Next Article in Journal
Normatively Irrelevant Affective Cues Affect Risk-Taking under Uncertainty: Insights from the Iowa Gambling Task (IGT), Skin Conductance Response, and Heart Rate Variability
Next Article in Special Issue
Inherited Neuromuscular Disorders: Which Role for Serum Biomarkers?
Previous Article in Journal
Evaluating the Involving Relationships between Temperament and Motor Coordination in Early Childhood: A Prognostic Measurement
Previous Article in Special Issue
COVID-19 and Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Vitamin D as a Biomarker in Alzheimer’s Disease

by
Giulia Bivona
1,
Bruna Lo Sasso
1,2,
Caterina Maria Gambino
1,
Rosaria Vincenza Giglio
1,
Concetta Scazzone
1,
Luisa Agnello
1 and
Marcello Ciaccio
1,2,*
1
Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
2
Department of Laboratory Medicine, AOUP “P. Giaccone”, 90127 Palermo, Italy
*
Author to whom correspondence should be addressed.
Brain Sci. 2021, 11(3), 334; https://doi.org/10.3390/brainsci11030334
Submission received: 15 February 2021 / Revised: 2 March 2021 / Accepted: 2 March 2021 / Published: 6 March 2021
(This article belongs to the Special Issue Biochemical Biomarkers and Neurodegenerative Diseases)

Abstract

:
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.

1. Introduction

If one searches for the keywords “Vitamin D” and “Cognition” in Pubmed.com, one finds over 1000 articles that have been published with no break in continuity for the past 50 years. The idea of a possible link between Vitamin D metabolism and brain function has been successfully proposed and then proved by a remarkable body of data. When assessing the Vitamin D circulating levels in Mild Cognitive Impairment and Alzheimer Disease (AD) patients, an association has yet been found. Nevertheless, the attempt to use Vitamin D as a biomarker of cognitive decline systematically failed and, furthermore, Vitamin D supplementation in these patients yielded controversial results. Many reasons can explain this debacle. First, the studies assessing Vitamin D levels and its serum biomarker 25(OH)D in AD patients have some limitations (different assay methods; heterogeneity of Vitamin D cut-offs; discrepancies among the measures used to define the cognitive function), which sharply limit the robustness of findings achieved. Second, discrepancies in the cut-offs and methods used to measure 25(OH)D across the studies, due to the lack of 25(OH)D measurement standardization, made the results difficult to interpret. Third, a specific biomarker’s clinical usefulness is defined as its capability to influence clinicians to diagnose the disease, predict prognosis, and guide treatment, which is nothing Vitamin D can do. Indeed, well-established diagnostic biomarkers for AD are currently available. Hence, there is no need for a marker for diagnosis, and, on the other hand, effective treatment for AD lacks so far. Therefore, it is unclear whether Vitamin D circulating levels can impact AD patients’ outcome until a question is addressed: is AD onset preventable by reaching the optimal Vitamin D levels? Based on the available literature data, this review aims to explain why this question’s answer could be no.
Vitamin D is a steroid hormone that can be synthesized endogenously. Primarily known to regulate calcium-phosphorus metabolism, it exerts several biological activities, counting brain function and immune response regulation [1,2,3].
In humans, Vitamin D is produced in a multi-step process that involves the ultraviolet B (UVB) rays irradiation of a cutaneous compound, the 7-dehydro-cholesterol (7-DHC).
Once UVB rays act on 7-DHC, the cholecalciferol is produced, needing two sequential hydroxylation steps to form the active Vitamin D. First hydroxylation occurs in the liver, by a 25 hydroxylase generating 25(OH)D, while the second mainly depends on a renal 1,25 hydroxylase, producing 1,25(OH)2D. 1,25 hydroxylase is present within various organs and cells; thus, Vitamin D’s active form can be produced in several tissues, including the lung, brain, prostate, placenta, and immune system cells. CYP2R1, CYP3A4, and CYP27A1 enzymes have 25-hydroxylase activities, while CYP27B1 is responsible for 1,25 hydroxylation. Kidney CYP27B1 gives rise to a hormone involved in calcium-phosphorus metabolism. Non-renal active Vitamin D is implicated in regulating some cellular processes, including cell differentiation and proliferation. While CYP27B1 is regulated by the parathyroid hormone (PTH), the fibroblast growth factor (FGF23) and 1,25(OH)2D, extra-renal CYP27B1 is regulated by interferon γ (IFN-γ) and tumour necrosis factor (TNF) [4,5].
Vitamin D binding protein (VDBP) conveys both 25(OH)D and 1,25(OH)2D from the liver and kidney to other tissues, where active Vitamin D binds the nuclear Vitamin D Receptor (VDR) [3,6,7,8,9], leading to the genomic and non-genomic actions (for more details on Vitamin D genomic and non-genomic actions see reference 1).
CYP24A1 enzyme, displaying 24 hydroxylase activity, carries out Vitamin D catabolism.
Vitamin D status is typically evaluated by measuring serum 25(OH)D [9]. A consensus on which 25(OH)D levels define Vitamin D sufficiency, deficiency, and insufficiency is lacking, also due to the standardization dearth in the past decades [10]. Most of the studies performed on Vitamin D’s role in various diseases report unstandardized data, and AD is no exception. Thus, Vitamin D’s reliability as a serum biomarker in AD has been considered a debatable issue, leading to controversial opinions across the scientific community [10].

