Next Article in Journal
Hepatotoxicity and Antimicrobial Resistance to Amoxicillin and Amoxicillin/Clavulanic Acid: Data Analysis from EudraVigilance
Previous Article in Journal
A Novel Schiff Base Probe Based on Fluorescein for Fluorometric and Colorimetric Dual-Mode Rapid Detection of Cu2+
Previous Article in Special Issue
Tricyclic Isatin Derivatives as Anti-Inflammatory Compounds with High Kinase Binding Affinity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin D Associated with Exercise Can Be Used as a Promising Tool in Neurodegenerative Disease Protection

Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(18), 3823; https://doi.org/10.3390/molecules30183823
Submission received: 22 July 2025 / Revised: 17 September 2025 / Accepted: 19 September 2025 / Published: 21 September 2025

Abstract

Neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease, multiple sclerosis, and Huntington’s disease, represent unmet medical and social needs. Still, no definitive cure exists for these illnesses, hence a therapeutic approach with molecules able to prevent/downtone/modify the disease seems highly attractive. Remarkably, a higher risk of neurodegenerative disease is associated with low vitamin D levels. Vitamin D is a multifaceted molecule able to target critical neuroinflammatory processes underlying neurodegeneration, acting through genomic or rapid signaling. This narrative review aims to focus on vitamin D’s potential to be an optimal neuroprotective molecule, based on its ability to target and counteract aberrant biomolecular processes involved in neuroinflammation/neurodegeneration. Noticeably, exercise can potentiate vitamin D’s protective effect through some anti-inflammatory actions exerted on shared biomolecular targets. Thus, although vitamin D is not strictly a drug, it could be potentially allocated within the therapeutic approach to neurodegenerative diseases in combination with adapted exercise, best as an early intervention. Topics on the complexity concerning the doses for supplementation and data discrepancy from trials are addressed. The urgent demand is to test and clarify vitamin D efficacy and safety, combined or not with exercise, in clinical settings.

Graphical Abstract

1. Introduction

Vitamin D-dependent regulation goes beyond classical bone phospho-calcium homeostasis and extends to other organ or tissue functions, including the central nervous system (CNS) [1]. Currently, vitamin D pleiotropic actions are acknowledged due to its ability to modulate many biological processes, including cell proliferation and differentiation, immune response, and inflammation [1].
Acting through the vitamin D receptor (VDR), which is expressed in almost all brain areas, vitamin D can behave as a neuroprotective molecule since it inhibits a variety of processes, i.e., oxidative stress, inflammation, and pathogenic protein formation and aggregation, which underlie and precede neurodegeneration [2]. In humans, vitamin D insufficiency increases the susceptibility to neurodegenerative diseases, such as Parkinson’s disease (PD), Alzheimer’s disease (AD), multiple sclerosis (MS), Huntington’s disease (HD), cognitive disorders, and dementia, although causative links are still to be fully elucidated [3]. Furthermore, lower vitamin D levels associate with higher neurodegenerative stages, whereas vitamin D sufficiency, i.e., reached after supplementation, seems to contribute to a better prognosis [4].
Currently, half of the people across the world exhibit poor vitamin D levels, with more than one billion people suffering from vitamin D deficiency—defined as less than 30 nmol/L (<12 ng/mL) [5,6].
Furthermore, the neurodegenerative occurrence, meaning both incidence and prevalence, increases with the increasing number of elderly people, representing a growing challenge in public health.
The aim of this narrative review is to focus on the potential neuroprotective role of vitamin D, which exhibits higher efficacy when combined with physical exercise (PE), since this molecule and PE share some neuroinflammatory/neurodegenerative pathways as common targets. Neuroinflammation and neurodegeneration are addressed as interacting parts of a vicious loop that needs to be counteracted as early as possible to control disease development. Based on its anti-inflammatory properties targeting the vicious loop, vitamin D is suggested as a complementary molecule to be allocated in therapeutic options for neurodegenerative diseases to ameliorate prevention, management, and outcome. Importantly, the synergistic effect between vitamin D and PE is discussed as an emerging attractive issue to focus on, hopefully suggesting new scenarios in a multidimensional approach to neurogenerative diseases.

2. The Versatile Identity of Vitamin D

This vitamin, classified as “D” since it is the fourth to be discovered, although belonging to the vitamin class, functions as a prototypical secosteroid hormone (a steroid subclass with a broken ring). In humans, the majority of vitamin D3, or cholecalciferol, originates mainly in skin from enzymatic conversion of the precursor 7-dehydrocholesterol under ultraviolet (UV) rays and only 20% from diet [7]. Although the 25(OH)D is the more abundant and stable metabolite in circulation—used, in fact, for vitamin D serum level determination—the 1.25(OH2)D metabolite retains 500 times higher affinity for VDR [8].
Upon VDR binding, vitamin D signals via a hormone-like classic mechanism, heterodimerizing with 9-cis-retinoic acid receptor X receptor (RXR) to form the dimeric complex VDR:RXR, which targets the promoter regions within vitamin D-responsive elements (VDREs) and starts gene transcription [9,10]. Vitamin D can also signal via a non-genomic rapid mechanism, in which the VDR translocates from cytoplasm to plasma membrane through caveolae, which are plasmalemma microdomains highly specialized for macromolecule transcytosis, and activates transmembrane rapid signal transduction paths within seconds to minutes [11].
A membrane-associated rapid-response steroid-binding protein (MARRS) dedicated to vitamin D non-genomic effects is described in [12]. Through these mechanisms, vitamin D exerts a fine-tuned regulation on an exceptionally broad spectrum of biological activities, highly impacting human health.
Indeed, besides the classical regulation of calcium-phosphorus for bone homeostasis, vitamin D status regulates several biological processes, i.e., energy metabolism and skeletal muscle function. Importantly, vitamin D drives the immune response to an anti-inflammatory profile, i.e., downregulating T helper (Th)1/Th17 phenotypes, both deeply involved in inflammatory overresponse [13,14], and shifting human brain microglial from proinflammatory M1 to anti-inflammatory M2 phenotype [15]. Hence, considering its anti-inflammatory role, vitamin D has gained high interest as a neuroprotective molecule.
Although neuroinflammation is usually described as the cause preceding neurodegeneration, these processes occur almost simultaneously and establish a mutual detrimental dialogue, as addressed in the following paragraph.

3. Neuroinflammation and Neurodegeneration: A Two-Way Route

The question of whether inflammation is the cause or consequence of neurodegeneration is still debated. Like the whole body, inflammation in the CNS is a physiological immune response to harmful stimuli, i.e., traumas or infections, behaving as a safeguard of the cell microenvironment [16]. CNS resident immune cells, the microglia, represent the main immune defense in the brain, which, under challenge, trigger a defensive neuroinflammatory response characterized by cellular and signaling overresponse, dysfunction of the brain–blood barrier (BBB), release of inflammatory mediators, such as reactive oxygen species (ROS), nitrogen species, cytokines, and chemokines [17,18].
Whenever this process is inappropriately protracted, a chronic neuroinflammatory state occurs and potentially represents the first step toward different neurodegenerative diseases, independently of their specific characteristics [17,19].

The Systemic-Local Detrimental Loop in CNS

The causes inducing chronic neuroinflammation are numerous and still far from being well elucidated. The immunocyte infiltration into CNS tissue from the peripheral areas likely occurs due to a consequence of a robust inflammatory peripheral response, triggered, i.e., by systemic exposure to inflammation inducers, such as lipopolysaccharides, or viruses [20]. This process would first prompt the activation of microglia to produce and release into the BBB inflammatory mediators, which, in turn, recall from the peripheral immune system additional infiltration of immune cells, such as T cells and macrophages, further weakening the barrier [21,22]. Thus, a vicious circle starts.
The activated microglia, astrocytes (a subtype of glial cells with homeostatic function), oligodendrocytes, pericytes, endothelial cells, and peripheral immunocytes interconnect and establish a complex web of proinflammatory molecules and aberrant signaling, contributing to inflammatory microenvironment maintenance and tissue injury [23,24]. These processes converge in a self-enhancing, detrimental circle between systemic and local areas.
In this scenario, aberrant signaling cascades engaging pattern recognition receptors (PRRs), such as toll-like receptors (TLRs), receptors for advanced glycation end (RAGE) products, nod-like receptor protein (NLRP)3 inflammasome, damage-associated molecular pattern (DAMP) profile, accumulation of reactive oxygen species (ROS), cytokines and chemokines, protein aggregates, adenosine triphosphate (ATP), and mitochondrial DNA released by damaged neurons, act as inflammation enhancers, as exhaustively described elsewhere [25].
Furthermore, a huge release of proinflammatory cytokines, including tumor necrosis factor (TNFα), interleukin (IL)-6, IL-23, IL-1β, interferon (IFN)γ, and granulocyte macrophage colony-stimulating factor (GM-CSF), further sustains and contributes to disease progression. As examples, IL-23, IL-6, or IL-1β, produced by CNS resident immunocytes, amplify T cell pathogenic activity in neuroinflammation, and GM-CSF promotes tissue damage through macrophage infiltration [26,27]. Noticeably, these cytokines are targets of vitamin D, as addressed in the following paragraph.
The large cytokine release, once activated, induces anomalous signaling cascades, which, in turn, trigger synaptic/neuronal loss and aberrant protein release/accumulation, which ends in plaque formation and worsens neuronal dysfunction, such as amyloid beta (Aβ) in AD [28]. Indeed, brain tissues and blood from AD patients show high levels of inflammatory cytokines derived from microglia, which, failing to efficiently remove Aβ, become chronically activated and intensify protein aggregation or plaque formation.
Similarly, in PD, the aberrant deposition of α-synuclein ends in aggregates (oligomers or fibrils), which further activate microglia [29]. The excess of superoxide, nitric oxide, and TNFα derived from chronically activated microglia culminates in neurotoxic effects, typical of the disease. Furthermore, the CNSs of PD patients show massive CD4+ and CD8+ T cell infiltration, which likely precedes pathological α-synuclein deposition [17]. The mechanisms underlying PD development are intricate and still not fully elucidated. Moreover, the gut–brain axis seems to contribute to the disease progression via gut dysbiosis-dependent inflammatory cascade and BBB permeability modification [30]. The latter gives permission for extra cell infiltration. Hence, the inflammatory loop is perpetuated and exhibits a mutual-promotion pattern to aggravate neurodegeneration, acknowledged as a pathological condition common to several neurodegenerative diseases exhibiting different pathogenetic mechanisms, protein aggregates, and genetic variations [19]. Figure 1 summarizes the link between neuroinflammation and neurodegeneration.
Undeniably, limiting the neuroinflammatory loop as soon as possible, ideally before clinical signs, would be of pivotal importance.
The current therapeutic approaches to neurodegenerative diseases such as AD, PD, HD, or MS include anti-inflammatory and antioxidant drugs, to limit inflammation, immune infiltration, and metabolic failure in CNS; molecules regulating myelin deposition and increasing autophagic flux in neurons; drugs enabling amyloid clearance; molecules targeting transcriptional regulation or epigenetic mechanisms, aimed at modulating widespread downstream effects [31].
To date, most of these drugs show promising results in experimental models, often not immediately translated to humans, showing, indeed, controversial results between preclinical models and trials. The discrepancy might also be due to species-specific features.
In addition, data from trials, whose primary outcome is aimed at targeting neurodegeneration-associated protein aggregates, are too often disappointing because the cognitive function is not reestablished, even when aggregates are eliminated by the treatment [32].
In this scenario, there is growing evidence and interest in vitamin D status that can contribute to brain health or disease, depending on sufficient or deficient levels, respectively.

