Skip to main content
American Journal of Alzheimer's Disease and Other Dementias logoLink to American Journal of Alzheimer's Disease and Other Dementias
. 2013 Jan 15;28(2):126–136. doi: 10.1177/1533317512473196

The Role of Vitamin D in Alzheimer’s Disease

Possible Genetic and Cell Signaling Mechanisms

Khanh Vinh Quốc Lương 1,, Lan Thi Hoàng Nguyễn 1
PMCID: PMC10852937  PMID: 23322908

Abstract

Alzheimer’s disease (AD) is the most common form of dementia in the elderly individuals and is associated with progressive memory loss and cognitive dysfunction. A significant association between AD and low levels of vitamin D has been demonstrated. Furthermore, vitamin D supplements appear to have a beneficial clinical effect on AD by regulating micro-RNA, enhancing toll-like receptors, modulating vascular endothelial factor expression, modulating angiogenin, and advanced glycation end products. Vitamin D also exerts its effects on AD by regulating calcium-sensing receptor expression, enhancing amyloid-β peptides clearance, interleukin 10, downregulating matrix metalloproteinases, upregulating heme oxygenase 1, and suppressing the reduced form of nicotinamide adenine dinucleotide phosphate expression. In conclusion, vitamin D may play a beneficial role in AD. Calcitriol is the best vitamin D supplement for AD, because it is the active form of the vitamin D3 metabolite and modulates inflammatory cytokine expression. Therefore, further investigation of the role of calcitriol in AD is needed.

Keywords: calcitriol, vitamin D, Alzheimer’s disease, dementia

Introduction

Alzheimer’s disease (AD) is the most common form of dementia in elderly individuals and is associated with progressive memory loss and cognitive dysfunction. There is a high prevalence of dementia in patients with low serum 25-hydroxyvitamin D3 (25OHD) levels. 1 In a 7-year longitudinal study, serum vitamin D deficiency predicted non-Alzheimer’s dementias in older women. 2 In addition, higher vitamin D dietary intake was associated with a lower risk of AD. 3 In a cross section of older adults, vitamin D deficiency was associated with low mood and impaired cognitive performance. 4 In older African Americans, vitamin D deficiency was also associated with impaired cognitive performance and low bone density of the hip. 5 A significant prevalence of vitamin D insufficiency has been reported in patients with AD. 6 Elderly women diagnosed with AD have an increased prevalence of vitamin D deficiency, 7 and this deficiency is also associated with low mood and impaired cognitive performance in older adults. 8 There is an association between Mini-Mental State Examination (MMSE) test scores and serum 25OHD levels. Vitamin D-sufficient patients had significantly higher MMSE scores compared to vitamin D-insufficient patients. 9 In another study, vitamin D deficiency was associated with increased odds of cognitive impairment in the elderly population of the United States. 10 A vitamin D-free regimen intensified the spatial learning deficit in AD animal models. 11 Long-term treatment with calcitriol resulted in a higher density of CA1 neurons in the middle regions of the hippocampus in aging rats, 12 which suggests that vitamin D can modulate the biological markers of brain aging. The membrane-associated rapid response steroid-binding receptor (1,25D3-MARRS) was reported to be expressed on the cell surface and to mediate the rapid response of calcitriol. 13 Diosgenin derivative, a plant-derived steroidal sapogenin, significantly reduced amyloid deposits and improved memory dysfunction in amyloid β peptide (Aβ)1-42-infused AD model rats. 14 Diosgenin also exhibited significantly improved performance of object recognition memory in AD model mice and significantly reduced amyloid plaques and neurofibrillary tangles in the cerebral cortex and hippocampus. 15 The 1,25D3-MARRS was shown to be a target of diosgenin; 1,25D3-MARRS knockdown completely inhibited diosgenin-induced axonal growth in cortical neurons and treatment with a neutralizing antibody against 1,25D3-MARRS diminished the axonal regeneration effect of diosgenin in Aβ1-42-induced axonal atrophy. 15 These reports suggest that the exogenous stimulator diosgenin activates the 1,25D3-MARRS pathway, which may be a very critical signaling target for anti-AD therapy. In our previous publication, genetic studies identified a number of proteins that link vitamin D to AD pathology; vitamin D can also act in AD through a number of nongenomic mechanisms. 16 In the present article, we will further discuss the relationship between vitamin D and AD.

The Role of Vitamin D in AD

Many studies have suggested a role for vitamin D in brain development and function. The expression of the vitamin D receptor (VDR) occurs early in the developing rodent brain.17,18 Both VDR and 1α-hydroxylase, the enzyme responsible for the formulating of active vitamin D in the human brain, were found in both neurons and glial cells in a regional and layer-specific pattern; VDR was restricted to the nucleus and 1α-hydroxylase was distributed throughout the cytoplasm. 19 These findings suggest that vitamin D may have autocrine/paracrine properties in the human brain and plays a role in cellular development within the nervous system. In patients with AD, VDR expression reportedly reduced in different layers of the hippocampus, 20 which is more vulnerable to degeneration in AD. Variations at the VDR locus have been associated with susceptibility and progression to several diseases. The VDR gene variants influence the susceptibility to age-related changes in cognitive functioning. 21 In Baltimore Longitudinal Study of Aging, sex-specific VDR gene variation can modify age-related cognitive decline among the US adults. 22 The VDR polymorphisms of Apa1 and Taq1 were associated with the AD risk, particularly in people under the age of 75. 23 Wang et al 24 also demonstrated an association between VDR polymorphisms and the risk of late-onset AD. The frequency of “TaubF” haplotype (alleles of TaqI, ApaI, Tru9I, BsmI, and FokI, respectively), which was determined by analyzing 5 polymorphisms together, was significantly higher in the patient group with AD compared to the controls. 25

Micro-RNAs (miRNAs) are small, noncoding, single-stranded RNA molecules that exist in cells and body fluids 26 and play important roles in the pathogenesis of various diseases.27,28 There is evidence that alterations in miRNA expression play a key role in AD pathogenesis. The miRNA-146a binds complementary to the 3’-untranslated region (3’-UTR) of complement factor H (CFH), an important repressor of the inflammatory response of the brain. Upregulation of miRNA-146a coupled to downregulation of CFH was observed in AD brain as a function of disease progression.29,30 A strong positive correlation between miRNA-125b abundance and the glial cell marker glial fibrillary acidic protein (GFAP) was noted in brains with advanced AD. 31 An analysis of the temporal-lobe neocortex-derived extracellular fluid and the cerebrospinal fluid (CSF) of patients with AD showed the abundance of miRNA, including proinflammatory miRNA-146a and miRNA-155 from stressed human primary neural cells. 32 The presence of stable miRNAs in CSF at high enough concentrations to transmit inflammatory signaling in AD and that miRNA bioactivities may extend well outside of the cell cytoplasm to further mediate homeostatic or pathogenic interneural communication. In another study, the levels of urinary miRNA-146a in patients with systemic lupus erythematosus (SLE) were significantly higher than those of the healthy controls. Calcitriol generally reduces the levels of urinary miRNA-146a, which significantly correlates with the estimated glomerular filtration rate in patients with SLE. 33 Serum miRNA-155 levels have also been reported to be lower in patients with SLE than in healthy controls, 33 although the urinary levels were significantly higher in the patients with SLE than the controls. Calcitriol also reduces the levels of urinary miRNA-155, which significantly correlate with proteinuria and the SLE disease activity index. 34 Furthermore, many additional studies have documented the relationship between vitamin D and miRNAs. Plasma miRNA-146 levels changed significantly after supplementation with high doses of vitamin D in healthy males compared to placebo. 35 Moreover, VDR 3′-UTR-luciferase activities decreased 40% to 50% in human embryonic kidney 293 cells after transfection with pS-miR-27b or pS-mmu-miR-298. 36 Human vitamin D3 hydroxylase (CYP24), which catalyzes the inactivation of calcitriol, is posttranscriptionally regulated by miRNA-125b. 37 A potential miRNA-125b recognition element was identified in the 3′-UTR of human VDR messenger RNA (mRNA). 38 Fifteen miRNAs are known to be differentially regulated by calcitriol in prostate cancer cells, 39 and the endogenous level of VDR mRNA is inversely correlated with the expression of miRNA-125b in melanoma cell lines. 40 These findings suggest that calcitriol may have an effect on AD by regulating the expression of miRNAs.