2. Vitamin D and Alzheimer Disease

A growing interest in Vitamin D role in both brain development and function in adulthood led several authors to investigate the 25(OH)D circulating levels in AD patients [11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26]. The brain displays the capability to produce and receive Vitamin D’s active form, which is deemed to support neurotransmission, synaptic plasticity, and neuroprotection [1,2,10]. From a pathophysiologic point of view, the relation between Vitamin D and AD onset and progression has been explained by impressive in vitro and in vivo studies. Given that amyloid plaques, along with neurofibrillary tangles, represent features of AD, it has been shown that 1,25(OH)2D can help the amyloid plaques phagocytosis and clearance by the innate immune cells [1,2,27,28,29,30,31]. For instance, MCI and AD patient-derived macrophages show enhanced capability to eliminate amyloid plaques after 1,25(OH)2D treatment [30], and a Vitamin D-enriched diet can decrease the number of plaques in AβPP-PS1 transgenic mice, an AD animal model [31]. Also, amyloid protein precursor (APP) metabolism involves some transcription factors, counting SMAD and transforming growth factor-beta (TGF-β), that, in turn, interact with VDR/ligand complex in the nucleus [29,32,33]. Finally, it should be considered that Vitamin D has a role in reducing cerebral microenvironment inflammation and oxidative stress, which are regarded as possible mechanisms underlying neurodegeneration and AD pathogenesis [1,10,29]. Table 1 summarises the characteristic of the studies considered.