4. Vitamin D Status: Towards Neuroprotection or Neurodegeneration

The putative role of vitamin D as a neuroprotectant mainly relies on its multitargeting effects within the CNS, since this molecule can target neuronal cells and non-neural cells, such as endothelial cells involved in neurovascular dysfunction [33,34,35].
Upon binding to the VDR, which is ubiquitously expressed in human brain areas—hypothalamus, hippocampal formation, amygdala, stria terminalis, and neocortex [36,37,38], vitamin D can substantially counteract homeostasis loss both with genomic and non-genomic mechanisms, exerting pro-survival/anti-inflammatory effects to prevent neuronal cell loss or demyelination [39,40,41].
Vitamin D-deficient status seems to contribute to the progression of neurodegeneration via Wnt signaling failure [42]. Interestingly, Wnt signaling is highly conserved and it is known to exert a critical role in synaptic maintenance and neuronal functions, acting either through canonical or non-canonical paths, whose deregulation is found in AD [43].
Vitamin D is documented to maintain the Wnt pathway to warrant brain plasticity and promote brain circuit regulation, both in early and late life [44]. Accordingly, a reduced expression of VDR impairs Wnt-dependent cascade affecting neural cell signaling and unbalancing the neurogenic homeostasis, such as in cognitive impairments, as observed in hippocampal cells of old brains [42].
It seems that hypovitaminosis D can promote cognitive decline, reducing neurogenesis by upregulating Wnt inhibitors, such as Dickkopf-1 (DKK1); conversely, adequate vitamin D levels sustain Wnt signaling, repressing its inhibitors. This latter effect likely prolongs the time of stem cell quiescence, preserving the staminal reservoir [45]. So far, in neural cells, vitamin D seems to act as a co-activator of Wnt, whereas in other cell types, i.e., cancer cells, this molecule acts as a Wnt antagonist [46]. To date, the complex interaction between vitamin D and Wnt is dependent on the microenvironment and not fully elucidated.
The risk of PD development seems to be closely associated with vitamin D deficiency and vitamin D–VDR axis failure [47]. Indeed, VDR-regulated genes, such as nuclear receptor-related 1 protein (Nurr1) gene (critical in dopaminergic system development and maintenance) and tyrosine hydroxylase (which catalyzes L-DOPA formation), are likely co-responsible for pathomechanisms of PD [48,49]. Within the nigrostriatal signaling, the substantia nigra, the area deeply involved in dopaminergic neuron degeneration, highly expresses both VDR and 1α-hydroxylase, specific for the enzymatic conversion to vitamin D’s active form, and it is likely that a deficient vitamin D/VDR system accelerates the disease [50]. Experimental in vitro studies document that the addition of calcitriol can restore pro-survival cell signaling, such as phosphatidylinositol 3-kinase (PI3K) pathway, and increase glial cell-derived neurotrophic factor (GDNF) [51,52]. Consequently, cell viability and proliferation are both improved, and oxidative stress/ROS production, α-synuclein aggregation, and neurotoxicity are reduced [53].
Remarkably, restoring the function of the vitamin D-VDR axis can inhibit aberrancies in NLRP3/caspase1 signaling, mitigate inflammation and membrane permeabilization, significantly attenuating microglia overactivation, as recently shown in a PD experimental study [54]. Nevertheless, there is evidence that while PD risk increases with D deficiency, vitamin D supplementation plays little role, especially in well-established disease [55].
Indeed, upon VDR binding, vitamin D significantly reduces the level of proinflammatory cytokines such as TNFα, IL-1β, IL-6, deeply involved in neuroinflammatory and pathogenetic mechanisms in PD, AD, or MS neurodegeneration, and protects neural tissues from damage through the increase in anti-inflammatory cytokine IL-10 [56,57]. Vitamin D-induced IL-10 increase counteracts inflammation through induction of suppressor of cytokine signaling 3 (SOCS3) [58]. In the PD mouse model, vitamin D has been shown to increase other anti-inflammatory molecules, such as transforming growth factor (TGF)β, IL-4, and cluster of differentiation (CD)163, CD202, and CD206, involved against oxidative damage and in the shift to a protolerogenic profile [59].
Furthermore, vitamin D can modulate tissue architecture through extracellular matrix deposition and fibrosis development, targeting the enzymatic activity of metalloproteinases (MMP) and TGFβ [41,60].
Vitamin D is known to modulate mitochondrial function and reduce neuronal apoptosis via AMP-activated protein kinase/protein kinase B/glycogen synthase kinase (AMPK/AKT/GSK)-3β signaling path activation, displaying, therefore, a reparatory activity besides the neurogenic effect [61,62].
Vitamin D-induced maintenance of mitochondrial function entails ROS reduction and regulates antioxidant expression, on one side, via vitamin D-Klotho-nuclear factor erythroid 2-related factor 2 (Nrf2) regulatory network, glutathione, and superoxide dismutase expression upregulation, on the other side, via nitric oxide (NO)/inducible nitric oxide synthase (iNOS) downregulation [63,64,65,66].
Altogether, the anti-inflammatory/antioxidant/trophic effects of vitamin D undeniably converge in maintaining as intact as possible the synaptic plasticity, cell cytoskeleton, and molecular transport, whereas vitamin D deficiency results in loss of function [67,68,69].
In line with in vitro data, results from experimental models show that vitamin D intake in deficient status reduces oxidative stress and dopaminergic loss, restores endothelial function, and improves neurogenesis and working memory, highlighting the importance of administration timing, with the earliest intervention being the most efficient, as shown in PD and AD [70,71,72,73,74]. Of note, the observation on early administration timing is in line with the concept of limiting the neuroinflammatory loop as soon as possible, as previously addressed.
In PD patients, vitamin D can restore motor functionality and rebalance the immune homeostasis [75].
Vitamin D can mitigate the Th1 proinflammatory effects, significantly reducing both circulating and local Th1-type cytokines and chemokines, such as interferon gamma (IFNγ), TNFα, IL-1, IL-2, IL-6, IL-1β, and C-C or C-X-C motif chemokines [76,77]. This cytokine inhibition is mostly due to nuclear factor-kB (NF-kB) p65 block via the upregulation of the inhibitory protein inhibitor of kappa B-alpha (IkBa) [78].
This vitamin D preserves vascular cells from injury mainly through local reduction of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), cyclooxygenase 2 (COX-2), E-selectin, or vascular tissue expression of MMP-2 and MMP-9 [41,79].
Thus far, vitamin D-induced neuroprotection is exerted at the systemic and local levels. Inhibiting inflammatory molecules at the cell/tissue level seems highly intriguing since this effect potentially interrupts the self-detrimental loop between systemic and local compartments, which aggravates neurodegeneration.
We have previously reported on VDR agonists as tools for the resolution of inflammation, directly targeting different human cell types, i.e., T cells, cardiac and muscular cells, prostate and bladder cells, and renal cells activated by a maximal inflammatory challenge [80,81,82,83,84]. The inhibition of the release of chemotactic mediators is a pivotal step to interrupt the vicious inflammatory circle.
Some of the vitamin D-induced local and systemic effects are reported in Figure 2.
Studies on humans and animals on HD document that vitamin D deficiency likely directly correlates with motor abnormalities, whereas vitamin D supplementation leads to clinical symptom improvement [85,86,87].
Furthermore, HD is reported to be associated with bone metabolism alteration, showing significantly lower bone mineral density and disease-specific osteoporosis, likely related to the number of CAG repeats—dominantly inherited CAG trinucleotide repeat in the huntingtin gene—that represents the most frequently evaluated marker of disease severity [88,89,90].
These relevant issues contribute to impairment of independent ambulation in HD, and, consequently, to falls, autonomy loss, and transfer to nursing homes. Nevertheless, there are no specific dedicated studies, which are potentially of great interest. To date, epidemiological or clinical studies focusing on the relationship between vitamin D status and HD are lacking, except for the explorative study from Chel, reporting a positive inverse association between functional impairment and vitamin D level [85]. In vitro studies show that vitamin D induces the neurotrophins nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) in several neuronal cells, including striatal neurons, the main affected cell population in HD [91,92].
Some investigators report a relationship between reduced vitamin D blood level and higher risk of MS; accordingly, MS patients taking vitamin D show reduced neuroinflammation and neurodegeneration, likely due to an increase in tissue oxygenation [93,94]. As per data in the MS murine model, proteolipid protein (PLP), myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), and CNP increase after vitamin D intake [95,96]. Levels of two isoforms of vitamin D binding proteins and apolipoprotein E in the cerebrospinal fluid are suggested as potential biomarkers for MS diagnosis, albeit some other studies evidence no relationship between vitamin D level and MS etiopathogenesis [97,98,99]. Thus, the need for further investigations into vitamin D therapeutic activity in MS is undeniable.
Similarly, few studies have been carried out on vitamin D in amyotrophic lateral sclerosis (ALS). ALS development seems associated with vitamin D status via the regulation of immune components, involving toll-like receptors (TLR), major histocompatibility complex (MHC) class II molecules, poly (ADP-ribose) polymerase 1 (PARP1), and heme oxygenase-1 (HO-1) [100]. Furthermore, vitamin D status affects ALS through the direct control of important cell signaling mechanisms, i.e., mitogen-activated protein kinase (MAPK), ROS, Wnt/β-catenin, MMP, and nitric oxidase synthase [100]. It should be underlined that, although vitamin D supplementation can improve motor functional capacity and decrease muscle weakness, and, therefore, improve the quality of life, it cannot modify disease prognosis or outcome [101]. Thus far, it must be underlined once more that the importance of further investigations on the potential role of vitamin D in ALS pathophysiology. However, the advantage of using vitamin D to supplement deficient/insufficient status appears undeniably as an optimal approach. Nevertheless, this issue opens a window on another important question: what is the best dose for supplementation?
It is acknowledged that the vitamin D dose may vary according to the individual’s need, which depends on several factors, including diet, sunlight exposure, sex, latitude, lifestyle, and inadequate renal conversion or intestinal absorption [102,103,104]. Thus, supplementation doses should be calibrated in response to skeletal and extra-skeletal needs.
According to guidelines, a 25(OH)D sufficient serum concentration should be 50–100 nmol/L; the Central European guideline recommends 75–125 nmol/L [104,105,106]. To date, the latest report, “Consensus Statement on vitamin D Status Assessment and Supplementation: Whys, Whens, and Hows,” evidences that the optimal 25(OH)D level remains to be clarified, particularly for extra-skeletal needs [107]. Concerning the effect of vitamin D supplementation in neurodegenerative diseases, data are conflicting as well. Trials on vitamin D supplementation in AD, PD, or MS patients report either no effect or some benefits on cognitive function, independently of the taken dose (from 800 IU/day to 1000 IU/day, or 6000 IU/day), as recently summarized [3]. It is mandatory to underline that the lack of reliable results from the trials also depends on the lack of assay standardization, high variability in methods, and, consequently, a non-homogeneous approach for vitamin D supplementation (different forms, metabolites, and doses) [5,6,107].
Remarkably, vitamin D’s effect on the CNS can be affected by sedentary or active behavior. This topic deserves particular attention, since physical exercise (PE) can affect, per se, the risk of neurodegenerative diseases and could exert a multiplicative effect when combined with vitamin D [108,109], as addressed in the following paragraph.

