC-Cog only tests
3MS 94.8 ± 4.9 94.9 ± 4.4
TMT-Part A (s) 29.2 ± 11.5 29.8 ± 9.4
TMT-Part B (s) 80.0 ± 42.9 82.5 ± 44.2
Vegetable naming test 15.6 ± 4.6 15.4 ± 4.5
Common tests across VITAL-Cog and CTSC-Cog
TICS 10-word list recall-immediate 4.6 (1.7) 4.7 (1.7) 4.7 ± 1.3 4.7 ± 1.3
TICS 10-word list recall-delayed 2.7 (1.9) 2.7 (1.9) 2.0 ± 1.8 1.9 ± 1.7
EBMT-immediate 9.6 (1.7) 9.6 (1.8) 9.7 ± 1.7 9.7 ± 1.6
EBMT-delayed 9.3 (1.8) 9.3 (1.9) 9.3 ± 1.7 9.3 ± 1.7
Animal naming test 19.4 (5.5) 19.7 (5.6) 21.1 ± 5.9 20.3 ± 6.1
Global composite score − 0.02 (0.57) 0.02 (0.57) 0.02 (0.63) − 0.02 (0.56)
Baseline serum 25(OH)D (ng/mL) 32.2 (9.8) 32.5 (9.6) 28.0 (8.3) 29.1 (9.1)
n (%)
Omega-3 assignment
Active group 844 (49.4%) 855 (49.9%) 198 (50.0%) 198 (49.8%)
Placebo group 866 (50.6%) 859 (50.1%) 198 (50.0%) 200 (50.3%)
Sex
Female 1011 (59.1%) 1005 (58.6%) 205 (51.8%) 195 (49.0%)
Male 699 (40.9%) 709 (41.4%) 191 (48.2%) 203 (51.0%)
Self-reported race/ethnicity
Non-Hispanic White 1184 (71.3%) 1245 (73.8%) 341 (88.1%) 345 (89.2%)
Black 387 (23.3%) 356 (21.1%) 18 (4.7%) 26 (6.7%)
Other race/ethnicityb90 (5.4%) 85 (5.0%) 28 (7.2%) 16 (4.1%)
Highest attained education
High school or under 189 (11.1%) 183 (10.7%) 29 (7.3%) 34 (8.5%)
College 678 (39.7%) 664 (38.9%) 138 (34.9%) 152 (38.2%)
Post-graduate studies 842 (49.3%) 861 (50.4%) 228 (57.7%) 212 (53.3%)
Depressionc
No 1363 (82.7%) 1383 (82.9%) 324 (83.7%) 309 (79.8%)
Yes 285 (17.3%) 285 (17.1%) 63 (16.3%) 78 (20.2%)
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
Discussion
In this randomized trial among generally healthy community-dwelling older participants followed for 2–3years,
supplementation with 2000IU/day of vitamin D3 was not associated with cognitive decline. No eects were
observed for the primary outcome as well as secondary outcomes of verbal memory, executive function/attention
and global cognition and for both substudies where cognition was assessed by phone or in person. However,
a pre-specied subgroup analysis showed cognitive benets over time for vitamin D3 supplementation versus
placebo among Black participants, but not by levels of 25(OH)D. Because the subgroup analyses by race may be
due to chance, the results should be interpreted with caution and conrmed in future studies.
Randomized trials of vitamin D and cognitive decline21,22,41 in relatively healthy populations have shown
conicting results, with most reporting no benet from supplementation, despite vitamin D’s potential neuropro-
tective anti-inammatory and antioxidant eects711,42. In the largest (n = 4143) and longest trial (7.8years dura-
tion) where women aged 65+ years received vitamin D3 (400IU/day) and calcium (1000mg/day) or placebo24,
Table 2. Cognitive function at two assessments by Vitamin D supplement assignment, for VITAL-Cog
participants aged 60 + years, (n = 3424) assessed by telephone and for CTSC-Cog participants aged 60 + years,
(n = 794) assessed in person. 3MS Modied Mini-Mental Status exam (range = 0–100)33; CI condence interval;
CTSC Clinical and Translational Science Collaborative center for VITAL in Boston, MA; TICS Telephone
Interview of Cognitive Status (range = 0–41)37. a In the VITAL-Cog, 2483 completed both assessments, 501
completed only the baseline, 440 completed only the 2nd assessment. In the CTSC-Cog, 497 completed both
assessments, 279 completed only the baseline and 17 completed only the 2nd assessment. b In the VITAL-
Cog: global score is a composite score representing the mean of the z-scores of 8 tests: TICS (range 0–41),
immediate and delayed recalls of the East Boston Memory Test, category uency (animal naming test), delayed
recall of the TICS 10-word list, oral trails making test A, oral trails making test B and digit span backwards.
Verbal memory score is a composite score representing the mean of the z-scores of 4 tests: the immediate and
delayed recalls of both the TICS 10-word list and the East Boston Memory Test. Executive function/attention
score is a composite score representing the mean of the z-scores of 4 tests: trails making test A and B, category
uency tests (naming animals), and digit-span backwards. In the CTSC-Cog: the global score is a composite
score representing the mean of the z-scores of 9 tests: 3MS, immediate and delayed recalls of the East Boston
Memory Test, category uency tests (naming animals and vegetables), the immediate and delayed recalls of
a 10-word list and trail-making tests A and B. Verbal memory score was dened the same way as in VITAL-
Cog. Executive function/attention score is a composite score representing the mean of the z-scores of 4 tests:
trails making tests A and B, category uency tests (naming animals and vegetables). c Least squares means
and standard errors and dierences of least squares means and standard errors were derived from univariate
models.
VITAL-COG (n = 3424; telephone assessments) CTSC-COG (n = 794; in-person assessments)
Vitamin D Group Placebo Group Dierence in score
at each timepoint
(Vitamin D
-Placebo; 95% CI)c
Vitamin D Group Placebo Group Dierence in
score at each
timepoint (Vitamin
D-Placebo; 95%
CI)c
N Mean (SE)cN Mean (SE)cN Mean (SE) N Mean (SE)
Primary outcome Primary outcome
Global composite
scorebDierence in scorecGlobal composite
scorebDierence in scorec
1st assessment
score 14,800 − 0.04 (0.01) 1504 − 0.01 (0.01) − 0.03 (− 0.07, 0.01) 1st assessment
scorec385 0.02 (0.03) 391 − 0.03 (0.03) 0.05 (− 0.04, 0.13)
2nd assessment
score 1466 − 0.28 (0.01) 1457 − 0.26 (0.02) − 0.02 (− 0.06, 0.02) 2nd assessment
scorec254 0.11 (0.03) 261 0.06 (0.03) 0.05 (− 0.04, 0.14)
Secondary outcomes Secondary outcomes
Verbal memory
composite scorebDierence in scorecVerbal memory
composite scorebDierence in scorec
1st assessment
score 14,800 − 0.02 (0.02) 1504 − 0.01 (0.02) − 0.01 (− 0.06, 0.04) 1st assessment
scorec385 0.01 (0.04) 391 − 0.02 (0.03) 0.03 (− 0.07, 0.13)
2nd assessment
score 1466 − 0.01 (0.02) 1457 − 0.02 (0.02) 0.01 (− 0.04, 0.07) 2nd assessment
scorec254 0.17 (0.04) 261 0.11 (0.04) 0.07 (− 0.05, 0.18)
Executive func-
tion/attention
compositeb scoreaDierence in scorecExecutive func-
tion/attention
compositeb scoreaDierence in scorec
1st assessment
score 14,800 − 0.05 (0.02) 1504 0.01 (0.02) − 0.05 (− 0.10,
− 0.01) 1st assessment
scorec385 0.03 (0.04) 391 − 0.05 (0.03) 0.08 (− 0.02, 0.18)
2nd assessment
score 1466 − 0.53 (0.02) 1457 − 0.49 (0.02) − 0.04 (− 0.08, 0.01) 2nd assessment
scorec254 0.04 (0.04) 261 − 0.03 (0.04) 0.06 (− 0.04, 0.16)
TICS Dierence in scorec3MS Dierence in scorec
1st assessment
score 14,800 33.81 (0.07) 1504 33.92 (0.07) − 0.12 (− 0.31, 0.08) 1st assessment
scorec385 94.80 (0.25) 391 94.86 (0.22) − 0.05 (− 0.71, 0.60)
2nd assessment
score 1466 33.92 (0.07) 1457 33.97 (0.07) − 0.05 (− 0.26, 0.16) 2nd assessment
scorec254 95.69 (0.23) 261 95.52 (0.24) 0.17 (− 0.48, 0.82)
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
no association was observed between vitamin D3/calcium treatment and incident cognitive impairment or
dementia, although the vitamin D3 dose was low, and was one h of our dose. us, our ndings are important
in suggesting that even much higher doses of vitamin D3 do not provide meaningful cognitive benets overall.
Similarly, in two European randomized trials of 2000IU/day of vitamin D3 compared to placebo43 (n = 2157) or
800IU/day44 (n = 273) of vitamin D3 assessed over 343 and 244years among community-dwelling older persons
observed no dierences in change in cognitive function by intervention. Four small studies (n < 400)23,4547 have
evaluated even higher doses of vitamin D3 but were short-term (≤ 1year of treatment) and also have not observed
signicant overall dierences in cognitive change. Among Black adults, in a 3-year study among 260 older women
(aged 65–73years) where higher doses of vitamin D3 (individualized doses of ≥ 2400IU/day needed to maintain
serum 25(OH)D ≥ 30ng/mL) and calcium (1200mg/day) was compared to placebo and calcium (1200mg/day),
Owusu etal.48 observed no dierence in change in MMSE performance, similar to our null nding for general
cognition for Black participants; change in executive function was not assessed in this study. us, our study
adds to the literature in that it was a long-term, large study (n > 4200) testing a relatively high dose of vitamin
D3 for long durations and had a relatively large representation of Black participants.
In subgroup analyses, we observed that in Black participants, vitamin D3 supplementation was signicantly
associated with better cognitive maintenance in the global score and executive function/attention score. is
is consistent with studies that have observed that vitamin D3 deciency is associated most prominently with
decits in executive function41,45,49. While the interaction by race and baseline blood 25(OH)D levels were pre-
specied, these subgroup ndings were not adjusted for multiple comparisons and thus, should be interpreted
with caution. We had hypothesized a priori that vitamin D3 might have particular benets in Black participants
who had lower 25(OH)D concentrations at baseline; yet, given the lack of a signicant eect modication by
Table 3. Meta-analysis of the mean dierences (95% CI) in change over time among VITAL-Cog participants
(n = 3424) and CTSC-Cog participants (n = 794), by Vitamin D supplement assignment. 3MS Modied
Mini-Mental Status exam (range = 0–100)33, CI condence interval; CTSC Clinical and Translational Science
Collaborative center for VITAL in Boston, MA; TICS Telephone Interview of Cognitive Status (range = 0–41)37.
a For denitions of the global scores and the key secondary outcomes for the two populations, see footnotes
for Table2. b From linear mixed models of cognitive performance: model 1 includes time since randomization
modelled as a continuous variable, vitamin D assignment, and their interaction. c From linear mixed models
of cognitive performance: model 2 is model 1 with adjustment for 6 additional variables, omega-3 assignment
(yes/no), sex (male/female), age at randomization (years), race/ethnicity (non-Hispanic white, black, other race/
ethnicity), education (high school or under, college, graduate school), history of depression (yes/no; see footnote
in Table1 for denition), and the six interaction terms (products with time since randomization). d Pooled
using Dersimonian and Laird xed-eects method for meta-analysis40 except for general cognition where the
p for heterogeneity across the two substudies was 0.04 and results were meta-analyzed with random-eects.
e Due to the dierences in scale between the TICS (0–41) used in VITAL-Cog and 3MS (range 0–100) used in
CTSC-Cog, for pooling purposes, the 3MS scores were multiplied by 0.41 for conversion to the same scale as the
TICS scores. As the p for heterogeneity across the two substudies was 0.04, the results were meta-analyzed with
Dersimonian and Laird method incorporating random-eects40. f None of the eects for the secondary outcomes
were signicant at Bonferroni-adjusted p-value of 0.0167 (= 0.05/3 secondary outcomes).