2.1. Observational Studies on 25(OH)D Serum Levels in AD Patients

Based on these considerations, Littlejohns et al. [14] enrolled, in 2014, 1658 subjects, of which 171 developed dementia (102 AD out of 171 all-cause dementia) over a 5.6-year follow-up period. Findings revealed that subjects with 25(OH)D serum levels <25 nm/L had a two-fold risk of AD onset compared to those with >50 nm/L. Authors defined Vitamin D deficiency <50 nm/L, distinguishing between deficiency and severe deficiency (25 to 50 nmol/L and <25 nm/L, respectively). The strength of the study was the use of procedures and materials certified by NIST. Within the Rotterdam Study [15], Licher et al. evaluated the role of Vitamin D levels as a risk factor for developing AD. Authors found that subjects with vitamin D <25 nmol/L (defined as the deficiency) had an increased risk of developing dementia, compared to those with ≥50 nmol/L (sufficiency), but this finding did not achieve statistical significance. However, the longitudinal analyses (follow-up period 13.3 years) revealed that the lower the baseline 25(OH)D levels, the higher the risk of developing AD. The Licher’s study has various plus points, consisting of robust methods: for instance, the first 5 year follow-up period was excluded from the analysis to avoid reverse causation; a sensitivity analysis excluding patients with stroke was performed; each analysis was adjusted for several confounders. Nonetheless, an electrochemiluminescence binding assay was used to measure Vitamin D, while liquid chromatography-tandem mass spectrometry (LC/MS-MS) is recommended as the gold standard assay method; also, the adoption of NIST-certified procedures and materials has been not reported.
Opposite results were obtained by Ulstein et al. [23], who reported no association between vitamin D levels and AD development. To note that the Ulstein study sample size was small (73 AD patients and 63 controls). Karakis et al. [25] analyzed 1663 non-demented subjects for a 9-years follow-up period, documenting that no association exists between 25(OH)D levels and incident AD. In this study, Vitamin D deficiency, insufficiency, and sufficiency were defined as <12 ng/mL, 12 to <20 ng/mL, and 20 to <50 ng/mL, respectively.
As it can be noted, a high heterogeneity among the cut-offs used to define Vitamin D status exists, as it has been confirmed by Balion et al. [26], who documented an association between 25(OH)D concentrations and the risk of developing AD in a meta-analysis of 35,000 subjects. However, the authors highlighted remarkable discrepancies among the studies reviewed, undermining the findings obtained.
The interpretation of the studies mentioned above should take into account some considerations. First, many drawbacks weaken the results of the studies performed, including the differences among the assay methods used to measure Vitamin D, the heterogeneity among the cut-offs used to define Vitamin D deficiency and insufficiency, the lack of internationally recognized procedures and materials, and the discrepancies among the measures used to define the cognitive function.

2.2. Interventional Studies

To establish a role for Vitamin D in AD, a key question is whether AD onset is preventable by increasing Vitamin D serum levels, since diagnostic biomarkers for AD are available, and predicting prognosis and treatment response is difficult due to the lack of effective therapies [34]. Randomized controlled trials (RCTs) are suitable tools to address this question, but, unfortunately, they are few and achieved debatable conclusions [35,36,37,38,39,40,41,42,43,44,45].
Generally, it could be stated that Vitamin D supplementation failed to prevent AD onset [35,36,37,38,42,43,45,46]. It is worth mentioning Rossom et al. on 4143 older women free from dementia, receiving 400 IU or placebo, reporting a similar cognitive decline incidence between the treatment and placebo groups. Authors proved that exogenous Vitamin D has no impact on dementia development risk [37]. Although Jia et al. gained opposite findings, it should be noted that the sample size of the Jia study was smaller (210 patients) and the follow-up period short (12 months vs. 7.8 years in Rossom’s study) [39]. Some authors reported that Vitamin D could improve cognitive function combined with other compounds, like memantine [40] and medium-chain triglycerides plus L-leucine-rich amino acids [41], but also these studies had limited populations and follow-up duration. Oppositely, the Cochrane Database of Systematic Reviews has recently published the exciting findings of Rutjes et al., who performed a meta-analysis to assess the impact of vitamins supplementation on cognition in healthy individuals. Authors found no evidence of a significant influence of vitamin supplementation in the risk of cognitive decline, and, importantly, revealed that many studies reporting an effect of Vitamin D in cognitive performance had a low grade of certainty, that is a marked difference between the estimated effect and the true one [45]. In 2020, Bischoff-Ferrari et al. carried out an RCT in 1900 subjects within the DO-HEALTH RTC, evaluating the impact of Vitamin D supplement on the Montreal Cognitive Assessment (MoCA) in a 3-year follow-up. Authors conclude that Vitamin D has no impact on cognitive function improvement [42]. Although other authors gained different results in the same year [44], here again, the study sample and follow-up sharply differ between the two studies, having Bischoff-Ferrari’s RCT a larger population and a longer follow-up.
When evaluating interventional studies, the impact of AD lengthy latency period should be taken into account, which further hinders univocal interpretation of the potential role for vitamin D in this disease. Indeed, during the course of AD, modifications of the mechanisms underlying the progression occur, which increases intricacy in understanding the pathophysiology and, in turn, of the candidate risk factors of the disease.
Taken together, RCTs suggest that Vitamin D supplementation does not influence cognition, regardless of the dose of the administration [46].