5. Vitamin D Status and Exercise: A Possible Synergy in Neuroprotection

There is growing evidence of some beneficial effects of PE against neurodegenerative disorders through several simultaneous actions [108].
PE is defined as designed, organized, and repeated aerobic/anaerobic physical activity characterized by defined frequency, duration, and intensity, with the specific goal of enhancing or maintaining physical fitness [110].
Interestingly, engaging in PE on a regular basis may slow aging-related cognitive decline [111,112]. An acknowledged effect of PE is the improvement of blood flow and, consequently, cerebral flow, which supports neuronal plasticity.
Clinical research shows that PE leads to increases in gray matter volume, particularly in the frontal cortex and hippocampus [113] and elevates the level of neurotrophic factors, such as peripheral BDNF [114].
Nowadays, adapted PE is fully recognized as an anti-inflammatory intervention since it significantly reduces the level of proinflammatory cytokines, including the cytokines resident in the CNS [115,116]. This effect is undeniably due to the direct regulation of PE onto immune cells, which is noted as “exercise immunology” [117,118,119] but also depends on the biomolecules released during skeletal muscle working, named myokines, which all exert anti-inflammatory and regulatory effects onto several body functions, acting within a network established among almost all body tissues and organs [120,121,122,123]. More than 600 have been found, although their functions are still to be clarified [124]. There are neurotrophic/anti-inflammatory/regulatory myokines able to cross the BBB and involved in muscle–brain cross-talk, i.e., BDNF, IL-6, irisin, cathepsin B, or muscle-derived insulin-like growth factor 1 (IGF-1), as previously reported, although their fine-tuned mechanisms are not yet elucidated [125,126].
Remarkably, recent evidence suggests a possible synergy between PE and vitamin D to counteract neuroinflammation through a merged action on several common targets.
To date, most of the myokines able to cross the BBB are sustained by vitamin D. As examples, vitamin D, through BDNF modulation, mitigates molecular derangements, improves neuronal outcomes in animal models with neurogeneration, and might support neuroprotection in vitamin D-deficient subjects (<30 ng/mL) [127,128]. Furthermore, higher BDNF levels are associated with the modulation of microglial activation by triggering a shift from the proinflammatory M1 phenotype to the anti-inflammatory and neuroprotective M2 phenotype, which, in turn, downregulates the secretion of inflammatory cytokines, including IL-1β and TNFα [129,130].
Muscle-derived irisin and IL-6 are increased by vitamin D level or VDR expression [131,132,133]; vitamin D upregulates cathepsin B expression and positively correlates with IGF-1 [134,135].
Another example comes from the observation that regular exercise can enhance VDR expression in the brain, particularly in regions essential for cognitive, memory, and emotional regulation, such as the hippocampus and prefrontal cortex [109,136]. This effect can trigger a virtuous neuroprotective loop since the increased VDR expression improves neuronal sensitivity to vitamin D, therefore enhancing PE-induced effects.
In addition, both PE and vitamin D target NF-κB, the central regulator of inflammatory responses, to inhibit the production of proinflammatory genes, such as TNFα [137]. Another example of targets shared by PE and vitamin D comes from the activation of the Nrf2 pathway, which plays a crucial role in antioxidant defense. Of note, Nrf2 activation increases the expression of enzymes such as superoxide dismutase, glutathione peroxidase, and catalase, and reduces oxidative stress and neuroinflammation [138,139].
Vitamin D exerts similar anti-inflammatory and antioxidant effects by activating the Nrf2 signaling pathway, by enhancing the expression of antioxidant genes such as heme oxygenase-1 (H-1) and glutathione peroxidase 4 (GPX4), and reducing ferroptosis in aging brains, all processes contributing to the reduction of neuroinflammation and CNS oxidative damage [139].
So far, these common molecular targets support the hypothesis of potential synergistic effects between vitamin D and exercise, which is useful for the prevention and treatment of neurodegenerative diseases. From a recent paper reporting data in an experimental model, the combination of exercise training and vitamin D results in a synergistic effect on brain health by improving cognitive function, BBB integrity, and mitochondrial efficiency [140].
Table 1 shows the common targets and pathways modulated by PE and vitamin D, together with the induced effects that may merge in a synergistic action.
Noticeably, it is generally acknowledged that PE increases testosterone in men depending on several factors, including types of exercise (endurance/resistance), training intensity and recovery times, and population (sedentary/active/athletes; young/old) [141]. PE can also induce the expression of androgen receptor (AR), resulting in general anti-inflammatory effects through STAT3, counteracting STAT3-induced sarcopenia [142]. Nowadays, androgens’ function is acknowledged to extend far beyond sexual function, reaching into the human brain to sustain neuroprotection, memory, and mitigate cognitive decline [143]. Indeed, androgens can directly modulate neuroplasticity, particularly in the hippocampus, and protect neurons from Aβ toxicity, involving several intracellular signaling cascades, such as extracellular signal-regulated kinase-cAMP response element-binding protein (ERK-CREB), MAPK/ERK, or protein tyrosine kinase (PTK) [143,144,145,146].
Androgen administration is hypothesized as a therapy for neurodegenerative diseases [147].
Whether or not vitamin D deficiency can impact testosterone levels in humans is still debated, as some studies reported a positive correlation between the levels of these hormones, while others did not, highlighting the need for further well-designed, long-term trials [148].
The research in humans on neuroprotection combining vitamin D and exercise and all related effects is still in its infancy and undeniably needs further investigations, both in vitro and in vivo.

6. Conclusions

Thus far, it is undeniable that vitamin D positively interferes with signal paths engaged in the pathogenesis of neurodegenerative diseases, i.e., targeting the cellular signals contributing to neuronal loss or abnormal protein aggregates, which represent the common features of neurodegenerative diseases. Indeed, vitamin D-induced multifaceted actions, such as anti-inflammatory, antioxidative, immunomodulatory, neurovascular, and trophic effects, merge and contribute to neuroprotection.
At the same time, the discrepancy reported in results from clinical studies makes the research on this topic jump a step back. To date, concerns about the conflicting data from clinical trials reflect the inconsistency of the methods used, including dose and duration variability of vitamin D treatment. Indeed, investigations are mandatory for well-designed clinical trials and further insights into molecular mechanisms. Finally, studies on regimens combining vitamin D supplementation and PE should be considered the main issues in a multidimensional therapeutic approach, which is currently quite neglected. Personalized interventions based on genetics, vitamin D levels, and fitness may represent the future of prevention strategies.
Neurodegenerative diseases are expanding—also due to the growing number of elderly populations—with no definite cure still found, and represent a critical unmet need impacting medical and social areas [149].
Noticeably, neurodegeneration has recently been acknowledged as a tumor-permissive/tumor-promoting process [150]. In this context, both vitamin D (suggested long ago as a cancer-limiting agent in immune and cancer cell proliferation/invasion/differentiation [151,152,153,154]) and PE (an emerging discipline in cancer care, noted as exercise oncology [155,156,157]) can be considered as mechanistically convergent against cancer secondary to neurodegenerative diseases.
In this scenario, the opportunity to think of vitamin D combined with PE in the treatment to reduce neurological illness burden seems highly attractive.