Difference in annual rate of change
Vitamin D – Placebo; 95% confidence interval)
Primary outcome: Global composite score*Model 1:
univariate
Model 2:
multivariable-ad
j
usted
VITAL-Cog 0.01 (-0.01, 0.02) 0.01 (-0.01, 0.02)
CTSC-Cog 0.003 (-0.03, 0.04) -0.004 (-0.04, 0.03)
POOLED§0.01 (-0.01, 0.02)
pooled p-value=0.39
Key secondary outcomes
p
ooled p-values≥0.33
Verbal memory composite score*
VITAL-Cog 0.01 (-0.01, 0.03) 0.01 (-0.01, 0.03)
CTSC-Cog 0.02 (-0.04, 0.08) 0.01 (-0.05, 0.06)
POOLED§0.01 (-0.01, 0.02)
Executive function/attention composite score*
VITAL-Cog 0.01 (-0.01, 0.03) 0.01 (-0.01, 0.02)
CTSC-Cog -0.01 (-0.04, 0.03) -0.01 (-0.05, 0.02)
POOLED§0.01 (-0.01, 0.02)
General cognition||
VITAL-Cog 0.03 (-0.04, 0.10) 0.03 (-0.04, 0.09)
CTSC-Cog 0.04 (-0.08, 0.17) 0.04 (-0.08, 0.17)
POOLED§0.03 (-0.03, 0.09)
Placebo better Vitamin D better
-0. 2-0.10 0. 10.2
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
Table 4. Mean dierence (95% CI) in rate of change in global score between vitamin D and placebo group:
eect modication by risk factors for cognitive decline. CI condence interval; CTSC-Cog, subset that received
in-person interviews at the Harvard Clinical and Translational Science Collaborative center for VITAL in
Boston, MA; CVD cardiovascular disease; VITAL-Cog subset that received telephone cognitive interviews in
VITAL. For denitions of the global scores for the two populations, see footnote for Table2. a Mean dierence
in annual rate of decline of vitamin D—placebo groups from multivariable-adjusted linear mixed models: see
footnotes for Table3. e stratied analyses were done among those with non-missing data on the eect modier.
b Interaction terms across the two substudies were pooled using Dersimonian and Laird xed-eects method for
meta-analysis. ere was signicant heterogeneity (p < 0.05) for two pooled p-interactions, and for these, random
eects were incorporated into the meta-analysis; the pooled p-for interaction was 0.48 for sex and 0.66 for
Vitamin D supplement use (< 800IU) outside of the trial. None of the interaction terms for the non-pre-specied
modiers were signicant at the Bonferroni-adjusted p-value of 0.0038 (= 0.05/13 subgroup analyses): pooled p
for interaction ≥ 0.18. c Stratum-specic estimates were pooled using Dersimonian and Laird xed-eects method
for meta-analysis. ere was signicant heterogeneity (p-het < 0.05) for three strata, and for these, random eects
were incorporated into the meta-analysis and presented in the Table. For reference, the xed eects meta-
analyzed pooled estimates were: 0.004 (95% CI − 0.01, 0.02) for females (p-het = 0.03), 0.003 (95% CI − 0.02, 0.02)
for those with multiple CVD risk factors (p-het = 0.04) and 0.01 (95% CI − 0.01, 0.02) for those using Vitamin D
supplements (< 800IU) outside of the trial (p-het = 0.03). d See footnote in Table1 for denition of depression.
e Median for the global score was 0.05 in both the VITAL-Cog and the CTSC-Cog. f Compliance is dened as
taking ≥ 2/3rd of pills on all of the follow-up questionnaires between the rst and the second cognitive assessment
and not initiating out-of-study sh oil supplementation.
DELOOPCSTCGOC-LATIV
C
haracteristics Difference (95%CI)N Difference (95%CI) N Difference (95%CI)†‡§
P
re-
s
pecified Modifie
r
10.0=noitcaretnirofpecardetroper-fleS
Non-Black 0.001 (-0.01, 0.01) 260
4
)10.0,10.0-(100.0-037)20.0,40.0-(10.0-
)80.0,10.0(40.044)04.0,10.0(02.0347)70.0,200.0-(40.0kcalB
48.0=noitcaretnirofpsleveldoolbD)HO(52enilesaB
)20.0,10.0-(200.0125)40.0,40.0-(100.0-7641)20.0,20.0-(300.0)Ld/gn23(naidem<
≥ )20.0,10.0-(10.0272)40.0,70.0-(20.0-7251)30.0,10.0-(10.0)Ld/gn23(naidem
N
on-pre-specified Modifiers
tnemssessatsriftaegA
)30.0,10.0-(10.0006)40.0,40.0-(200.01871)30.0,400.0-(10.0sraey07<
≥ )20.0,20.0-(200.0-491)30.0,01.0-(40.0-3461)20.0,20.0-(1000.0sraey07
tnemngissa3-agemO
)20.0,10.0-(10.0893)30.0,70.0-(20.0-5271)30.0,10.0-(10.0oN
)20.0,10.0-(300.0693)60.0,40.0-(10.09961)20.0,20.0-(200.0seY
§xeS
Female 0.01 (-0.01, 0.03) 201
6
)40.0,70.0-(20.0-004)100.0,01.0-(50.0-
)20.0,10.0-(10.0493)70.0,10.0-(30.08041)20.0,20.0-(200.0elaM
noitacudE
High School/College -0.003 (-0.02, 0.02) 171
4
)20.0,20.0-(200.0-353)60.0,40.0-(10.0
)30.0,10.0-(10.0044)30.0,60.0-(10.0-3071)30.0,200.0-(20.0seidutsegelloc-tsoP
||noisserpeD
N
o 0.01 ( -0.01, 0.02) 274
6
)20.0,10.0-(10.0336)40.0,30.0-(200.0
Ye s 0.01 (-0.02, 0.04) 57
0
)40.0,20.0-(10.0141)11.0,01.0-(100.0
Body mass index (kg/m2)
)10.0,30.0-(10.0-832)40.0,90.0-(20.0-7011)20.0,30.0-(10.0-52<
)40.0,400.0(20.0533)70.0,30.0-(20.03231)40.0,300.0(20.092-52
≥ )20.0,30.0-(10.0-122)30.0,90.0-(30.0-909)30.0,30.0-(3000.0-03
setebaiD
No 0.01 (-0.01, 0.02) 2955 -0.0002 (-0.03, 0.03)711 0.004 (-0.01, 0.02)
)50.0,20.0-(10.028)70.0,51.0-(40.0-564)60.0,20.0-(20.0seY
noisnetrepyH
N
)20.0,10.0-(10.0914)40.0,50.0-(300.0-7551)20.0,10.0-(10.0o
Ye s 0.01 (-0.01, 0.02) 1852 )20.0,10.0-(300.0173)30.0,70.0-(20.0-
loretselohchgiH
)20.0,10.0-(10.0764)60.0,20.0-(20.09491)20.0,10.0-(10.0oN
)20.0,20.0-(200.0423)10.0,01.0-(40.0-5441)30.0,10.0-(10.0seY
Multiple CVD risk factors§
<2 risk factors 0.004 (-0.01, 0.02) 183
6
)20.0,10.0-(10.0574)50.0,30.0-(10.0
≥2 risk factors 0.01 (-0.01, 0.03) 141
4
)50.0,80.0-(20.0-603)300.0,11.0-(50.0-
§esutnemelppus3DnimatiV
)20.0,20.0-(200.0604)80.0,20.0-(30.03171)20.0,20.0-(200.0-oN
)40.0,60.0-(10.0-883)10.0,80.0-(40.0-1171)30.0,200.0-(20.0)UI008<(seY
Baseline global score
≤median of baseline score 0.01 (-0.01, 0.03) 1492 )30.0,10.0-(10.0883)60.0,40.0-(10.0
>median of baseline score -0.0001 (-0.02, 0.02) 1492 )10.0,20.0-(200.0-883)20.0,60.0-(20.0-
Compliance to treatment*
*
Compliant 0.01 (-0.01, 0.02) 295
4
)20.0,10.0-(10.0557)30.0,40.0-(400.0-
)50.0,30.0-(10.092)03.0,61.0-(70.0704)50.0,30.0-(10.0tnailpmoc-noN
Placebo better Vitamin D better
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
baseline blood 25(OH)D levels in this substudy and the main trial28, the reasons for the specic benets in Black
participants remain unclear. A future evaluation using novel biomarkers of vitamin D (e.g., vitamin D binding
protein (VDBP) or free 25(OH)D)29,30,50,51 and genetic variants for VDBP5254 that show dierence in distribution
across race/ethnicity groups may be insightful. Also, it is notable that Black participants had a higher prevalence
of diabetes and other cardiovascular risk factors, and lower baseline blood 25(OH)D levels, education and base-
line cognitive scores, which were characteristics of subgroups for which vitamin D had a suggestively stronger
Table 5. Pooled results across VITAL-Cog and CTSC-Cog for mean dierence in annual rate for the secondary
outcomes for vitamin D-Placebo: eect modication by race and blood 25(OH)D levels for cognitive decline.
3MS Modied Mini-Mental Status exam (range = 0–100)33; CI condence interval; CTSC Clinical and
Translational Science Collaborative center for VITAL in Boston, MA; TICS Telephone Interview of Cognitive
Status (range = 0–41)37. For denitions of the secondary outcomes for the two populations, see footnotes for
Table2. a From multivariable-adjusted linear mixed models of cognitive performance (model 2) as described
in footnote in Table3. b Pooled using Dersimonian and Laird xed-eects method for meta-analysis40 unless
otherwise noted. c Pooled using Dersimonian and Laird random-eects method for meta-analysis40 as the p for
heterogeneity was 0.001; if xed eects methods are used, the pooled estimate was 0.07 (95% CI 0.03, 0.12).
d Not signicant at Bonferroni-corrected p-value of 0.0167 (= 0.05/3 outcomes). e Due to the dierences in scale
between the TICS (0–41) used in VITAL-Cog and 3MS (range 0–100) used in CTSC-Cog, for pooling purposes,
the 3MS scores were multiplied by 0.41 for conversion to the same scale as the TICS scores.