Toll-like receptors (TLRs) are a group of glycoproteins that function as surface transmembrane receptors. These receptors are involved in the innate immune response to exogenous pathogenic microorganisms. Substantial evidence exists supporting the important role of TLRs in the pathogenesis and outcome of AD. The TLR-2 and TLR-4 expression is increased in the peripheral blood mononuclear cells (PBMCs) of patients with AD. 41 In an AD model, fibrillary Aβ were TLR-4-dependent and stimulated microglial activation and the upregulation of cytokines, such as tumor necrosis factor (TNF)-α, IL-1β, IL-10, and IL-17.42,43 The TLR-2 is a primary receptor that mediates the neuroinflammatory activation triggered by Aβ.44,45 In mice, active immunization with Aβ impairs memory performance via the TLR-2/4-dependent activation of the innate immune system. 46 The TLR-4 signaling increases the vulnerability of neurons to damage by Aβ and oxidative stress in AD. 47 These findings suggest that TLR2/4 signaling increases the vulnerability of neurons to Aβ and oxidative stress in AD. Furthermore, variations at the TLR locus have been associated with susceptibility and progression to AD. The TLR-2 polymorphisms are associated with AD in Han Chinese.48,49 The TLR-4 polymorphisms are associated with AD in Han Chinese and Italian populations and in a mouse model of AD.5053 Soscia et al 54 demonstrated that whole AD brain homogenates have significantly higher antimicrobial activity (antimicrobial peptide [AMP]) than age-matched non-AD samples and that AMP action correlated with Aβ-positive tissue. High levels of AMP (LL-37) are also associated with atherosclerotic plaques and induce death in vascular smooth muscle cells. 55 In contrast, vitamin D deficiency increases the expression of hepatic mRNA levels of TLR-2, TLR-4, and TLR-9 in obese rats. 56 However, calcitriol suppresses the expression of TLR-2 and TLR-4 mRNA and protein in human monocytes and triggers hyporesponsiveness to pathogen-associated molecular patterns. 57 Calcitriol has also been shown to downregulate intracellular TLR-2, TLR-4, and TLR-9 expression in human monocytes. 58 Interestingly, the TLR activation upregulated the expression of VDR and 1α-vitamin D hydroxylase in human monocytes. 59 In addition, calcitriol can increase the vitamin D-induced expression of cathelicidin in bronchial epithelial cells 60 and may enhance cathelicidin LL-37 production. 61 The addition of a VDR antagonist has been reported to inhibit the induction of cathelicidin mRNA by more than 80%, which consequently reduces protein expression of this antimicrobial agent by approximately 70%. 60 Taken together, vitamin D may have a role in patients with AD by enhancing TLR pathways.

Angiogenesis is a complex process that involves the coordinated steps of endothelial cell activation, proliferation, migration, tube formation, and capillary sprouting. In addition, angiogenesis requires the participation of intracellular signaling pathways. Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis. Pathological angiogenesis could be a key event in the pathogenesis of AD. The abnormal regulation of VEGF expression has been reported in AD pathogenesis. Homogenates in the brains of APP23 mice, a transgenic model of AD, induced the formation of new vessels during in vivo angiogenesis, and could be blocked by a VEGF antagonist. 62 Angiopoietin 2 and VEGF are highly expressed in the microcirculatory system of patients with AD compared to controls. 63 Enhanced VEGF immunoreactivity in clusters of reactive astrocytes was found in the neocortex of patients with AD but not in elderly controls. 64 Increased VEGF levels in CSF were observed in patients with AD and vascular dementia compared to healthy controls. 65 These findings suggest that angiogenic changes occur in the microcirculation of the AD brain and may contribute to disease pathogenesis. The VEGF interacts with Aβ and coaccumulates with Aβ in the brains of patients with AD. 66 The Aβ also inhibits the VEGF-induced migration of endothelial cells, as well as VEGF-induced permeability in an in vitro model of the blood brain barrier. 67 The VEGF gene variability was suggested as genetic factors influencing the lifespan in the cohort of Italian. 68 The VEGF polymorphisms have been associated with AD in Italian, Han Chinese, and Tunisian populations.6972 Interestingly, calcitriol has been reported to inhibit angiogenesis both in vitro and in vivo. 73 Calcitriol significantly inhibits VEGF-induced endothelial cell spouting and elongation in a dose-dependent manner and decreases the production of VEGF. 74 Calcitriol has also been shown to be a potent inhibitor of retinal neovascularization in a mouse model of oxygen-induced ischemic retinopathy. 75 In addition, vitamin D and its analogs have been shown to reduce VEGF expression in various cancer cell lines.76,77 These findings suggested that vitamin D may modulate VEGF expression in AD.

Angiogenin is endocytosed by subconfluent endothelial cells, translocated to the nucleus, accumulates in the nucleolus, and is a potent inducer of neovascularization. 78 Angiogenin protects cultured motor neurons against excito-toxic injury, whereas the knockdown of angiogenin levels potentiated excitotoxic motorneuron death. 79 The patients with AD exhibited lower serum angiogenin and higher VEGF levels than control participants. Cognitive function was positively correlated with serum levels of angiogenin. 80 Mouse and human angiogenin are induced during inflammatory process. 81 These findings implicate angiogenin may be involved in the pathogenesis of AD. However, vitamin D-deficient mice showed significantly decreased angiotensin and impaired colonic antibacterial activity, and were predisposed to colitis. 82 Lagishetty et al 82 also revealed that putative vitamin D response elements are located within the promoter for the angiogenin gene. Taken together, vitamin D may modulate angiogenin in AD.

Glyoxalase 1 (Glyo-1) catalyzes the first and rate-limiting step of methylglyoxal (MG) removal, which is the major precursor to advanced glycation end product (AGE) formation. The AGEs are a heterogeneous group of macromolecules that are formed by the nonenzymatic glycation of proteins, lipids, and nucleic acids. The receptors for AGE (RAGEs) are multiligand receptors, and their ligands are also likely to recognize several receptors while mediating their biological effects. 83 Glyoxalase I-specific anti-serum revealed many intensely stained flame-shaped neurons in AD brains compared to brains from controls without dementia. 84 Moreover, Glyo-I protein levels increase 1.5 times in patients with early AD and continuously decrease in the intermediate and late stages of AD. 85 The accumulation of AGEs occurs in the brain during the aging process and might be involved in the pathogenesis of AD. The level of CSF AGEs was 1.7 times higher in patients with AD than in a normal age-matched group. 86 The plasma AGE levels were almost twice as high in patients with AD as in controls. 87 In AD, most astrocytes contained both AGE- and RAGE-positive granules and their distribution was almost the same. 88 Micro-vascular RAGE levels increase in conjunction with the onset of AD and continue to increase linearly as a function of the pathologic severity of AD. 89 The Aβ has been demonstrated to serve as a binding ligand for RAGE, which triggers a cascade of events leading to the generation of reactive oxygen species (ROS) in microglial cells.90,91 Variations at the RAGE locus have been associated with susceptibility and progression to AD. There is an association between the RAGE G82S polymorphism and AD in Chinese and European populations.92,93 Anti-RAGE and anti-Aβ IGs correlate strongly with global scores of dementia. 94 These findings suggest potential for anti-Aβ and anti-RAGE immunoglobulins G as blood biomarkers for AD. Moreover, high levels of AGEs are thought to play a role in the development of chronic vasculopathies in advanced chronic kidney disease (CKD). The addition of calcitriol normalized the expression, secretion, and activity of RAGEs and IL-6 in cultured endothelial cells that were incubated in a CKD-like environment. 95 Calcitriol may act as a vascular protective agent counteracting the deleterious effects of AGEs on endothelial cell activities. 96 Calcitriol also decreased the expression of adhesion molecules, as well as the lipopolysaccharide (LPS)-induced mRNA expression of RAGEs and IL-6. In addition, calcitriol blunted the proatherosclerotic parameters via nuclear factor κB (NF-κB) and p38 in vitro. 97 These findings suggest that vitamin D may protect against the delirious effects of AGEs in AD.

The calcium-sensing receptor (CaSR) is a G-protein-coupled, transmembrane receptor that responds to changes in Ca2+ levels. The CaSR plays a central role in calcium homeostasis, primarily by regulating parathyroid hormone (PTH) secretion and renal tubular calcium reabsorption,98,99 and CaSR is also expressed in the brain, particularly in parts of the hippocampus, hypothalamus, and cerebellum. 100 Calcium dysregulation increases the susceptibility to neuronal cell damage, and CaSR is altered in patients with AD. In AD brains, the intracellular immunostaining for the neuronal calcium sensor proteins, vicilin-like protein 1 and -3 (VILIP-1 and -3), was reduced compared to the controls. 101 Also, significantly fewer VILIP-1-immunoreactive neurons were found in the temporal cortex of patients with AD compared to the controls. 101 The VILIP-1 expression enhanced the hyper-phosphorylation of tau protein compared to the nontransfected or calbindin-D28K-transfected cells and was associated with amyloid plaques and fibrillary tangles in AD brains. 102 The Aβ deposited on hippocampal neurons in AD overactivated CaSR, resulting in neuronal dysfunction.103,104 These morphological and neurochemical findings suggest an involvement of these neuronal calcium sensor proteins in pathology and possibly pathophysiology of changed calcium homeostasis in AD. In CaSR-null mice, CaSR activation was reported to be critical for the expansion and differentiation of oligodendrocyte progenitor cells. 105 The CaSR polymorphism was significantly associated with AD susceptibility, particularly in individuals without the Apolipoprotein E4 allele. 106 These findings demonstrate that the CASR has a role in AD susceptibility. However, CaSR expression is upregulated by calcitriol in the kidney, parathyroid gland, adipocyte, and in genetic hypercalciuric stone forming rats.107110 In addition, calcitriol regulates CaSR in human colorectal cancer. 111 Taken together, vitamin D may have a role in AD by regulating CaSR expression.