Author Contributions

G.F. draft writing, figure/table assembling, reference update; C.C. conceptualization, final revision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Autier, P.; Boniol, M.; Pizot, C.; Mullie, P. Vitamin D status and ill health: A systematic review. Lancet. Diabetes Endocrinol. 2014, 2, 76–89. [Google Scholar] [CrossRef]
  2. Eyles, D.W.; Smith, S.; Kinobe, R.; Hewison, M.; McGrath, J.J. Distribution of the vitamin D receptor and 1 alpha-hydroxylase in human brain. J. Chem. Neuroanat. 2005, 29, 21–30. [Google Scholar] [CrossRef]
  3. Wang, W.; Li, Y.; Meng, X. Vitamin D and neurodegenerative diseases. Heliyon 2023, 9, e12877. [Google Scholar] [CrossRef]
  4. Eyles, D.W.; Burne, T.H.; McGrath, J.J. Vitamin D, effects on brain development, adult brain function and the links between low levels of vitamin D and neuropsychiatric disease. Front. Neuroendocrinol. 2013, 34, 47–64. [Google Scholar] [CrossRef] [PubMed]
  5. Holick, M.F. The vitamin D deficiency pandemic: Approaches for diagnosis, treatment and prevention. Rev. Endocr. Metab. Disord. 2017, 18, 153–165. [Google Scholar] [CrossRef]
  6. Giustina, A.; Bouillon, R.; Binkley, N.; Sempos, C.; Adler, R.A.; Bollerslev, J.; Dawson-Hughes, B.; Ebeling, P.R.; Feldman, D.; Heijboer, A.; et al. Controversies in Vitamin D: A Statement From the Third International Conference. JBMR Plus 2020, 4, e10417. [Google Scholar] [CrossRef] [PubMed]
  7. Holick, M.F. Vitamin D: Evolutionary, physiological and health perspectives. Curr. Drug Targets 2011, 12, 4–18. [Google Scholar] [CrossRef]
  8. Bouillon, R.; Okamura, W.H.; Norman, A.W. Structure-function relationships in the vitamin D endocrine system. Endocr. Rev. 1995, 16, 200–257. [Google Scholar] [CrossRef] [PubMed]
  9. Goltzman, D. Functions of vitamin D in bone. Histochem. Cell Biol. 2018, 149, 305–312. [Google Scholar] [CrossRef] [PubMed]
  10. Pike, J.W.; Meyer, M.B.; Bishop, K.A. Regulation of target gene expression by the vitamin D receptor—An update on mechanisms. Rev. Endocr. Metab. Disord. 2012, 13, 45–55. [Google Scholar] [CrossRef]
  11. Buitrago, C.; Boland, R. Caveolae and caveolin-1 are implicated in 1alpha,25(OH)2-vitamin D3-dependent modulation of Src, MAPK cascades and VDR localization in skeletal muscle cells. J. Steroid Biochem. Mol. Biol. 2010, 121, 169–175. [Google Scholar] [CrossRef]
  12. Khanal, R.C.; Nemere, I. The ERp57/GRp58/1,25 D3-MARRS receptor: Multiple functional roles in diverse cell systems. Curr. Med. Chem. 2007, 14, 1087–1093. [Google Scholar] [CrossRef]
  13. Penna, G.; Adorini, L. 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J. Immunol. 2000, 164, 2405–2411. [Google Scholar] [CrossRef]
  14. Berer, A.; Stöckl, J.; Majdic, O.; Wagner, T.; Kollars, M.; Lechner, K.; Geissler, K.; Oehler, L. 1,25-Dihydroxyvitamin D(3) inhibits dendritic cell differentiation and maturation in vitro. Exp. Hematol. 2000, 28, 575–583. [Google Scholar] [CrossRef]
  15. Thu, V.T.A.; Hoang, T.X.; Kim, J.Y. 1,25-Dihydroxy Vitamin D3 Facilitates the M2 Polarization and β-Amyloid Uptake by Human Microglia in a TREM2-Dependent Manner. BioMed Res. Int. 2023, 2023, 3483411. [Google Scholar] [CrossRef]
  16. Ransohoff, R.M.; Brown, M.A. Innate immunity in the central nervous system. J. Clin. Investig. 2012, 122, 1164–1171. [Google Scholar] [CrossRef] [PubMed]
  17. Adamu, A.; Li, S.; Gao, F.; Xue, G. The role of neuroinflammation in neurodegenerative diseases: Current understanding and future therapeutic targets. Front. Aging Neurosci. 2024, 16, 1347987. [Google Scholar] [CrossRef]
  18. Ahmad, M.A.; Kareem, O.; Khushtar, M.; Akbar, M.; Haque, M.R.; Iqubal, A.; Haider, M.F.; Pottoo, F.H.; Abdulla, F.S.; Al-Haidar, M.B.; et al. Neuroinflammation: A Potential Risk for Dementia. Int. J. Mol. Sci. 2022, 23, 616. [Google Scholar] [CrossRef]
  19. Mayne, K.; White, J.A.; McMurran, C.E.; Rivera, F.J.; de la Fuente, A.G. Aging and Neurodegenerative Disease: Is the Adaptive Immune System a Friend or Foe? Front. Aging Neurosci. 2020, 12, 572090. [Google Scholar] [CrossRef] [PubMed]
  20. Sharma, R.; Zamani, A.; Dill, L.K.; Sun, M.; Chu, E.; Robinson, M.J.; O’Brien, T.J.; Shultz, S.R.; Semple, B.D. A systemic immune challenge to model hospital-acquired infections independently regulates immune responses after pediatric traumatic brain injury. J. Neuroinflammation 2021, 18, 72. [Google Scholar] [CrossRef] [PubMed]
  21. Qin, J.; Ma, Z.; Chen, X.; Shu, S. Microglia activation in central nervous system disorders: A review of recent mechanistic investigations and development efforts. Front. Neurol. 2023, 14, 1103416. [Google Scholar] [CrossRef] [PubMed]
  22. Ronaldson, P.T.; Davis, T.P. Regulation of blood-brain barrier integrity by microglia in health and disease: A therapeutic opportunity. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2020, 40 (Suppl. 1), S6–S24. [Google Scholar] [CrossRef]
  23. Paul, B.D.; Snyder, S.H.; Bohr, V.A. Signaling by cGAS-STING in Neurodegeneration, Neuroinflammation, and Aging. Trends Neurosci. 2021, 44, 83–96. [Google Scholar] [CrossRef] [PubMed]
  24. Costantini, E.; D’Angelo, C.; Reale, M. The Role of Immunosenescence in Neurodegenerative Diseases. Mediat. Inflamm. 2018, 2018, 6039171. [Google Scholar] [CrossRef]
  25. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  26. Hamilton, J.A. GM-CSF in inflammation. J. Exp. Med. 2020, 217, e20190945. [Google Scholar] [CrossRef]
  27. Codarri, L.; Gyülvészi, G.; Tosevski, V.; Hesske, L.; Fontana, A.; Magnenat, L.; Suter, T.; Becher, B. RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat. Immunol. 2011, 12, 560–567. [Google Scholar] [CrossRef]
  28. Sobue, A.; Komine, O.; Yamanaka, K. Neuroinflammation in Alzheimer’s disease: Microglial signature and their relevance to disease. Inflamm. Regen. 2023, 43, 26. [Google Scholar] [CrossRef]
  29. Stefanis, L. α-Synuclein in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009399. [Google Scholar] [CrossRef]
  30. Pajares, M.; IRojo, A.; Manda, G.; Boscá, L.; Cuadrado, A. Inflammation in Parkinson’s Disease: Mechanisms and Therapeutic Implications. Cells 2020, 9, 1687. [Google Scholar] [CrossRef] [PubMed]
  31. de la Fuente, A.G.; Pelucchi, S.; Mertens, J.; Di Luca, M.; Mauceri, D.; Marcello, E. Novel therapeutic approaches to target neurodegeneration. Br. J. Pharmacol. 2023, 180, 1651–1673. [Google Scholar] [CrossRef] [PubMed]
  32. Landel, V.; Annweiler, C.; Millet, P.; Morello, M.; Féron, F. Vitamin D, Cognition and Alzheimer’s Disease: The Therapeutic Benefit is in the D-Tails. J. Alzheimer’s Dis. JAD 2016, 53, 419–444. [Google Scholar] [CrossRef] [PubMed]
  33. Laso’ n, W.; Jantas, D.; Leśkiewicz, M.; Regulska, M.; Basta-Kaim, A. The Vitamin D Receptor as a Potential Target for the Treatment of Age-Related Neurodegenerative Diseases Such as Alzheimer’s and Parkinson’s Diseases: A Narrative Review. Cells 2023, 12, 660. [Google Scholar] [CrossRef]
  34. Lang, F.; Ma, K.; Leibrock, C.B. 1,25(OH)2D3 in Brain Function and Neuropsychiatric Disease. Neuro Signals 2019, 27, 40–49. [Google Scholar] [CrossRef]
  35. Lasoń, W.; Jantas, D.; Leśkiewicz, M.; Regulska, M.; Basta-Kaim, A. Vitamin D3 and Ischemic Stroke: A Narrative Review. Antioxidants 2022, 11, 2120. [Google Scholar] [CrossRef]
  36. Smolders, J.; Schuurman, K.G.; van Strien, M.E.; Melief, J.; Hendrickx, D.; Hol, E.M.; van Eden, C.; Luchetti, S.; Huitinga, I. Expression of vitamin D receptor and metabolizing enzymes in multiple sclerosis-affected brain tissue. J. Neuropathol. Exp. Neurol. 2013, 72, 91–105. [Google Scholar] [CrossRef]
  37. Landel, V.; Stephan, D.; Cui, X.; Eyles, D.; Feron, F. Differential expression of vitamin D-associated enzymes and receptors in brain cell subtypes. J. Steroid Biochem. Mol. Biol. 2018, 177, 129–134. [Google Scholar] [CrossRef] [PubMed]
  38. Stumpf, W.E.; O’Brien, L.P. 1,25 (OH)2 vitamin D3 sites of action in the brain. An autoradiographic study. Histochemistry 1987, 87, 393–406. [Google Scholar] [CrossRef]
  39. Baas, D.; Prüfer, K.; Ittel, M.E.; Kuchler-Bopp, S.; Labourdette, G.; Sarliève, L.L.; Brachet, P. Rat oligodendrocytes express the vitamin D(3) receptor and respond to 1,25-dihydroxyvitamin D(3). Glia 2000, 31, 59–68. [Google Scholar] [CrossRef]
  40. Zorrilla Veloz, R.I.; McKenzie, T.; Palacios, B.E.; Hu, J. Nuclear hormone receptors in demyelinating diseases. J. Neuroendocrinol. 2022, 34, e13171. [Google Scholar] [CrossRef]