Multivariable-adjusted difference in annual rate of change (Vitamin D – Placebo; 95% CI)†
SELF-REPORTED RACE BASELINE 25(OH)D BLOOD LEVELS
P
OOLED p-interactions|| p-interactions ≥0.20p-interactions≥0.12
Verbal memory composite scor
e
Non-Blac
k
< median (32 ng/dL)
V
ITAL-Cog 0.001 (-0.02, 0.02) -0.01 (-0.04, 0.02)
C
TSC-Cog 0.01 (-0.06, 0.07)0.01 (-0.06, 0.09)
P
OOLED
0.002 (-0.02, 0.02) -0.01 (-0.03, 0.02)
Blac
k
median (32 ng/dL)
V
ITAL-Cog 0.01 (-0.04, 0.06)0.03 (-0.002, 0.05)
C
TSC-Cog 0.10 (-0.17, 0.36) -0.02 (-0.12, 0.08)
P
OOLED
0.01 (-0.04, 0.07)0.02 (-0.004, 0.05)
E
xecutive function/attention composite score
Non-Blac
k
< median (32 ng/dL)
V
ITAL-Cog 0.004 (-0.01, 0.02) 0.02 (-0.01, 0.04)
C
TSC-Cog -0.03 (-0.06, 0.01) -0.01 (-0.05, 0.04)
P
OOLED
-0.002 (-0.02, 0.01) 0.01 (-0.01, 0.03)
Blac
k
median (32 ng/dL)
V
ITAL-Cog 0.06 (0.02, 0.10) -0.0004 (-0.02, 0.02)
C
TSC-Cog 0.40 (0.20, 0.59) -0.03 (-0.09, 0.03)
P
OOLED
0.21 (-0.12, 0.54)
§
-0.01 (-0.03, 0.02)
General cognitio
n
Non-Blac
k
< median (32 ng/dL)
V
ITAL-Cog 0.02 (-0.06, 0.09) -0.02 (-0.12, 0.09)
C
TSC-Cog 0.05 (-0.08, 0.18)0.01 (-0.15, 0.16)
P
OOLED
0.02 (-0.04, 0.09) -0.01 (-0.09, 0.08)
Blac
k
median (32 ng/dL)
V
ITAL-Cog 0.06 (-0.15, 0.26)0.09 (-0.02, 0.19)
C
TSC-Cog 0.02 (-0.65, 0.69)0.09 (-0.14, 0.31)
P
OOLED
0.05 (-0.14, 0.25)0.09 (-0.01, 0.18)
-0.7 00.7 -0.7 00.7
Placebo better Vitamin D better Placebo better Vitamin D better
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
Table 6. Pooled results across VITAL-Cog and CTSC-Cog for mean dierence in annual rate for the secondary
outcomes for vitamin D-Placebo: eect modication by risk factors for cognitive decline. 3MS Modied Mini-
Mental Status exam; CI condence interval; CTSC-Cog subset that received in-person interviews at the Harvard
Clinical and Translational Science Collaborative center for VITAL in Boston, MA; TICS Telephone Interview of
Cognitive Status; VITAL-Cog subset that received telephone interviews in VITAL. From multivariable-adjusted
linear mixed models of cognitive performance (model 2) as described in footnote in Table3. a For denitions
of the verbal memory and executive function scores for the two populations, see footnotes for Table2. For
general cognition, due to the dierences in scale between the TICS (0–41) and 3MS (range 0–100), for
pooling purposes, the 3MS scores were multiplied by 0.41 for conversion to the same scale as the TICS scores.
b Interaction terms across the two substudies were pooled using Dersimonian and Laird xed-eects method for
meta-analysis. For a few interactions where there was signicant heterogeneity (p < 0.05) for the estimate across
the two substudies, random eects were incorporated into the meta-analysis. Among these non-pre-specied
modiers for secondary outcomes, none of the pooled p-interactions were signicant at Bonferroni-adjusted
p-value of 0.0038 (= 0.05/13 subgroup analyses), except for three nominally signicant pooled p-interactions
for education for verbal memory (p = 0.04), diabetes for executive function/attention (p = 0.04); compliance
for general cognition (p = 0.01). c Stratum-specic estimates were pooled using Dersimonian and Laird xed-
eects method for meta-analysis. For a few strata where there was signicant heterogeneity (p < 0.05) for the
estimate across the two substudies, random eects were incorporated into the meta-analysis. For reference, the
xed eects pooled estimate for executive function/attention is 0.002 (95% CI − 0.02, 0.02) for those without
hypertension; 0.01 (95% CI − 0.01, 0.03) for those taking Vitamin D supplements (< 800IU) outside of the trial;
and the pooled estimate for general cognition is − 0.001 (95% CI − 0.08, 0.08) for those without hypertension
and 0.03 (95% CI − 0.06, 0.13) for those with multiple CVD risk factors. d For the denition of depression, see
footnote in Table1. e For the verbal memory score, the median was -0.02 standard units in VITAL-Cog and 0.02
in the CTSC-Cog; for the executive memory/attention score, the median was 0.04 in VITAL-Cog and 0.02 in
the CTSC-Cog; for TICS, the median was 34 in VITAL-Cog and for the 3MS in CTSC-Cog, the median was
96 (equivalent to 39 on the transformed variable to have the same range as the TICS). f Compliance is dened
as taking ≥ 2/3rd of pills on all of the follow-up questionnaires between the rst and the second cognitive
assessment and not initiating out-of-study sh oil supplementation.
VERBAL MEMORY
EXECUTIVE
FUNCTION/ATTENTIONGENERAL COGNITION
Char
acteristics
‡§
Difference (95%CI)Difference (95%CI)Difference (95%CI)
Age at first assess
ment
)11.0,40.0-(40.0)30.0,10.0-(10.0)40.0,10.0-(20.0sraey07<
≥ )31.0,60.0-(30.0)30.0,20.0-(300.0)20.0,30.0-(10.0-sraey07
Om
ega-3 assignment
)11.0,50.0-(30.0)20.0,20.0-(300.0)40.0,10.0-(20.0oN
)11.0,70.0-(20.0)30.0,10.0-(10.0)20.0,30.0-(300.0-seY
Sex
)11.0,60.0-(30.0)30.0,10.0-(10.0)30.0,20.0-(100.0elameF
)31.0,40.0-(40.0)20.0,20.0-(200.0-)40.0,10.0-(10.0elaM
Education
College/High School -0.01 (-0.04, 0.01)0.01 (-0.01, 0.03)-0.002 (-0.09, 0.09)
Post-graduate studies 0.03 ( 0.001, 0.05) -0.0003 (-0.02, 0.02
)0
.06 (-0.02, 0.14)
Depr
ession
||
)31.0,200.0(70.0)20.0,10.0-(10.0)30.0,10.0-(10.0oN
)60.0,72.0-(11.0-)40.0,30.0-(10.0)60.0,40.0-(10.0seY
Body ma
ss index (kg/m
2
)
)90.0,31.0-(20.0-)10.0,40.0-(20.0-)40.0,20.0-(10.052<
)12.0,30.0(21.0)50.0,10.0(30.0)40.0,20.0-(10.092-52
≥ )50.0,81.0-(70.0-)30.0,30.0-(100.0)30.0,50.0-(10.0-03
Diabetes
)01.0,30.0-(40.0)20.0,10.0-(2000.0)30.0,10.0-(10.0oN
)51.0,02.0-(20.0-)01.0,200.0(50.0)30.0,70.0-(20.0-seY
Hypertension
)22.0,51.0-(30.0)50.0,80.0-(20.0-)30.0,20.0-(10.0oN
)41.0,40.0-(50.0)30.0,10.0-(10.0)30.0,20.0-(10.0seY
High cholesterol
N
)11.0,50.0-(30.0)20.0,20.0-(300.0-)50.0,300.0-(20.0o
)31.0,60.0-(30.0)40.0,10.0-(20.0)20.0,40.0-(10.0-seY
Multiple
CVD risk factors
)01.0,50.0-(30.0)10.0,20.0-(400.0-)40.0,10.0-(20.0srotcafksir2<
≥ )22.0,62.0-(20.0-)40.0,10.0-(10.0)20.0,30.0-(10.0-srotcafksir2
V
itamin D supplement use
)60.0,21.0-(30.0-)20.0,20.0-(10.0)30.0,20.0-(400.0oN
)71.0,10.0(90.0)50.0,70.0-(10.0-)30.0,20.0-(10.0)UI008<(seY
Baselin
e score
≤median of baseline scor
e
0.01 (-0.02, 0.04)0.01 (-0.02, 0.03
)0
.04 (-0.05, 0.12)
>median of baseline scor
e
0.01 (-0.01, 0.03)-0.01 (-0.03, 0.01)0.01 (-0.06, 0.08)
Co
mpliance to treatment**
)21.0,10.0-(60.0)20.0,10.0-(200.0)30.0,10.0-(10.0tnailpmoC
)10.0,93.0-(91.0-)01.0,10.0-(50.0)30.0,80.0-(20.0-tnailpmoc-noN
Placebo better Vitamin D better Placebo better Vitamin D better Placebo better Vitamin D bet
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
benecial eect, particularly for executive function; thus, the concentration of a multitude of risk factors for
cognitive decline may have led to stronger benets of vitamin D in Black participants.
Limitations of our study warrant consideration. First, in VITAL-Cog, cognitive assessments were conducted
over the phone; however, our validation of the telephone cognitive assessment with in-person assessment showed
reasonable validity, and the main results were similar in CTSC-Cog, with in-person cognitive assessments.
While telephone interviews increased participation, it is possible that there was more misclassication in out-
come assessment and that subtle changes were missed compared to in-person assessments. Our trial included
mostly healthy, well-educated individuals (> 50% had post-graduate studies); this likely led to modest observed
cognitive decline and few participants being vitamin D decient. Both factors may have limited our ability to
detect modest eects of vitamin D3 supplements on cognition. Also, while a dose of 2000IU/day was used in
the study, it is possible that the optimal dose for brain health might be higher, although the literature has been
inconsistent43,48,55. Finally, the follow-up period of 2–3years, with only two assessments, may have been too short
to detect eects of vitamin D3 supplementation, particularly in a healthy population at relatively lower risk for
cognitive decline. Although in VITAL-Cog, we did observe cognitive decline in the placebo group over 2.8years
follow-up, additional studies with longer durations of follow-up and more cognitive assessments among those
at highest risk of vitamin D deciency and cognitive decline would be important.
Our study had several strengths. is was a randomized trial including > 4200 participants, with high rates of
follow-up and adherence to the assigned treatment group. In particular, there was a relatively high proportion of
Black participants (19%), who are at high risk for vitamin D insuciency30,5658. Also, we were able to investigate
the eect of vitamin D3 supplements on multiple cognitive domains.
In conclusion, among generally well-educated healthy adults aged 60+ years, supplementation of vitamin
D3 (2000IU/day) did not slow cognitive decline over 2–3years, although there were modest benets observed
specically in Black older adults that should be conrmed in future studies.
Methods
Study design, randomization and masking, and procedures. VITAL trial. VITAL2628 is a com-
pleted large randomized, double-blind, placebo-controlled, 2 × 2 factorial clinical trial of vitamin D3 (vitamin
D3[cholecalciferol], 2000IU/day) and marine omega-3 fatty acid (Omacor® sh oil, eicosapentaenoic acid + do-
cosahexaenoic acid, 1g/day) oral supplements in the primary prevention of cancer and cardiovascular dis-
ease. Participants were free of cancer (except non-melanoma skin cancer) and cardiovascular disease. Partici-
pants (n = 25,871 US men aged ≥ 50 and women aged ≥ 55 years) were randomized from 2011 to 2014 and
were required to limit using out-of-study supplemental vitamin D3 to ≤ 800IU/day, supplemental calcium to
1200mg/day, and to avoid using omega-3 fatty acid supplements. Supplementation with 2000IU/day vitamin
D3 for one year in VITAL led to a 40% mean increase in 25-hydroxyvitamin D (25(OH)D) levels (from 29.8
to 41.8ng/mL)28. e VITAL trial main phase has been completed, and its trial design (including details on
randomization and masking)26 and main ndings have been published27,28. e marine n-3 arm results for the
cognitive substudies have been analyzed separately59.
Participants. We used data from two distinct subsets of VITAL participants. Although cognitive function
was not the main planned outcome to be evaluated in the parent VITAL trial, assessing cognitive function was
planned before the start of the trial and baseline cognitive function assessments were planned to occur before
randomization as much as possible. One subset (VITAL-Cog; NCT01669915); n = 3424) completed cognitive
assessments by phone with randomization and again 2.8years later. Another subset (CTSC-Cog; n = 794 in an
ancillary study of depression (VITAL-DEP; NCT01696435)) completed in-person cognitive assessments with
randomization and again 2.0years later.