The Aβ is produced in excess in AD and contribute to neuronal dysfunction and degeneration. The Aβ triggers neurodegeneration not only by inducing L-type voltage-sensitive calcium channels expression and nerve growth factor (NGF) levels but also by suppressing VDR expression. 112 Administration of vitamin D to animal AD model protected neurons by preventing cytotoxicity and apoptosis, and also by downregulating LVSCC A1C and upregulating VDR. 113 In patients with AD, PBMCs are defective in the phagocytosis and degradation of Aβ. 114 The transcription of MGAT3 stimulated Aβ and distinguished macrophages into types 0-2. In vivo, the immune cell function was improved by calcitriol in both type I and type II macrophages.114117 In both type I and type II macrophages, calcitriol strongly stimulated Aβ phagocytosis and clearance while protecting against apoptosis. 117 In aged mice, vitamin D administration significantly reduced the levels of Aβ accumulation in the retinal inflammation and in the numbers of retinal macrophage and marked a shift in macrophage morphology. 118 The overexpression of VDR or vitamin D treatment suppressed the amyloid precursor protein (APP) transcription in neuroblastoma cells. 24 A vitamin D-enriched diet is correlated with a decrease in the number of amyloid plaques, a decrease in Aβ, a decrease in inflammation, and an increase in NGF in the brains of Aβ precursor protein transgenic mice. 119 These observations suggest that a vitamin D3-enriched diet may benefit patients with AD. Calcitriol also enhances the cerebral clearance of human Aβ across the blood–brain barrier of mouse brains and appears to enhance brain-to-blood Aβ1-40 efflux transport by both genomic and nongenomic actions. 120 Taken together, vitamin D may benefit to AD by enhancing Aβ clearance.

It is known that inflammation plays an important role in the pathogenesis of AD. Certain proinflammatory cytokines and chemokines are closely related to cognitive dysfunction and neurodegeneration. Interleukin-10 (IL-10) is an anti-inflammatory cytokine that may modulate disease progression. Compared with the controls, IL-10 production was reduced in patients with AD after stimulating the PMBC with Aβ. 121 In blood cells stimulated with LPS, IL-10 release was reduced in patients with multiinfarct dementia compared to normal participants. 122 The IL-10 enhances neurogenesis and cognitive function in APP + PS1 transgenic mice. 123 The findings suggest that decreased IL-10 production point to a reduction of suppressor cell function in patients with AD. Positive associations between AD and the IL-10 polymorphism are reported in Chinese and Italian populations.124,125 The homozygous A allele of the -1082 polymorphism of IL-10 increased the risk of AD and reduced the IL-10 generation in peripheral cells after amyloid cleavage. 126 A systemic meta-analysis suggested an association between IL-19 polymorphisms and AD.127,128 These reports suggest that polymorphisms of cytokine genes can affect neurodegeneration. Notably, IL-10 signaling is essential for the calcitriol-mediated inhibition of experimental autoimmune encephalomyelitis. 129 In addition, the genes encoding the cytokines IL-2, IL-19, and IL-12B are primary calcitriol target genes. 130 Note that low vitamin D status is associated with low cord blood levels of the immune-suppressive cytokine IL-10. 131 Calcitriol and calcipotriol have been shown to induce IL-10 receptor gene expression in human epidermal cells, 132 and calcitriol directly affects Treg cell growth and promotes IL-10 production. 133 Vitamin D treatment downregulated TLR-4, TLR-5, and triggering receptor expressed on myeloid cells (TREM)-2; upregulated Toll interacting protein, CD-80, TREM-1, and the anti-inflammatory cytokine IL-10 in human myometrial cells. 134 Taken together, vitamin D may have a role in AD by modulating IL-10.

Matrix metalloproteinases (MMPs) are proteolytic enzymes that are responsible for remodeling the extracellular matrix and regulating the leukocyte migration through the extracellular matrix. This migration is an important step in inflammatory and infectious pathophysiology. The MMPs are produced by many cell types, including lymphocytes, granulocytes, astrocytes, and activated macrophages. There is growing evidence that MMPs play an important role in the pathogenesis of AD. Leake et al 135 identified an approximately 50% increase in the cortex levels of MMP-1 in AD. This finding is consistent with the presence of an inflammatory state within the brain in AD and contributes to the blood–brain barrier dysfunction seen in AD. Plasma MMP-3 was significantly elevated in patients with AD. 136 The MMP-3 was expressed predominantly in white matter of brain and was also expressed in senile plaques in the cortices of patients with AD. 137 The MMP-9 levels were significantly elevated in the plasma of patients with AD compared to the controls. 138 In the brain tissue of patients with AD, MMP-9 expression was found in the neurons’ cytoplasm, neurofibrillary tangles, senile plaques, and vascular walls. 139 There were inverse correlations between the Global Cognitive and MMSE scores and MMP-9 activity. 140 The Aβ is a potent stimulator of MMP-9 and MMP-2 activity in mixed hippocampal astrocyte cultures. 141 The interaction of Aβ and RAGEs induces MMP-2 expression in brain endothelial cells. 142 The MMP genotypes may influence the risk of dementia. The MMP gene polymorphisms reportedly associated with vascular dementia and AD. 143 The MMP-3 variants are associated with changes in the Aβ levels in humans and an increased risk of dementia.144,145 An MMP-9 inhibitor improves the Aβ-mediated cognitive impairment and neurotoxicity in mice. 146 These finding further suggest that MMP-9 plays a causal role in Aβ-induced cognitive impairment and neurotoxicity. Moreover, VDR-knock-out mice were shown to have an influx of inflammatory cells, phospho-acetylation of NF-κB, and upregulated expression of MMP-2, MMP-9 and MMP-12 in the lungs. 147 The VDR TaqI polymorphism is associated with the decreased production of TIMP-1, a natural MMP-9 inhibitor. 148 In addition, calcitriol modulates tissue MMP expression under experimental conditions, 149 downregulates MMP-9 levels in keratinocytes, and may attenuate the deleterious effects of excessive TNF-α-induced proteolytic activity, which is associated with cutaneous inflammation. 150 Calcitriol also inhibits both the basal levels and the staphylococcus-stimulated production of MMP-9 in human blood monocytes and alveolar macrophages. 151 Moreover, a vitamin D analog has also been reported to reduce the expression of MMP-2, MMP-9, VEGF, and PTH-related peptide in Lewis lung carcinoma cells. 152 Furthermore, calcitriol significantly attenuates the Mycobacterium tuberculosis-induced expression of MMP-7 and MMP-10, and suppresses the secretion of MMP-7 by M tuberculosis-infected PBMCs. The MMP-9 gene expression, secretion, and activity were also shown to be significantly inhibited by calcitriol, irrespective of the infection status. 153 In another study, calcitriol also suppressed the production of MMPs (MMP-7 and MMP-9) and increased the level of TMP-1 in patients with tuberculosis. 154 In human articular chondrocytes, calcitriol was shown to significantly suppress the increased production of MMP-9-induced by phorbol myristate acetate. 155 Together, these studies suggest that calcitriol may play an important role in the pathological processes of AD by downregulating the level of MMPs and regulating the level of TIMPs.

Heme oxygenase-1 (HO-1) is a stress protein that may confer cytoprotection by enhancing the catabolism of prooxidant heme to the radical scavenging bile pigments biliverdin and bilirubin. In various models of oxidative tissue injuries, the induction of HO-1 protects tissues from further damage by removing the heme. 156 Several studies have suggested that HO-1 may be involved in the pathogenesis of neurodegenerative diseases, such as AD. The HO-1 mRNA and its transcripted proteins were increased in the neocortical and cerebral vessels of patients with AD compared to the controls. 157 The percentage of GFAP-positive astrocytes that coexpress HO-1 in the hippocampus in AD is significantly increased relative to age-matched controls. 158 In rat astrocytes, human HO-1 overexpression resulted in significant oxidative damage to mitochondrial lipids, proteins, and nucleic acids and increased cell death. 159 The long-term overexpression of HO-1 promotes tau aggregation in mouse brains. 160 Astroglial HO-1 expression in the temporal cortex was associated with decreased scores of global cognition, semantic, and working memory, and was correlated with the burden of neurofibrillary pathology. 161 Another study found that exposing a group of obese rats to a Westernized diet (high fat/high fructose corn syrup) deficient in vitamin D exacerbated nonalcoholic fatty liver disease and resulted in an increased expression of the oxidative stress marker HO-1. 162 Additionally, pretreatment with vitamin D3 ameliorates systemic IL-6 levels following ischemia and reperfusion of bilaterally occluded vessels in rats improves lung and muscle injury, and results in a significant increase in leukocytes HO-1 expression. 163 Moreover, following the focal cortical ischemia that is elicited by photothrombosis, calcitriol treatment results in a transient but significant upregulation of glial HO-1 immunoreactivity. This upregulation was concomitant with a reduction in GFAP in remote cortical regions that are affected by the secondary spread of injury in glial cells. 164 These results support the protective role of calcitriol in postcellular injury.

The reduced form of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) enzyme complex mediates critical physiological and pathological processes including cell signaling, inflammation, and mitogenesis, by generating ROS from molecular oxygen. The NOX is widely expressed in various immune cells, including microglia, macrophages, and neutrophils. In AD, NOX is activated in microglia, resulting in the formation of ROS that are toxic to neighboring neurons. 165 The Aβ induces mitochondrial dysfunction and oxidative stress in astrocytes and the death of neurons via NOX activation.166,167 The NOX expression and activity are upregulated specifically in vulnerable brain regions of mildly cognitively impaired patients. 168 The NOX is also upregulated in the frontal and temporal cortex and contributes to AD progression. 169 The inhibition of NOX or the gene deletion of its functional p47phox subunit promotes alternative and anti-inflammatory microglial activation during neuroinflammation. 170 However, vitamin D deprivation in rats decreased the activity of cytosolic NADPH-dependent 3,5,3′-triodo-l-thyronine binding in the liver. This decrease was restored by administering calcitriol. 171 In heart mitochondria, NAD+-dependent isocitrate dehydrogenase decreased notably in vitamin D-deficient rats, but calcitriol subsequently restored normal values. 172 In rat centrilobular hepatocytes, a vitamin D-deficient diet induced a significant increase in NADPH. 173 Husain et al 174 reported that cardiac NOX activity increased by 300% in uremic rats compared to the normal controls. Treatment with paricalcitol protected the uremic rats from cardiac oxidative stress by inhibiting NOX activity (by 50%), thus lowering superoxide production in the heart. Taken together, vitamin D may have a role in AD by suppressing NADPH expression.

Conclusions

Vitamin D may play a role in AD. Genetic studies have provided the opportunity to determine which proteins link vitamin D to the pathology of AD, including miRNA, TLRs, VEGF, angiogenin, and AGEs. Vitamin D also exerts its effects on AD via CaSR, Aβ, IL-10, MMPs, HO-1, and the reduced form of NADP. Calcitriol is the best potential vitamin D supplement for patients with AD because it is an active form of vitamin D3 metabolite, suppresses PTH (a cellular growth factor), and binds to the VDR receptors on human nerve cells. The monitor of serum 25OHD3 levels after calcitriol administration is unnecessary because calcitriol inhibits the liver production of serum 25OHD3.175,176 The main limitations to the widespread use of calcitriol are its hypercalcemic side effects.

Footnotes

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

References

  • 1.Buell JS, Dawson-Hughes B, Scott TM, et al. 25-Hydroxyvitamin D, dementia, and cerebrovascular pathology in elders receiving home services. Neurology. 2010;74(1):18-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Annweiler C, Rolland Y, Schott AM, Blain H, Vellas B, Beauchet O. Serum vitamin D deficiency as a predictor of incident non-Alzheimer dementias: a 7-year longitudinal study. Dement Geriatr Cogn Disord. 2011;32(4):273-278. [DOI] [PubMed] [Google Scholar]
  • 3.Annweiler C, Rolland Y, Schott AM, et al. Higher vitamin d dietary intake is associated with lower risk of Alzheimer’s disease: A 7-year follow-up [published online April 13, 2012]. J Gerontol A Biol Sci Med Sci. 2012. [DOI] [PubMed] [Google Scholar]
  • 4.Wilkins CH, Sheline YI, Roe CM, Birge SJ, Morris JC. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry. 2006;14(12):1032-1040. [DOI] [PubMed] [Google Scholar]
  • 5.Wilkins CH, Birge SJ, Sheline YI, Morris JC. Vitamin D deficiency is associated with worse cognitive performance and lower bone density in older African Americans. J Natl Med Assoc. 2009;101(4):349-354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Evatt ML, DeLong MR, Khazai N, Triche S, Tangpricha V. Prevalence of vitamin D insufficiency in patients with Parkinson disease and Alzheimer disease. Arch Neurol. 2008;65:1348-1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sato Y, Asoh T, Oizumi K. High prevalence of vitamin D deficiency and reduced bone mass in elderly women with Alzheimer’s disease. Bone. 1998;23(6):555-557. [DOI] [PubMed] [Google Scholar]
  • 8.Wilkins CH, Sheline YI, Roe CM, Birge SJ, Morris JC. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry. 2006;14(12):1032-1040. [DOI] [PubMed] [Google Scholar]
  • 9.Oudshoorn C, Mattace-Raso FUS, van der Velde N, Colin EM, van der Cammen TJM. Higher serum vitamin D3 levels are associated with better cognitive test performance in patients with Alzheimer’s disease. Dement Geriatr Cogn Disord. 2008;25(6):539-543. [DOI] [PubMed] [Google Scholar]
  • 10.Llewellyn DJ, Lang IA, Langa KM, Melzer D. Vitamin D and cognitive impairment in the elderly U.S. population. J Gerontol A Biol Sci Med Sci. 2011;66(1):59-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Taghizadeh M, Djazayery A, Salami M, Eshraghian MR, Zavareh SA. Vitamin-D-free regimen intensifies the spatial learning deficit in Alzheimer’s disease. Int J Neurosci. 2011;121(1):16-24. [DOI] [PubMed] [Google Scholar]
  • 12.Landfield PW, Cadwallader-Neal L. Long-term treatment with calcitriol (1,25(OH)2 vit D3) retards a biomarker of hippocampal aging in rats. Neurobiol Aging. 1998;19(5):469-77. [DOI] [PubMed] [Google Scholar]
  • 13.Larsson B, Nemere I. Effect of growth and maturation on membrane-initiated actions of 1,25-dihydroxyvitamin D3-II: calcium transport, receptor kinetics, and signal transduction in intestine of female chickens. J Cell Biochem. 2003;90(5):901-913. [DOI] [PubMed] [Google Scholar]
  • 14.Lecanu L, Rammouz G, McCourty A, Sidahmed EK, Greeson J, Papadopoulos V. Caprospinol reduces amyloid deposits and improves cognitive function in a rat model of Alzheimer’s disease. Neuroscience. 2010;165(2):427-435. [DOI] [PubMed] [Google Scholar]
  • 15.Tohda C, Urano T, Umezaki M, Nemere I, Kuboyama T. Diosgenin is an exogenous activator of 1,25D3-MARRS/Pdia3/ERp57 and improves Alzheimer’s disease pathologies in 5XFAD mice. Sci Rep. 2012;2:535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lu’o’ng KV, Nguyên LT. The beneficial role of vitamin D in Alzheimer’s disease. Am J Alzheimers Dis Other Demen. 2011;26(7):511-520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Veenstra TD, Prüfer K, Koenigsberger C, Brimijoin SW, Grande JP, Kumar R. 1,25-Dihydroxyvitamin D3 receptors in the central nervous system of the rat embryo. Brain Res. 1998;804(2):193-205. [DOI] [PubMed] [Google Scholar]
  • 18.Prüfer K, Veenstra TD, Jirikowski GF, Kumar R. Distribution of 1,25-dihydroxyvitamin D3 receptor immunoreactivity in the rat brain and spinal cord. J Chem Neuroanat. 1999;16(2):135-145. [DOI] [PubMed] [Google Scholar]
  • 19.Eyles DW, Smith S, Kinobe R, Hewison M, McGrath JJ. Distribution of the vitamin D receptor and 1 alpha-hydroxylase in human brain. J Chem Neuroanat. 2005;29(1):21-30. [DOI] [PubMed] [Google Scholar]
  • 20.Sutherland MK, Somerville MJ, Yoong LKK, Bergeron C, Haussler MR, Mclachlan DRC. Reduction of vitamin D hormone receptor mRNA levels in Alzheimer as compared to Huntington hippocampus: correlation with calbindin-28 k mRNA levels. Mol Brain Res. 1992;13(3):239-250. [DOI] [PubMed] [Google Scholar]
  • 21.Kuningas M, Mooijaart SP, Jolles J, Slagboom PE, Westendorp RG, van Heemst D. VDR gene variants associate with cognitive function and depressive symptoms in old age. Neurobiol Aging. 2009;30(3):466-473. [DOI] [PubMed] [Google Scholar]
  • 22.Beydoun MA, Ding EL, Beydoun HA, Tanaka T, Ferrucci L, Zonderman AB. Vitamin D receptor and megalin gene polymorphisms and their associations with longitudinal cognitive change in US adults. Am J Clin Nutr. 2012;95(1):163-178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lehmann DJ, Refsum H, Warden DR, Medway C, Wilcock GK, Smith AD. The vitamin D receptor gene is associated with Alzheimer’s disease. Neurosci Lett. 2011;504(2):79-82. [DOI] [PubMed] [Google Scholar]
  • 24.Wang L, Hara K, Van Baaren JM, et al. Vitamin D receptor and Alzheimer’s disease: a genetic and functional study. Neurobiol Aging. 2012;33(8):1844.e1-1849.e1. [DOI] [PubMed] [Google Scholar]
  • 25.Gezen-Ak D, Dursun E, Bilgiç B, et al. Vitamin d receptor gene haplotype is associated with late-onset Alzheimer’s disease. Tohoku J Exp Med. 2012;228(3):189-196. [DOI] [PubMed] [Google Scholar]
  • 26.Gilad S, Meiri E, Yogev Y, et al. Serum microRNAs are promising novel biomarkers. PLoS One. 2008;3(9):e3148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pauley KM, Cha S, Chan EK. MicroRNA in autoimmunity and autoimmune diseases. J Autoimmun. 2009;32(3-4):189-194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lu LF, Liston A. MicroRNA in the immune system, microRNA as an immune system. Immunology. 2009;127(3):291-298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lukiw WJ, Zhao Y, Cui JG. An NF-kappaB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem. 2008;283(46):31315-31322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cui JG, Li YY, Zhao Y, Bhattacharjee S, Lukiw WJ. Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK-1) and IRAK-2 by microRNA-146a and NF-kappaB in stressed human astroglial cells and in Alzheimer disease. J Biol Chem. 2010;285(50):38951-38960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pogue AI, Cui JG, Li YY, Zhao Y, Culicchia F, Lukiw WJ. Micro RNA-125b (miRNA-125b) function in astrogliosis and glial cell proliferation. Neurosci Lett. 2010;476(1):18-22. [DOI] [PubMed] [Google Scholar]
  • 32.Lukiw WJ, Alexandrov PN, Zhao Y, Hill JM, Bhattacharjee S. Spreading of Alzheimer’s disease inflammatory signaling through soluble micro-RNA. Neuroreport. 2012;23(10):621-626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang G, Tam LS, Kwan BC, et al. Expression of miR-146a and miR-155 in the urinary sediment of systemic lupus erythematosus. Clin Rheumatol. 2012;31(3):435-440. [DOI] [PubMed] [Google Scholar]
  • 34.Wang Y, Borchert ML, Deluca HF. Identification of the vitamin D receptor in various cells of the mouse kidney. Kidney Int. 2012;81(10):993-1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jorde R, Svartberg J, Joakimsen RM, Coucheron DH. Plasma profile of microRNA after supplementation with high doses of vitamin D3 for 12 months. BMC Res Notes. 2012;5:245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pan YZ, Gao W, Yu AM. MicroRNAs regulate CYP3A4 expression via direct and indirect targeting. Drug Metab Dispos. 2009;37(10):2112-2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Komagata S, Nakajima M, Takagi S, Mohri T, Taniya T, Yokoi T. Human CYP24 catalyzing the inactivation of calcitriol is post-transcriptionally regulated by miR-125b. Mol Pharmacol. 2009;76(4):702-709. [DOI] [PubMed] [Google Scholar]
  • 38.Mohri T, Nakajima M, Takagi S, Komagata S, Yokoi T. MicroRNA regulates human vitamin D receptor. Int J Cancer. 2009;125:1328-1333. [DOI] [PubMed] [Google Scholar]
  • 39.Wang WL, Chatterjee N, Chittur SV, Welsh J, Tenniswood MP. Effects of 1α,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011;10:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Essa S, Denzer N, Mahlknecht U, et al. VDR microRNA expression and epigenetic silencing of vitamin D signaling in melanoma cells. J Steroid Biochem Mol Biol. 2010;121(1-2):110-113. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang W, Wang LZ, Yu JT, Chi ZF, Tan L. Increased expressions of TLR2 and TLR4 on peripheral blood mononuclear cells from patients with Alzheimer’s disease. J Neurol Sci. 2012;315(1-2):67-71. [DOI] [PubMed] [Google Scholar]
  • 42.Reed-Geaghan EG, Savage JC, Hise AG, Landreth GE. CD14 and toll-like receptors 2 and 4 are required for fibrillar Aβ-stimulated microglial activation. J Neurosci. 2009;29(38):11982-11892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jin JJ, Kim HD, Maxwell JA, Li L, Fukuchi K. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2008;5:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Liu S, Liu Y, Hao W, et al. TLR2 is a primary receptor for Alzheimer’s amyloid β peptide to trigger neuroinflammatory activation. J Immunol.2012;188(3):1098-1107. [DOI] [PubMed] [Google Scholar]
  • 45.Jana M, Palencia CA, Pahan K. Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer’s disease. J Immunol. 2008;181(10):7254-7262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Vollmar P, Kullmann JS, Thilo B, et al. Active immunization with amyloid-beta 1-42 impairs memory performance through TLR2/4-dependent activation of the innate immune system. J Immunol. 2010;185(10):6338-6347. [DOI] [PubMed] [Google Scholar]
  • 47.Tang SC, Lathia JD, Selvaraj PK, et al. Toll-like receptor-4 mediates neuronal apoptosis induced by amyloid beta-peptide and the membrane lipid peroxidation product 4-hydroxynonenal. Exp Neurol. 2008;213(1):114-121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang LZ, Tian Y, Yu JT, et al. Association between late-onset Alzheimer’s disease and microsatellite polymorphisms in intron II of the human toll-like receptor 2 gene. Neurosci Lett. 2011;489(3):164-167. [DOI] [PubMed] [Google Scholar]
  • 49.Yu JT, Mou SM, Wang LZ, Mao CX, Tan L. Toll-like receptor 2 -196 to -174 del polymorphism influences the susceptibility of Han Chinese people to Alzheimer’s disease. J Neuroinflammation. 2011;8:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang LZ, Yu JT, Miao D, et al. Genetic association of TLR4/11367 polymorphism with late-onset Alzheimer’s disease in a Han Chinese population. Brain Res. 2011;1381:202-207. [DOI] [PubMed] [Google Scholar]
  • 51.Yu JT, Miao D, Cui WZ, et al. Common variants in toll-like receptor 4 confer susceptibility to Alzheimer’s disease in a Han Chinese population. Curr Alzheimer Res. 2012;9(4):458-466. [DOI] [PubMed] [Google Scholar]
  • 52.Minoretti P, Gazzaruso C, Vito CD, et al. Effect of the functional toll-like receptor 4 Asp299Gly polymorphism on susceptibility to late-onset Alzheimer’s disease. Neurosci Lett. 2006;391(3):147-149. [DOI] [PubMed] [Google Scholar]
  • 53.Song M, Jin J, Lim JE, et al. TLR4 mutation reduces microglial activation, increases Aβ deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. J Neuroinflammation. 2011;8:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Soscia SJ, Kirby JE, Washicosky KJ, et al. The Alzheimer’s disease-associated amyloid beta-protein is an antimicrobial peptide. PLoS One. 2010;5(3):e9505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ciornei CD, Tapper H, Bjartell A, Sternby NH, Bodelsson M. Human antimicrobial peptide LL-37 is present in atherosclerotic plaques and induces death of vascular smooth muscle cells: a laboratory study. BMC Cardiovasc Disord. 2006;6:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Roth CL, Elfers CT, Figlewicz DP, et al. Vitamin D deficiency in obese rats exacerbates NAFLD and increases hepatic resistin and toll-like receptor activation. Hepatology. 2012;55:1103-1111. [DOI] [PubMed] [Google Scholar]
  • 57.Sadeghi K, Wessner B, Laggner U, et al. Vitamin D3 down-regulates monocyte TLR expression and triggers hyporesponsiveness to pathogen-associated molecular patterns. Eur J Immunol. 2006;36(2):361-370. [DOI] [PubMed] [Google Scholar]
  • 58.Dickie L, Church L, Coulthard L, Mathews R, Emery P, McDermott M. Vitamin D3 downregulates intracellular toll-like receptor 9 expression and toll-like receptor 9-induced IL-6 production in human monocytes. Rheumatology. 2010;49(8):1466-1471. [DOI] [PubMed] [Google Scholar]
  • 59.Liu PT, Stenger S, Li H, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311(5768):1770-1773. [DOI] [PubMed] [Google Scholar]
  • 60.Yim S, Dhawan P, Ragunath C, Christakos S, Diamond G. Induction of cathelicidin in normal and CF bronchial epithelial cells by 1,25-dihydroxyvitamin D3. J Cys Fibros. 2007;6(6):403-410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Rivas-Santiago B, Hernandez-Pando R, Carranza C, et al. Expression of cathelicidin LL-37 during Mycobacterium tuberculosis infection in human alveolar macrophages, monocytes, neutrophils, and epithelial cells. Infect Immunity. 2008;76(3):935-941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Schultheiss C, Blechert B, Gaertner FC, Drecoll E, Mueller J. In vivo characterization of endothelial cell activation in a transgenic mouse model of Alzheimer’s disease. Angiogenesis. 2006;9:59-65. [DOI] [PubMed] [Google Scholar]
  • 63.Thirumangalakudi L, Samany PG, Owoso A, Wiskar B, Grammas P. Angiogenic proteins are expressed by brain blood vessels in Alzheimer’s disease. J Alzheimers Dis. 2006;10:111-118. [DOI] [PubMed] [Google Scholar]
  • 64.Kalaria RN, Cohen DL, Premkumar DR, Nag S, LaManna JC, Lust WD. Vascular endothelial growth factor in Alzheimer’s disease and experimental cerebral ischemia. Brain Res Mol Brain Res. 1998;62(1):101-105. [DOI] [PubMed] [Google Scholar]
  • 65.Tarkowski E, Issa R, Sjögren M, et al. Increased intrathecal levels of the angiogenic factors VEGF and TGF-beta in Alzheimer’s disease and vascular dementia. Neurobiol Aging. 2002;23(2):237-243. [DOI] [PubMed] [Google Scholar]
  • 66.Yang SP, Bae DG, Kang HJ, Gwag BJ, Gho YS, Chae CB. Co-accumulation of vascular endothelial growth factor with beta-amyloid in the brain of patients with Alzheimer’s disease. Neurobiol Aging. 2004;25(3):283-290. [DOI] [PubMed] [Google Scholar]
  • 67.Patel NS, Mathura VS, Bachmeier C, et al. Alzheimer’s beta-amyloid peptide blocks vascular endothelial growth factor mediated signaling via direct interaction with VEGFR-2. J Neurochem. 2010;112(1):66-76. [DOI] [PubMed] [Google Scholar]
  • 68.Del Bo R, Ghezzi S, Scarlato M, et al. Role of VEGF gene variability in longevity: a lesson from the Italian population. Neurobiol Aging. 2008;29(12):1917-1922. [DOI] [PubMed] [Google Scholar]
  • 69.Del Bo R, Scarlato M, Ghezzi S, et al. Vascular endothelial growth factor gene variability is associated with increased risk for AD. Ann Neurol. 2005;57(3):373-380. [DOI] [PubMed] [Google Scholar]
  • 70.Chiappelli M, Borroni B, Archetti S, et al. VEGF gene and phenotype relation with Alzheimer’s disease and mild cognitive impairment. Rejuvenation Res. 2006;9(4):485-493. [DOI] [PubMed] [Google Scholar]
  • 71.Yuan Q, Zuo X, Jia J. Association between promoter polymorphisms of vascular endothelial growth factor gene and sporadic Alzheimer’s disease among Northern Chinese Han. Neurosci Lett. 2009;457(3):133-136. [DOI] [PubMed] [Google Scholar]
  • 72.Smach MA, Charfeddine B, Othman LB, et al. -1154G/A and -2578C/A polymorphisms of the vascular endothelial growth factor gene in Tunisian Alzheimer patients in relation to beta-amyloid (1-42) and total tau protein. Neurosci Lett. 2010;472(2):139-142. [DOI] [PubMed] [Google Scholar]
  • 73.Mantell D, Owens P, Bundred N, Mawer E, Canfield A. 1α,25-dihydroxyvitamin D3 inhibits angiogenesis in vitro and in vivo. Circ Res. 2000;87(3):214-220. [DOI] [PubMed] [Google Scholar]
  • 74.Gruber H, Hoelscher G, Ingram J, Chow Y, Loffler B, Hanley E. 1α,25-dihydroxyvitamin D3 inhibits proliferation and decreases production of monocyte chemoattractant protein-1, thrombopoietin, VEGF, and angiogenin by human annulus cells in vitro. Spine. 2008;33(7):755-765. [DOI] [PubMed] [Google Scholar]
  • 75.Albert D, Scheef E, Wang S, et al. Calcitriol is a potent inhibitor of retinal neovascularization. Invest Ophthalmol Vis Sci. 2007;48(5):2327-2334. [DOI] [PubMed] [Google Scholar]
  • 76.Nakagawa K, Sasaki Y, Kato S, et al. 22-Oxa-1α,25-dihydroxyvitamin D3 inhibits metastasis and angiogenesis in lung cancer. Carcinogenesis. 2005;26(6):1044-1054. [DOI] [PubMed] [Google Scholar]
  • 77.Ben-Shoshan M, Amir S, Dang D, Dang L, Weisman Y, Mabjeesh N. 1α,25-dihydroxyvitamin D3 (Calcitriol) inhibits hypoxia-inducible factor-1/vascular endothelial growth pathway in human cancer cells. Mol Cancer Ther. 2007;6(4):1433-1439. [DOI] [PubMed] [Google Scholar]
  • 78.Moroianu J, Riordan JF. Identification of the nucleolar targeting signal of human angiogenin. Biochem Biophys Res Commun. 1994;203(3):1765-1772. [DOI] [PubMed] [Google Scholar]
  • 79.Kieran D, Sebastia J, Greenway MJ, et al. Control of motoneuron survival by angiogenin. J Neurosci. 2008;28(52):14056-14061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kim YN, Kim do H. Decreased serum angiogenin level in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2012;38(2):116-120. [DOI] [PubMed] [Google Scholar]
  • 81.Hooper LV, Stappenbeck TS, Hong CV, Gordon JI. Angiogenins: a new class of microbicidal proteins involved in innate immunity. Nat Immunol. 2003;4(3):269-273. [DOI] [PubMed] [Google Scholar]
  • 82.Lagishetty V, Misharin AV, Liu NQ, et al. Vitamin D deficiency in mice impairs colonic antibacterial activity and predisposes to colitis. Endocrinology. 2010;151(6):2423-2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bierhaus A, Humpert PM, Morcos M, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl). 2005;83(11):876-886. [DOI] [PubMed] [Google Scholar]
  • 84.Chen F, Wollmer MA, Hoerndli F, et al. Role for glyoxalase I in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2004;101(20):7687-7692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kuhla B, Boeck K, Schmidt A, et al. Age- and stage-dependent glyoxalase I expression and its activity in normal and Alzheimer’s disease brains. Neurobiol Aging. 2007;28(1):29-41. [DOI] [PubMed] [Google Scholar]
  • 86.Shuvaev VV, Laffont I, Serot JM, Fujii J, Taniguchi N, Siest G. Increased protein glycation in cerebrospinal fluid of Alzheimer’s disease. Neurobiol Aging. 2001;22(3):397-402. [DOI] [PubMed] [Google Scholar]
  • 87.Riviere S, Birlouez-Aragon I, Vellas B. Plasma protein glycation in Alzheimer’s disease. Glycoconj J. 1998;15:1039-1042. [DOI] [PubMed] [Google Scholar]
  • 88.Sasaki N, Toki S, Chowei H, et al. Immunohistochemical distribution of the receptor for advanced glycation end products in neurons and astrocytes in Alzheimer’s disease. Brain Res. 2001;888(2):256-262. [DOI] [PubMed] [Google Scholar]
  • 89.Miller MC, Tavares R, Johanson CE, et al. Hippocampal RAGE immunoreactivity in early and advanced Alzheimer’s disease. Brain Res. 2008;1230:273-280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lue LF, Walker DG, Brachova L, et al. Involvement of microglial receptor for advanced glycation endproducts ( RAGE ) in Alzheimer’s disease: identification of a cellular activation mechanism. Exp Neurol. 2001;171(1):29-45. [DOI] [PubMed] [Google Scholar]
  • 91.Du Yan S, Zhu H, Fu J, et al. Amyloid-beta peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. Proc Natl Acad Sci U S A. 1997;94(10):5296-5301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Li K, Dai D, Zhao B, et al. Association between the RAGE G82 S polymorphism and Alzheimer’s disease. J Neural Transm. 2010;117(1):97-104. [DOI] [PubMed] [Google Scholar]
  • 93.Daborg J, von Otter M, Sjölander A, et al. Association of the RAGE G82 S polymorphism with Alzheimer’s disease. J Neural Transm. 2010;117(7):861-867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wilson JS, Mruthinti S, Buccafusco JJ, et al. Anti-RAGE and Abeta immunoglobulin levels are related to dementia level and cognitive performance. J Gerontol A Biol Sci Med Sci. 2009;64(2):264-271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Talmor-Barkan Y, Bernheim J, Green J, Benchetrit S, Rashid G. Calcitriol counteracts endothelial cell pro-inflammatory processes in a chronic kidney disease-like environment. J Steroid Biochem Mol Biol. 2011;124:19-24. [DOI] [PubMed] [Google Scholar]
  • 96.Talmor Y, Golan E, Benchetrit S, et al. Calcitriol blunts the deleterious impact of advanced glycation end products on endothelial cells. Am J Physiol Renal Physiol. 2008;294(5):F1059-F1064. [DOI] [PubMed] [Google Scholar]
  • 97.Talmor Y, Bernheim J, Klein O, Green J, Rashid G. Calcitriol blunts pro-atherosclerotic parameters through NFkappaB and p38 in vitro. Eur J Clin Invest. 2008;38:548-554. [DOI] [PubMed] [Google Scholar]
  • 98.Brown EM, MacLeod RJ. Extracellular calcium sensing and extracellular calcium signaling. Physiol Rev. 2001;81(1):239-297. [DOI] [PubMed] [Google Scholar]
  • 99.Magno AL, Ward BK, Ratajczak T. The calcium-sensing receptor: a molecular perspective. Endocr Rev. 2011;32:3-30. [DOI] [PubMed] [Google Scholar]
  • 100.Yano S, Brown EM, Chattopadhyay N. Calcium-sensing receptor in the brain. Cell Calcium. 2004;35(3):257-264. [DOI] [PubMed] [Google Scholar]
  • 101.Braunewell K, Riederer P, Spilker C, Gundelfinger ED, Bogerts B, Bernstein HG. Abnormal localization of two neuronal calcium sensor proteins, visinin-like proteins (vilips)-1 and -3, in neocortical brain areas of Alzheimer disease patients. Dement Geriatr Cogn Disord. 2001;12(2):110-116. [DOI] [PubMed] [Google Scholar]
  • 102.Schnurra I, Bernstein HG, Riederer P, Braunewell KH. The neuronal calcium sensor protein VILIP-1 is associated with amyloid plaques and extracellular tangles in Alzheimer’s disease and promotes cell death and tau phosphorylation in vitro: a link between calcium sensors and Alzheimer’s disease? Neurobiol Dis. 2001;8(5):900-909. [DOI] [PubMed] [Google Scholar]
  • 103.Ye C, Ho-Pao CL, Kanazirska M, et al. Amyloid-beta proteins activate Ca2+-permeable channels through calcium-sensing receptors. J Neurosci Res. 1997;47(5):547-554. [DOI] [PubMed] [Google Scholar]
  • 104.Chiarini A, Dal Pra I, Marconi M, Chakravarthy B, Whitfield JF, Armato U. Calcium-sensing receptor (CaSR) in human brain’s pathophysiology: roles in late-onset Alzheimer’s disease (LOAD). Curr Pharm Biotechnol. 2009;10(3):317-326. [DOI] [PubMed] [Google Scholar]
  • 105.Chattopadhyay N, Espinosa-Jeffrey A, Tfelt-Hansen J, et al. Calcium receptor expression and function in oligodendrocyte commitment and lineage progression: potential impact on reduced myelin basic protein in CaR-null mice. J Neurosci Res. 2008;86(10):2159-2167. [DOI] [PubMed] [Google Scholar]
  • 106.Conley YP, Mukherjee A, Kammerer C, et al. Evidence supporting a role for the calcium-sensing receptor in Alzheimer disease. Am J Med Genet B Neuropsychiatr Genet. 2009;150B(5):703-709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Brown AJ, Zhong M, Finch J, et al. Rat calcium-sensing receptor is regulated by vitamin D but not by calcium. Am J Physiol. 1996;270(3 pt 2):F454-F460. [DOI] [PubMed] [Google Scholar]
  • 108.Canaff L, Hendy GN. Human calcium-sensing receptor gene. Vitamin D response elements in promoters P1 and P2 confer transcriptional responsiveness to 1,25-dihydroxyvitamin D. J Biol Chem. 2002;277(33):30337-3050. [DOI] [PubMed] [Google Scholar]
  • 109.He YH, Song Y, Liao XL, et al. The calcium-sensing receptor affects fat accumulation via effects on antilipolytic pathways in adipose tissue of rats fed low-calcium diets. J Nutr. 2011;141(11):1938-1946. [DOI] [PubMed] [Google Scholar]
  • 110.Yao JJ, Bai S, Karnauskas AJ, Bushinsky DA, Favus MJ. Regulation of renal calcium receptor gene expression by 1,25-dihydroxyvitamin D3 in genetic hypercalciuric stone-forming rats. J Am Soc Nephrol. 2005;16(5):1300-1308. [DOI] [PubMed] [Google Scholar]
  • 111.Chakrabarty S, Wang H, Canaff L, Hendy GN, Appelman H, Varani J. Calcium sensing receptor in human colon carcinoma: interaction with Ca2+ and 1,25-dihydroxyvitamin D3. Cancer Res. 2005;65:493-498. [PubMed] [Google Scholar]
  • 112.Gezen-Ak D, Dursun E, Yilmazer S. The effects of vitamin D receptor silencing on the expression of LVSCC-A1C and LVSCC-A1D and the release of NGF in cortical neurons. PLoS One. 2011;6(3):e17553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Dursun E, Gezen-Ak D, Yilmazer S. A novel perspective for Alzheimer’s disease: vitamin D receptor suppression by amyloid-β and preventing the amyloid-β induced alterations by vitamin D in cortical neurons. J Alzheimers Dis. 2011;23(2):207-219. [DOI] [PubMed] [Google Scholar]
  • 114.Fiala M, Porter V. Delineating Alzheimer’s disease progression with MGAT3, a biomarker for improved prognosis and personalized therapy. Biomark Med. 2011;5(5):645-647. [DOI] [PubMed] [Google Scholar]
  • 115.Fiala M, Mahanian M, Rosenthal M, et al. MGAT3 mRNA: a biomarker for prognosis and therapy of Alzheimer’s disease by vitamin D and curcuminoids. J Alzheimers Dis. 2011;25(1):135-144. [DOI] [PubMed] [Google Scholar]
  • 116.Masoumi A, Goldenson B, Ghirmai S, et al. 1alpha,25-dihydroxyvitamin D3 interacts with curcuminoids to stimulate amyloid-beta clearance by macrophages of Alzheimer’s disease patients. J Alzheimers Dis. 2009;17(3):703-717. [DOI] [PubMed] [Google Scholar]
  • 117.Mizwicki MT, Menegaz D, Zhang J, et al. Genomic and nongenomic 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. [DOI] [PubMed] [Google Scholar]
  • 118.Lee V, Rekhi E, Hoh Kam J, Jeffery G. Vitamin D rejuvenates aging eyes by reducing inflammation, clearing amyloid beta and improving visual function. Neurobiol Aging. 2012;33(10):2382-2389. [DOI] [PubMed] [Google Scholar]
  • 119.Yu J, Gattoni-Celli M, Zhu H, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ito S, Ohtsuki S, Nezu Y, Koitabashi Y, Murata S, Terasaki T. 1α,25-Dihydroxyvitamin D3 enhances cerebral clearance of human amyloid-β peptide(1-40) from mouse brain across the blood-brain barrier. Fluids Barriers CNS. 2011;8:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Speciale L, Calabrese E, Saresella M, et al. Lymphocyte subset patterns and cytokine production in Alzheimer’s disease patients. Neurobiol Aging. 2007;28(8):1163-1169. [DOI] [PubMed] [Google Scholar]
  • 122.De Luigi A, Fragiacomo C, Lucca U, et al. Inflammatory markers in Alzheimer’s disease and multi-infarct dementia. Mech Ageing Dev. 2001;122(16):1985-1995. [DOI] [PubMed] [Google Scholar]
  • 123.Kiyota T, Ingraham KL, Swan RJ, Jacobsen MT, Andrews SJ, Ikezu T. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP+PS1 mice. Gene Ther. 2012;19(7):724-733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Ma SL, Tang NL, Lam LC, Chiu HF. The association between promoter polymorphism of the interleukin-10 gene and Alzheimer’s disease. Neurobiol Aging. 2005;26(7):1005-1010. [DOI] [PubMed] [Google Scholar]
  • 125.Bagnoli S, Cellini E, Tedde A, et al. Association analysis of the paraoxonase-1 gene with Alzheimer’s disease. Neurosci Lett. 2006;408(3):199-202. [DOI] [PubMed] [Google Scholar]
  • 126.Arosio B, Mastronardi L, Vergani C, Annoni G. Intereleukin-10 promoter polymorphism in mild cognitive impairment and in its clinical evolution. Int J Alzheimers Dis. 2010;2010.pii-854527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zhang Y, Zhang J, Tian C, et al. The -1082G/A polymorphism in IL-10 gene is associated with risk of Alzheimer’s disease: a meta-analysis. J Neurol Sci. 2011;303(1-2):133-138. [DOI] [PubMed] [Google Scholar]
  • 128.Di Bona D, Rizzo C, Bonaventura G, Candore G, Caruso C. Association between interleukin-10 polymorphisms and Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2012;29(4):751-759. [DOI] [PubMed] [Google Scholar]
  • 129.Spach KM, Nashold FE, Dittel BN, Hayes CE. IL-10 signaling is essential for 1,25-dihydroxyvitamin D3-mediated inhibition of experimental autoimmune encephalomyelitis. J Immunol. 2006;177(9):6030-6037. [DOI] [PubMed] [Google Scholar]
  • 130.Matilainen JM, Räsänen A, Gynther P, Väisänen S. The genes encoding cytokines IL-2, IL-10 and IL-12B are primary 1alpha,25(OH)2D3 target genes. J Steroid Biochem Mol Biol. 2010;121(1-2):142-145. [DOI] [PubMed] [Google Scholar]
  • 131.Zittermann A, Dembinski J, Stehle P. Low vitamin D status is associated with low cord blood levels of the immunosuppressive cytokine interleukin-10. Pediatr Allergy Immunol. 2004;15(3):242-246. [DOI] [PubMed] [Google Scholar]
  • 132.Michel G, Gailis A, Jarzebska-Deussen B, Müschen A, Mirmohammadsadegh A, Ruzicka T. 1,25-(OH)2-vitamin D3 and calcipotriol induce IL-10 receptor gene expression in human epidermal cells. Inflamm Res. 1997;46(1):32-34. [DOI] [PubMed] [Google Scholar]
  • 133.Khoo AL, Joosten I, Michels M, et al. 1,25-Dihydroxyvitamin D3 inhibits proliferation but not the suppressive function of regulatory T cells in the absence of antigen-presenting cells. Immunology. 2011;134(4):459-468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Thota C, Farmer T, Garfield RE, Menon R, Al-Hendy A. Vitamin D elicits anti-inflammatory response, inhibits contractile-associated proteins, and modulates toll-like receptors in human myometrial cells [published online September 25]. Reprod Sci. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Leake A, Morris CM, Whateley J. Brain matrix metalloproteinase 1 levels are elevated in Alzheimer’s disease. Neurosci Lett. 2000;291(3):201-203. [DOI] [PubMed] [Google Scholar]
  • 136.Horstmann S, Budig L, Gardner H, et al. Matrix metalloproteinases in peripheral blood and cerebrospinal fluid in patients with Alzheimer’s disease. Int Psychogeriatr. 2010;22(6):966-972. [DOI] [PubMed] [Google Scholar]
  • 137.Yoshiyama Y, Asahina M, Hattori T. Selective distribution of matrix metalloproteinase-3 (MMP-3) in Alzheimer’s disease brain. Acta Neuropathol. 2000;99(2):91-95. [DOI] [PubMed] [Google Scholar]
  • 138.Lorenzl S, Albers DS, Relkin N, et al. Increased plasma levels of matrix metalloproteinase-9 in patients with Alzheimer’s disease. Neurochem Int. 2003;43(3):191-196. [DOI] [PubMed] [Google Scholar]
  • 139.Asahina M, Yoshiyama Y, Hattori T. Expression of matrix metalloproteinase-9 and urinary-type plasminogen activator in Alzheimer’s disease brain. Clin Neuropathol. 2001;20(2):60-63. [PubMed] [Google Scholar]
  • 140.Bruno MA, Mufson EJ, Wuu J, Cuello AC. Increased matrix metalloproteinase 9 activity in mild cognitive impairment. J Neuropathol Exp Neurol. 2009;68(12):1309-1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Deb S, Gottschall PE. Increased production of matrix metalloproteinases in enriched astrocyte and mixed hippocampal cultures treated with beta-amyloid peptides. J Neurochem. 1996;66(4):1641-1647. [DOI] [PubMed] [Google Scholar]
  • 142.Du H, Li P, Wang J, Qing X, Li W. The interaction of amyloid β and the receptor for advanced glycation endproducts induces matrix metalloproteinase-2 expression in brain endothelial cells. Cell Mol Neurobiol. 2012;32:141-147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Flex A, Gaetani E, Proia AS, et al. Analysis of functional polymorphisms of metalloproteinase genes in persons with vascular dementia and Alzheimer’s disease. J Gerontol A Biol Sci Med Sci. 2006;61(10):1065-1069. [DOI] [PubMed] [Google Scholar]
  • 144.Reitz C, van Rooij FJ, Soares HD, et al. Matrix metalloproteinase 3 haplotypes and plasma amyloid beta levels: the Rotterdam Study. Neurobiol Aging. 2010;31(4):715-718. [DOI] [PubMed] [Google Scholar]
  • 145.Helbecque N, Cottel D, Hermant X, Amouyel P. Impact of the matrix metalloproteinase MMP-3 on dementia. Neurobiol Aging. 2007;28(8):1215-1220. [DOI] [PubMed] [Google Scholar]
  • 146.Mizoguchi H, Takuma K, Fukuzaki E, et al. Matrix metalloprotease-9 inhibition improves amyloid beta-mediated cognitive impairment and neurotoxicity in mice. J Pharmacol Exp Ther. 2009;331(1):14-22. [DOI] [PubMed] [Google Scholar]
  • 147.Sundar I, Hwang J, Wu S, Sun J, Rahman I. Deletion of vitamin D receptor leads to premature emphysema/COPD by increased matrix metalloproteinase and lymphoid aggregates formation. Biochem Biophys Res Commun. 2011;406(1):127-133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Timms PM, Mannan N, Hitman GA, et al. Circulating MMP9, vitamin D and variation in the TIMP-1 response with VDR genotype: mechanisms for inflammatory damage in chronic disorders? Q J Med. 2002;95(12):787-796. [DOI] [PubMed] [Google Scholar]
  • 149.Dean DD, Schwartz Z, Schmitz J, et al. Vitamin D regulation of metalloproteinase activity in matrix vesicles. Connect Tissue Res. 1996;35(1-4):331-336. [DOI] [PubMed] [Google Scholar]
  • 150.Bahar-Shany K, Ravid A, Koren R. Upregulation of MMP-production by TNFalpha in keratinocytes and its attenuation by vitamin D. J Cell Physiol. 2010;222(3):729-737. [DOI] [PubMed] [Google Scholar]
  • 151.Lacraz S, Dayer J, Nicod L, Welgus H. 1,25-dihydroxyvitamin D3 dissociates production of interstitial collagenase and 92-kDa gelatinase in human mononuclear phagocytes. J Biol Chem. 1994;269(9):6485-6490. [PubMed] [Google Scholar]
  • 152.Nakagawa K, Sasaki Y, Kato S, Kubodera N, Okano T. 22-Oxa-1α,25-dihydroxyvitamin D3 inhibits metastasis and angiogenesis in lung cancer. Carcinogenesis. 2005;26(6):1044-1054. [DOI] [PubMed] [Google Scholar]
  • 153.Coussens A, Timms P, Boucher B, et al. 1α,25-dihydroxyvitamin D3 inhibits matrix metalloproteinases induced by Mycobacterium tuberculosis infection. Immunology. 2009;127(4):539-548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Anand S, Selvaraj P. Effect of 1,25 dihydroxyvitamin D3 on matrix metalloproteinases MMP-7, MMP-9 and the inhibitor TIMP-1 in pulmonary tuberculosis. Clin Immunol. 2009;133(1):126-131. [DOI] [PubMed] [Google Scholar]
  • 155.Tetlow L, Woolley D. Expression of vitamin D receptors and matrix metalloproteinases in osteoarthritic cartilage and human articular chondrocytes in vitro. Osteoarthritis Cartilage. 2001;9(5):423-431. [DOI] [PubMed] [Google Scholar]
  • 156.Takahashi T, Morita K, Akagi R, Sassa S. Protective role of heme oxygenase-1 in renal ischemia. Antioxid Redox Signal. 2004;6(5):867-877. [DOI] [PubMed] [Google Scholar]
  • 157.Premkumar DR, Smith MA, Richey PL, et al. Induction of heme oxygenase-1 mRNA and protein in neocortex and cerebral vessels in Alzheimer’s disease. J Neurochem. 1995;65(3):1399-1402. [DOI] [PubMed] [Google Scholar]
  • 158.Schipper HM. Glial HO-1 expression, iron deposition and oxidative stress in neurodegenerative diseases. Neurotox Res. 1999;1(1):57-70. [DOI] [PubMed] [Google Scholar]
  • 159.Song W, Su H, Song S, Paudel HK, Schipper HM. Over-expression of heme oxygenase-1 promotes oxidative mitochondrial damage in rat astroglia. J Cell Physiol. 2006;206(3):655-663. [DOI] [PubMed] [Google Scholar]
  • 160.Hui Y, Wang D, Li W, et al. Long-term overexpression of heme oxygenase 1 promotes tau aggregation in mouse brain by inducing tau phosphorylation. J Alzheimers Dis. 2011;26(2):299-313. [DOI] [PubMed] [Google Scholar]
  • 161.Schipper HM, Bennett DA, Liberman A, et al. Glial heme oxygenase-1 expression in Alzheimer disease and mild cognitive impairment. Neurobiol Aging. 2006;27(2):252-261. [DOI] [PubMed] [Google Scholar]
  • 162.Roth CL, Elfers CT, Figlewicz DP, et al. Vitamin D deficiency in obese rats exacerbates NAFLD and increases hepatic resistin and toll-like receptor activation. Hepatology. 2012;55(4):1103-1111. [DOI] [PubMed] [Google Scholar]
  • 163.Shih PK, Chen YC, Huang YC, Chang YT, Chen JX, Cheng CM. Pretreatment of vitamin D3 ameliorates lung and muscle injury induced by reperfusion of bilateral femoral vessels in a rat model. J Surg Res. 2011;171(1):323-328. [DOI] [PubMed] [Google Scholar]
  • 164.Oermann E, Bidmon HJ, Witte OW, Zilles K. Effects of 1α,25-didroxyvitamin D3 on the expression of HO-1 and GFAP in glial cells of the photothrombotically lesioned cerebral cortex. J Chem Neuroanat. 2004;28(4):225-238. [DOI] [PubMed] [Google Scholar]
  • 165.Shimohama S, Tanino H, Kawakami N, et al. Activation of NADPH oxidase in Alzheimer’s disease brains. Biochem Biophys Res Commun. 2000;273(1):5-9. [DOI] [PubMed] [Google Scholar]
  • 166.Jana A, Pahan K. Fibrillar amyloid-beta-activated human astroglia kill primary human neurons via neutral sphingomyelinase: implications for Alzheimer’s disease. J Neurosci. 2010;30(38):12676-12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Abramov AY, Canevari L, Duchen MR. Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J Neurosci. 2004;24(2):565-575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Bruce-Keller AJ, Gupta S, Parrino TE, et al. NOX activity is increased in mild cognitive impairment. Antioxid Redox Signal. 2010;12(12):1371-1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Ansari MA, Scheff SW. NADPH-oxidase activation and cognition in Alzheimer disease progression. Free Radic Biol Med. 2011;51(1):171-178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Choi SH, Aid S, Kim HW, Jackson SH, Bosetti F. Inhibition of NADPH oxidase promotes alternative and anti-inflammatory microglial activation during neuroinflammation. J Neurochem. 2012;120(2):292-301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Hashizume K, Suzuki S, Ichikawa K, Takeda T, Kobayashi M. Effect of active vitamin D3 on the levels of NADPH-dependent cytosolic 3,5,3’-triiodo-L-thyronine-binding protein. Biochem Biophys Res Commun. 1991;177(1):388-394. [DOI] [PubMed] [Google Scholar]
  • 172.Stio M, Lunghi B, Iantomasi T, Vincenzini MT, Treves C. Effect of vitamin D deficiency and 1,25-dihydroxyvitamin D3 on rat heart metabolism. J Mol Cell Cardiol. 1994;26(11):1421-1428. [DOI] [PubMed] [Google Scholar]
  • 173.Bachelet M, Bader C, Merlot AM, Laborde K, Snarska J, Ulmann A. Cellular utilization of cytosolic NADPH in kidney and liver cells from rats fed a normal or a vitamin D-deficient diet. Cell Biochem Funct. 1983;1(1):25-29. [DOI] [PubMed] [Google Scholar]
  • 174.Husain K, Ferder L, Mizobuchi M, Finch J, Slatopolsky E. Combination therapy with paricalcitol and enalapril ameliorates cardiac oxidative injury in uremic rats. Am J Nephrol. 2009;29(5):465-472. [DOI] [PubMed] [Google Scholar]
  • 175.Bell NH, Shaw S, Turner RT. Evidence that 1,25-dihydroxyvitamin D3 inhibits the hepatic production of 25-hydroxyvitamin D in man. J Clin Invest. 1984;74(4):1540-1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Luong KV, Nguyen LT. Coexisting hyperparathyroidism and primary hyperparathyroidism with vitamin D-deficient osteomalacia in a Vietnamese immigrant. Endocr Pract. 1996;2(4):250-254. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Alzheimer's Disease and Other Dementias are provided here courtesy of SAGE Publications

RESOURCES