  41. Holick, M.F.; Mazzei, L.; García Menéndez, S.; Martín Giménez, V.M.; Al Anouti, F.; Manucha, W. Genomic or Non-Genomic? A Question about the Pleiotropic Roles of Vitamin D in Inflammatory-Based Diseases. Nutrients 2023, 15, 767. [Google Scholar] [CrossRef]
  42. Gómez-Oliva, R.; Geribaldi-Doldán, N.; Domínguez-García, S.; Carrascal, L.; Verástegui, C.; Nunez-Abades, P.; Castro, C. Vitamin D deficiency as a potential risk factor for accelerated aging, impaired hippocampal neurogenesis and cognitive decline: A role for Wnt/β-catenin signaling. Aging 2020, 12, 13824–13844. [Google Scholar] [CrossRef]
  43. Arrázola, M.S.; Silva-Alvarez, C.; Inestrosa, N.C. How the Wnt signaling pathway protects from neurodegeneration: The mitochondrial scenario. Front. Cell. Neurosci. 2015, 9, 166. [Google Scholar] [CrossRef]
  44. Uthaiah, C.A.; Beeraka, N.M.; Rajalakshmi, R.; Ramya, C.M.; Madhunapantula, S.V. Role of Neural Stem Cells and Vitamin D Receptor (VDR)-Mediated Cellular Signaling in the Mitigation of Neurological Diseases. Mol. Neurobiol. 2022, 59, 4065–4105. [Google Scholar] [CrossRef] [PubMed]
  45. Ziebell, F.; Dehler, S.; Martin-Villalba, A.; Marciniak-Czochra, A. Revealing age-related changes of adult hippocampal neurogenesis using mathematical models. Development 2018, 145, dev153544. [Google Scholar] [CrossRef]
  46. González-Sancho, J.M.; Larriba, M.J.; Muñoz, A. Wnt and Vitamin D at the Crossroads in Solid Cancer. Cancers 2020, 12, 3434. [Google Scholar] [CrossRef]
  47. Agliardi, C.; Guerini, F.R.; Zanzottera, M.; Bolognesi, E.; Meloni, M.; Riboldazzi, G.; Zangaglia, R.; Sturchio, A.; Casali, C.; Di Lorenzo, C.; et al. The VDR FokI (rs2228570) polymorphism is involved in Parkinson’s disease. J. Neurol. Sci. 2021, 428, 117606. [Google Scholar] [CrossRef] [PubMed]
  48. Decressac, M.; Volakakis, N.; Björklund, A.; Perlmann, T. NURR1 in Parkinson disease—From pathogenesis to therapeutic potential. Nat. Rev. Neurol. 2013, 9, 629–636. [Google Scholar] [CrossRef] [PubMed]
  49. Haavik, J.; Toska, K. Tyrosine hydroxylase and Parkinson’s disease. Mol. Neurobiol. 1998, 16, 285–309. [Google Scholar] [CrossRef]
  50. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
  51. Shirazi, H.A.; Rasouli, J.; Ciric, B.; Rostami, A.; Zhang, G.X. 1,25-Dihydroxyvitamin D3 enhances neural stem cell proliferation and oligodendrocyte differentiation. Exp. Mol. Pathol. 2015, 98, 240–245. [Google Scholar] [CrossRef]
  52. da Silva Teixeira, S.; Harrison, K.; Uzodike, M.; Rajapakshe, K.; Coarfa, C.; He, Y.; Xu, Y.; Sisley, S. Vitamin D actions in neurons require the PI3K pathway for both enhancing insulin signaling and rapid depolarizing effects. J. Steroid Biochem. Mol. Biol. 2020, 200, 105690. [Google Scholar] [CrossRef]
  53. Zhang, Y.; Ji, W.; Zhang, S.; Gao, N.; Xu, T.; Wang, X.; Zhang, M. Vitamin D Inhibits the Early Aggregation of α-Synuclein and Modulates Exocytosis Revealed by Electrochemical Measurements. Angew. Chem. 2022, 61, e202111853. [Google Scholar] [CrossRef]
  54. Zheng, Z.; Chen, M.; Feng, S.; Zhao, H.; Qu, T.; Zhao, X.; Ruan, Q.; Li, L.; Guo, J. VDR and deubiquitination control neuronal oxidative stress and microglial inflammation in Parkinson’s disease. Cell Death Discov. 2024, 10, 150. [Google Scholar] [CrossRef]
  55. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Selim, H.M.; Alexiou, A.; Papadakis, M.; Negm, W.A.; Batiha, G.E. Does vitamin D protect or treat Parkinson’s disease? A narrative review. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 397, 33–40. [Google Scholar] [CrossRef]
  56. Sharma, S.; Borski, C.; Hanson, J.; Garcia, M.A.; Link, C.D.; Hoeffer, C.; Chatterjee, A.; Nagpal, P. Identifying an Optimal Neuroinflammation Treatment Using a Nanoligomer Discovery Engine. ACS Chem. Neurosci. 2022, 13, 3247–3256. [Google Scholar] [CrossRef]
  57. Kim, M.S.; Kim, Y.H.; Kim, M.S.; Kwon, B.; Cho, H.R. Efficacy and Safety of Early Anti-inflammatory Drug Therapy for Secondary Injury in Traumatic Brain Injury. World Neurosurg. 2023, 172, e646–e654. [Google Scholar] [CrossRef] [PubMed]
  58. Boontanrart, M.; Hall, S.D.; Spanier, J.A.; Hayes, C.E.; Olson, J.K. Vitamin D3 alters microglia immune activation by an IL-10 dependent SOCS3 mechanism. J. Neuroimmunol. 2016, 292, 126–136. [Google Scholar] [CrossRef] [PubMed]
  59. Calvello, R.; Cianciulli, A.; Nicolardi, G.; De Nuccio, F.; Giannotti, L.; Salvatore, R.; Porro, C.; Trotta, T.; Panaro, M.A.; Lofrumento, D.D. Vitamin D Treatment Attenuates Neuroinflammation and Dopaminergic Neurodegeneration in an Animal Model of Parkinson’s Disease, Shifting M1 to M2 Microglia Responses. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2017, 12, 327–339. [Google Scholar] [CrossRef] [PubMed]
  60. Sayeed, I.; Turan, N.; Stein, D.G.; Wali, B. Vitamin D deficiency increases blood-brain barrier dysfunction after ischemic stroke in male rats. Exp. Neurol. 2019, 312, 63–71. [Google Scholar] [CrossRef]
  61. Li, Y.; Li, X.; Xu, S.; Zhao, Y.; Pang, M.; Zhang, X.; Wang, X.; Wang, Y. 1,25-D3 attenuates cerebral ischemia injury by regulating mitochondrial metabolism via the AMPK/AKT/GSK3β pathway. Front. Aging Neurosci. 2022, 14, 1015453. [Google Scholar] [CrossRef] [PubMed]
  62. Rastegar-Moghaddam, S.H.; Alipour, F.; Hosseini, M.; Ebrahimzadeh-Bideskan, A. Anti-apoptotic and neurogenic properties in the hippocampus as possible mechanisms for learning and memory improving impacts of vitamin D in hypothyroid rats during the growth period. Life Sci. 2023, 312, 121209. [Google Scholar] [CrossRef] [PubMed]
  63. Calton, E.K.; Keane, K.N.; Soares, M.J. The potential regulatory role of vitamin D in the bioenergetics of inflammation. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 367–373. [Google Scholar] [CrossRef]
  64. Garcion, E.; Sindji, L.; Montero-Menei, C.; Andre, C.; Brachet, P.; Darcy, F. Expression of inducible nitric oxide synthase during rat brain inflammation: Regulation by 1,25-dihydroxyvitamin D3. Glia 1998, 22, 282–294. [Google Scholar] [CrossRef]
  65. Garcion, E.; Sindji, L.; Leblondel, G.; Brachet, P.; Darcy, F. 1,25-dihydroxyvitamin D3 regulates the synthesis of gamma-glutamyl transpeptidase and glutathione levels in rat primary astrocytes. J. Neurochem. 1999, 73, 859–866. [Google Scholar] [CrossRef]
  66. Berridge, M.J. Vitamin D, reactive oxygen species and calcium signalling in ageing and disease. Philos. Trans. R. Soc. London Ser. B Biol. Sci. 2016, 371, 20150434. [Google Scholar] [CrossRef] [PubMed]
  67. Eyles, D.; Almeras, L.; Benech, P.; Patatian, A.; Mackay-Sim, A.; McGrath, J.; Féron, F. Developmental vitamin D deficiency alters the expression of genes encoding mitochondrial, cytoskeletal and synaptic proteins in the adult rat brain. J. Steroid Biochem. Mol. Biol. 2007, 103, 538–545. [Google Scholar] [CrossRef]
  68. Almeras, L.; Eyles, D.; Benech, P.; Laffite, D.; Villard, C.; Patatian, A.; Boucraut, J.; Mackay-Sim, A.; McGrath, J.; Féron, F. Developmental vitamin D deficiency alters brain protein expression in the adult rat: Implications for neuropsychiatric disorders. Proteomics 2007, 7, 769–780. [Google Scholar] [CrossRef]
  69. de Abreu, D.A.F.; Eyles, D.; Féron, F. Vitamin D, a neuro-immunomodulator: Implications for neurodegenerative and autoimmune diseases. Psychoneuroendocrinology 2009, 34 (Suppl. 1), S265–S277. [Google Scholar] [CrossRef]
  70. Kim, H.A.; Perrelli, A.; Ragni, A.; Retta, F.; De Silva, T.M.; Sobey, C.G.; Retta, S.F. Vitamin D Deficiency and the Risk of Cerebrovascular Disease. Antioxidants 2020, 9, 327. [Google Scholar] [CrossRef]
  71. Farghali, M.; Ruga, S.; Morsanuto, V.; Uberti, F. Can Brain Health Be Supported by Vitamin D-Based Supplements? A Critical Review. Brain Sci. 2020, 10, 660. [Google Scholar] [CrossRef]
  72. Morello, M.; Landel, V.; Lacassagne, E.; Baranger, K.; Annweiler, C.; Féron, F.; Millet, P. Vitamin D Improves Neurogenesis and Cognition in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 6463–6479. [Google Scholar] [CrossRef]
  73. Pal, R.; Choudhury, S.; Kumar, H.; Dey, S.; Das, N.; Basu, B.R. Vitamin D deficiency and genetic polymorphisms of vitamin D-associated genes in Parkinson’s disease. Eur. J. Neurosci. 2023, 58, 3362–3377. [Google Scholar] [CrossRef]