In VITAL-Cog, the baseline cognitive interview was conducted from September 2011 through April 2014
(mean = 1month before randomization; range of 1.2years before to 0.5years aer randomization (1.31% done
> 1month aer randomization); Fig.1a). Of 3658 eligible people as of April 2014 and we attempted to contact,
241 (7%) were unreachable, and of 3417 contacted, 3271 (96%) participated. We further excluded 262 participants
who were also in the CTSC-Cog, leaving 3009 participants (2984, including 317 Black participants, with complete
scores on all tests and 25 with scores missing on some tests). For the 2nd cognitive assessment (February 2013 to
June 2016), of the 3009 who participated in the 1st assessment, 58 died (2%) and 322 were unreachable (11%).
Of the 2629 contacted, 100 (4%) refused, and 2529 (96%) participated (2501 with complete scores on all tests
and 28 with scores missing on some tests).
To allow for enough time for follow-up assessments within the trial period and because we had reached the
target of 3000 participants, we stopped administering baseline cognitive assessments in April 2014, even though
there were additional eligible participants. However, to increase the number of Black participants, at the initiation
of 2nd assessments, we invited 618 additional eligible Black participants (Fig.1a: aged 60+ years at randomiza-
tion and willing to be part of VITAL-Cog). Of 618 Black participants, 141 (23%) could not be contacted. Of
477 contacted, 48 refused (10%) and 429 (90%) participated (November 2014 to June 2016; 422 with complete
scores on all tests; and 7 with scores missing on some tests). us, the total number of unique individuals in
VITAL-Cog was 3424: 2984 with complete baseline assessments and 2923 (= 2501 + 422 new Black participants)
with complete follow-up assessments.
In CTSC-Cog, the baseline assessment occurred from January 2012 to March 2014 (mean = 0.5month before
randomization; range of 3.0months before to within 1month aer randomization). For CTSC-Cog (Fig.1b),
we excluded 229 participants aged < 60years and four people who refused participation, leaving 821 partici-
pants (776 with complete scores on all tests and 45 with scores missing on some tests). A 2-year follow-up in-
person interview was conducted from January 2014 through April 2016. Of 821 who participated in the baseline
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
assessment, 3 died (0.4%), 217 (26%) were ineligible for VITAL-DEP due to their baseline assessments showing
neuropsychiatric disorders, 6 (1%) showed neuropsychiatric disorders and possible dementia, 55 (7%) refused;
and 540 participated (515 with complete scores on all tests and 25 with scores missing on some tests). e total
number of unique individuals in CTSC-Cog was 794 (including 44 Black participants): 776 with complete base-
line assessments and 515 with complete follow-up assessments.
Standard protocol approvals, registrations, and patient consents. e research followed the Dec-
laration of Helsinki, and this substudy protocol was approved by the institutional review board of the Brigham
and Womens Hospital. Written informed consent was obtained directly from VITAL participants and CTSC-
Cog participants or from their legally authorized representatives/next of kin26; for VITAL-Cog, completion of
cognitive tests was considered as implied consent.
Figure1. (a) Flow of Participants in the VITAL-Cog Ancillary Study to the VITAL Trial. (b) Flow of
Participants in the subset of CTSC-Cog participants in the VITAL-DEP Ancillary Study to the VITAL Trial.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
Participants and outcomes: VITAL‑Cog study population and telephone cognitive function
assessments. In VITAL-Cog substudy, the eligibility criteria were age 60+ years and in the screening ques-
tionnaire, being willing to participate in cognitive function assessments. Cognition was assessed by telephone
by trained interviewers, with eight neuropsychological tests assessing general cognition (Telephone Interview
of Cognitive status (TICS; range = 0–41 points)), verbal memory, and executive function/attention (see details
in the “Supplementary Methods”). We derived a global composite score by averaging the z-scores of the eight
individual tests (based on the baseline test distributions). When generating composite scores for the 2nd assess-
ment, the baseline means and SDs of scores from VITAL-Cog were used. Our primary, pre-specied outcome
was the annual rate of change of the global composite score, and for secondary outcomes, we also evaluated the
TICS and composite scores for verbal memory and executive function/attention (“Supplementary Methods”).
Participants and outcomes: CTSC‑Cog study population and in‑person cognitive function
assessments. A subgroup of 1054 VITAL participants received in-person health assessments, including
cognitive assessments as part of VITAL-DEP60, by trained interviewers at the CTSC in Boston with randomi-
zation (CTSC-Cog). e in-person cognitive battery included nine cognitive tests assessing general cognition
(Modied Mini-Mental State (3MS; range = 0–100)33), verbal memory and executive function/attention. e
CTSC global composite score, the primary outcome, was calculated as the average of the z-scores for the nine
assessments, using the CTSC-Cog baseline means and SDs, for both baseline and follow-up; secondary out-
comes included the 3MS and verbal memory and executive function/attention composite scores (“Supplemen-
tary Methods”).
Validation study of the VITAL‑Cog telephone cognitive assessment. Cognitive assessment by
phone has been extensively validated61,62. In VITAL-Cog, we validated our telephone cognitive assessment
against in-person assessments among a subset of 181 of the 262 CTSC participants with both assessments who
had the two within 1month of each other. We compared the global composite score derived from scores on the
eight tests administered by telephone versus a similar score derived from the nine tests administered in-person.
e intraclass correlation between the two modes was 0.64, supporting the validity of our telephone cognitive
interview (“Supplementary Methods”).
Statistical analyses. We compared characteristics at randomization by treatment group using Wilcoxons
rank-sum tests for continuous variables and chi-square tests for proportions. Primary analyses were conducted
using the intention to treat principle. For each substudy, linear mixed-eects models with random intercepts
were used to estimate the mean change in participants’ scores as a function of time (years between randomiza-
tion and each assessment), treatment assignment, and their interaction63. We tted models by maximum likeli-
hood, incorporating the longitudinal correlation within participants (using unstructured covariance structure);
for statistical testing, we used Wald tests. We calculated multivariable-adjusted mean dierences in annual rate
of decline and 95% condence intervals (CIs); information on covariates at pre-randomization were collected by
questionnaires. We used two models: model 1 included just the treatment group, while model 2 was additionally
adjusted for age at randomization (years), sex, highest attained education, race, omega-3 treatment arm assign-
ment, and depression history.
In secondary analyses, we evaluated potential eect modication by race and baseline blood vitamin D levels,
which were pre-specied given that supplementation may have stronger eects on subgroups with relatively lower
blood vitamin D levels such as Black participants29,30. We also evaluated eect modication by testing the 3-way
interaction terms in multivariable-adjusted linear mixed models for 13 possible risk factors of cognitive decline
(based on self-report on pre-randomization questionnaires): age, sex, omega-3 fatty acid assignment, education,
depression, body mass index, diabetes, hypertension, high cholesterol, multiple CVD risk factors, out-of-study
vitamin D3 supplement use, baseline score and compliance (over the entire follow-up period).
For the primary outcome of global score and for the two pre-specied subgroup analyses, the signicance tests
were 2-sided, and the signicance level was p-value < 0.05. For the secondary outcomes and subgroup analyses,
multiple comparisons were adjusted using Bonferroni corrections.
We rst evaluated associations separately by substudy and then pooled the substudy-specic results using
the Dersimonian and Laird meta-analytic approach incorporating xed-eects40. Because the TICS and 3MS
had dierent scales, for pooling, we multiplied the 3MS scores by 0.41 to generate the same scale as the TICS.
In sensitivity analyses, we restricted the analyses in both substudies to those who reported no hearing impair-
ment (68% in VITAL-Cog; 86% in CTSC-Cog), restricted the analyses to those enrolled from the 1st assessment
in VITAL-Cog (to ascertain whether missingness in the data can be assumed to be missing at random), restricted
the analyses in CTSC-Cog to those who did not have neuropsychiatric disorders or possible dementia at baseline
(72%), and restricted the analyses in both substudies to those who were in the top 90% of performance in each
outcome. In additional analyses, we additionally adjusted for practice eects by adjusting for the number of prior
assessments and in alternate models, we additionally adjusted for the season of cognitive assessment as vitamin
D levels may depend on season of the year.
For statistical analyses, we used SAS (SAS release 9.4; SAS Institute Inc, Cary, NC). For the cognitive ancillary
substudies, there was no data monitoring committee. is study is registered with ClinicalTrials.gov VITAL-Cog
(NCT01669915), VITAL-DEP (NCT01696435) and VITAL (NCT01169259).
Data availability
e corresponding author can be contacted for de-identied data requests. Analysis proposal requests will require
review and approval by the VITAL Publications & Presentations Committee and appropriate IRB approval. Once
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
approved and a data access agreement has been executed, deidentied data generated from this research will be
made available to aliated investigators through secure databases for the prespecied analysis.
Received: 1 August 2021; Accepted: 1 November 2021
References
1. Annweiler, C. et al. Vitamin D and ageing: Neurological issues. Neuropsychobiology 62, 139–150. https:// doi. org/ 10. 1159/ 00031
8570 (2010).
2. Llewellyn, D. J. et al. Vitamin D and risk of cognitive decline in elderly persons. Arch. Intern. Med. 170, 1135–1141. https:// doi.
org/ 10. 1001/ archi ntern med. 2010. 173 (2010).
3. Buell, J. S. & Dawson-Hughes, B. Vitamin D and neurocognitive dysfunction: preventing “D”ecline?. Mol. Aspects Med. 29, 415–422.
https:// doi. org/ 10. 1016/j. mam. 2008. 05. 001 (2008).
4. Slusarczyk, J. et al. Nanocapsules with polyelectrolyte shell as a platform for 1,25-dihydroxyvitamin d3 neuroprotection: Study in
organotypic hippocampal slices. Neurotox. Res. 30, 581–592. https:// doi. org/ 10. 1007/ s12640- 016- 9652-2 (2016).
5. Garcion, E., Wion-Barbot, N., Montero-Menei, C. N., Berger, F. & Wion, D. New clues about vitamin D functions in the nervous
system. Trends Endocrinol. Metab. 13, 100–105. https:// doi. org/ 10. 1016/ s1043- 2760(01) 00547-1 (2002).
6. Peterson, C. A. & Heernan, M. E. Serum tumor necrosis factor-alpha concentrations are negatively correlated with serum 25(OH)
D concentrations in healthy women. J. Inamm. (Lond.) 5, 10. https:// doi. org/ 10. 1186/ 1476- 9255-5- 10 (2008).
7. Bivona, G. et al. Non-skeletal activities of vitamin D: From physiology to brain pathology. Medicina (Kaunas) https:// doi. org/ 10.
3390/ medic ina55 070341 (2019).
8. Bivona, G. et al. Standardized measurement of circulating vitamin D [25(OH)D] and its putative role as a serum biomarker in
Alzheimer’s disease and Parkinson’s disease. Clin. Chim. Acta 497, 82–87. https:// doi. org/ 10. 1016/j. cca. 2019. 07. 022 (2019).
9. Bivona, G. et al. e role of vitamin D as a biomarker in Alzheimer’s disease. Brain Sci. https:// doi. org/ 10. 3390/ brain sci11 030334
(2021).
10. Gambino, C. M., Sasso, B. L., Bivona, G., Agnello, L. & Ciaccio, M. Aging and neuroinammatory disorders: New biomarkers and
therapeutic targets. Curr. Pharm. Des. 25, 4168–4174. https:// doi. org/ 10. 2174/ 13816 12825 66619 11120 93034 (2019).
11. Bivona, G., Gambino, C. M., Iacolino, G. & Ciaccio, M. Vitamin D and the nervous system. Neurol. Res. 41, 827–835. https:// doi.
org/ 10. 1080/ 01616 412. 2019. 16228 72 (2019).
12. Annweiler, C. et al. Dietary intake of vitamin D and cognition in older women: A large population-based study. Neurology 75,
1810–1816. https:// doi. org/ 10. 1212/ WNL. 0b013 e3181 fd6352 (2010).