3. Conclusions

There is no uncertainty that Vitamin D takes part in normal brain function, and low Vitamin D levels can occur among demented patients. However, this finding’s clinical and laboratory significance remains unclear, also due to several drawbacks of the available studies, weakening their results and hampering concluding. Taken the evidence of past and recent literature with the appropriate cautions, Vitamin D cannot be considered a reliable biomarker of AD, since measuring the biomarker does not improve diagnosis and prognosis in these patients. Also, no clear evidence on the role of low Vitamin D levels as a risk factor for the disease exists since interventional studies on this topic are few and findings are inconsistent. Preventing the onset of AD by modifying Vitamin D levels seems too good to be true.

Author Contributions

G.B. conceived and wrote the manuscript; B.L.S. and C.S. contributed to the literature research; C.M.G., R.V.G. and L.A. contributed to writing the manuscript; M.C. revised the manuscript and supervised the entire process. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bivona, G.; Gambino, C.M.; Iacolino, G.; Ciaccio, M. Vitamin D and the nervous system. Neurol. Res. 2019, 41, 827–835. [Google Scholar] [CrossRef]
  2. Bivona, G.; Agnello, L.; Bellia, C.; Iacolino, G.; Scazzone, C.; Lo Sasso, B.; Ciaccio, M. Non-Skeletal Activities of Vitamin D: From Physiology to Brain Pathology. Medicina 2019, 55, 341. [Google Scholar] [CrossRef] [Green Version]
  3. Bikle, D.; Christakos, S. New aspects of vitamin D metabolism and action—Addressing the skin as source and target. Nat. Rev. Endocrinol. 2020, 16, 234–252. [Google Scholar] [CrossRef] [PubMed]
  4. Bikle, D.D.; Patzek, S.; Wang, Y. Physiologic and pathophysiologic roles of extra renal CYP27b1: Case report and review. Bone Rep. 2018, 8, 255–267. [Google Scholar] [CrossRef]
  5. Bivona, G.; Agnello, L.; Ciaccio, M. The immunological implication of the new vitamin D metabolism. Cent. Eur. J. Immunol. 2018, 43, 331–334. [Google Scholar] [CrossRef] [PubMed]
  6. Bikle, D.D.; Schwartz, J. Vitamin D binding protein, total and free vitamin D levels in different physiological and pathophysiological conditions. Front. Endocrinol. 2019, 10, 317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Prabhu, A.V.; Luu, W.; Li, D.; Sharpe, L.J.; Brown, A.J. DHCR7: A vital enzyme switch between cholesterol and vitamin D production. Prog. Lipid Res. 2016, 64, 138–151. [Google Scholar] [CrossRef] [PubMed]
  8. Meyer, M.B.; Benkusky, N.A.; Kaufmann, M.; Lee, S.M.; Onal, M.; Jones, G.; Pike, J.W. A kidney-specific genetic control module in mice governs endocrine regulation of the cytochrome P450 gene Cyp27b1 essential for vitamin D3 activation. J. Biol. Chem. 2017, 292, 17541–17558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M. Evaluation, treatment, and prevention of vitamin D deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011, 96, 1911–1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Bivona, G.; Lo Sasso, B.; Iacolino, G.; Gambino, C.M.; Scazzone, C.; Agnello, L.; Ciaccio, M. 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 2019, 497, 82–87. [Google Scholar] [CrossRef]
  11. Duchaine, C.S.; Talbot, D.; Nafti, M.; Giguère, Y.; Dodin, S.; Tourigny, A.; Carmichael, P.H.; Laurin, D. Vitamin D status, cognitive decline and incident dementia: The Canadian Study of Health and Aging. Can. J. Public Health 2020, 111, 312–321. [Google Scholar] [CrossRef]