  74. Mirarchi, A.; Albi, E.; Beccari, T.; Arcuri, C. Microglia and Brain Disorders: The Role of Vitamin D and Its Receptor. Int. J. Mol. Sci. 2023, 24, 11892. [Google Scholar] [CrossRef]
  75. Li, D.; Ma, X.; Zhang, W.; Zhong, P.; Li, M.; Liu, S. Impact of vitamin D3 supplementation on motor functionality and the immune response in Parkinson’s disease patients with vitamin D deficiency. Sci. Rep. 2025, 15, 25154. [Google Scholar] [CrossRef]
  76. Alcalá-Santiago, Á.; Toscano-Sánchez, R.; Márquez-López, J.C.; González-Jurado, J.A.; Fernández-Pachón, M.S.; García-Villanova, B.; Pedroche, J.; Rodríguez-Martín, N.M. The Synergic Immunomodulatory Effect of Vitamin D and Chickpea Protein Hydrolysate in THP-1 Cells: An In Vitro Approach. Int. J. Mol. Sci. 2024, 25, 12628. [Google Scholar] [CrossRef]
  77. Zha, X.; Liu, M.; Ruan, S.; Jiang, Y.; Wang, S. Relationship between serum vitamin D levels and pro-inflammatory cytokines in patients with rheumatoid arthritis combined with cardiovascular disease. Clin. Rheumatol. 2025, 44, 3467–3475. [Google Scholar] [CrossRef] [PubMed]
  78. Colotta, F.; Jansson, B.; Bonelli, F. Modulation of inflammatory and immune responses by vitamin D. J. Autoimmun. 2017, 85, 78–97. [Google Scholar] [CrossRef]
  79. Naeini, A.E.; Moeinzadeh, F.; Vahdat, S.; Ahmadi, A.; Hedayati, Z.P.; Shahzeidi, S. The Effect of Vitamin D Administration on Intracellular Adhesion Molecule-1 and Vascular Cell Adhesion Molecule-1 Levels in Hemodialysis Patients: A Placebo-controlled, Double-blinded Clinical Trial. J. Res. Pharm. Pract. 2017, 6, 16–20. [Google Scholar] [CrossRef] [PubMed]
  80. Scolletta, S.; Colletti, M.; Di Luigi, L.; Crescioli, C. Vitamin D receptor agonists target CXCL10: New therapeutic tools for resolution of inflammation. Mediat. Inflamm. 2013, 2013, 876319. [Google Scholar] [CrossRef] [PubMed]
  81. Sottili, M.; Cosmi, L.; Borgogni, E.; Sarchielli, E.; Maggi, L.; Francalanci, M.; Vannelli, G.B.; Ronconi, E.; Adorini, L.; Annunziato, F.; et al. Immunomodulatory effects of BXL-01-0029, a less hypercalcemic vitamin D analogue, in human cardiomyocytes and T cells. Exp. Cell Res. 2009, 315, 264–273. [Google Scholar] [CrossRef]
  82. Crescioli, C.; Sottili, M.; Bonini, P.; Cosmi, L.; Chiarugi, P.; Romagnani, P.; Vannelli, G.B.; Colletti, M.; Isidori, A.M.; Serio, M.; et al. Inflammatory response in human skeletal muscle cells: CXCL10 as potential therapeutic target. Eur. J. Cell Biol. 2012, 91, 139–149. [Google Scholar] [CrossRef]
  83. Morelli, A.; Vignozzi, L.; Filippi, S.; Vannelli, G.B.; Ambrosini, S.; Mancina, R.; Crescioli, C.; Donati, S.; Fibbi, B.; Colli, E.; et al. BXL-628, a vitamin D receptor agonist effective in benign prostatic hyperplasia treatment, prevents RhoA activation and inhibits RhoA/Rho kinase signaling in rat and human bladder. Prostate 2007, 67, 234–247. [Google Scholar] [CrossRef]
  84. Sagrinati, C.; Sottili, M.; Mazzinghi, B.; Borgogni, E.; Adorini, L.; Serio, M.; Romagnani, P.; Crescioli, C. Comparison between VDR analogs and current immunosuppressive drugs in relation to CXCL10 secretion by human renal tubular cells. Transpl. Int. 2010, 23, 914–923. [Google Scholar] [CrossRef] [PubMed]
  85. Chel, V.G.; Ooms, M.E.; van der Bent, J.; Veldkamp, F.; Roos, R.A.; Achterberg, W.P.; Lips, P. High prevalence of vitamin D deficiency and insufficiency in patients with manifest Huntington disease: An explorative study. Derm. Endocrinol. 2013, 5, 348–351. [Google Scholar] [CrossRef]
  86. Molnár, M.F.; Török, R.; Szalárdy, L.; Sümegi, E.; Vécsei, L.; Klivényi, P. High-dose 1,25-dihydroxyvitamin D supplementation elongates the lifespan of Huntington’s disease transgenic mice. Acta Neurobiol. Exp. 2016, 76, 176–181. [Google Scholar] [CrossRef]
  87. Rai, S.N.; Singh, P.; Steinbusch, H.W.M.; Vamanu, E.; Ashraf, G.; Singh, M.P. The Role of Vitamins in Neurodegenerative Disease: An Update. Biomedicines 2021, 9, 1284. [Google Scholar] [CrossRef]
  88. de Miranda, R.C.; Di Lorenzo, N.; Andreoli, A.; Romano, L.; De Santis, G.L.; Gualtieri, P.; De Lorenzo, A. Body composition and bone mineral density in Huntington’s disease. Nutrition 2019, 59, 145–149. [Google Scholar] [CrossRef] [PubMed]
  89. Goodman, A.O.; Barker, R.A. Body composition in premanifest Huntington’s disease reveals lower bone density compared to controls. PLoS Curr. 2011, 3, RRN1214. [Google Scholar] [CrossRef] [PubMed]
  90. Duyao, M.; Ambrose, C.; Myers, R.; Novelletto, A.; Persichetti, F.; Frontali, M.; Folstein, S.; Ross, C.; Franz, M.; Abbott, M. Trinucleotide repeat length instability and age of onset in Huntington’s disease. Nat. Genet. 1993, 4, 387–392. [Google Scholar] [CrossRef]
  91. Zuccato, C.; Ciammola, A.; Rigamonti, D.; Leavitt, B.R.; Goffredo, D.; Conti, L.; MacDonald, M.E.; Friedlander, R.M.; Silani, V.; Hayden, M.R.; et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington’s disease. Science 2001, 293, 493–498. [Google Scholar] [CrossRef]
  92. Manjari, S.K.V.; Maity, S.; Poornima, R.; Yau, S.Y.; Vaishali, K.; Stellwagen, D.; Komal, P. Restorative Action of Vitamin D3 on Motor Dysfunction Through Enhancement of Neurotrophins and Antioxidant Expression in the Striatum. Neuroscience 2022, 492, 67–81. [Google Scholar] [CrossRef] [PubMed]
  93. Smolders, J.; Damoiseaux, J.; Menheere, P.; Hupperts, R. Vitamin D as an immune modulator in multiple sclerosis, a review. J. Neuroimmunol. 2008, 194, 7–17. [Google Scholar] [CrossRef] [PubMed]
  94. Sintzel, M.B.; Rametta, M.; Reder, A.T. Vitamin D and Multiple Sclerosis: A Comprehensive Review. Neurol. Ther. 2018, 7, 59–85. [Google Scholar] [CrossRef]
  95. Mashayekhi, F.; Salehi, Z. Administration of vitamin D3 induces CNPase and myelin oligodendrocyte glycoprotein expression in the cerebral cortex of the murine model of cuprizone-induced demyelination. Folia Neuropathol. 2016, 54, 259–264. [Google Scholar] [CrossRef]
  96. Savran, Z.; Baltaci, S.B.; Aladag, T.; Mogulkoc, R.; Baltaci, A.K. Vitamin D and Neurodegenerative Diseases Such as Multiple Sclerosis (MS), Parkinson’s Disease (PD), Alzheimer’s Disease (AD), and Amyotrophic Lateral Sclerosis (ALS): A Review of Current Literature. Curr. Nutr. Rep. 2025, 14, 77. [Google Scholar] [CrossRef]
  97. Müller, T.; Lohse, L.; Blodau, A.; Frommholz, K. Vitamin D rise enhances blood perfusion in patients with multiple sclerosis. J. Neural Transm. 2019, 126, 1631–1636. [Google Scholar] [CrossRef]
  98. Perga, S.; Giuliano Albo, A.; Lis, K.; Minari, N.; Falvo, S.; Marnetto, F.; Caldano, M.; Reviglione, R.; Berchialla, P.; Capobianco, M.A.; et al. Vitamin D Binding Protein Isoforms and Apolipoprotein E in Cerebrospinal Fluid as Prognostic Biomarkers of Multiple Sclerosis. PLoS ONE 2015, 10, e0129291. [Google Scholar] [CrossRef]
  99. Disanto, G.; Ramagopalan, S.V.; Para, A.E.; Handunnetthi, L. The emerging role of vitamin D binding protein in multiple sclerosis. J. Neurol. 2011, 258, 353–358. [Google Scholar] [CrossRef]
  100. Long, K.V.; Nguyễn, L.T. Roles of vitamin D in amyotrophic lateral sclerosis: Possible genetic and cellular signaling mechanisms. Mol. Brain 2013, 6, 16. [Google Scholar] [CrossRef] [PubMed]
  101. Libonati, L.; Onesti, E.; Gori, M.C.; Ceccanti, M.; Cambieri, C.; Fabbri, A.; Frasca, V.; Inghilleri, M. Vitamin D in amyotrophic lateral sclerosis. Funct. Neurol. 2017, 32, 35–40. [Google Scholar] [CrossRef] [PubMed]
  102. Holick, M.F. Vitamin D deficiency. N. Engl. J. Med. 2007, 357, 266–281. [Google Scholar] [CrossRef]
  103. Cranney, A.; Horsley, T.; O’Donnell, S.; Weiler, H.; Puil, L.; Ooi, D.; Atkinson, S.; Ward, L.; Moher, D.; Hanley, D.; et al. Effectiveness and safety of vitamin D in relation to bone health. Evid. Rep. Technol. Assess. 2007, 158, 1–235. [Google Scholar]
  104. Bouillon, R.; van Schoor, N.M.; Gielen, E.; Boonen, S.; Mathieu, C.; Vanderschueren, D.; Lips, P. Optimal vitamin D status: A critical analysis on the basis of evidence-based medicine. J. Clin. Endocrinol. Metab. 2013, 98, E1283–E1304. [Google Scholar] [CrossRef]
  105. Lips, P.; Cashman, K.D.; Lamberg-Allardt, C.; Bischoff-Ferrari, H.A.; Obermayer-Pietsch, B.; Bianchi, M.L.; Stepan, J.; El-Hajj Fuleihan, G.; Bouillon, R. Current vitamin D status in European and Middle East countries and strategies to prevent vitamin D deficiency: A position statement of the European Calcified Tissue Society. Eur. J. Endocrinol. 2019, 180, P23–P54. [Google Scholar] [CrossRef]