13. Llewellyn, D. J., Lang, I. A., Langa, K. M. & Melzer, D. Vitamin D and cognitive impairment in the elderly US population. J. Gerontol.
A Biol. Sci. Med. Sci. 66, 59–65. https:// doi. org/ 10. 1093/ gerona/ glq185 (2011).
14. Tarcin, O. et al. Eect of vitamin D deciency and replacement on endothelial function in asymptomatic subjects. J. Clin. Endo-
crinol. Metab. 94, 4023–4030. https:// doi. org/ 10. 1210/ jc. 2008- 1212 (2009).
15. Keisala, T. et al. Premature aging in vitamin D receptor mutant mice. J. Steroid. Biochem. Mol. Biol. 115, 91–97. https:// doi. org/ 10.
1016/j. jsbmb. 2009. 03. 007 (2009).
16. Zou, J. et al. Progressive hearing loss in mice with a mutated vitamin D receptor gene. Audiol. Neurootol. 13, 219–230. https:// doi.
org/ 10. 1159/ 00011 5431 (2008).
17. Brewer, L. D., Porter, N. M., Kerr, D. S., Landeld, P. W. & ibault, O. Chronic 1alpha,25-(OH)2 vitamin D3 treatment reduces
Ca2+-mediated hippocampal biomarkers of aging. Cell Calcium 40, 277–286. https:// doi. org/ 10. 1016/j. ceca. 2006. 04. 001 (2006).
18. Holick, M. F. & Chen, T. C. Vitamin D deciency: A worldwide problem with health consequences. Am. J. Clin. Nutr. 87,
1080S-1086S. https:// doi. org/ 10. 1093/ ajcn/ 87.4. 1080S (2008).
19. Shah, I. et al. Low 25OH vitamin D2 levels found in untreated Alzheimer’s patients, compared to acetylcholinesterase-inhibitor
treated and controls. Curr. Alzheimer Res. 9, 1069–1076. https:// doi. org/ 10. 2174/ 15672 05128 03568 975 (2012).
20. Wang, L. et al. Circulating vitamin D levels and Alzheimer’s disease: A mendelian randomization study in the IGAP and UK
Biobank. J. Alzheimers. Dis. 73, 609–618. https:// doi. org/ 10. 3233/ JAD- 190713 (2020).
21. Sommer, I. et al. Vitamin D deciency as a risk factor for dementia: A systematic review and meta-analysis. BMC Geriatr. 17, 16.
https:// doi. org/ 10. 1186/ s12877- 016- 0405-0 (2017).
22. Aghajafari, F., Pond, D., Catzikiris, N. & Cameron, I. Quality assessment of systematic reviews of vitamin D, cognition and demen-
tia. BJPsych. Open 4, 238–249. https:// doi. org/ 10. 1192/ bjo. 2018. 32 (2018).
23. Dean, A. J. et al. Eects of vitamin D supplementation on cognitive and emotional functioning in young adults—A randomised
controlled trial. PLoS ONE 6, e25966. https:// doi. org/ 10. 1371/ journ al. pone. 00259 66 (2011).
24. Rossom, R. C. et al. Calcium and vitamin D supplementation and cognitive impairment in the women’s health initiative. J. Am.
Geriatr. Soc. 60, 2197–2205. https:// doi. org/ 10. 1111/ jgs. 12032 (2012).
25. Stein, M. S., Scherer, S. C., Ladd, K. S. & Harrison, L. C. A randomized controlled trial of high-dose vitamin D2 followed by
intranasal insulin in Alzheimer’s disease. J. Alzheimers Dis. 26, 477–484. https:// doi. org/ 10. 3233/ JAD- 2011- 110149 (2011).
26. Manson, J. E. et al.e VITamin D and OmegA-3 TriaL (VITAL): rationale and design of a large randomized controlled trial of
vitamin D and marine omega-3 fatty acid supplements for the primary prevention of cancer and cardiovascular disease. Contemp.
Clin. Trials 33, 159–171. https:// doi. org/ 10. 1016/j. cct. 2011. 09. 009 (2012).
27. Manson, J. E. et al. Marine n-3 fatty acids and prevention of cardiovascular disease and cancer. N. Engl. J. Med. 380, 23–32. https://
doi. org/ 10. 1056/ NEJMo a1811 403 (2019).
28. Manson, J. E. et al. Vitamin D supplements and prevention of cancer and cardiovascular disease. N. Engl. J. Med. 380, 33–44.
https:// doi. org/ 10. 1056/ NEJMo a1809 944 (2019).
29. Gutierrez, O. M., Farwell, W. R., Kermah, D. & Taylor, E. N. Racial dierences in the relationship between vitamin D, bone min-
eral density, and parathyroid hormone in the National Health and Nutrition Examination Survey. Osteoporos. Int. 22, 1745–1753.
https:// doi. org/ 10. 1007/ s00198- 010- 1383-2 (2011).
30. Powe, C. E. et al. Vitamin D-binding protein and vitamin D status of black Americans and white Americans. N. Engl. J. Med. 369,
1991–2000. https:// doi. org/ 10. 1056/ NEJMo a1306 357 (2013).
31. Canevelli, M. et al. Race reporting and disparities in clinical trials on Alzheimer’s disease: A systematic review. Neurosci. Biobehav.
Rev. 101, 122–128. https:// doi. org/ 10. 1016/j. neubi orev. 2019. 03. 020 (2019).
32. Manson, J. E. et al. Vitamin D, marine n-3 fatty acids, and primary prevention of cardiovascular disease current evidence. Circ.
Res. 126, 112–128. https:// doi. org/ 10. 1161/ CIRCR ESAHA. 119. 314541 (2020).
33. Teng, E. L. & Chui, H. C. e Modied Mini-Mental State (3MS) examination. J. Clin. Psychiatry 48, 314–318 (1987).
34. Albert, M. et al. Use of brief cognitive tests to identify individuals in the community with clinically diagnosed Alzheimer’s disease.
Int. J. Neurosci. 57, 167–178 (1991).
35. Abraham, E., Axelrod, B. N. & Ricker, J. H. Application of the oral trail making test to a mixed clinical sample. Arch. Clin. Neu-
ropsychol. 11, 697–701 (1996).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
13
Vol.:(0123456789)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
36. Bastug, G. et al. Oral trail making task as a discriminative tool for dierent levels of cognitive impairment and normal aging. Arch.
Clin. Neuropsychol. 28, 411–417. https:// doi. org/ 10. 1093/ arclin/ act035 (2013).
37. Brandt, J. & Folstein, M. F. Telephone Interview for Cognitive Status: Professional Manual (Psychological Assessment Resources,
Inc, 2003).
38. Army Individual Test Battery. Manual of Directions and Scoring. (War Department, Adjutant General’s Oce, 1944).
39. Reitan, R. M. & Wolfson, D. e Halstead-Reitan Neuropsycholgical Test Battery: erapy and Clinical Interpretation (Neuropsy-
chological Press, 1985).
40. DerSimonian, R. & Laird, N. Meta-analysis in clinical trials. Control Clin. Trials 7, 177–188. https:// doi . org/ 10. 1016/ 0197- 2456(86)
90046-2 (1986).
41. Annweiler, C. et al. Meta-analysis of memory and executive dysfunctions in relation to vitamin D. J. Alzheimers Dis. 37, 147–171.
https:// doi. org/ 10. 3233/ JAD- 130452 (2013).
42. Annweiler, C. Vitamin D in dementia prevention. Ann. NY Acad. Sci. 1367, 57–63. https:// doi. org/ 10. 1111/ nyas. 13058 (2016).
43. Bischo-Ferrari, H. A. et al. Eect of vitamin D supplementation, omega-3 fatty acid supplementation, or a strength-training
exercise program on clinical outcomes in older adults: e DO-HEALTH randomized clinical trial. JAMA 324, 1855–1868. https://
doi. org/ 10. 1001/ jama. 2020. 16909 (2020).
44. Schietzel, S. et al. Eect of 2000 IU compared with 800 IU vitamin D on cognitive performance among adults age 60 years and
older: A randomized controlled trial. Am. J. Clin. Nutr. 110, 246–253. https:// doi. org/ 10. 1093/ ajcn/ nqz081 (2019).
45. Pettersen, J. A. Does high dose vitamin D supplementation enhance cognition? A randomized trial in healthy adults. Exp. Gerontol.
90, 90–97. https:// doi. org/ 10. 1016/j. exger. 2017. 01. 019 (2017).
46. Castle, M. et al. ree doses of vitamin D and cognitive outcomes in older women: A double-blind randomized controlled trial.
J. Gerontol. A Biol. Sci. Med. Sci. 75, 835–842. https:// doi. org/ 10. 1093/ gerona/ glz041 (2020).
47. Jorde, R. et al. Vitamin D supplementation has no eect on cognitive performance aer four months in mid-aged and older subjects.
J. Neurol. Sci. 396, 165–171. https:// doi. org/ 10. 1016/j. jns. 2018. 11. 020 (2019).
48. O wusu, J. E. et al. Cognition and vitamin D in older African–American women-physical performance and osteoporosis prevention
with vitamin D in older African Americans Trial and Dementia. J. Am. Geriatr. Soc. 67, 81–86. https:// doi. org/ 10. 1111/ jgs. 15607
(2019).
49. Lipowski, M. et al. Improvement of attention, executive functions, and processing speed in elderly women as a result of involvement
in the nordic walking training program and vitamin D supplementation. Nutrients https:// doi. org/ 10. 3390/ nu110 61311 (2019).
50. Bikle, D. D. et al. Assessment of the free fraction of 25-hydroxyvitamin D in serum and its regulation by albumin and the vitamin
D-binding protein. J. Clin. Endocrinol. Metab. 63, 954–959. https:// doi. org/ 10. 1210/ jcem- 63-4- 954 (1986).
51. Brown, L. L. et al. e vitamin D paradox in Black Americans: a systems-based approach to investigating clinical practice, research,
and public health—Expert panel meeting report. BMC Proc. 12, 6. https:// doi. org/ 10. 1186/ s12919- 018- 0102-4 (2018).
52. Braun, A., Bichlmaier, R. & Cleve, H. Molecular analysis of the gene for the human vitamin-D-binding protein (group-specic
component): Allelic dierences of the common genetic GC types. Hum. Genet. 89, 401–406. https:// doi. org/ 10. 1007/ BF001 94311
(1992).
53. Kamboh, M. I. & Ferrell, R. E. Ethnic variation in vitamin D-binding protein (GC): A review of isoelectric focusing studies in
human populations. Hum. Genet. 72, 281–293. https:// doi. org/ 10. 1007/ BF002 90950 (1986).
54. Cooke, N. E. & Haddad, J. G. Vitamin D binding protein (Gc-globulin). Endocr. Rev. 10, 294–307. https:// doi. org/ 10. 1210/ edrv-
10-3- 294 (1989).
55. Byrn, M. A., Adams, W., Penckofer, S. & Emanuele, M. A. Vitamin D supplementation and cognition in people with type 2 diabetes:
A randomized control trial. J. Diabetes Res. 2019, 5696391. https:// doi. org/ 10. 1155/ 2019/ 56963 91 (2019).
56. Moore, C. E., Murphy, M. M. & Holick, M. F. Vitamin D intakes by children and adults in the United States dier among ethnic
groups. J. Nutr. 135, 2478–2485. https:// doi. org/ 10. 1093/ jn/ 135. 10. 2478 (2005).
57. Holick, M. F. Bioavailability of vitamin D and its metabolites in black and white adults. N. Engl. J. Med. 369, 2047–2048. https://
doi. org/ 10. 1056/ NEJMe 13122 91 (2013).
58. Harris, S. S. Vitamin D and African Americans. J. Nu tr. 136, 1126–1129. https:// doi. org/ 10. 1093/ jn/ 136.4. 1126 (2006).