  12. Manzo, C.; Castagna, A.; Palummeri, E.; Traini, E.; Cotroneo, A.M.; Fabbo, A.; Natale, M.; Gareri, P.; Putignano, S. Relationship between 25-hydroxy vitamin D and cognitive status in older adults: The COGNIDAGE study. Recenti Prog. Med. 2016, 107, 75–83. [Google Scholar] [CrossRef]
  13. Lee, D.H.; Chon, J.; Kim, Y.; Seo, Y.K.; Park, E.J.; Won, C.W.; Soh, Y. Association between vitamin D deficiency and cognitive function in the elderly Korean population: A Korean frailty and aging cohort study. Medicine 2020, 99, e19293. [Google Scholar] [CrossRef]
  14. Littlejohns, T.J.; Henley, W.E.; Lang, I.A.; Annweiler, C.; Beauchet, O.; Chaves, P.H.; Fried, L.; Kestenbaum, B.R.; Kuller, L.H.; Langa, K.M.; et al. Vitamin D and the risk of dementia and Alzheimer disease. Neurology 2014, 83, 920–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Licher, S.; de Bruijn, R.F.A.G.; Wolters, F.J.; Zillikens, M.C.; Ikram, M.A.; Ikram, M.K. Vitamin D and the risk of dementia: The Rotterdam study. J. Alzheimers Dis. 2017, 60, 989–997. [Google Scholar] [CrossRef] [PubMed]
  16. Buell, J.S.; Dawson-Hughes, B.; Scott, T.M.; Weiner, D.E.; Dallal, G.E.; Qui, W.Q.; Bergethon, P.; Rosenberg, I.H.; Folstein, M.F.; Patz, S.; et al. 25-Hydroxyvitamin D, dementia, and cerebrovascular pathology in elders receiving home services. Neurology 2010, 74, 18–26. [Google Scholar] [CrossRef]
  17. Feart, C.; Helmer, C.; Merle, B.; Herrmann, F.R.; Annweiler, C.; Dartigues, J.F.; Delcourt, C.; Samieri, C. Associations of lower vitamin D concentrations with cognitive decline and long-term risk of dementia and Alzheimer’s disease in older adults. Alzheimers Dement. 2017, 13, 1207–1216. [Google Scholar] [CrossRef]
  18. Ouma, S.; Suenaga, M.; BölükbaşıHatip, F.F.; Hatip-Al-Khatib, I.; Tsuboi, Y.; Matsunaga, Y. Serum vitamin D in patients with mild cognitive impairment and Alzheimer’s disease. Brain Behav. 2018, 8, e00936. [Google Scholar] [CrossRef] [Green Version]
  19. Afzal, S.; Bojesen, S.E.; Nordestgaard, B.G. Reduced 25-hydroxyvitamin D and risk of Alzheimer’s disease and vascular dementia. Alzheimers Dement. 2014, 10, 296–302. [Google Scholar] [CrossRef] [PubMed]
  20. Ertilav, E.; Barcin, N.E.; Ozdem, S. Comparison of Serum Free and Bioavailable 25-Hydroxyvitamin D Levels in Alzheimer’s Disease and Healthy Control Patients. Lab. Med. 2020, lmaa066. [Google Scholar] [CrossRef]
  21. Aguilar-Navarro, S.G.; Mimenza-Alvarado, A.J.; Jiménez-Castillo, G.A.; Bracho-Vela, L.A.; Yeverino-Castro, S.G.; Ávila-Funes, J.A. Association of Vitamin D with Mild Cognitive Impairment and Alzheimer’s Dementia in Older Mexican Adults. Rev. Investig. Clin. 2019, 71, 381–386. [Google Scholar] [CrossRef] [Green Version]
  22. Shih, E.J.; Lee, W.J.; Hsu, J.L.; Wang, S.J.; Fuh, J.L. Effect of vitamin D on cognitive function and white matter hyperintensity in patients with mild Alzheimer’s disease. Geriatr. Gerontol. Int. 2020, 20, 52–58. [Google Scholar] [CrossRef]
  23. Ulstein, I.; Bøhmer, T. Normal Vitamin Levels and Nutritional Indices in Alzheimer’s Disease Patients with Mild Cognitive Impairment or Dementia with Normal Body Mass Indexes. J. Alzheimers Dis. 2017, 55, 717–725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Olsson, E.; Byberg, L.; Karlström, B.; Cederholm, T.; Melhus, H.; Sjögren, P.; Kilander, L. Vitamin D is not associated with incident dementia or cognitive impairment: An 18-y follow-up study in community-living old men. Am. J. Clin. Nutr. 2017, 105, 936–943. [Google Scholar] [CrossRef]