  106. El-Hajj Fuleihan, G.; Bouillon, R.; Clarke, B.; Chakhtoura, M.; Cooper, C.; McClung, M.R.; Singh, R. Serum 25-hydroxyvitamin D levels: Variability, knowledge gaps and the concept of a desirable range. J. Bone Miner. Res. 2015, 30, 1119–1133. [Google Scholar] [CrossRef] [PubMed]
  107. Giustina, A.; Bilezikian, J.P.; Adler, R.A.; Banfi, G.; Bikle, D.D.; Binkley, N.C.; Bollerslev, J.; Bouillon, R.; Brandi, M.L.; Casanueva, F.F.; et al. Consensus Statement on Vitamin D Status Assessment and Supplementation: Whys, Whens, and Hows. Endocr. Rev. 2024, 45, 625–654. [Google Scholar] [CrossRef]
  108. Ben Ezzdine, L.; Dhahbi, W.; Dergaa, I.; Ceylan, H.İ.; Guelmami, N.; Ben Saad, H.; Chamari, K.; Stefanica, V.; El Omri, A. Physical activity and neuroplasticity in neurodegenerative disorders: A comprehensive review of exercise interventions, cognitive training, and AI applications. Front. Neurosci. 2025, 19, 1502417. [Google Scholar] [CrossRef]
  109. Guo, J.; Mo, H.; Zuo, L.; Zhang, X. Association of physical activity and vitamin D deficiency with cognitive impairment in older adults: A population based cross-sectional analysis. Front. Nutr. 2024, 11, 1390903. [Google Scholar] [CrossRef] [PubMed]
  110. World Health Organization, T. Global recommendations on physical activity for health. In Global Recommendations on Physical Activity for Health; World Health Organization: Geneva, Switzerland, 2010; p. 60. [Google Scholar]
  111. Bherer, L.; Erickson, K.I.; Liu-Ambrose, T. A review of the effects of physical activity and exercise on cognitive and brain functions in older adults. J. Aging Res. 2013, 2013, 657508. [Google Scholar] [CrossRef]
  112. Hamer, M.; Chida, Y. Physical activity and risk of neurodegenerative disease: A systematic review of prospective evidence. Psychol. Med. 2009, 39, 3–11. [Google Scholar] [CrossRef]
  113. Colcombe, S.J.; Erickson, K.I.; Scalf, P.E.; Kim, J.S.; Prakash, R.; McAuley, E.; Elavsky, S.; Marquez, D.X.; Hu, L.; Kramer, A.F. Aerobic exercise training increases brain volume in aging humans. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2006, 61, 1166–1170. [Google Scholar] [CrossRef]
  114. Hötting, K.; Schickert, N.; Kaiser, J.; Röder, B.; Schmidt-Kassow, M. The Effects of Acute Physical Exercise on Memory, Peripheral BDNF, and Cortisol in Young Adults. Neural Plast. 2016, 2016, 6860573. [Google Scholar] [CrossRef]
  115. Beltran, G.; Park, K.S.; Arellano, J.; Reyes, S.; Park, J.; Cho, S.K. Effects of exercise intensities on changes in pro-/anti-inflammatory cytokines following 8-weeks of aerobic exercise training. Physiology 2024, 39 (Suppl. 1), 570. [Google Scholar] [CrossRef]
  116. Seo, D.Y.; Heo, J.W.; Ko, J.R.; Kwak, H.B. Exercise and Neuroinflammation in Health and Disease. Int. Neurourol. J. 2019, 23 (Suppl. 2), S82–S92. [Google Scholar] [CrossRef] [PubMed]
  117. Nieman, D.C.; Wentz, L.M. The compelling link between physical activity and the body’s defense system. J. Sport Health Sci. 2019, 8, 201–217. [Google Scholar] [CrossRef]
  118. Nieman, D.C.; Pence, B.D. Exercise immunology: Future directions. J. Sport Health Sci. 2020, 9, 432–445. [Google Scholar] [CrossRef] [PubMed]
  119. Forte, P.; Branquinho, L.; Ferraz, R. The Relationships between Physical Activity, Exercise, and Sport on the Immune System. Int. J. Environ. Res. Public Health 2022, 19, 6777. [Google Scholar] [CrossRef]
  120. Lee, J.H.; Jun, H.S. Role of Myokines in Regulating Skeletal Muscle Mass and Function. Front. Physiol. 2019, 10, 42. [Google Scholar] [CrossRef]
  121. Whitham, M.; Parker, B.L.; Friedrichsen, M.; Hingst, J.R.; Hjorth, M.; Hughes, W.E.; Egan, C.L.; Cron, L.; Watt, K.I.; Kuchel, R.P.; et al. Extracellular Vesicles Provide a Means for Tissue Crosstalk during Exercise. Cell Metab. 2018, 27, 237–251.e4. [Google Scholar] [CrossRef]
  122. Jin, L.; Han, S.; Lv, X.; Li, X.; Zhang, Z.; Kuang, H.; Chen, Z.; Lv, C.A.; Peng, W.; Yang, Z.; et al. The muscle-enriched myokine Musclin impairs beige fat thermogenesis and systemic energy homeostasis via Tfr1/PKA signaling in male mice. Nat. Commun. 2023, 14, 4257. [Google Scholar] [CrossRef]
  123. Severinsen, M.C.K.; Pedersen, B.K. Muscle-Organ Crosstalk: The Emerging Roles of Myokines. Endocr. Rev. 2020, 41, 594–609. [Google Scholar] [CrossRef]
  124. Bettariga, F.; Taaffe, D.R.; Galvão, D.A.; Lopez, P.; Bishop, C.; Markarian, A.M.; Natalucci, V.; Kim, J.S.; Newton, R.U. Exercise training mode effects on myokine expression in healthy adults: A systematic review with meta-analysis. J. Sport Health Sci. 2024, 13, 764–779. [Google Scholar] [CrossRef]
  125. Kostka, M.; Morys, J.; Małecki, A.; Nowacka-Chmielewska, M. Muscle-brain crosstalk mediated by exercise-induced myokines—Insights from experimental studies. Front. Physiol. 2024, 15, 1488375. [Google Scholar] [CrossRef]
  126. Pedersen, B.K. Physical activity and muscle-brain crosstalk. Nat. Rev. Endocrinol. 2019, 15, 383–392. [Google Scholar] [CrossRef] [PubMed]
  127. Aladdin, N.; Ghareib, S.A. Vitamin D3 Exerts a Neuroprotective Effect in Metabolic Syndrome Rats: Role of BDNF/TRKB/Akt/GS3Kβ Pathway. J. Biochem. Mol. Toxicol. 2024, 38, e70082. [Google Scholar] [CrossRef] [PubMed]
  128. Skoczek-Rubińska, A.; Cisek-Woźniak, A.; Molska, M.; Heyser, M.; Trocholepsza, M.; Pietrzak, S.; Mruczyk, K. Impact of Vitamin D Status and Supplementation on Brain-Derived Neurotrophic Factor and Mood-Cognitive Outcomes: A Structured Narrative Review. Nutrients 2025, 17, 2655. [Google Scholar] [CrossRef] [PubMed]
  129. Jiang, T.; Sun, Q.; Chen, S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson’s disease and Alzheimer’s disease. Prog. Neurobiol. 2016, 147, 1–19. [Google Scholar] [CrossRef]
  130. Guo, S.; Wang, H.; Yin, Y. Microglia Polarization From M1 to M2 in Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 815347. [Google Scholar] [CrossRef]
  131. Sanesi, L.; Dicarlo, M.; Pignataro, P.; Zerlotin, R.; Pugliese, F.; Columbu, C.; Carnevale, V.; Tunnera, S.; Scillitani, A.; Grano, M.; et al. Vitamin D Increases Irisin Serum Levels and the Expression of Its Precursor in Skeletal Muscle. Int. J. Mol. Sci. 2023, 24, 4129. [Google Scholar] [CrossRef]
  132. Crescioli, C. Targeting Age-Dependent Functional and Metabolic Decline of Human Skeletal Muscle: The Geroprotective Role of Exercise, Myokine IL-6, and Vitamin D. Int. J. Mol. Sci. 2020, 21, 1010. [Google Scholar] [CrossRef] [PubMed]
  133. Pojednic, R.M.; Ceglia, L.; Lichtenstein, A.H.; Dawson-Hughes, B.; Fielding, R.A. Vitamin D receptor protein is associated with interleukin-6 in human skeletal muscle. Endocrine 2015, 49, 512–520. [Google Scholar] [CrossRef]
  134. Mathiasen, I.S.; Foghsgaard, L.; Hansen, C.M.; Jäättelä, M. Sensitization to TNF-induced apoptosis by 1,25-dihydroxy vitamin D3 involves up-regulation of the TNF receptor 1 and cathepsin B. Int. J. Cancer 2001, 93, 224–231. [Google Scholar] [CrossRef] [PubMed]
  135. Ameri, P.; Giusti, A.; Boschetti, M.; Murialdo, G.; Minuto, F.; Ferone, D. Interactions between vitamin D and IGF-I: From physiology to clinical practice. Clin. Endocrinol. 2013, 79, 457–463. [Google Scholar] [CrossRef]
  136. da Costa, R.O.; Gadelha-Filho, C.V.J.; de Aquino, P.E.A.; Lima, L.A.R.; de Lucena, J.D.; Ribeiro, W.L.C.; Lima, F.A.V.; Neves, K.R.T.; de Barros Viana, G.S. Vitamin D (VD3) Intensifies the Effects of Exercise and Prevents Alterations of Behavior, Brain Oxidative Stress, and Neuroinflammation, in Hemiparkinsonian Rats. Neurochem. Res. 2023, 48, 142–160. [Google Scholar] [CrossRef]
  137. Al-Rasheed, N.M.; Al-Rasheed, N.M.; Bassiouni, Y.A.; Hasan, I.H.; Al-Amin, M.A.; Al-Ajmi, H.N.; Mohamad, R.A. Vitamin D attenuates pro-inflammatory TNF-α cytokine expression by inhibiting NF-κB/p65 signaling in hypertrophied rat hearts. J. Physiol. Biochem. 2015, 71, 289–299. [Google Scholar] [CrossRef]
  138. Radak, Z.; Chung, H.Y.; Goto, S. Systemic adaptation to oxidative challenge induced by regular exercise. Free Radic. Biol. Med. 2008, 44, 153–159. [Google Scholar] [CrossRef]
  139. Li, J.; Cao, Y.; Xu, J.; Li, J.; Lv, C.; Gao, Q.; Zhang, C.; Jin, C.; Wang, R.; Jiao, R.; et al. Vitamin D Improves Cognitive Impairment and Alleviates Ferroptosis via the Nrf2 Signaling Pathway in Aging Mice. Int. J. Mol. Sci. 2023, 24, 15315. [Google Scholar] [CrossRef]
  140. Kim, T.; Kim, D.; Kim, Y.; Kim, J.; Kang, S.; Cho, J. Combined Effects of Exercise and Vitamin D on Neuroinflammation, Blood-Brain Barrier Integrity, Oxidative Stress, and Cognitive Function in Nonpathological Mice. Int. Neurourol. J. 2025, 29 (Suppl. 1), S22–S34. [Google Scholar] [CrossRef] [PubMed]
  141. Riachy, R.; McKinney, K.; Tuvdendorj, D.R. Various Factors May Modulate the Effect of Exercise on Testosterone Levels in Men. J. Funct. Morphol. Kinesiol. 2020, 5, 81. [Google Scholar] [CrossRef]
  142. Hashimoto, S.; Hosoi, T.; Yakabe, M.; Matsumoto, S.; Hashimoto, M.; Akishita, M.; Ogawa, S. Exercise-induced vitamin D receptor and androgen receptor mediate inhibition of IL-6 and STAT3 in muscle. Biochem. Biophys. Rep. 2023, 37, 101621. [Google Scholar] [CrossRef] [PubMed]
  143. Kuwahara, N.; Nicholson, K.; Isaacs, L.; MacLusky, N.J. Androgen Effects on Neural Plasticity. Androg. Clin. Res. Ther. 2021, 2, 216–230. [Google Scholar] [CrossRef]
  144. Guo, G.; Kang, L.; Geng, D.; Han, S.; Li, S.; Du, J.; Wang, C.; Cui, H. Testosterone modulates structural synaptic plasticity of primary cultured hippocampal neurons through ERK—CREB signalling pathways. Mol. Cell. Endocrinol. 2020, 503, 110671. [Google Scholar] [CrossRef]
  145. Foradori, C.D.; Weiser, M.J.; Handa, R.J. Non-genomic actions of androgens. Front. Neuroendocrinol. 2008, 29, 169–181. [Google Scholar] [CrossRef]
  146. Pike, C.J.; Nguyen, T.V.; Ramsden, M.; Yao, M.; Murphy, M.P.; Rosario, E.R. Androgen cell signaling pathways involved in neuroprotective actions. Horm. Behav. 2008, 53, 693–705. [Google Scholar] [CrossRef]
  147. Bianchi, V.E.; Rizzi, L.; Bresciani, E.; Omeljaniuk, R.J.; Torsello, A. Androgen Therapy in Neurodegenerative Diseases. J. Endocr. Soc. 2020, 4, bvaa120. [Google Scholar] [CrossRef]
  148. Monson, N.R.; Klair, N.; Patel, U.; Saxena, A.; Patel, D.; Ayesha, I.E.; Nath, T.S. Association Between Vitamin D Deficiency and Testosterone Levels in Adult Males: A Systematic Review. Cureus 2023, 15, e45856. [Google Scholar] [CrossRef] [PubMed]
  149. Wang, S.; Jiang, Y.; Yang, A.; Meng, F.; Zhang, J. The Expanding Burden of Neurodegenerative Diseases: An Unmet Medical and Social Need; Advance online publication. Aging Dis. 2024, 16, 2937–2952. [Google Scholar] [CrossRef] [PubMed]
  150. Bautista, J.; Coral-Riofrio, E.C.; Suárez Urresta, S.; Palacios-Zavala, D.; Echeverría, C.E.; Araujo-Abad, S.; López-Cortés, A. Neurodegeneration rewires the tumor microenvironment via the neuro–immune–cancer axis; Advance online publication. iScience 2025. [Google Scholar] [CrossRef]
  151. Reichel, H.; Koeffler, H.P.; Norman, A.W. The role of the vitamin D endocrine system in health and disease. N. Engl. J. Med. 1989, 320, 980–991. [Google Scholar] [CrossRef]
  152. Dallavalasa, S.; Tulimilli, S.V.; Bettada, V.G.; Karnik, M.; Uthaiah, C.A.; Anantharaju, P.G.; Nataraj, S.M.; Ramashetty, R.; Sukocheva, O.A.; Tse, E.; et al. Vitamin D in Cancer Prevention and Treatment: A Review of Epidemiological, Preclinical, and Cellular Studies. Cancers 2024, 16, 3211. [Google Scholar] [CrossRef]
  153. Marchiani, S.; Bonaccorsi, L.; Ferruzzi, P.; Crescioli, C.; Muratori, M.; Adorini, L.; Forti, G.; Maggi, M.; Baldi, E. The vitamin D analogue BXL-628 inhibits growth factor-stimulated proliferation and invasion of DU145 prostate cancer cells. J. Cancer Res. Clin. Oncol. 2006, 132, 408–416. [Google Scholar] [CrossRef]
  154. Carlberg, C.; Muñoz, A. An update on vitamin D signaling and cancer. Semin. Cancer Biol. 2022, 79, 217–230. [Google Scholar] [CrossRef] [PubMed]
  155. Watson, G.; Coyne, Z.; Houlihan, E.; Leonard, G. Exercise oncology: An emerging discipline in the cancer care continuum. Postgrad. Med. 2022, 134, 26–36. [Google Scholar] [CrossRef] [PubMed]
  156. Mizrahi, D.; Rees-Punia, E.; Newton, R.U.; Sandler, C.X. The expanding role of exercise oncology in cancer care: An editorial highlighting emerging research. JSAMS Plus 2024, 4, 100078. [Google Scholar] [CrossRef]
  157. Avancini, A.; Borsati, A.; Toniolo, L.; Ciurnelli, C.; Belluomini, L.; Budolfsen, T.; Lillelund, C.; Milella, M.; Quist, M.; Pilotto, S. Physical activity guidelines in oncology: A systematic review of the current recommendations. Crit. Rev. Oncol. Hematol. 2025, 210, 104718. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the mutual loop between neuroinflammation and neurodegeneration. The initial steps trigger a proinflammatory signaling cascade that leads to neurodegenerative processes, which contribute to and perpetuate neuroinflammation.
Figure 1. Schematic representation of the mutual loop between neuroinflammation and neurodegeneration. The initial steps trigger a proinflammatory signaling cascade that leads to neurodegenerative processes, which contribute to and perpetuate neuroinflammation.
Molecules 30 03823 g001
Figure 2. Vitamin D status drives toward neuroinflammation or neuroprotection. Adequate levels of vitamin D result in an overall anti-inflammatory effect, targeting both local and systemic mechanisms involved in neuroinflammation and neurodegeneration; up and down arrows indicate up- and downregulation, respectively.
Figure 2. Vitamin D status drives toward neuroinflammation or neuroprotection. Adequate levels of vitamin D result in an overall anti-inflammatory effect, targeting both local and systemic mechanisms involved in neuroinflammation and neurodegeneration; up and down arrows indicate up- and downregulation, respectively.
Molecules 30 03823 g002
Table 1. Common Targets of Physical Exercise and Vitamin D.
Table 1. Common Targets of Physical Exercise and Vitamin D.
Targets/PathwaysPE EffectsVitamin D EffectsSynergistic Effects
Myokines↑ BDNF, ↑ Irisin, ↑ IL-6, ↑ Cathepsin B, ↑ IGF-1 [125,126]
↑ Plasticity, gray matter volume [113], anti-inflammatory effects [114]
↑ BDNF [127,128], ↑ Irisin [131],
↑ IL-6 [132,133], ↑ Cathepsin B [134],
↑ IGF-1 [135]
↓ molecular derangements [127,128]
↑ neurogeneration and neuroprotection [127,128]
↑ Microglial shift M1→M2 [129,130]
↓ IL-1β, TNFα, CN Sinflammation [129,130]
↑ neuronal sensitivity via exercise-induced
↑ VDR/Vitamin D [136]
VDR↑ VDR in the brain (hippocampus, PFC) [136]↑ VDR expression in cognitive areas [136]↑ neuronal sensitivity to vitamin D and PE-induced effects [136]
NF-κB↓ proinflammatory signaling [137]↓ inflammatory gene expression [137]↓ TNFα, CNS inflammation [137]
Nrf2↑ antioxidant enzymes [138,139]↑ HO-1, GPX4 [139]
↓ ferroptosis [139]
↓ oxidative and neuroinflammatory stress [138,139]
Proinflammatory cytokines↓ via immunoregulation [117,118,119]
↑ myokines [120,121,122,123]
↓ via NF-κB [137], ↑ Nrf2 [138,139]↓ TNFα, IL-1β [129,130]
Plasticity/Cerebral blood flow↑ plasticity and flow [111,112]↑ VDR-supported [136]↑ multidimensional neuroprotective effects [140]
Molecular and signaling paths as common targets modulated by vitamin D and PE, potentially resulting in a synergistic effect; up and down arrows indicate up- and downregulation, respectively. Abbreviations are reported. PE: Physical Exercise; BDNF: Brain-Derived Neurotrophic Factor; IL-6: Interleukin-6; IGF-1: Insuline-like Growth Factor 1; VDR: Vitamin D Receptor; IL-1β: Interleukin-1 beta; TNFα: Tumor Necrosis Factor alpha; CNS: Central Nervous System; NF-κB: Nuclear Factor-kB; Nrf2: Nuclear Factor Erythroid 2-Related Factor 2; HO-1: Heme Oxygenase 1; GPX4: Glutathione Peroxidase 4; M1/M2: Microglial phenotypes: M1 (proinflammatory), M2 (anti-inflammatory); PFC: Prefrontal Cortex.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Farina, G.; Crescioli, C. Vitamin D Associated with Exercise Can Be Used as a Promising Tool in Neurodegenerative Disease Protection. Molecules 2025, 30, 3823. https://doi.org/10.3390/molecules30183823

AMA Style

Farina G, Crescioli C. Vitamin D Associated with Exercise Can Be Used as a Promising Tool in Neurodegenerative Disease Protection. Molecules. 2025; 30(18):3823. https://doi.org/10.3390/molecules30183823

Chicago/Turabian Style

Farina, Gabriele, and Clara Crescioli. 2025. "Vitamin D Associated with Exercise Can Be Used as a Promising Tool in Neurodegenerative Disease Protection" Molecules 30, no. 18: 3823. https://doi.org/10.3390/molecules30183823

APA Style

Farina, G., & Crescioli, C. (2025). Vitamin D Associated with Exercise Can Be Used as a Promising Tool in Neurodegenerative Disease Protection. Molecules, 30(18), 3823. https://doi.org/10.3390/molecules30183823

Article Metrics

Back to TopTop