59. Kang, J. H. et al. Marine n-3 fatty acids and cognitive change among older adults in the VITAL randomized trial.Accepted abstract
#55393 at the 2021 Alzheimer’s Association International Conference.
60. Okereke, O. I. et al. Eect of long-term vitamin d3 supplementation vs placebo on risk of depression or clinically relevant depressive
symptoms and on change in mood scores: a randomized clinical trial. JAMA 324, 471–480. https:// doi. org/ 10. 1001/ jama. 2020.
10224 (2020).
61. Evans, D. A., Grodstein, F., Loewenstein, D., Kaye, J. & Weintraub, S. Reducing case ascertainment costs in US population studies
of Alzheimer’s disease, dementia, and cognitive impairment-Part 2. Alzheimers Dement. 7, 110–123. https:// doi. org/ 10. 1016/j. jalz.
2010. 11. 008 (2011).
62. Rapp, S. R. et al. Validation of a cognitive assessment battery administered over the telephone. J. Am. Geriatr. Soc. 60, 1616–1623.
https:// doi. org/ 10. 1111/j. 1532- 5415. 2012. 04111.x (2012).
63. Fitzmaurice, G. M., Laird, N. M. & Ware, J. H. Applied Longitudinal Analysis 103–139 (Wiley, 2004).
Acknowledgements
VITAL (NCT01169259), VITAL-DEP (NCT01696435) and VITAL-Cog (NCT01669915) are registered with
ClinicalTrials.gov. VITAL-Cog was supported by R01 AG036755; VITAL-DEP was supported by R01 MH091448;
and VITAL was supported by Grants U01 CA138962, R01 CA138962, and UL1TR001102 including support from
the National Cancer Institute, National Heart, Lung and Blood Institute, Oce of Dietary Supplements, National
Institute of Neurological Disorders and Stroke, and the National Centre for Complementary and Integrative
Health. e ancillary studies are supported by grants from multiple Institutes, including the National Heart,
Lung and Blood Institute; the National Institute of Diabetes and Digestive and Kidney Diseases; the National
Institute on Aging; the National Institute of Arthritis and Musculoskeletal and Skin Diseases; the National Insti-
tute of Mental Health; and others. e funding sources had no role in the study design; the collection, analysis,
and interpretation of data; in the writing of the report; or in the decision to submit the paper for publication.
Author contributions
J.H.K., O.I.O., F.G., J.E.M. conceptualized the study design, investigation; J.H.K., O.I.O., D.D., F.G., J.E.M. curated
the data and provided project administration; J.H.K., N.R.C. and C.M.V. did the formal analysis; J.H.K., O.I.O.,
F.G., J.E.M. acquired the funding; J.H.K., O.I.O., C.M.V., S.O., J.E.M. wrote the original dra; J.H.K. and C.V.M.
accessed and veried the data, and all authors (J.H.K., C.M.V., O.I.O., S.O., M.A., I.L., D.D., J.E.B., N.R.C., F.G.,
J.E.M.) provided input on the interpretation of the data, reviewed and edited the nal submitted dra of the
manuscript.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
14
Vol:.(1234567890)
Scientic Reports | (2021) 11:23253 | https://doi.org/10.1038/s41598-021-02485-8
www.nature.com/scientificreports/
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 021- 02485-8.
Correspondence and requests for materials should be addressed to J.H.K.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is 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. e images or other third party material in this
article are included in the articles 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:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2021
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... www.nature.com/scientificreports/ (VITAL-Cog), suggested VitD3 supplementation to have no significant impact on cognitive functioning in older adults 13 . The endogenous production of VitD is greatly dependent on the geographical region in which an individual resides. ...
... According to the results of the present study, low VitD levels did not affect global cognition as evidenced from nonsignificant HMSE and ACE-III total scores, but individuals with insufficient levels of VitD showed decreased level of attention. Several previous studies have reported that VitD did not affect global cognition, neither improved cognitive functioning on supplementation 13,40,42 . Deficits in attention is a part of aging and it has been found that attention training interventions improved cognitive functioning in individuals with MCI or mild to moderate dementia 57 . ...
Article
Full-text available
Vitamin D (VitD) is a naturally occurring, fat-soluble vitamin which regulates calcium and phosphate homeostasis in the human body and is also known to have a neuroprotective role. VitD deficiency has often been associated with impaired cognition and a higher risk of dementia. In this study, we aimed to explore the relationship between levels of VitD and cognitive functioning in adult individuals. 982 cognitively healthy adults (≥ 45 years) were recruited as part of the CBR-Tata Longitudinal Study for Aging (TLSA). Addenbrooke’s cognitive examination-III (ACE-III) and Hindi mental status examination (HMSE) were used to measure cognitive functioning. 25-hydroxyvitamin D [25(OH)D] levels were measured from the collected serum sample and classified into three groups— deficient (< 20 ng/ml), insufficient (20–29 ng/ml) and normal (≥ 30 ng/ml). Statistical analysis was done using IBM SPSS software, version 28.0.1.1(15). The mean age of the participants was 61.24 ± 9 years. Among 982 participants, 572 (58%) were deficient, 224 (23%) insufficient and only 186 (19%) had normal levels of VitD. Kruskal–Wallis H test revealed a significant difference in age (p = 0.015) and education (p = 0.021) across VitD levels and the Chi-square test revealed a significant association between gender (p = 0.001) and dyslipidemia status (p = 0.045) with VitD levels. After adjusting for age, education, gender and dyslipidemia status, GLM revealed that individuals with deficient (p = 0.038) levels of VitD had lower scores in ACE-III verbal fluency as compared to normal. Additionally, we also found that 91.2% individuals who had VitD deficiency were also having dyslipidemia. It is concerning that VitD deficiency impacts lipid metabolism. Lower levels of VitD also negatively impacts verbal fluency in adult individuals. Verbal fluency involves higher order cognitive functions and this result provides us with a scope to further investigate the different domains of cognition in relation to VitD deficiency and other associated disorders.
... Hence, these results reaffirm that a deficiency in vitamin D is strongly connected to a markedly higher risk of developing all-cause dementia and Alzheimer's disease. This evidence further fuels the ongoing debate surrounding the impact of vitamin D on health conditions beyond those related to the skeletal system [68][69][70][71]. Therapeutic Potential of VDR Activation in Alzheimer's Disease As per the recent emerging trend various researches has spotlighted Vitamin D and Vitamin D Receptor (VDR), as potential therapeutic targets in AD [72]. ...
Article
Full-text available
Through antioxidant, anti-inflammatory, and neuroprotective actions, vitamin D, a fat-soluble vitamin, and its receptor, VDR, support brain health. VDR alters mechanisms associated with AD pathogenesis, including tau protein hyperphosphorylation and amyloid-beta (Aβ) metabolism, which are characteristics of the illness. According to this review, VDR activation promotes the clearance of Aβ and inhibits the enzymes that lead to tau and Aβ pathology. VDR also affects neuroinflammatory responses by inhibiting pro-inflammatory pathways and increasing anti-inflammatory cytokines. These outcomes demonstrate how VDR preserves cognitive function and synaptic plasticity by lowering oxidative stress, neuroinflammation, and neuronal death. Polymorphisms in the VDR gene have been linked to varying risks of developing AD across populations, suggesting genetic and environmental interactions in disease progression. Clinical and preclinical studies demonstrate that Vitamin D supplementation, through VDR activation, can improve cognitive outcomes, reduce Aβ deposition, and mitigate neuroinflammation. However, variability in these results necessitates further research to determine optimal dosages and therapeutic strategies. The evidence positions VDR as a promising therapeutic target for AD. Future studies should focus on unraveling the intricate pathways of VDR-related neuroprotection, exploring its genetic implications, and refining Vitamin D-based interventions to maximize therapeutic benefits for neurodegenerative diseases like Alzheimer's.
... Curcumin is one of the most extensively studied compounds (167,168), with other common treatments including quercetin (169), resveratrol (170) and luteolin (171). Despite the potential of dietary nutrients like vitamin D and omega-3 fatty acids for neuroprotection (172), findings from studies have yielded mixed results without consistent beneficial effects (173,174). Exploring the effect of nutraceuticals as treatments for neurologic disorders remains a significant area of interest. ...
Article
Full-text available
Neuroinflammation in response to environmental stressors is an important common pathway in a number of neurological and psychiatric disorders. Responses to immune-mediated stress can lead to epigenetic changes and the development of neuropsychiatric disorders. Isothiocyanates (ITC) have shown promise in combating oxidative stress and inflammation in the nervous system as well as organ systems. While sulforaphane from broccoli is the most widely studied ITC for biomedical applications, ITC and their precursor glucosinolates are found in many species of cruciferous and other vegetables including moringa. In this review, we examine both clinical and pre-clinical studies of ITC on the amelioration of neuropsychiatric disorders (neurodevelopmental, neurodegenerative, and other) from 2018 to the present, including documentation of protocols for several ongoing clinical studies. During this time, there have been 16 clinical studies (9 randomized controlled trials), most of which reported on the effect of sulforaphane on autism spectrum disorder and schizophrenia. We also review over 80 preclinical studies examining ITC treatment of brain-related dysfunctions and disorders. The evidence to date reveals ITC have great potential for treating these conditions with minimal toxicity. The authors call for well-designed clinical trials to further the translation of these potent phytochemicals into therapeutic practice.
... Several studies across diverse regions have reported that lower vitamin D is associated with cognitive impairment in the elderly (25,26), which was consistent with our hypothesis. However, other studies showed that there is no association between vitamin D status and cognitive impairment (27)(28)(29). Given the conflicting conclusions in existing research, it's plausible that the relationship between vitamin D and cognitive function is intricate, potentially influenced by mediating factors and different subgroups. ...
Article
Full-text available
Background The relationship between vitamin D levels, depressive symptoms, and cognitive function has yet to be definitively understood in the elderly, particularly when considering the impact of chronic diseases. This study focuses on how depression mediates the impact of 25-hydroxyvitamin D3 (25(OH)D3) on cognitive performance in older U.S. adults. Methods We analyzed data from 2,745 elderly individuals extracted from the NHANES 2011–2014 cycles, applying weighted processing to account for the complex multi-stage sampling design characteristic of NHANES data. Utilizing weighted data for covariate and model selection, we conducted mediation analyses on both the overall population and subgroup data. Significant mediation pathways were validated using a stratified weighted bootstrap approach. For significant subgroup pathways, we explored interactive mechanisms through interactive mediation analysis. Results Mediation analyses, thoroughly accounting for the impact of chronic conditions, revealed significant pathways in both the weighted overall population and the weighted diabetes subgroup. After 1,000 stratified weighted bootstrap replications, the proportion of mediation effects were 10.6% [0.040, 0.268] and 20.9% [0.075, 0.663], respectively. Interactive mediation analysis for diabetes indicated that the interaction between diabetes and depression was not significant in the direct pathway (estimates = 0.050, p = 0.113) but was significant in the mediation pathway, yielding the largest effect size compared to other covariates (estimates = 0.981, p < 0.001). Conclusion This study highlights the mediating role of depression in the relationship between vitamin D levels and cognitive function in the elderly, particularly emphasizing diabetes as a key moderator. Our findings suggest targeted interventions addressing both vitamin D sufficiency and depression could significantly benefit cognitive health, especially in diabetic individuals.
... Several studies across diverse regions have reported that lower vitamin D is associated with cognitive impairment in the elderly (25,26), which was consistent with our hypothesis. However, other studies showed that there is no association between vitamin D status and cognitive impairment (27)(28)(29). Given the conflicting conclusions in existing research, it's plausible that the relationship between vitamin D and cognitive function is intricate, potentially influenced by mediating factors and different subgroups. ...
... Wmedian, weighted median. [25,42,43]. How 25(OH)D may protect against AD in humans is unknown. ...