  25. Karakis, I.; Pase, M.P.; Beiser, A.; Booth, S.L.; Jacques, P.F.; Rogers, G.; DeCarli, C.; Vasan, R.S.; Wang, T.J.; Himali, J.J.; et al. Association of serum vitamin D with the risk of incident dementia and subclinical indices of brain aging: The Framingham Heart Study. J. Alzheimers Dis. 2016, 51, 451–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Balion, C.; Griffith, L.E.; Strifler, L.; Henderson, M.; Patterson, C.; Heckman, G.; Llewellyn, D.J.; Raina, P. Vitamin D, cognition, and dementia: A systematic review and meta-analysis. Neurology 2012, 79, 1397–1405. [Google Scholar] [CrossRef] [Green Version]
  27. Scazzone, C.; Agnello, L.; Ragonese, P.; Lo Sasso, B.; Bellia, C.; Bivona, G.; Schillaci, R.; Salemi, G.; Ciaccio, M. Association of CYP2R1 rs10766197 with MS risk and disease progression. J. Neurosci. Res. 2018, 96, 297–304. [Google Scholar] [CrossRef]
  28. Mizwicki, M.T.; Menegaz, D.; Zhang, J.; Barrientos-Durán, A.; Tse, S.; Cashman, J.R.; Griffin, P.R.; Fiala, M. Genomic and non genomic signaling induced by 1α,25(OH)2-vitamin D3 promotes the recovery of amyloid-β phagocytosis by Alzheimer’s disease macrophages. J. Alzheimers Dis. 2012, 29, 51–62. [Google Scholar] [CrossRef] [Green Version]
  29. Banerjee, A.; Khemka, V.K.; Ganguly, A.; Roy, D.; Ganguly, U.; Chakrabarti, S. Vitamin D and Alzheimer’s Disease: Neurocognition to Therapeutics. Int. J. Alzheimers. Dis. 2015, 2015, 192747. [Google Scholar] [CrossRef] [Green Version]
  30. Masoumi, A.; Goldenson, B.; Ghirmai, S.; Avagyan, H.; Zaghi, J.; Abel, K.; Zheng, X.; Espinosa-Jeffrey, A.; Mahanian, M.; Liu, P.T.; et al. 1α,25-dihydrox-yvitamin D3 interacts with curcuminoids to stimulate amyloid-βclearance by macrophages of alzheimer’s disease patients. J. Alzheimers Dis. 2009, 17, 703–717. [Google Scholar] [CrossRef]
  31. Yu, J.; Gattoni-Celli, M.; Zhu, H.; Bhat, N.R.; Sambamurti, K.; Gattoni-Celli, S.; Kindy, M.S. Vitamin D3-enriched diet correlates with a decrease of amyloid plaques in the brain of AβPP transgenic mice. J. Alzheimers Dis. 2011, 25, 295–307. [Google Scholar] [CrossRef] [Green Version]
  32. Carlberg, C. The concept of multiple vitamin D signaling pathways. J. Investig. Dermatol. Symp. Proc. 1996, 1, 10–14. [Google Scholar]
  33. Yanagisawa, J.; Yanagi, Y.; Masuhiro, Y.; Suzawa, M.; Watanabe, M.; Kashiwagi, K.; Toriyabe, T.; Kawabata, M.; Miyazono, K.; Kato, S. Convergence of transforming growth factor-βand vitamin D signaling pathways on SMAD transcriptional coactivators. Science 1999, 283, 1317–1321. [Google Scholar] [CrossRef] [PubMed]
  34. Agnello, L.; Piccoli, T.; Vidali, M.; Cuffaro, L.; Lo Sasso, B.; Iacolino, G.; Giglio, R.V.; Lupo, F.; Alongi, P.; Bivona, G.; et al. Diagnostic accuracy of cerebrospinal fluid biomarkers measured by chemiluminescent enzyme immunoassay for Alzheimer disease diagnosis. Scand. J. Clin. Lab. Investig. 2020, 80, 313–317. [Google Scholar] [CrossRef]
  35. 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. 2011, 26, 477–484. [Google Scholar] [CrossRef]