Article
Full-text available
Background Observational studies have found that vitamin D supplementation is associated with improved cognition. Further, recent Mendelian randomization (MR) studies have shown that higher vitamin D levels, 25(OH)D, may protect against Alzheimer’s disease. Thus, it is possible that 25(OH)D may protect against Alzheimer’s disease by improving cognition. Objective We assessed this hypothesis, by examining the relationship between 25(OH)D levels and seven cognitive measurements. Methods To mitigate bias from confounding, we performed two-sample MR analyses. We used instruments from three publications: Manousaki et al. (2020), Sutherland et al. (2022), and the Emerging Risk Factors Collaboration/EPIC-CVD/Vitamin D Studies Collaboration (2021). Results Our observational studies suggested a protective association between 25(OH)D levels and cognitive measures. An increase in the natural log of 25(OH)D by 1 SD was associated with a higher PACC score (BetaPACC score = 0.06, 95% CI = (0.04–0.08); p = 1.8×10⁻¹⁰). However, in the MR analyses, the estimated effect of 25(OH)D on cognitive measures was null. Specifically, per 1 SD increase in genetically estimated natural log of 25(OH)D, the PACC scores remained unchanged in the overall population, (BetaPACC score = –0.01, 95% CI (–0.06 to 0.03); p = 0.53), and amongst individuals aged over 60 (BetaPACC score = 0.03, 95% CI (–0.028 to 0.08); p = 0.35). Conclusions In conclusion, our MR study found no clear evidence to support a protective role of increased 25(OH)D concentrations on cognitive performance in European ancestry individuals. However, our study cannot entirely dismiss the potential beneficial effect on PACC for individuals over the age of 60.
Article
Metabolic syndrome (MetS) is usually associated with cognitive impairment, neuropathic pain, and reduced brain‐derived neurotrophic factor (BDNF) levels. BDNF via tropomyosin receptor kinase B (TrkB) exerts neuroprotection by activating protein kinase B (Akt) to inhibit glycogen synthase kinase‐3β (GSK3β). Although Vitamin D3 (VitD3) has demonstrated favorable metabolic and neuronal outcomes in MetS, the precise molecular mechanisms underlying its neuroprotective effects remain poorly elucidated. We aimed to test the hypothesis that VitD3 mitigates MetS‐induced cognition deficits and neuropathic pain via modulating the BDNF/TRKB/Akt/GS3Kβ signaling pathway. MetS was induced in male rats by 10% fructose‐supplemented water and 3% salt‐enriched diet. After 6 weeks, normal and MetS rats received either vehicle or VitD3 (10 µg/kg/day) for an additional 6 weeks. Glycemic status, lipid profile, and behavioral changes were assessed. The advanced glycation end products (AGEs), and markers of inflammation (TNF‐α and NF‐κB), oxidative stress (malondialdehyde), and apoptosis (caspase3), as well as BDNF, TrkB, PI3K, Akt, GSK3β, phosphorylated tau, and amyloid beta (Aβ) were assessed in the cerebral cortex. MetS rats had deteriorated glycemic and lipid profiles, higher AGEs, reduced levels of BDNF, TrkB, PI3K, and active Akt, along with increased GSK3β levels, inflammation, oxidative stress, and apoptosis. These changes were associated with higher levels of cognitive impairment markers phosphorylated tau and Aβ, as well as behavioral changes indicative of cognitive impairment and neuropathic pain. VitD3 improved the cognitive and behavioral alterations, while mitigating the associated molecular derangements. Our results indicate that VitD3 may exert neuroprotective effects by modulating the BDNF/TrkB/PI3K/Akt/GSK3β signaling pathway.
Chapter
Nutrition-related disorders and malnutrition are prevalent at all stages of dementia, and early identification and treatment are crucial elements of dementia care. Therefore, it is important to consider screening, conducting comprehensive nutritional assessments, and implementing nutrition management for early nutritional support. This chapter provides insights into modifiable causes of malnutrition, screening tools, and assessment strategies. Furthermore, the consumption of a diet that provides adequate energy, macronutrients, and micronutrients has been linked to improved cognitive function in patients with dementia. Nutrients can have positive effects on maintaining lean body mass, enhancing synaptic plasticity and cognition, and safeguarding against chronic diseases associated with dementia. Adhering to a healthy and balanced dietary pattern has been associated with a reduced risk of overall dementia. Special dietary patterns, such as the Mediterranean, Dietary Approaches to Stop Hypertension (DASH), MedDiet-DASH Intervention for Neurodegenerative Delay (MIND), and ketogenic diets, are explored as they may offer improved clinical outcomes for different forms of dementia. Lastly, this chapter addresses the management of eating behavior and feeding route options for patients with advanced dementia, along with ethical considerations.
Article
Full-text available
Background Dementia poses great health and social challenges in China. Dementia prevalence may vary across geographic areas, while comparable estimations on provincial level is lacking. This study aims to estimate dementia prevalence by provinces across China, taking into account risk factors of individual level and potential spatial correlation of provinces. Methods In this study, 17,176 adults aged 50 years or older were included from the fourth wave of the China Health and Retirement Longitudinal Study (CHARLS 2018), covering 28 provinces, autonomous regions and municipalities. To improve provincial representativeness, we constructed provincial survey weights based on China 7th census (2020). The prevalence of dementia and 95% Bayesian credible intervals (BCIs) were estimated using a Bayesian conditional autoregressive (CAR) model with spatially varying coefficients of covariates. Findings The weighted prevalence of dementia at provincial level in China in 2018 ranged from 2.62% (95%BCI: 1.70%, 3.91%) to 13.53% (95%BCI: 8.82%, 20.93%). High dementia prevalence was concentrated in North China, with a prominent high–high cluster, while provinces of low prevalence were concentrated on East and South China, characterized by a low–low cluster. Ordered by the median estimation of prevalence, the top 10% of provinces, include Xinjiang, Jilin, and Beijing. Meanwhile, Fujian, Zhejiang, and Guangdong rank among the last. The association between dementia prevalence and drinking, smoking, social isolation, physical inactivity, hearing impairment, hypertension, and diabetes exhibits provincial variation. Interpretation Our study identifies a geospatial disparity in dementia prevalence and risk factor effects across China’s provinces, with high–high and low–low clusters in some northern and southern provinces, respectively. The findings emphasize the need for targeted strategies, such as addressing hypertension and hearing impairment, in specific regions for more effective dementia prevention and treatment. Funding 10.13039/501100001809National Science Foundation of China/the 10.13039/501100000269Economic and Social Research Council, UK Research and Innovation joint call: Understanding and Addressing Health and Social Challenges for Ageing in the UK and China. UK-China Health And Social Challenges Ageing Project (UKCHASCAP): present and future burden of dementia, and policy responses (grant number 72061137003, ES/T014377/1).
Article
Full-text available
Background: Regulatory T cells (Tregs) alterations have been implicated in the pathogenesis of Multiple Sclerosis (MS). Recently, a crucial role of the X-Linked Forkhead Box P3 (FoxP3) for the development and the stability of Tregs has emerged, and FOXP3 gene polymorphisms have been associated with the susceptibility to autoimmune diseases. The expression of Foxp3 in Tregs is regulated by the transcription factor GATA binding-protein 3 (GATA3) and vitamin D3. The aim of this retrospective case-control study was to investigate the potential association between FOXP3 and GATA3 genetic variants, Vitamin D3, and MS risk. Methods: We analyzed two polymorphisms in the FOXP3 gene (rs3761547 and rs3761548) and a polymorphism in the GATA3 gene (rs3824662) in 106 MS patients and 113 healthy controls. Serum 25(OH)D3 was also measured in all participants. Results: No statistically significant genotypic and allelic differences were found in the distribution of FOXP3 rs3761547 and rs3761548, or GATA3 rs3824662 in the MS patients, compared with controls. Patients that were homozygous for rs3761547 had lower 25(OH)D3 levels. Conclusions: Our findings did not show any association among FOXP3 and GATA3 SNPs, vitamin D3, and MS susceptibility.
Article
Full-text available
Vitamin D and cognition is a popular association, which led to a remarkable body of literature data in the past 50 years. The brain can synthesize, catabolize, and receive Vitamin D, which has been proved to regulate many cellular processes in neurons and microglia. Vitamin D helps synaptic plasticity and neurotransmission in dopaminergic neural circuits and exerts anti-inflammatory and neuroprotective activities within the brain by reducing the synthesis of pro-inflammatory cytokines and the oxidative stress load. Further, Vitamin D action in the brain has been related to the clearance of amyloid plaques, which represent a feature of Alzheimer Disease (AD), by the immune cell. Based on these considerations, many studies have investigated the role of circulating Vitamin D levels in patients affected by a cognitive decline to assess Vitamin D’s eventual role as a biomarker or a risk factor in AD. An association between low Vitamin D levels and the onset and progression of AD has been reported, and some interventional studies to evaluate the role of Vitamin D in preventing AD onset have been performed. However, many pitfalls affected the studies available, including substantial discrepancies in the methods used and the lack of standardized data. Despite many studies, it remains unclear whether Vitamin D can have a role in cognitive decline and AD. This narrative review aims to answer two key questions: whether Vitamin D can be used as a reliable tool for diagnosing, predicting prognosis and response to treatment in AD patients, and whether it is a modifiable risk factor for preventing AD onset.
Article
Full-text available
Observational studies strongly supported the association of low levels of circulating 25-hydroxyvitamin D (25OHD) and cognitive impairment or dementia in aging populations. However, randomized controlled trials have not shown clear evidence that vitamin D supplementation could improve cognitive outcomes. In fact, some studies reported the association between vitamin D and cognitive impairment based on individuals aged 60 years and over. However, it is still unclear that whether vitamin D levels are causally associated with Alzheimer’s disease (AD) risk in individuals aged 60 years and over. Here, we performed a Mendelian randomization (MR) study to investigate the causal association between vitamin D levels and AD using a large-scale vitamin D genome-wide association study (GWAS) dataset and two large-scale AD GWAS datasets from the IGAP and UK Biobank with individuals aged 60 years and over. Our results showed that genetically increased 25OHD levels were significantly associated with reduced AD risk in individuals aged 60 years and over. Hence, our findings in combination with previous literature indicate that maintaining adequate vitamin D status in older people especially aged 60 years and over, may contribute to slow down cognitive decline and forestall AD. Long-term randomized controlled trials are required to test whether vitamin D supplementation may prevent AD in older people especially those aged 60 years and may be recommended as preventive agents.
Article
Full-text available
Aim: Type 2 diabetes increases the risk of cognitive decline which adversely impacts self-management of the disease. Evidence also supports a relationship between low serum 25(OH)D levels and poor cognition. The purpose of this trial was to assess vitamin D supplementation on cognitive executive functioning in persons living with type 2 diabetes. Methods: This was a double-blinded RCT where participants were randomized to receive either weekly vitamin D3 supplementation (50,000 IUs) or a matching comparator (5,000 IUs) for three months. The primary outcome was a battery of neuropsychological tests. Serum 25(OH)D was measured by liquid chromatography/tandem mass spectrometry. Repeated assessments of cognitive measures were collected over 12 weeks using alternative testing forms to minimize practice effects. Results: Thirty participants were randomized to either the low-dose allocation (n = 15) or the high-dose allocation (n = 15). Most participants were female (83%) and identified as Black (57%). For all cognition measures, there was no statistically significant finding between participants who received high-dose vitamin D supplementation and those who received low-dose supplementation. However, when assessing cognitive function in both groups over time, minimal improvement on the Symbol-Digits, the Stroop Interference Test, and the Trail Making Test Part B was observed. Conclusions: To our knowledge, this is the first randomized control trial to examine the effects of vitamin D supplementation on cognitive function in people with type 2 diabetes. However, no significant differences in cognitive outcomes between participants who received high-dose therapy and those who received low dose were found.