  36. Przybelski, R.; Agrawal, S.; Krueger, D.; Engelke, J.A.; Walbrun, F.; Binkley, N. Rapid correction of low vitamin D status in nursing home residents. Osteoporos Int. 2008, 19, 1621–1628. [Google Scholar] [CrossRef]
  37. Rossom, R.C.; Espeland, M.A.; Manson, J.E.; Dysken, M.W.; Johnson, K.C.; Lane, D.S.; LeBlanc, E.S.; Lederle, F.A.; Masaki, K.H.; Margolis, K.L. Calcium and vitamin D supplementation and cognitive impairment in the women’s health initiative. J. Am. Geriatr. Soc. 2012, 60, 2197–2205. [Google Scholar] [CrossRef] [PubMed]
  38. Moran, C.; Scotto di Palumbo, A.; Bramham, J.; Moran, A.; Rooney, B.; De Vito, G.; Egan, B. Effects of a six-month multi-ingredient nutrition supplement intervention of omega-3 polyunsaturated fatty acids, vitamin D, resveratrol, and whey protein on cognitive function in older adults: A randomized, double-blind, controlled trial. J. Prev. Alzheimers Dis. 2018, 5, 175–183. [Google Scholar] [CrossRef] [PubMed]
  39. Jia, J.; Hu, J.; Huo, X.; Miao, R.; Zhang, Y.; Ma, F. Effects of vitamin D supplementation on cognitive function and blood Aβ-related biomarkers in older adults with Alzheimer’s disease: A randomized, double-blind, placebo-controlled trial. Neurol. Neurosurg. Psychiatry 2019, 90, 1347–1352. [Google Scholar] [CrossRef] [PubMed]
  40. Annweiler, C.; Herrmann, F.R.; Fantino, B.; Brugg, B.; Beauchet, O. Effectiveness of the combination of memantine plus vitamin D on cognition in patients with Alzheimer disease: A pre-post pilot study. Cogn. Behav. Neurol. 2012, 25, 121–127. [Google Scholar] [CrossRef]
  41. Abe, S.; Ezaki, O.; Suzuki, M. Medium-Chain Triglycerides in Combination with Leucine and Vitamin D Benefit Cognition in Frail Elderly Adults: A Randomized Controlled Trial. J. Nutr. Sci. Vitaminol. 2017, 63, 133–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Bischoff-Ferrari, H.A.; Vellas, B.; Rizzoli, R.; Kressig, R.W.; da Silva, J.A.P.; Blauth, M.; Felson, D.T.; McCloskey, E.V.; Watzl, B.; Hofbauer, L.C.; et al. Effect of Vitamin D Supplementation, Omega-3 Fatty Acid Supplementation, or a Strength-Training Exercise Program on Clinical Outcomes in Older Adults: The DO-HEALTH Randomized Clinical Trial. JAMA 2020, 324, 1855–1868. [Google Scholar] [CrossRef]
  43. Jorde, R.; Kubiak, J.; Svartberg, J.; Fuskevåg, O.M.; Figenschau, Y.; Martinaityte, I.; Grimnes, G. Vitamin D supplementation has no effect on cognitive performance after four months in mid-aged and older subjects. J. Neurol. Sci. 2019, 396, 165–171. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, T.; Wang, H.; Xiong, Y.; Chen, C.; Duan, K.; Jia, J.; Ma, F. Vitamin D Supplementation Improves Cognitive Function Through Reducing Oxidative Stress Regulated by Telomere Length in Older Adults with Mild Cognitive Impairment: A 12-Month Randomized Controlled Trial. J. Alzheimers Dis. 2020, 78, 1509–1518. [Google Scholar] [CrossRef] [PubMed]
  45. Rutjes, A.W.; Denton, D.A.; Di Nisio, M.; Chong, L.Y.; Abraham, R.P.; Al-Assaf, A.S.; Anderson, J.L.; Malik, M.A.; Vernooij, R.W.; Martínez, G.; et al. Vitamin and mineral supplementation for maintaining cognitive function in cognitively healthy people in mid and late life. Cochrane Database Syst. Rev. 2018, 12, CD011906. [Google Scholar] [CrossRef] [PubMed]
  46. Bode, L.E.; McClester Brown, M.; Hawes, E.M. Vitamin D Supplementation for Extraskeletal Indications in Older Persons. J. Am. Med. Dir. Assoc. 2020, 21, 164–171. [Google Scholar] [CrossRef] [PubMed]