Article
Background High intake of marine omega‐3 (n‐3) fatty acids has been associated with lower risk of cognitive decline; however, few large‐scale, long‐term randomized trials have been conducted. We evaluated n‐3 supplementation (1 gram/day, including 840 mg of EPA + DHA) and change in cognitive function over 2‐3 years. Method Community‐dwelling participants aged 60+ years (mean[SD]=70.9[5.8]y) in the Vitamin D and Omega‐3 Trial (VITAL) were included: 3424 had cognitive function assessment by phone (VITAL‐Cog; 8 neuropsychological tests; mean follow‐up=2.8 years) and 794 were evaluated in‐person at the Harvard Clinical and Translational Science Center (CTSC‐Cog; 9 neuropsychological tests; mean follow‐up=2.0 years). The primary, pre‐specified outcome was a global composite score (average z‐scores across all tests) of change over two assessments. We used multivariable‐adjusted linear mixed models and meta‐analyzed the results for the two settings. Result We observed no significant effect of n‐3 supplementation on the global composite score: the mean difference in annual rate of change for the n‐3 versus placebo group was ‐0.01 standard units (95% CI:‐0.02,0.003) in VITAL‐Cog and ‐0.002 (95% CI:‐0.04,0.03) in CTSC‐Cog; the pooled mean difference was ‐0.01 (95%CI:‐0.02,0.003; p=0.15). For secondary outcomes of change in verbal memory, executive function/attention and general cognition, we observed null results to trends of adverse associations with n‐3 treatment versus placebo that were not statistically significant after multiple‐testing correction. In pre‐specified secondary analyses, we observed no significant interaction of the intervention with baseline plasma omega‐3 fatty acid levels for the global composite score (pooled p‐interaction=0.37). Conclusion Marine n‐3 supplementation (1 gram/day) did not confer cognitive benefits over 2‐3 years in community‐dwelling older adults.
Article
Importance The benefits of vitamin D, omega-3 fatty acids, and exercise in disease prevention remain unclear. Objective To test whether vitamin D, omega-3s, and a strength-training exercise program, alone or in combination, improved 6 health outcomes among older adults. Design, Setting, and Participants Double-blind, placebo-controlled, 2 × 2 × 2 factorial randomized clinical trial among 2157 adults aged 70 years or older who had no major health events in the 5 years prior to enrollment and had sufficient mobility and good cognitive status. Patients were recruited between December 2012 and November 2014, and final follow-up was in November 2017. Interventions Participants were randomized to 3 years of intervention in 1 of the following 8 groups: 2000 IU/d of vitamin D3, 1 g/d of omega-3s, and a strength-training exercise program (n = 264); vitamin D3 and omega-3s (n = 265); vitamin D3 and exercise (n = 275); vitamin D3 alone (n = 272); omega-3s and exercise (n = 275); omega-3s alone (n = 269); exercise alone (n = 267); or placebo (n = 270). Main Outcomes and Measures The 6 primary outcomes were change in systolic and diastolic blood pressure (BP), Short Physical Performance Battery (SPPB), Montreal Cognitive Assessment (MoCA), and incidence rates (IRs) of nonvertebral fractures and infections over 3 years. Based on multiple comparisons of 6 primary end points, 99% confidence intervals are presented and P < .01 was required for statistical significance. Results Among 2157 randomized participants (mean age, 74.9 years; 61.7% women), 1900 (88%) completed the study. Median follow-up was 2.99 years. Overall, there were no statistically significant benefits of any intervention individually or in combination for the 6 end points at 3 years. For instance, the differences in mean change in systolic BP with vitamin D vs no vitamin D and with omega-3s vs no omega-3s were both −0.8 (99% CI, –2.1 to 0.5) mm Hg, with P < .13 and P < .11, respectively; the difference in mean change in diastolic BP with omega-3s vs no omega-3s was –0.5 (99% CI, –1.2 to 0.2) mm Hg; P = .06); and the difference in mean change in IR of infections with omega-3s vs no omega-3s was –0.13 (99% CI, –0.23 to –0.03), with an IR ratio of 0.89 (99% CI, 0.78-1.01; P = .02). No effects were found on the outcomes of SPPB, MoCA, and incidence of nonvertebral fractures). A total of 25 deaths were reported, with similar numbers in all treatment groups. Conclusions and Relevance Among adults without major comorbidities aged 70 years or older, treatment with vitamin D3, omega-3s, or a strength-training exercise program did not result in statistically significant differences in improvement in systolic or diastolic blood pressure, nonvertebral fractures, physical performance, infection rates, or cognitive function. These findings do not support the effectiveness of these 3 interventions for these clinical outcomes. Trial Registration ClinicalTrials.gov Identifier: NCT01745263
Article
Importance Low levels of 25-hydroxyvitamin D have been associated with higher risk for depression later in life, but there have been few long-term, high-dose large-scale trials. Objective To test the effects of vitamin D3 supplementation on late-life depression risk and mood scores. Design, Setting, and Participants There were 18 353 men and women aged 50 years or older in the VITAL-DEP (Vitamin D and Omega-3 Trial-Depression Endpoint Prevention) ancillary study to VITAL, a randomized clinical trial of cardiovascular disease and cancer prevention among 25 871 adults in the US. There were 16 657 at risk for incident depression (ie, no depression history) and 1696 at risk for recurrent depression (ie, depression history but no treatment for depression within the past 2 years). Randomization occurred from November 2011 through March 2014; randomized treatment ended on December 31, 2017, and this was the final date of follow-up. Intervention Randomized assignment in a 2 × 2 factorial design to vitamin D3 (2000 IU/d of cholecalciferol) and fish oil or placebo; 9181 were randomized to vitamin D3 and 9172 were randomized to matching placebo. Main Outcomes and Measures The primary outcomes were the risk of depression or clinically relevant depressive symptoms (total of incident and recurrent cases) and the mean difference in mood scores (8-item Patient Health Questionnaire depression scale [PHQ-8]; score range, 0 points [least symptoms] to 24 points [most symptoms]; the minimal clinically important difference for change in scores was 0.5 points). Results Among the 18 353 randomized participants (mean age, 67.5 [SD, 7.1] years; 49.2% women), the median treatment duration was 5.3 years and 90.5% completed the trial (93.5% among those alive at the end of the trial). Risk of depression or clinically relevant depressive symptoms was not significantly different between the vitamin D3 group (609 depression or clinically relevant depressive symptom events; 12.9/1000 person-years) and the placebo group (625 depression or clinically relevant depressive symptom events; 13.3/1000 person-years) (hazard ratio, 0.97 [95% CI, 0.87 to 1.09]; P = .62); there were no significant differences between groups in depression incidence or recurrence. No significant differences were observed between treatment groups for change in mood scores over time; mean change in PHQ-8 score was not significantly different from zero (mean difference for change in mood scores, 0.01 points [95% CI, −0.04 to 0.05 points]). Conclusions and Relevance Among adults aged 50 years or older without clinically relevant depressive symptoms at baseline, treatment with vitamin D3 compared with placebo did not result in a statistically significant difference in the incidence and recurrence of depression or clinically relevant depressive symptoms or for change in mood scores over a median follow-up of 5.3 years. These findings do not support the use of vitamin D3 in adults to prevent depression. Trial Registration ClinicalTrials.gov Identifiers: NCT01169259 and NCT01696435
Article
Whether marine omega-3 fatty acid (n-3 FA) or vitamin D supplementation can prevent cardiovascular disease (CVD) in general populations at usual risk for this outcome is unknown. A major goal of VITAL (Vitamin D and Omega-3 Trial) was to fill this knowledge gap. In this article, we review the results of VITAL, discuss relevant mechanistic studies regarding n-3 FAs, vitamin D, and vascular disease, and summarize recent meta-analyses of the randomized trial evidence on these agents. VITAL was a nationwide, randomized, placebo-controlled, 2×2 factorial trial of marine n-3 FAs (1 g/d) and vitamin D 3 (2000 IU/d) in the primary prevention of CVD and cancer among 25 871 US men aged ≥50 and women aged ≥55 years, including 5106 blacks. Median treatment duration was 5.3 years. Supplemental n-3 FAs did not significantly reduce the primary cardiovascular end point of major CVD events (composite of myocardial infarction, stroke, and CVD mortality; hazard ratio [HR], 0.92 [95% CI, 0.80–1.06]) but were associated with significant reductions in total myocardial infarction (HR, 0.72 [95% CI, 0.59–0.90]), percutaneous coronary intervention (HR, 0.78 [95% CI, 0.63–0.95]), and fatal myocardial infarction (HR, 0.50 [95% CI, 0.26–0.97]) but not stroke or other cardiovascular end points. For major CVD events, a treatment benefit was seen in those with dietary fish intake below the cohort median of 1.5 servings/wk (HR, 0.81 [95% CI, 0.67–0.98]) but not in those above ( P interaction=0.045). For myocardial infarction, the greatest risk reductions were in blacks (HR, 0.23 [95% CI, 0.11–0.47]; P interaction by race, 0.001). Vitamin D supplementation did not reduce major CVD events (HR, 0.97 [95% CI, 0.85–1.12]) or other cardiovascular end points. Updated meta-analyses that include VITAL and other recent trials document coronary risk reduction from supplemental marine n-3 FAs but no clear CVD risk reduction from supplemental vitamin D. Additional research is needed to determine which individuals may be most likely to derive net benefit from supplementation. Clinical Trial Registration URL: http://www.clinicaltrials.gov . Unique identifier: NCT01169259.
Article
Chronic neuroinflammation is a common feature of the pathogenic mechanisms involved in various neurodegenerative age-associated disorders, such as Alzheimer's disease, multiple sclerosis, Parkinson’s disease, and dementia. In particular, persistent low-grade inflammation may disrupt the brain endothelial barrier and cause a significant increase of pro-inflammatory cytokines and immune cells into the cerebral tissue that, in turn, leads to microglia dysfunction and loss of neuroprotective properties. Nowadays, growing evidence highlights a strong association between persistent peripheral inflammation, as well as metabolic alterations, and neurodegenerative disorder susceptibility. The identification of common pathways involved in the development of these diseases, which modulate the signalling and immune response, is an important goal of ongoing research. The aim of this review is to elucidate which inflammation-related molecules are robustly associated with the risk of neurodegenerative diseases. Of note, peripheral biomarkers may represent direct measures of pathophysiologic processes common of aging and neuroinflammatory processes. In addition, molecular changes associated with the neurodegenerative process might be present many decades before the disease onset. Therefore, the identification of a comprehensive markers panel, closely related to neuroinflammation, could be helpful for the early diagnosis, and the identification of therapeutic targets to counteract the underlying chronic inflammatory processes.
Article
The current review provides an overview on the development of 25(OH)D measurement standardization tools over the last three decades and clarifies whether there is a role as a serum biomarker for vitamin D in neurological diseases. In the past, a lack of internationally recognized 25(OH)D reference measurement procedures and reference standard materials led to unstandardized serum total 25(OH)D results among research and clinical care laboratories. The vitamin D Standardization Program (VDSP) has been introduced in 2010 to address this problem, however, vitamin D External Quality Assessment Scheme (DEQAS) reports still show substantial sample- to- sample variability. Further, immunoassays, which are mainly used in clinical care laboratories, display analytical issues, including matrix-effects interferences, which cannot be overcome by the standardization process. Hence, liquid chromatography-tandem mass spectrometry (LC/MS-MS) methods should be used to measure 25(OH)D. Low vitamin D serum levels have been found in patients affected by Alzheimer's disease and Parkinson's disease, suggesting a role for vitamin D as a serum biomarker in these diseases. However, few studies reported 25(OH)D standardized results, thus, no clear evidence on the potential role of 25(OH)D serum levels in these diseases exists.