Table 1. Characteristics of studies included in the analysis of vitamin D deficiency and the risk developing Alzheimer Disease.
Table 1. Characteristics of studies included in the analysis of vitamin D deficiency and the risk developing Alzheimer Disease.
Author & Publication YearRef.Study TypeNo. Patients (Total)Follow-Up DurationVitamin D Deficiency Cut-OffVitamin D Assessment MethodUse of Procedure NISTConclusion
Afzal, 2014, Denmark[19]Prospective1018630 years25 nmol/LECLIANot reportedLower vitamin D concentrations increase the risk of developing AD
Aguilar-Navarro, 2019, Mexico[21]Cross-sectional208Not reported20 ng/mLCMIANot reportedVitamin D deficiency is associated with AD
Buell, 2010, France[16]Cross-sectional318Not reported10 ng/mLRIANot reportedVitamin D deficiency is associated with AD
Duchaine, 2020, Canada[11]Prospective6615.4 years50 nmol/LCLIANot reportedNo association between 25(OH)D and AD
Feart, 2017, France[17]Prospective91612 years25 nmol/LCMIANot reportedAssociation between lower vitamin D concentrations and increased risk of AD
Karakis, 2016,[25]Prospective 16639 years12 ng/mLRIANot reportedNo associations between vitamin D levels and incident of AD
Lee, 2020, Korea[13]Prospective2990Not reported10 nmol/LCMIANot reportedNo direct correlation between VitD deficiency and cognitive impairment
Licher, 2017, Netherlands[15]Prospective622013.3 years25 nmol/LECLIANot reportedLower vitamin D concentrations increase the risk of developing AD
Littlejohns, 2014, US[14]Prospective16585.6 years50 nmol/LLC-MSSRM certified by NISTVitamin D deficiency increases the risk of developing AD
Manzo, 2016, Italy[12]Cross-sectional132Not reported10 ng/mLNot reportedNot reportedNo association between vitamin D deficiency and cognitive impairment
Olsson, 2017, Sweden[24]Prospective118218 years50 nmol/LHPLC-MSNot reportedNo association between baseline vitamin D status and long-term risk of dementia
Shih, 2020, China[22]Cross-sectional146Not reported20 ng/mLRIANot reportedReduced serum 25(OH)D levels are associated with lower MMSE scores in patients with mild AD
CLIA: Chemiluminescence-immunoassay; CMIA: ChemiluminescentMicroparticle immunoassay; ECLIA: electrochemiluminescent immunoassay; HPLC: High-performance liquid chromatography-mass spectrometry; LC-MS: Liquid chromatography tandem mass spectrometry; MMSE: Mini-Mental State Examination; NIST: National Institute for Standard and Technology; RIA: Radioimmunoassay; SRM:standard reference materials.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bivona, G.; Lo Sasso, B.; Gambino, C.M.; Giglio, R.V.; Scazzone, C.; Agnello, L.; Ciaccio, M. The Role of Vitamin D as a Biomarker in Alzheimer’s Disease. Brain Sci. 2021, 11, 334. https://doi.org/10.3390/brainsci11030334

AMA Style

Bivona G, Lo Sasso B, Gambino CM, Giglio RV, Scazzone C, Agnello L, Ciaccio M. The Role of Vitamin D as a Biomarker in Alzheimer’s Disease. Brain Sciences. 2021; 11(3):334. https://doi.org/10.3390/brainsci11030334

Chicago/Turabian Style

Bivona, Giulia, Bruna Lo Sasso, Caterina Maria Gambino, Rosaria Vincenza Giglio, Concetta Scazzone, Luisa Agnello, and Marcello Ciaccio. 2021. "The Role of Vitamin D as a Biomarker in Alzheimer’s Disease" Brain Sciences 11, no. 3: 334. https://doi.org/10.3390/brainsci11030334

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop