Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain‐derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone‐derived hormones, bone marrow‐derived cells, bone‐derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment.
Highlights
The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis.
Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms.
Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.
Keywords: Alzheimer's disease, amyloid beta, bone, bone‐derived proteins, brain, crosstalk, extracellular vesicles, intervention strategies
1. INTRODUCTION
Alzheimer's disease (AD) is the most common type of dementia. It is clinically characterized by progressive memory loss and cognitive decline in older adults. With an increasing number of patients being diagnosed with AD, it constitutes a serious burden to caregivers and society. 1 , 2 Senile plaques composed of amyloid beta (Aβ) peptide and neurofibrillary tangles consisting of hyperphosphorylated tau are the main neuropathological features of AD. 3 AD is conventionally regarded as a neurodegenerative disease of the brain. In recent years, numerous studies have shown that abnormalities in systemic organs and tissues contribute to brain AD‐type pathologies and cognitive impairment. 4 , 5 , 6 , 7 , 8 , 9 , 10 Large epidemiological studies have shown the associations of systemic comorbidities with AD and dementia risk. 11 , 12 , 13 Systemic factors from the peripheral system can enter the brain and mediate brain aging, neurodegeneration, and AD pathogenesis. 14 , 15 , 16 , 17 Thus, systemic organs and tissues are involved in the pathogenesis and progression of AD. Revealing the mechanisms by which the peripheral system affects AD occurrence and development could provide new insights for developing novel intervention strategies. 18
The skeletal system is one of the largest organs in the body and is traditionally viewed as a static organ that primarily provides support, protection, and movement. Bone functions are accomplished by osteoblasts (derived from mesenchymal cells with osteogenic functions), osteocytes (also thought to be bone‐resident cells), and osteoclasts (also thought to be bone resorption cells). In addition, bone marrow mesenchymal stem cells (BMSCs) and the bone microenvironment composed of inflammatory cells, endothelial cells, and Schwann cells may also be involved in maintaining bone homeostasis and bone repair. 19 , 20 Bone can be directly innervated by efferent nerves originating from the central nervous system (CNS), while various central neurohormones (eg, follicle‐stimulating hormone, thyroid‐stimulating hormone, adrenocorticotrophic hormone), neuropeptides (eg, neuropeptide Y, kisspeptin, vasoactive intestinal peptide), neurotransmitters (eg, glutamate, acetylcholine, dopamine), and their intracellular signaling pathways are also implicated in the regulation of bone metabolism. 21 , 22 , 23 , 24 Moreover, increasing evidence has indicated the importance of bone as an endocrine organ that connects systemic organs of the whole body including the brain. 25 , 26 The relationships between AD and bone diseases in mice and humans have been reported, emphasizing the crosstalk between bone and the brain in AD. 27 , 28 , 29 , 30 , 31 , 32 Therefore, we reviewed the clinical evidence for the associations between bone diseases and AD and investigated the impact of AD on bone metabolism and the underlying mechanisms involved. Here, we also discuss the underlying mechanisms through which bone influences AD pathogenesis, including mechanisms involving bone‐derived proteins, bone marrow‐derived cells, bone‐derived extracellular vesicles (EVs), and inflammation. In light of the crosstalk between bone and brain, we propose some bone‐based intervention strategies for the prevention and treatment of AD.
RESEARCH IN CONTEXT
Systematic review: Recently, increasing evidence has indicated a relationship between Alzheimer's disease (AD) and bone diseases in mice and humans. This literature review was performed by searching PubMed, Embase, and Web of Science, aiming to discuss the crosstalk between AD and bone and propose potential bone‐based intervention strategies for the prevention and treatment of AD.
Interpretation: In this review, we summarize the effects of AD from the perspectives of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain‐derived extracellular vesicles (EVs), and the autonomic nervous system. Moreover, bone‐derived hormones, bone marrow‐derived cells, bone‐derived EVs, and inflammation can influence brain AD pathology and cognitive function.
Future directions: More animal and clinical studies are needed to investigate the contribution of bone diseases and bone‐derived substances to AD pathogenesis. The identification of key molecules that promote AD in different bone diseases is also important. The active prevention and timely treatment of bone diseases in elderly people may be beneficial for the prevention and treatment of AD.
2. CLINICAL EVIDENCE FOR ASSOCIATIONS BETWEEN BONE DISEASES AND AD
Osteoporosis is a prevalent bone disorder characterized by a decrease in bone density and bone mass due to a variety of etiologies. A cohort study in 2005 demonstrated that low femoral neck bone mineral density (BMD) was associated with approximately twice the risk of AD and dementia in women, but no such association was observed in men. 33 Subsequent studies and meta‐analyses further confirmed an increased incidence of cognitive decline in osteoporosis patients. 34 , 35 , 36 , 37 , 38 Therefore, it can be inferred that certain pathophysiological changes occurring in bones may impact brain function. Conversely, AD is also a risk factor for osteoporosis. Clinical research has indicated that individuals with dementia have a greater likelihood of poor bone health and are more prone to developing fractures. 39 BMD was lower in an early AD group and was related to brain volume and memory ability, and higher BMD was associated with better memory performance in AD patients. 40 , 41 These findings suggest that changes in the brain of AD patients might influence bone functions. Recently, a Mendelian randomization analysis of large‐scale BMD and AD gene datasets revealed that low BMD was a risk factor of AD. 42 Systematic reviews and meta‐analyses have shown associations among osteoarthritis, bone loss, and AD. 43 , 44 , 45 A longitudinal study revealed that osteoarthritis was associated with earlier Aβ deposition and greater Aβ‐dependent tau deposition during follow‐up. 28 Alterations in hippocampal functional connectivity and levels of brain‐derived neurotrophic factor (BDNF) are associated with future cognitive decline in patients with osteoarthritis. 46 Therefore, a large amount of clinical evidence has associated bone diseases with AD and dementia (Table 1).
TABLE 1.
Clinical studies of an association between bone degenerative changes, cognitive impairment, and Alzheimer's disease pathologies.
| Study | Type | Participants | Variables | Correlations |
|---|---|---|---|---|
| [4] | Cross‐sectional study | 650 Koreans | BMD, MMSE | Positive |
| [28] | Observational study | 374 participants from ADNI database | Osteoarthritis, Aβ and tau accumulation in primary motor and somatosensory regions | Positive |
| [32] | Prospective cohort study | 3651 participants from Rotterdam Study | BMD, risk of developing AD | Negative |
| [33] | Prospective cohort study | 987 participants from the Framingham study | BMD, risk of developing all‐cause dementia and AD | Negative |
| [34] | Retrospective cohort study | 59966 Germans | Osteoporosis diagnosis, risk of developing dementia | Positive |
| [35] | Retrospective cohort study | 157988 Koreans | Osteoporosis diagnosis, risk of developing AD | Positive |
| [36] | Prospective cohort study | 2019 Chinese | BMD, risk of developing AD | Negative |
| [37] | Cross‐sectional study | 535 Chinese | BMD, MMSE | Positive |
| [39] | Retrospective cohort study | 8448 Chinese | Dementia diagnosis, risk of developing bone fracture | Positive |
| [40] | Cohort study | 140 Americans | BMD, cerebral atrophy and cognitive decline | Negative |
| [41] | Cohort study | 13113 participants from UK Biobank | BMD, gray matter volume | Positive |
| [42] | Mendelian randomization study | 394929 participants from UK Biobank | Heel BMD, risk of developing AD | Positive |
Abbreviations: AD, Alzheimer's disease; Aβ, amyloid beta; BMD, bone mineral density; MMSE, Mini‐Mental State Examination.
3. THE EFFECTS OF AD ON BONE FUNCTIONS
The brain is the central control center of systemic tissues and organs. The brain has been shown to control the growth and differentiation of bone cells and impact bone functions through chemical and electrical signals. 47 AD patients exhibit brain atrophy, neural circuit damage, and neurotransmitter imbalance. 48 , 49 , 50 , 51 Functional magnetic resonance imaging (MRI) revealed that AD patients exhibited no or less activation of the hippocampus and other medial temporal lobe structures during the completion of memory tasks. 52 Additionally, increased bone fracture rates and reduced BMD are commonly observed in patients with AD. 39 , 42 Therefore, neuropathological changes in the AD brain may lead to bone dysfunction.
3.1. Effect of AD pathogenic proteins on bone
Aβ peptides are proteolytic fragments of amyloid precursor protein (APP). Aβ42 is located mainly in the membrane and cytoplasm of osteocytes and the extracellular matrix. 53 In one study, the levels of APP and Aβ42 were markedly elevated in the osteoporotic bone tissues of humans and animals compared with those of controls. 53 Moreover, the Aβ42 levels in bone tissues were negatively correlated with BMD in patients. 53 Aβ42 was reported to enhance osteoclast differentiation and activation, suggesting that Aβ may play an important role in the pathogenesis of osteoporosis. 53 The Swedish mutation of APP (APPswe) could cause early‐onset AD. 54 APPswe can suppress osteoblast differentiation and cause osteoporosis, indicating that AD and osteoporosis may share conserved pathogenic mechanisms. 55 The osteoporotic deficit in young APPswe mice could be prevented by treatment with cognition‐improving antioxidants, suggesting that reactive oxygen species may be involved in the underlying mechanism of APPswe‐induced osteoporosis. 55 , 56 , 57 A subsequent study revealed that APP plays a physiological role in osteoblast survival and bone formation by regulating mitochondrial function. 58 Osteoclast activity was increased in younger APPswe mice (<4 months old) and decreased in older Tg2576 mice (>4 months old) compared to that in age‐ and sex‐matched wild‐type (WT) mice. 58 However, bone loss can still be observed in Tg2576 mice after 4 months of age or even at older ages. The decrease in osteoclast formation and activity in aged Tg2576 mice may be due to an increase in soluble advanced glycation end products. This dysregulation of bone remodeling in aged mice may lead to an increased incidence of fractures without altering overall bone density. 58 However, another study revealed that Aβ42 did not influence receptor activators of NF‐κB ligand (RANKL)‐induced osteoclastogenesis. 59 Instead, it promoted osteoclast differentiation by enhancing RANKL‐mediated activation of NF‐κB and TRAF6‐MAPK signaling pathways and increasing RANKL‐induced calcium oscillation. 59 In addition to amyloid plaques, intracellular neurofibrillary tangles consisting of hyperphosphorylated tau are also pathological features of AD. 60 Related studies indicated that tau may affect AD through Aβ‐dependent and Aβ‐independent pathways (eg, apolipoprotein E [APOE], the endocytic system, and cholesterol metabolism. 61 , 62 , 63 Early BMD reduction was observed in tauopathy‐specific AD model mice. 64 Another study revealed that arthritis model mice with tau gene knockout exhibited less bone loss, cartilage damage, and osteoclast activity due to reduced macrophage polarization. 65 Therefore, it can be inferred that two common pathological hallmarks of AD are involved in regulating bone homeostasis. So far, there are no reports about skeletal manifestations in familial AD, to which more attention should be paid in the future.
3.2. Effect of AD risk genes on bone
Several genetic factors have also been implicated in the association between AD and bone‐related diseases. The APOE gene is involved in more than half of AD cases, and its gene polymorphisms are considered the primary genetic risk factors for late‐onset AD. 66 , 67 APOE ε4 increases the risk of AD in a dose‐dependent manner by enhancing toxicity and inhibiting protective function. 67 A previous meta‐analysis indicated insufficient evidence linking the APOE genotype to BMD and fracture incidence. 68 In another cohort study, APOE ε4 was indicated to be associated with osteoporosis and an increased risk of fractures, whereas APOE ε3 had the opposite effect. 69 While APOE‐deficient mice and WT mice exhibited similar morphological changes in bone tissue, APOE‐deficient mice showed greater bone resorption than bone formation. 70 Another study demonstrated that the uptake of triglyceride‐rich lipoproteins by osteoblasts was reduced in APOE‐deficient mice. 71 This reduction resulted in elevated serum osteocalcin levels and ultimately led to increased bone formation. 71 A single‐cell atlas of the human infrapatellar fat pad and synovium implicated APOE signaling in osteoarthritis pathology, and inhibiting APOE signaling decreased the progression of osteoarthritis. 72 The complex role played by genetic polymorphisms of APOE underscores its plausible connection between AD and bones from various perspectives.
Triggering receptor expressed on myeloid cells 2 (TREM2) is expressed in microglia in the brain and in macrophages in the periphery, and it has been indicated to be involved in the pathogenesis of AD. 73 In bone tissue, TREM2 is expressed in osteoclasts and is a key regulator of osteoclast production. 74 , 75 In the periodontitis model, the TREM2‐mediated reactive oxygen species signal amplification cascade is important for osteoclast generation, and the accumulation of soluble Aβ42 oligomers in the periodontitis microenvironment further enhances this signal by direct interaction with TREM2. 74 Additionally, there is evidence that the TREM2 R47H variant can cause significant age‐ and sex‐dependent musculoskeletal changes in mice. 76 Nasu–Hakola disease, also called polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL), is caused by mutations in either the TREM2 or TYROBP gene and manifests primarily as progressive presenile dementia along with bone cysts and fractures. 77 , 78 Given that TREM2 expression predominantly occurs within microglia in the brain, some researchers have questioned whether Nasu–Hakola disease could be considered a “microglial disease.” 77 In conclusion, AD risk genes including APOE and TREM2 may be involved in AD, but the underlying mechanisms need further study.
3.3. Neurohormones connect AD and bone
Several studies have suggested that osteoporosis and AD often occur during perimenopause and menopause in women. 79 , 80 , 81 , 82 , 83 , 84 Perimenopause is characterized by increased follicle‐stimulating hormone (FSH) levels and decreased estrogen levels. With aging, a decrease in estrogen receptor α may contribute to cognitive impairment. 85 Additionally, estrogen was found to be involved in inducing APP degradation and regulating oxidative damage and energy metabolism. 85 , 86 In addition, estrogen can also exert neuroprotective effects by attenuating Aβ‐induced lipid peroxidation and glutamatergic excitotoxicity. 86 Overall, estrogen is important for maintaining normal brain function, and estrogen deficiency is associated with an increased risk of AD. 83 , 87 , 88 , 89 Similarly, estrogen can exert an anti‐osteoporotic effect by decelerating the rate of bone remodeling, inducing osteoclast apoptosis, and inhibiting osteoblast and osteocyte apoptosis. 90 , 91 , 92 Compared with senile osteoporosis, postmenopausal osteoporosis is characterized by an increased number of osteoclasts, leading to enhanced bone resorption. 93 Furthermore, in addition to directly impacting bone remodeling, a decrease in estrogen levels can result in elevated levels of certain inflammatory cytokines (such as interleukin 1 [IL‐1], IL‐6, and tumor necrosis factor α [TNF‐α]) and alterations in the immune cell profile (including increased T cells), ultimately affecting bone homeostasis. 93 , 94 In addition, studies have also demonstrated that mice with estrogen deficiency exhibit cartilage degeneration, subchondral bone damage, and other changes, which ultimately lead to osteoarthritis. 95 As a superior regulator of estrogen, FSH levels were elevated in the blood of AD patients. 96 FSH can directly act on brain neurons to accelerate APP and tau cleavage through the C/EBPβ‐δ‐secretase pathway, leading to AD pathologies and cognitive deficits. 97 FSH can promote bone loss through the stimulation of osteoclast production, but it is difficult to separate the action of FSH from that of estrogen. 98 Thus, estrogen and FSH are involved in the pathogenesis of both AD and bone diseases.
Thyroid‐stimulating hormone (TSH) is a pituitary gland‐secreted hormone that not only regulates thyroid function but also plays a role in bone remodeling. Clinical studies have shown that increased levels of free T3 and free T4 are associated with decreased BMD and that increased levels of TSH are protective for bone. 99 , 100 , 101 The anti‐osteoclastic effect of TSH reduces bone resorption, although its impact on osteoblasts varies depending on the stage of cell differentiation. 101 , 102 Similarly, low TSH levels are associated with a high incidence of AD. 103 A Mendelian randomization study suggested that genetically predicted elevated levels of TSH were associated with a reduced risk of AD, even when these levels were within normal ranges. 104 However, it has also been proposed that thyroid dysfunction may be a consequence rather than a cause of AD since the decrease in TSH could be related to pituitary degeneration due to AD pathology. 105 , 106 Similarly, hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis and elevated glucocorticoids can exacerbate the pathological changes in AD and contribute to bone loss. 107 , 108 , 109 , 110 , 111 , 112 , 113 Based on previous studies, the glucocorticoid cascade is hypothesized to have a causal relationship with hippocampal injury, and overactivation of the HPA axis in AD may contribute to tau phosphorylation and enhance Aβ toxicity. 114 , 115 In bone tissue, adrenocorticotropin is thought to stimulate osteoblast proliferation, and glucocorticoid can inhibit osteoblast differentiation, increase osteoclast production, and promote osteocyte apoptosis. 101 , 116 The hypothalamic–pituitary–target organ axis plays an important role in neuroendocrine regulation, and it may also play a ubiquitous and important role in the brain–bone axis, which requires further investigation.
3.4. Neuropeptides connect AD and bone
As endogenous active substances, neuropeptides such as neuropeptide Y (NPY) and kisspeptin are involved in several pathophysiological processes. Within the CNS, NPY is predominantly expressed in the hypothalamus and exerts neuroprotective effects against AD. 117 Clinical studies have consistently demonstrated a general decline in NPY levels among AD patients. 118 , 119 , 120 , 121 Animal models of AD have shown conflicting findings regarding cortical and hippocampal NPY levels. Some studies reported an increase, but others indicated a reduction in NPY‐responsive neurons during the early stages. 122 , 123 , 124 , 125 However, NPY is generally believed to play a neuroprotective role in AD by increasing the expression of nerve growth factor, alleviating the neurotoxic effects of Aβ, modulating neurogenesis, regulating calcium homeostasis, and alleviating neuroinflammation. 126 , 127 , 128 , 129 , 130 , 131 , 132 In bone tissue, receptors for NPY are expressed, for example, in osteoblasts and osteocytes. 117 NPY appears to play a paradoxical role in maintaining skeletal homeostasis. Researchers discovered that NPY mediated glucocorticoid‐induced trabecular bone degeneration through post‐translational modification of peroxisome proliferator‐activated receptor γ. 133 Senescent osteocytes secrete NPY, which can inhibit the expression of pro‐osteogenic and anti‐adipogenic transcription factors that enable the differentiation of BMSCs from osteoblasts to adipocytes. 134 A recent study reported that exogenous overexpression of NPY aggravated postmenopausal osteoporosis by regulating gut microbiota. 135 Downregulation of hypothalamic NPY expression can promote bone formation. 136 In addition, NPY receptor antagonists promoted bone formation and suppressed bone resorption in a rat model of osteoporosis. 135 , 137 Thus, NPY could inhibit osteogenesis and promote bone resorption. Moreover, NPY can enhance BMSC differentiation into osteoblasts by activating the Wnt pathway. 138 A study using rat models of femoral fractures revealed that antagonists targeting NPY receptors impeded fracture healing. 139 Furthermore, NPY can ameliorate osteoporosis by reducing osteoclast numbers through mobilizing hematopoietic stem/progenitor cells. 140 Therefore, NPY could also promote bone formation to some extent.
Kisspeptin, a hypothalamic neuropeptide, has traditionally been thought to be important for the regulation of reproduction. 141 An in vitro study showed that Aβ could stimulate the release of kisspeptin, which can bind to Aβ and antagonize its neurotoxicity. 142 In bone, loss of function of the kisspeptin signaling pathway was associated with delayed skeletal maturation. 143 In addition, kisspeptin increased osteoblast production and inhibited bone resorption by osteoclasts in vitro, and the effect of kisspeptin on osteoblasts was verified in healthy men. 144 Another study demonstrated that the deletion of estrogen receptor α with the Kiss1‐Cre knock‐in allele increased bone density in female mice. 145 Therefore, it can be inferred that kisspeptin is involved in the regulation of the brain–bone axis.
3.5. Regulation of peripheral nerves and neurotransmitters on bone in AD
Bone also receives innervation from the autonomic nervous system, including the sympathetic nervous system and parasympathetic nervous system. 30 , 47 , 146 Clinical evidence has shown that AD patients may have varying degrees of autonomic dysfunction, including orthostatic hypotension, decreased heart‐rate variability, constipation, and urinary incontinence. 147 With aging, bone density decreases, and degenerative changes in autonomic distribution occur. 148 In peripheral nerves, acetylcholine serves as the main neurotransmitter of parasympathetic nerves, and cholinergic receptors include muscarinic acetylcholine receptors (mAChRs) and nicotinic acetylcholine receptors (nAChRs). The cholinergic hypothesis is one of the pathogenic hypotheses for AD, postulating that the degeneration of cholinergic neurons in the brain, along with the depletion of presynaptic cholinergic transmitters and reduced activity of cholinesterase, leads to AD development. 149 , 150 It has been demonstrated that osteocytes express both mAChR and nAChR and that acetylcholine regulates osteocyte function through interactions with the AChR. 151 The AChR is composed of multiple subunits, and studies have suggested that M3 subunit of the mAChR has a positive influence on cancellous bone microstructure, flexural stiffness, and bone matrix synthesis. 152 , 153 Additionally, the α9 subunit of the nAChR has been implicated in maintaining osteocyte homeostasis and regulating bone mass. 154 Studies have shown that nAChR can increase bone mass by reducing osteoclast production. 155 , 156
The bone region with the highest metabolic activity has the richest sympathetic distribution. 146 The main neurotransmitter of sympathetic nerves is norepinephrine, which activates α and β adrenergic receptors (ARs). 157 The βAR subtype may be the primary ARs that mediate sympathetic remodeling of bone. 157 Stimulation of βARs in osteoblasts can enhance osteoclast production while inhibiting osteoblast proliferation through circadian genes, ultimately leading to the development of low bone mass. 157 , 158 , 159 In addition, the activation of paracrine proinflammatory factors in sympathetic nerves impaired the osteogenic ability of mesenchymal stem cells. 160 Another study revealed that sympathetic nerves induced osteoporosis by upregulating microRNA 21 expression in osteoblasts via βAR activation and stimulating osteoclasts through exosome‐mediated transport. 161 These findings indicate that dysregulated sympathetic activity in AD may lead to bone loss.
Neurotransmitters are also thought to be important in the brain–bone axis. Glutamate is an excitatory neurotransmitter responsible for excitatory synapse activity that is involved in the maintenance of neuronal function through the crucial glutamate–glutamine cycle. Promoting the glutamate–glutamine cycle in neuron‐astrocytes has been shown to be beneficial in alleviating AD. 162 Neurons exhibiting heightened baseline activity levels exhibit hyperactivity in response to Aβ, which disrupts glutamate reuptake and leads to neuronal overstimulation. 163 , 164 Numerous studies have shown the dysregulation of glutamate in AD patients and animal models. 165 In bone tissue, glutamate receptors are expressed by osteoblasts and osteoclasts. 166 The differentiation of osteoblasts is upregulated by glutamate through the alpha‐amino‐3‐hydroxy‐5‐methylisoxazole‐4‐propionic acid receptor and N‐methyl‐D‐aspartate (NMDA) receptor, and the NMDA receptor is also involved in the regulation of osteoclast production. 167 , 168 Furthermore, serotonin and dopamine are also implicated in the regulation of both brain and bone physiology. 30
3.6. Effect of brain‐derived EVs on bone
EVs are a heterogeneous group of membrane‐bound vesicles released by cells and include various subtypes such as exosomes, microvesicles, oncosomes, and apoptotic bodies. 169 Among these subtypes, exosomes have been extensively investigated due to their wide distribution and heterogeneous functions. Exosomes can be secreted by many tissues and contain membrane proteins, cytosolic and nuclear proteins, extracellular matrix proteins, metabolites, and nucleic acids. Injury to hippocampal neurons can release small EVs containing microRNAs, which subsequently target bone progenitor cells to promote osteogenesis and accelerate bone healing. 170 Therefore, it can be hypothesized that brain‐derived EVs have the ability to traverse the blood–brain barrier (BBB) and reach bone tissue to regulate its function. Recently, a study confirmed the BBB‐crossing potential of brain‐derived EVs in both AD and WT mice. 171 This study revealed that both brain‐derived EVs and plasma‐derived EVs in AD exert a significant inhibitory effect on osteogenic differentiation while promoting adipogenic differentiation of BMSCs, which ultimately leads to bone loss and marrow adiposity. 171
4. REGULATORY ROLES OF BONE IN AD
We have already discussed the possible effects of AD on bone. As patients with bone disorders including osteoporosis and osteoarthritis have a greater incidence of AD, we further investigated the possible effects of bone‐derived hormones, bone marrow‐derived cells, bone‐derived EVs, and inflammation on AD pathogenesis and cognitive function.
4.1. Bone‐derived hormones
4.1.1. Osteocalcin
Osteocalcin (OCN) is a 5‐kDa protein composed of 49 amino acids that is encoded by the Bglap gene. 172 OCN is synthesized by osteoblasts and serves as a marker for bone formation. 172 It exists in both carboxylated and undercarboxylated forms of OCN. 173 , 174 cOCN is primarily deposited in the bone matrix, and ucOCN is released into circulation to perform regulatory functions on multiple organs. 173 , 174 Clinical studies have shown that plasma OCN levels are correlated with cognitive function and brain microstructural changes. 29 , 175 A recent Mendelian randomization study also suggested that OCN may exert a protective effect against AD. 176 However, whether OCN levels are altered in the AD brain remains unknown. Several studies have been conducted to investigate the role and underlying mechanism of OCN in cognition. OCN can traverse the BBB and ultimately modulate spatial learning and memory in mice by influencing neurotransmitter synthesis. 177 G protein‐coupled receptor 158 (GPR158) was demonstrated to be the receptor of OCN in neurons of the hippocampus and mediated the regulation of hippocampal‐dependent memory by OCN through BDNF. 178 Moreover, the OCN/GPR158 signaling pathway has been shown to mediate a regulatory loop controlling the histone‐binding protein RbAp48, which in turn modulates the expression of GPR158. 179 Activation of the OCN/GPR158 pathway elicits elevated RbAp48 levels in middle‐aged animals and rescues age‐related cognitive decline. 179 A recent study showed that OCN could enhance the energy metabolism of astrocytes and microglia through the GPR158 protein, thereby promoting glycolysis, reducing Aβ deposition in the brain, and alleviating cognitive deficits in AD mice. 180 However, a previous study demonstrated that GPR158 is mainly expressed in neurons and is largely absent in glia. 181 In addition, OCN can regulate oligodendrocyte differentiation and myelin homeostasis via GPR37 in the mouse brain. 182 As myelin dysfunction is involved in AD pathogenesis, improving oligodendrocyte health and myelin integrity could alleviate cognitive impairment of AD. 183 , 184 Thus, OCN may protect against AD by enhancing myelination. Considering the function of OCN in bone‐related diseases and the brain, some researchers have proposed that OCN could be identified as an antigeronic hormone. 26 This also implies the role of aging in bone and AD. However, the underlying mechanisms need further study.
4.1.2. Lipocalin‐2
Lipocalin‐2 (LCN‐2) is a glycoprotein consisting of 198 amino acids and has been identified as a molecule secreted by osteoblasts. 47 The expression level of LCN‐2 is at least 10‐fold greater than that in other tissues. 47 The extensive literature on LCN‐2 has shown that it is involved in various pathophysiological processes, including innate immunity, inflammation, metabolism, and apoptosis. 185 The expression of LCN‐2 is upregulated during osteoblast differentiation and related to bone development and turnover. 186 , 187 In addition, LCN‐2 is also highly expressed in the osteoblasts of AD. 188 Osteoblast‐derived LCN‐2 can cross the BBB and suppress appetite by binding to the melanocortin‐4 receptor in the hypothalamus. 187 In the preclinical stage of AD, blood LCN‐2 levels are elevated and correlated with Aβ42 levels in cerebrospinal fluid (CSF). 189 Another study also revealed increased blood LCN‐2 levels in AD patients compared with healthy control individuals and patients with other types of dementia; however, no correlation was found between plasma LCN‐2 and CSF AD biomarkers or cognitive function. 190 LCN‐2 levels in osteoblasts can promote AD development by upregulating hippocampus collapsin response mediator protein 2. 188 Moreover, decreasing the expression of LCN‐2 in osteoblasts via the overexpression of Fork‐head box O1 could prevent AD progression. 188 These findings highlight that LCN‐2 may be a potential therapeutic target for AD. Due to ongoing controversies surrounding the study of LCN‐2 in the brain, including questions about which cells produce it and its role during inflammation, 191 more studies are warranted to explore the relationship between bone‐derived LCN‐2 and AD.
4.1.3. Fibroblast growth factor 23
Fibroblast growth factor 23 (FGF23), which is mainly secreted by osteocytes and osteoblasts in bone, 192 is thought to be involved in bone mineralization. 193 It can interact with the klotho protein, which has been implicated in aging and cognition. 194 However, limited research has been conducted on the role of FGF‐23 in AD. A clinical study revealed an association between elevated circulating FGF23 levels and increased dementia risk. 195 AD patients exhibit increased concentrations of FGF23, which are correlated with inflammatory cytokine levels. 196 An animal study showed that overexpression of FGF23 led to impairment of long‐term potentiation in the hippocampal CA1 region. 197 These findings suggest that FGF23 may be involved in the development of AD, but the underlying mechanism is not fully understood.
4.1.4. Osteoprotegerin
Osteoprotegerin (OPG), a soluble glycoprotein of the TNF receptor superfamily, is synthesized by osteoblasts, B‐lymphocytes, and articular chondrocytes and plays a role in bone homeostasis. 198 , 199 , 200 The role of OPG in AD is unclear. While one study reported significantly elevated blood levels of OPG in AD patients, another study reported no statistically significant difference in blood OPG levels between AD patients and healthy controls individuals. 201 , 202 Elevated circulating OPG levels were reported to be associated with decreased total brain volume. 203 Increasing evidence suggests that OPG plays an important role in vascular injury and inflammatory processes within the CNS. 204 However, more clinical and experimental studies are needed to investigate the role of OPG in AD.
4.1.5. Osteopontin
Osteopontin (OPN) was the first extracellular matrix component identified in bone tissue and is widely distributed across various tissues. 205 OPN plays an important role in bone remodeling. 206 High levels of blood OPN in postmenopausal women were associated with low BMD and osteoporotic vertebral fractures. 207 , 208 Clinical investigations demonstrated elevated levels of OPN in both the plasma and CSF of AD patients, 209 , 210 , 211 suggesting that OPN might play some role in the pathophysiology of AD. An experimental study revealed that microglia‐derived OPN levels were increased in both AD mouse models and AD patients and contributed to AD pathology. 212 Moreover, blocking OPN inhibited the microglial proinflammatory response and decreased Aβ plaque pathology in AD mice. 212 Whether bone‐derived OPN contributes to AD progression remains largely unknown and warrants further investigation.
4.1.6. Osteocrin
Osteocrin (OSTN, also known as musclin) is a bone‐derived protein consisting of 103 amino acids with a molecular weight of 11.4 kDa. While mice express OSTN primarily in bones and muscles rather than in the brain, primate brains express OSTN within the neocortex. 213 In bone, OSTN is predominantly expressed in osteoblasts and osteocytes, particularly during the early stages of bone formation, and its expression gradually declines with age. 214 Due to its homology to natriuretic peptide, it has been proposed that periosteal osteoblast‐produced OSTN may modulate growth plate development by augmenting C‐type natriuretic peptide (CNP)‐dependent proliferation and maturation of chondrocytes. 215 By binding to the natriuretic peptide receptor 3 (NPR3) in periosteal bone progenitor cells, OSTN impedes NPR3‐mediated CNP clearance, thereby promoting bone growth. 215 , 216 Additionally, overexpression of OSTN partially mitigated glucocorticoid‐induced osteocyte degeneration in mice. 217 Furthermore, overexpression of OSTN can rescue osteocyte dendritic defects caused by Sp7 gene deficiency, and Sp7‐dependent genes that mark osteocytes are highly enriched in neurons, suggesting potential associations between Sp7‐dependent traits and neuronal development. 218 The evolution of the OSTN gene led to the emergence of a novel primate‐specific enhancer sequence capable of binding to myocyte enhancer factor 2 (MEF2). 213 MEF2 and OSTN act as regulators of dendritic growth in the developing cerebral cortex in response to sensory experience. 213 Recent literature has shown that OSTN can prevent depressive‐like behavior in male mice by activating urinary corticin‐2 signaling in the hypothalamus. 219 In addition, OSTN ameliorated the expression of inflammatory markers in monocytes, 220 suggesting a protective role against neuroinflammation. However, whether bone‐derived OSTN is involved in AD pathogenesis remains unclear and needs to be investigated in the future.
4.1.7. Sclerostin
Sclerostin (SOST), a protein expressed by mature bone cells, is an inhibitor of the Wnt signaling pathway. 221 SOST can bind to low‐density‐lipoprotein receptor‐related proteins on the surface of osteoblasts, block the Wnt signaling pathway, and inhibit osteoblast proliferation and differentiation. 222 Elevated blood SOST levels were correlated with low BMD in patients with prediabetes 223 and with worse fracture healing in patients after surgical stabilization of long bone fractures. 224 SOST levels in blood and CSF are positively correlated with age. 225 , 226 Blood SOST was positively associated with brain Aβ burden and cognitive impairment in both aged individuals and patients with AD. 225 , 226 A recent study demonstrated that bone‐derived SOST can cross the BBB, increase Aβ production through β‐catenin–β‐secretase 1 signaling, and impair synaptic plasticity and memory function in mice. 226 More clinical and experimental studies are needed to investigate the role of SOST in AD.
4.1.8. Bone morphogenetic proteins
Bone morphogenetic proteins (BMPs) are the largest subgroup of the transforming growth factor‐beta superfamily, with more than 20 identified BMPs. 227 BMPs are widely expressed in various tissues throughout the body including bone and brain and are important for bone development and regeneration and maintenance of bone homeostasis. 228 Specifically, BMP4 is implicated in bone repair and regeneration; however, it can inhibit neurogenesis and oligodendrogenesis in induced pluripotent stem cells (iPSCs) of AD patients. 228 , 229 Decreased hippocampal cell proliferation was associated with increased expression of BMP4 in AD. 230 In addition, BMP4 transgenic mice exhibit increased levels of APP and tau proteins along with cognitive impairment, and in vitro experiments have shown that the downregulation of BMP4 reduces the levels of AD pathogenic proteins. 231 Furthermore, BMP2 and BMP4 can upregulate the expression of aquaporin‐4 in astrocytes, which is also involved in AD pathogenesis. 232 A clinical study revealed that AD patients had lower blood BMP6 levels, which are positively associated with cognitive function. 233 However, the role of bone‐derived BMPs in the pathogenesis and progression of AD needs further study.
4.1.9. Bone‐derived platelet‐derived growth factor‐BB
Platelet‐derived growth factor (PDGF)‐BB, a member of the PDGF family, has been widely studied in the field of orthopedics. 234 , 235 , 236 A study demonstrated that osteoclasts are the primary source of elevated circulating levels of PDGF‐BB during aging. 236 , 237 Bone‐derived PDGF‐BB is implicated in the regulation of angiogenesis, arteriosclerosis, bone remodeling, and joint structure damage. 238 , 239 A study revealed that aged male mice had increased blood PDGF‐BB levels, which could induce brain vascular calcification. 240 Furthermore, elevated concentrations of bone‐derived PDGF‐BB could induce hippocampal BBB impairment and memory deficits by promoting the shedding of PDGF receptor β from the pericytes' surface. 241 Therefore, future investigations should explore the role of bone‐derived PDGF‐BB in other age‐related diseases such as AD.
4.2. Bone marrow‐derived cells
4.2.1. Bone marrow‐derived immune cells
Dysfunction of microglia in the AD brain is closely related to the occurrence and development of AD. There is evidence that bone marrow‐derived macrophages can migrate into the brain to become microglia under physiological conditions, but these microglia account for only approximately 1% of the total number of microglia in the brain. 242 The effect of bone marrow‐derived immune cells on AD is controversial. Early studies revealed that infiltrating monocytes in the blood of AD patients did not perform long‐term functions in clearing Aβ in the brain. 243 A recent study found that increased infiltration of bone marrow‐derived grancalcin (GCA)‐positive immune cell subpopulation in the brain parenchyma of AD model mice promoted the progression of AD through the secretion of GCA, which could inhibit the phagocytosis and clearance of Aβ in microglia by competitively binding to lipoprotein receptor‐related protein 1. 244 Therefore, the complicated roles of bone marrow immune cells in the pathogenesis of AD remain unclear and need further study.
4.2.2. Bone marrow mesenchymal stem cells
As pluripotent stem cells, BMSCs are also involved in the bone microenvironment and have extensive applications in skeletal system diseases and AD. 245 , 246 , 247 One study demonstrated that intracerebral injection of BMSCs resulted in a significant reduction in Aβ accumulation and an increase in proteins facilitating synaptic transmission. 248 Additionally, intracerebral administration of BMSCs enhanced microglial phagocytosis of Aβ, possibly through microglial activation, upregulation of Aβ‐degrading enzymes, and increased expression of Aβ binding receptors. 249 , 250 Furthermore, transplantation of BMSCs was found to alleviate tau hyperphosphorylation in the brains of AD model mice. 249 , 251 BMSC transplantation prevents microglia in the classical M1 state from secreting proinflammatory factors and stimulates M2 microglia to produce anti‐inflammatory cytokines. 252 Nevertheless, both injection methods resulted in decreased expression of inflammation‐related genes such as TNF‐α and IL‐1β and alleviated pathological changes associated with AD. 249 , 253 , 254 Co‐culture of BMSCs with Aβ‐treated primary cortical and hippocampal neurons in vitro also demonstrated elevated levels of anti‐inflammatory factors, increased telomere length and telomerase activity, and activated signaling pathways that have been linked to aging. 255 In conclusion, BMSC transplantation can alleviate cognitive deficits by regulating Aβ clearance, tau hyperphosphorylation, neurogenesis, neuroinflammation, immunomodulation, apoptosis, autophagy, and angiogenesis in AD mouse models. 248 , 249 , 250 , 253 , 254 , 256 . Moreover, the effect of BMSCs on AD has been verified in different transplantation approaches and different animal models. 247 Microscopic vascular channels were found between the bone marrow of the skull and the brain. 257 , 258 These channels allow CSF to enter the skull bone marrow and can also affect the migration of cells in the bone marrow to the brain. 257 , 259 Therefore, whether the changes in CSF of AD patients affect the skull bone marrow cells to enter the brain and participate in the neuroimmune process of AD needs to be further studied. 260
4.3. Bone‐derived EVs
Osteoblasts, osteoclasts, and osteocytes can release EVs, which are widely involved in the regulation of bone metabolism. 261 , 262 It was observed that plasma levels of osteocyte‐derived EVs were greater in AD patients than in cognitively normal individuals. 263 Compared with aged osteocyte‐derived EVs, young osteocyte‐derived EVs exhibited an enhanced ability to cross the BBB. 263 In vivo and in vitro experiments demonstrated that young osteocyte‐derived EVs from WT mice can mitigate brain Aβ deposition and cognitive impairment in both the early and late stages of AD. 263 Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that multiple functional factors related to Aβ degradation and mitochondrial energy metabolism were highly enriched in young osteocyte‐derived EVs compared with aged osteocyte‐derived EVs. 263 This study suggested that young osteocyte‐derived EVs played a protective role in AD by preventing Aβ deposition and neuronal damage. However, whether aged osteocyte‐derived EVs from AD transgenic mice exacerbate AD progression remains unknown and deserves further investigation. In addition, EVs derived from aged bone matrix have been found to promote adipogenesis and exacerbate vitamin D3‐induced vascular calcification, 264 which could contribute to AD dementia. 265 , 266 BMSC‐derived EVs are also involved in the pathogenesis of AD. Studies have indicated that BMSC‐derived EVs effectively reduce Aβ accumulation in the brains of AD mice, with a more pronounced effect observed in young mice, suggesting the potential of BMSC‐derived EVs to mitigate or prevent Aβ formation during the early stages of AD. 267 , 268 In addition, EVs from BMSCs can attenuate the expression of inducible nitric oxide synthase induced by Aβ oligomers, potentially ameliorating hippocampal synaptic plasticity deficits and cognitive impairment. 269 Additionally, BMSC‐derived exosomes alleviate neuroinflammation in AD animal models by reducing neurotoxicity resulting from the activation of microglial and astrocytes in the hippocampus and significantly increasing BDNF expression levels. 270 Therefore, we speculate that bone‐derived EVs may play complicated roles in the crosstalk between bone and brain in AD.
4.4. Inflammation
Chronic inflammation is a hallmark feature of both bone diseases and AD. During the inflammatory response, the NF‐κB signaling pathway is activated, promoting osteoclasts and simultaneously inhibiting osteoblasts. 271 , 272 The complement system associated with NF‐κB signaling activation is also involved in regulating bone loss. 273 In addition, proinflammatory factors such as IL‐1, IL‐6, and TNF have also been found to be associated with bone‐related diseases such as osteoporosis or osteoarthritis. 274 , 275 , 276 , 277 , 278 Similarly, in AD, the activation of inflammation‐related signaling pathways including the NF‐κB pathway, the complement system, and proinflammatory factors are also activated. 279 , 280 , 281 A genome‐wide association study also revealed that some risk genes for AD are also associated with inflammation. 282 In general, inflammation is involved in the pathogenesis of bone‐related diseases and AD. Investigations into whether bone‐related diseases are related to systemic inflammation have been conducted. 277 , 283 , 284 , 285 Peripheral inflammation has been suggested to exacerbate AD‐type pathologies and induce cognitive impairment. 280 , 286 , 287 , 288 Therefore, we speculate that systemic inflammation could be regarded as a common risk factor for bone disease and AD. Peripheral inflammation in osteoarthritis exacerbates brain inflammation and subsequent AD pathology in AD mice. 289 , 290 Therefore, the inflammatory mediators produced during bone diseases may contribute to AD by promoting neuroinflammation and neurodegeneration through the activation of glial cells. However, the recently reported relevant research is limited and needs to be further explored.
Microglia and osteoclasts are tissue‐specific macrophages and functionally similar cell types in the brain and bone. The inflammatory pathway induced by microglia and osteoclasts may mediate shared risk factors for AD and osteoporosis. 291 A recent transcriptional sequencing study of microglia in AD mice revealed that seven of the differentially regulated genes in microglia were related to osteoclast generation, recruitment, and inflammation, 292 suggesting that osteoclasts and microglia may represent a common pathological cell type linking osteoporosis and AD.
5. PERSPECTIVE ON NOVEL AD INTERVENTION STRATEGIES TARGETING THE BONE–BRAIN AXIS
5.1. Prevention and management of bone diseases
AD frequently coexists with bone disorders, which could promote brain AD pathology and increase the risk of AD, as discussed earlier. Thus, preventing and managing bone diseases such as osteoporosis and osteoarthritis has become increasingly crucial. There is evidence indicating a negative association between sedentary behavior and BMD. 293 Another study suggested that increased time spent in sedentary behavior was significantly associated with a greater incidence of dementia among older individuals. 294 In fact, one review concluded that sedentary behavior could impact protein homeostasis, mitochondrial function, cell‐to‐cell communication, and other aspects to accelerate biological aging. 295 This link between bone diseases and AD underscores the importance for AD patients to avoid sedentariness and increase physical activity in AD patients. Studies have indicated that physical exercise can enhance BMD in patients with osteoporosis 296 , 297 and improve fracture resistance and bone strength in AD model mice. 298 Furthermore, a meta‐analysis also corroborated the notion that exercise could enhance cognitive function, physical capability, and functional independence in individuals with AD. 299 Consequently, exercise is generally considered the most cost‐effective and accessible strategy for mitigating osteoporosis and AD.
In addition, some anti‐osteoporotic therapies show great potential for the prevention and treatment of AD. Bisphosphonates are available therapeutic drugs for the management of osteoporosis that inhibit bone resorption. At present, bisphosphonates are believed to prevent AChR inactivation and inhibit acetylcholinesterase activity, thereby exerting a protective effect against AD. 300 , 301 , 302 , 303 However, the other underlying mechanism remains unclear and needs further research. Studies have shown that vitamin D plays a role in the absorption of calcium and phosphate and that vitamin D supplementation can reduce bone turnover and increase BMD. 304 In addition, vitamin D deficiency has also been associated with several psychiatric disorders (eg, schizophrenia, autism spectrum disorder) and neurodegenerative diseases (eg, AD, Parkinson's disease). 305 , 306 , 307 , 308 Research has indicated that vitamin D is associated with processes such as neurological development, oxidative stress, calcium homeostasis, and inflammation. 305 , 309 One study showed that vitamin D supplementation exacerbated Aβ deposition in animal models and increased the risk of death in dementia patients. 310 However, some scholars noted that this study equated vitamin D and calcitriol and drew inappropriate clinical conclusions. 311 The potential benefits of vitamin D supplementation in young and middle‐aged individuals should not be disregarded, as previous studies also indicated its potential efficacy in the early stages prior to Aβ formation. 312 , 313 , 314 , 315 Recently, a prospective cohort study that included 269,229 participants in the UK Biobank showed that vitamin D supplementation could decrease by 17% the risk of AD. 308 In general, although vitamin D connects bones and the brain, the effects of vitamin D in the brain are diverse. Thus, its role in different pathophysiological processes of AD remains unclear. It may be important to investigate whether vitamin D supplementation could decrease AD risk in patients with osteoporosis in the future. A previous study revealed that aged patients who used heavy amounts of non‐steroidal anti‐inflammatory drugs (NSAIDs) had a greater incidence of dementia and AD. 316 A meta‐analysis suggested a correlation between exposure to NSAIDs and a reduced risk of AD. 317 In addition, two NSAIDs have been found to improve cognitive dysfunction and attenuate Aβ deposition in AD mice. 318 The contradictory results of clinical studies may be related to the study population, 316 , 319 but the relationship between NSAID use and AD risk in bone‐related diseases still needs further investigation. Consequently, the prevention and timely treatment of bone diseases may be essential for AD management.
5.2. Targeting bone‐derived proteins
Based on the dual role of bone‐derived proteins, we propose that modulating the levels of bone‐derived proteins may be a potential therapeutic approach for AD. As discussed earlier, OCN has been verified as a neuroprotective and cognition‐enhancing peptide. Thus, the potential of OCN‐based therapy for AD should be investigated in in‐depth basic and clinical studies. The antagonism of bone‐derived proteins that exacerbate AD may also be promising for AD intervention. Animal experiments have shown that reducing bone‐derived SOST could reduce synaptic damage. 27 Romosozumab, a monoclonal antibody that inhibits sclerostin, has gained approval for osteoporosis treatment, but there remains a lack of research on its treatment of AD or dementia. 320 , 321 , 322 Similarly, other bone‐derived proteins have similar limitations. Nevertheless, it can be speculated that augmenting protective bone‐derived proteins and inhibiting destructive bone‐derived proteins may have an impact on AD from the perspective of the bone‐brain axis.
5.3. BMSCs‐based therapy
The strong differentiation potential of BMSCs and the close communication between bone and brain led researchers to investigate whether BMSCs could be used to treat AD. A recent study revealed that transplantation of normal bone marrow could replace more than 90% of the genetically mutated microglia in the mouse brain, restore the function of microglia, and reduce the pathological and cognitive function of AD in the mouse brain. 323 Several promising clinical trials of BMSC intervention in AD are currently under way (NCT02795052; NCT03724136). Compared with BMSC transplantation, BMSC‐derived EVs have greater stability, lack of immunity, lack of carcinogenicity, lack of ethical controversy, and easier access and preservation. 324 , 325 , 326 However, perhaps due to the complexity of the process of extracting EVs from BMSCs and the lack of standardized isolation, storage, and purification protocols, there are no relevant clinical trials on BMSC‐derived EV intervention in AD. Overall, to some extent, BMSC‐based therapy is a promising treatment for AD but still needs further development.
FIGURE 1.

Crosstalk between bone and brain in Alzheimer's disease (AD). AD can affect bone functions through various pathways, including AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain‐derived extracellular vesicles (EVs), and the autonomic nervous system. On the other hand, bone‐derived proteins, bone marrow‐derived cells, EVs, and inflammation may also influence brain AD pathology and cognitive function. Some intervention strategies targeting the bone–brain axis may be beneficial for the comprehensive management of AD. Aβ, amyloid beta; APOE, apolipoprotein E; APP, amyloid precursor protein; AD, Alzheimer's disease; BBB, blood–brain barrier; BMP, bone morphogenetic protein; BMSC, bone marrow mesenchymal stem cell; EV, extracellular vesicle; FGF23, fibroblast growth factor 23; FSH, follicle‐stimulating hormone; LCN‐2, lipocalin‐2; NPY, neuropeptide Y; NSAID; nonsteroidal anti‐inflammatory drug; OCN; osteocalcin; OPG, osteoprotegerin; OPN, osteopontin; OSTN, Osteocrin; PDGF, platelet‐derived growth factor; SOST, sclerostin; TREM2, triggering receptor expressed on myeloid cells 2.
6. CONCLUSION AND PERSPECTIVES
In this review, we provided an overview of clinical evidence supporting a strong link between AD and bone health. We discussed the bidirectional effects between AD and bone. AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, the autonomic nervous system, neurotransmitters, and brain‐derived EVs can impact bone health, and bone‐derived proteins, bone marrow‐derived cells, EVs, and inflammation may also influence brain AD pathology and cognitive function. In addition, the common risk factors for both bone diseases and AD, including aging, female sex, and sedentariness, may also be the connections and promoters of these two diseases. Therefore, the crosstalk between bone and brain provides new insight into dealing with AD and bone disease, and the two may interact to promote each other. The active prevention and timely treatment of bone diseases in elderly people may be beneficial to the prevention and treatment of AD. We summarized the potential mutual mechanisms of bone and brain in AD as well as related intervention strategies in Figure 1.
The crosstalk between bone and brain in AD holds great importance in the prevention and management of AD, so we propose possible perspectives for future research in this area. Additional animal and clinical studies are needed to investigate the contribution of bone homeostasis and bone diseases to AD pathogenesis. First, because the recently reported relevant clinical studies are heterogeneous, larger prospective cohort studies are warranted to elucidate the causal relationship between bone diseases and AD pathology as well as AD progression. Second, considering the significant disparities between animal models and humans, macaque and organoid models may be better able to explain the roles of bone‐derived substances in AD. Third, single‐cell transcriptome and protein sequencing could be used to clarify the key brain regions, cell types, and molecular pathways that link bone and AD. Fourth, the potential utilization of bone‐derived proteins or their antibodies as therapeutic targets for AD also needs to be validated in AD patients to translate the findings in animal experiments to clinical studies. In addition, considering the complex crosstalk between bone and brain, the identification of key molecules that promote AD in different bone diseases will be important for future treatments.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflicts of interest. Author disclosures are available in the Supporting information.
Supporting information
Supporting Information
ACKNOWLEDGMENTS
The figure was created with Biorender.com. This study was supported by the National Science Foundation of China (Nos. 82122023 and U22A20294), the Science and Technology Innovation 2030 Major Projects (No. 2022ZD0211600), and the Natural Science Foundation of Chongqing Municipality (No. CSTB2023NSCQ‐JQX0019).
Liu Z‐T, Liu M‐H, Xiong Y, Wang Y‐J, Bu X‐L. Crosstalk between bone and brain in Alzheimer's disease: Mechanisms, applications, and perspectives. Alzheimer's Dement. 2024;20:5720–5739. 10.1002/alz.13864
Zhuo‐Ting Liu, Ming‐Han Liu, and Yan Xiong authors contributed equally to this work.
Contributor Information
Yan‐Jiang Wang, Email: yanjiang_wang@tmmu.edu.cn.
Xian‐Le Bu, Email: buxianle@tmmu.edu.cn.
REFERENCES
- 1. Ren R, Qi J, Lin S, et al. The China Alzheimer Report 2022. Gen Psychiatr. 2022;35:e100751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Jia LF, Quan MN, Fu Y, et al. Dementia in China: epidemiology, clinical management, and research advances. Lancet Neurol. 2020;19:81‐92. [DOI] [PubMed] [Google Scholar]
- 3. Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid beta‐based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther. 2023;8:248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Kang HG, Park HY, Ryu HU, Suk SH. Bone mineral loss and cognitive impairment: the PRESENT project. Medicine (Baltimore). 2018;97:e12755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Sun HL, Chen SH, Yu ZY, et al. Blood cell‐produced amyloid‐β induces cerebral Alzheimer‐type pathologies and behavioral deficits. Mol Psychiatry. 2021;26:5568‐5577. [DOI] [PubMed] [Google Scholar]
- 6. Chen C, Zhou Y, Wang H, et al. Gut inflammation triggers C/EBPβ/δ‐secretase‐dependent gut‐to‐brain propagation of Aβ and Tau fibrils in Alzheimer's disease. Embo j. 2021;40:e106320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Tian DY, Cheng Y, Zhuang ZQ, et al. Physiological clearance of amyloid‐beta by the kidney and its therapeutic potential for Alzheimer's disease. Mol Psychiatry. 2021;26:6074‐6082. [DOI] [PubMed] [Google Scholar]
- 8. Yu ZY, Chen DW, Tan CR, et al. Physiological clearance of Aβ by spleen and splenectomy aggravates Alzheimer‐type pathogenesis. Aging Cell. 2022;21:e13533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Liu ZH, Bai YD, Yu ZY, et al. Improving blood monocyte energy metabolism enhances its ability to phagocytose amyloid‐β and prevents Alzheimer's disease‐type pathology and cognitive deficits. Neurosci Bull. 2023;39:1775‐1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Cheng Y, He CY, Tian DY, et al. Physiological β‐amyloid clearance by the liver and its therapeutic potential for Alzheimer's disease. Acta Neuropathol. 2023;145:717‐731. [DOI] [PubMed] [Google Scholar]
- 11. Grande G, Marengoni A, Vetrano DL, et al. Multimorbidity burden and dementia risk in older adults: the role of inflammation and genetics. Alzheimers Dement. 2021;17:768‐776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Ben Hassen C, Fayosse A, Landre B, et al. Association between age at onset of multimorbidity and incidence of dementia: 30 year follow‐up in Whitehall II prospective cohort study. BMJ. 2022;376:e068005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Hu HY, Zhang YR, Aerqin Q, et al. Association between multimorbidity status and incident dementia: a prospective cohort study of 245,483 participants. Transl Psychiatry. 2022;12:505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Pluvinage JV, Wyss‐Coray T. Systemic factors as mediators of brain homeostasis, ageing and neurodegeneration. Nat Rev Neurosci. 2020;21:93‐102. [DOI] [PubMed] [Google Scholar]
- 15. Liu CC, Zhao J, Fu Y, et al. Peripheral apoE4 enhances Alzheimer's pathology and impairs cognition by compromising cerebrovascular function. Nat Neurosci. 2022;25:1020‐1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Giannisis A, Patra K, Edlund AK, et al. Brain integrity is altered by hepatic ε4 in humanized‐liver mice. Mol Psychiatr. 2022;27:3533‐3543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Schroer AB, Ventura PB, Sucharov J, et al. Platelet factors attenuate inflammation and rescue cognition in ageing. Nature. 2023;620(7976):1071‐1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Liu ZH, Wang YJ, Bu XL. Alzheimer's disease: targeting the peripheral circulation. Mol Neurodegener. 2023;18(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Bio. 2020;21:696‐711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Yuan J, Meloni BP, Shi TX, et al. The potential influence of bone‐derived modulators on the progression of Alzheimer's disease. J Alzheimers Dis. 2019;69:59‐70. [DOI] [PubMed] [Google Scholar]
- 21. Huang S, Li Z, Liu Y, et al. Neural regulation of bone remodeling: identifying novel neural molecules and pathways between brain and bone. J Cell Physiol. 2019;234:5466‐7547. [DOI] [PubMed] [Google Scholar]
- 22. Otto E, Knapstein PR, Jahn D, et al. Crosstalk of brain and bone‐clinical observations and their molecular bases. Int J Mol Sci. 2020;21(14):4946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Herber CB, Krause WC, Wang L, et al. Estrogen signaling in arcuate Kiss1 neurons suppresses a sex‐dependent female circuit promoting dense strong bones. Nat Commun. 2019;10:163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Zhang L, Liu N, Shao J, et al. Bidirectional control of parathyroid hormone and bone mass by subfornical organ. Neuron. 2023;111:1914‐1932. [DOI] [PubMed] [Google Scholar]
- 25. Zhou RY, Guo QY, Xiao Y, et al. Endocrine role of bone in the regulation of energy metabolism. Bone Res. 2021;9:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Obri A, Khrimian L, Karsenty G, Oury F. Osteocalcin in the brain: from embryonic development to age‐related decline in cognition. Nat Rev Endocrinol. 2018;14:174‐182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Shi TS, Shen SY, Shi Y, et al. Osteocyte‐derived sclerostin impairs cognitive function during ageing and Alzheimer's disease progression. Nat Metab. 2024;6:531‐549. [DOI] [PubMed] [Google Scholar]
- 28. Du J, Li A, Shi D, et al. Association of APOE‐ε4, osteoarthritis, β‐amyloid, and tau accumulation in primary motor and somatosensory regions in Alzheimer disease. Neurology. 2023;101:e40‐e49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Bradburn S, McPhee JS, Bagley L, et al. Association between osteocalcin and cognitive performance in healthy older adults. Age Ageing. 2016;45:844‐849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Gerosa L, Lombardi G. Bone‐to‐Brain: a round trip in the adaptation to mechanical stimuli. Front Physiol. 2021;12:623893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. LLabre JE, Gil C, Amatya N, Lagalwar S, Possidente B, Vashishth D. Degradation of bone quality in a transgenic mouse model of Alzheimer's disease. J Bone Miner Res. 2022;37:2548‐2565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Xiao T, Ghatan S, Mooldijk SS, et al. Association of bone mineral density and dementia the Rotterdam study. Neurology. 2023;100:E2125‐E2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Tan ZS, Seshadri S, Beiser A, et al. Bone mineral density and the risk of Alzheimer disease. Arch Neurol. 2005;62:107‐111. [DOI] [PubMed] [Google Scholar]
- 34. Kostev K, Hadji P, Jacob L. Impact of osteoporosis on the risk of dementia in almost 60,000 patients followed in general practices in Germany. J Alzheimers Dis. 2018;65:401‐407. [DOI] [PubMed] [Google Scholar]
- 35. Kwon MJ, Kim JH, Kim JH, Cho SJ, Nam ES, Choi HG. The occurrence of Alzheimer's disease and Parkinson's disease in individuals with osteoporosis: a longitudinal follow‐up study using a national health screening database in Korea. Front Aging Neurosci. 2021;13:786337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Zhou R, Deng J, Zhang M, Zhou HD, Wang YJ. Association between bone mineral density and the risk of Alzheimer's disease. J Alzheimers Dis. 2011;24:101‐108. [DOI] [PubMed] [Google Scholar]
- 37. Lin SF, Fan YC, Pan WH, Bai CH. Bone and lean mass loss and cognitive impairment for healthy elder adults: analysis of the nutrition and health survey in Taiwan 2013‐2016 and a validation study with structural equation modeling. Front Nutr. 2021;8:747877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Zhao Y, Chen H, Qiu F, He J, Chen J. Cognitive impairment and risks of osteoporosis: a systematic review and meta‐analysis. Arch Gerontol Geriatr. 2023;106:104879. [DOI] [PubMed] [Google Scholar]
- 39. Wang HK, Hung CM, Lin SH, et al. Increased risk of hip fractures in patients with dementia: a nationwide population‐based study. BMC Neurol. 2014;14:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Loskutova N, Honea RA, Vidoni ED, Brooks WM, Burns JM. Bone density and brain atrophy in early Alzheimer's disease. J Alzheimers Dis. 2009;18:777‐785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Kalc P, Dahnke R, Hoffstaedter F, Gaser C. Low bone mineral density is associated with gray matter volume decrease in UK Biobank. Front Aging Neurosci. 2023;15:1287304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Gao F, Pan R, Fan T, Liu L, Pan H. Identification of heel bone mineral density as a risk factor of Alzheimer's disease by analyzing large‐scale genome‐wide association studies datasets. Front Cell Dev Biol. 2023;11:1247067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Weber A, Mak SH, Berenbaum F, et al. Association between osteoarthritis and increased risk of dementia: a systemic review and meta‐analysis. Medicine (Baltimore). 2019;98:e14355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Guo R, Ou YN, Hu HY, Ma YH, Tan L, Yu JT. The association between osteoarthritis with risk of dementia and cognitive impairment: a meta‐analysis and systematic review. J Alzheimers Dis. 2022;89:1159‐1172. [DOI] [PubMed] [Google Scholar]
- 45. Lary CW, Ghatan S, Gerety M, et al. Bone mineral density and the risk of incident dementia: a meta‐analysis. J Am Geriatr Soc. 2024;72:194‐200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Liu J, Tao J, Cai G, et al. The altered hippocampal functional connectivity and serum brain‐derived neurotrophic factor level predict cognitive decline in patients with knee osteoarthritis. Cereb Cortex. 2023;33:10584‐10594. [DOI] [PubMed] [Google Scholar]
- 47. Zeng WW, Yang F, Shen WL, Zhan C, Zheng P, Hu J. Interactions between central nervous system and peripheral metabolic organs. Sci China Life Sci. 2022;65:1929‐1958. [DOI] [PubMed] [Google Scholar]
- 48. Yuan P, Zhang MY, Tong L, et al. PLD3 affects axonal spheroids and network defects in Alzheimer's disease. Nature. 2022;612:328‐337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Harris SS, Wolf F, De Strooper B, Busche MA. Tipping the scales: peptide‐dependent dysregulation of neural circuit dynamics in Alzheimer's disease. Neuron. 2020;107:417‐435. [DOI] [PubMed] [Google Scholar]
- 50. Xu YQ, Yan JQ, Zhou P, et al. Neurotransmitter receptors and cognitive dysfunction in Alzheimer's disease and Parkinson's disease. Prog Neurobiol. 2012;97:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Pini L, Pievani M, Bocchetta M, et al. Brain atrophy in Alzheimer's Disease and aging. Ageing Res Rev. 2016;30:25‐48. [DOI] [PubMed] [Google Scholar]
- 52. Chandra A, Dervenoulas G, Politis M. Magnetic resonance imaging in Alzheimer's disease and mild cognitive impairment. J Neurol. 2019;266:1293‐1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Li SF, Liu B, Zhang LM, Rong LM. Amyloid beta peptide is elevated in osteoporotic bone tissues and enhances osteoclast function. Bone. 2014;61:164‐175. [DOI] [PubMed] [Google Scholar]
- 54. Haass C, Lemere CA, Capell A, et al. The Swedish mutation causes early‐onset Alzheimer's disease by beta‐secretase cleavage within the secretory pathway. Nat Med. 1995;1:1291‐1296. [DOI] [PubMed] [Google Scholar]
- 55. Xia WF, Jung JU, Shun C, et al. Swedish mutant APP suppresses osteoblast differentiation and causes osteoporotic deficit, which are ameliorated by N‐Acetyl‐L‐Cysteine. J Bone Miner Res. 2013;28:2122‐2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Xiong WC, Xia WF, Jung J, Cui S, Xiong S, Xiong L. Swedish mutant APP suppresses osteoblast differentiation and causes osteoporotic deficit, which are ameliorated by n‐acetyl‐l‐cysteine. J Bone Miner Res. 2013;28:2122‐2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Pan JX, Tang FL, Zhao K, et al. APP promotes osteoblast survival and bone formation by regulating mitochondrial function and preventing oxidative stress. J Bone Miner Res. 2019;34:91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Cui S, Xiong F, Hong Y, et al. APPswe/Aβ regulation of osteoclast activation and RAGE expression in an age‐dependent manner. J Bone Miner Res. 2011;26:1084‐1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Li S, Yang B, Teguh D, Zhou L, Xu J, Rong L. Amyloid β peptide enhances RANKL‐induced osteoclast activation through NF‐κB, ERK, and calcium oscillation signaling. Int J Mol Sci. 2016;17:1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Basheer N, Smolek T, Hassan I. Does modulation of tau hyperphosphorylation represent a reasonable therapeutic strategy for Alzheimer's disease? From preclinical studies to the clinical trials. Mol Psychiatry. 2023;28:2197‐2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Bloom GS. Amyloid‐β and tau the trigger and bullet in alzheimer disease pathogenesis. Jama Neurol. 2014;71:505‐508. [DOI] [PubMed] [Google Scholar]
- 62. Chong FP, Ng KY, Koh RY, Chye SM. Tau proteins and tauopathies in Alzheimer's disease. Cell Mol Neurobiol. 2018;38:965‐980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. van der Kant R, Goldstein LSB, Ossenkoppele R. Amyloid‐β‐independent regulators of tau pathology in Alzheimer disease. Nat Rev Neurosci. 2020;21:21‐35. [DOI] [PubMed] [Google Scholar]
- 64. Dengler‐Crish CM, Smith MA, Wilson GN. Early evidence of low bone density and decreased serotonergic synthesis in the dorsal raphe of a tauopathy model of Alzheimer's disease. J Alzheimers Dis. 2017;55:1605‐1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Chen M, Fu WY, Xu HY, Liu CJ. Tau deficiency inhibits classically activated macrophage polarization and protects against collagen‐induced arthritis in mice. Arthritis Res Ther. 2023;25:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu GJ. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15:501‐518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Raulin AC, Doss S, Trottier ZA, Ikezu TC, Bu GJ, Liu CC. ApoE in Alzheimer's disease: pathophysiology and therapeutic strategies. Mol Neurodegener. 2022;17:72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Peter I, Crosier MD, Yoshida M, et al. Associations of APOE gene polymorphisms with bone mineral density and fracture risk: a meta‐analysis. Osteoporosis Int. 2011;22:1199‐1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Souza LS, Rochette NF, Pedrosa DF, et al. Role of APOE gene in bone mineral density and incidence of bone fractures in brazilian postmenopausal women. J Clin Densitom. 2018;21:227‐235. [DOI] [PubMed] [Google Scholar]
- 70. Carmona‐Fernandes D, Casimiro RI, Silva A, et al. Bone disturbances and progression of atherosclerosis in ApoE knockout mice. Clin Exp Rheumatol. 2023;41:1746‐1753. [DOI] [PubMed] [Google Scholar]
- 71. Schilling AF, Schinke T, Münch C, et al. Increased bone formation in mice lacking apolipoprotein E. J Bone Miner Res. 2005;20:274‐282. [DOI] [PubMed] [Google Scholar]
- 72. Tang S, Yao L, Ruan J, et al. Single‐cell atlas of human infrapatellar fat pad and synovium implicates APOE signaling in osteoarthritis pathology. Sci Transl Med. 2024;16:eadf4590. [DOI] [PubMed] [Google Scholar]
- 73. Li RY, Qin Q, Yang HC, et al. TREM2 in the pathogenesis of AD: a lipid metabolism regulator and potential metabolic therapeutic target. Mol Neurodegener. 2022;17:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Weng YT, Wang HC, Li L, Feng YHZ, Xu SY, Wang ZL. Trem2 mediated Syk‐dependent ROS amplification is essential for osteoclastogenesis in periodontitis microenvironment. Redox Biol. 2021;40:101849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Otero K, Shinohara M, Zhao H, et al. TREM2 and β‐catenin regulate bone homeostasis by controlling the rate of osteoclastogenesis. J Immunol. 2012;188:2612‐2621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Essex AL, Huot JR, Deosthale P, et al. Triggering receptor expressed on myeloid cells 2 (TREM2) R47H variant causes distinct age‐ and sex‐dependent musculoskeletal alterations in mice. J Bone Miner Res. 2022;37:1366‐1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Bianchin MM, Martin KC, de Souza AC, de Oliveira MA, Rieder CRD. Nasu‐Hakola disease and primary microglial dysfunction. Nat Rev Neurol. 2010;6:1‐2. [DOI] [PubMed] [Google Scholar]
- 78. Dardiotis E, Siokas V, Pantazi E. A novel mutation in TREM2 gene causing Nasu‐Hakola disease and review of the literature. Neurobiol Aging. 2017;53:194.e13‐194.e22. [DOI] [PubMed] [Google Scholar]
- 79. Sowers MR, Finkelstein JS, Ettinger B, et al. The association of endogenous hormone concentrations and bone mineral density measures in pre‐ and perimenopausal women of four ethnic groups: sWAN. Osteoporosis Int. 2003;14:44‐52. [DOI] [PubMed] [Google Scholar]
- 80. Chapurlat RD, Garnero P, Sornay‐Rendu E, Arlot ME, Claustrat B, Delmas PD. Longitudinal study of bone loss in pre‐ and perimenopausal women: evidence for bone loss in perimenopausal women. Osteoporos Int. 2000;11:493‐498. [DOI] [PubMed] [Google Scholar]
- 81. Sowers MR, Jannausch M, McConnell D, et al. Hormone predictors of bone mineral density changes during the menopausal transition. J Clin Endocr Metab. 2006;91:1261‐1267. [DOI] [PubMed] [Google Scholar]
- 82. Recker R, Lappe J, Davies K, Heaney R. Characterization of perimenopausal bone loss: a prospective study. J Bone Miner Res. 2000;15:1965‐1973. [DOI] [PubMed] [Google Scholar]
- 83. Sochocka M, Karska J, Pszczolowska M, et al. Cognitive decline in early and premature menopause. Int J Mol Sci. 2023;24:6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. McCarthy M, Raval AP. The peri‐menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflamm. 2020;17:317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Rettberg JR, Yao J, Brinton RD. Estrogen: a master regulator of bioenergetic systems in the brain and body. Front Neuroendocrinol. 2014;35:8‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Uddin MS, Rahman MM, Jakaria M, et al. Estrogen signaling in Alzheimer's disease: molecular insights and therapeutic targets for Alzheimer's. Dementia Mol Neurobiol. 2020;57:2654‐2670. [DOI] [PubMed] [Google Scholar]
- 87. Brinton RD, Yao J, Yin F, Mack WJ. Cadenas E. Perimenopause as a neurological transition state. Nat Rev Endocrinol. 2015;11:393‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Albert KM, Newhouse PA. Estrogen, stress, and depression: cognitive and biological interactions. Annu Rev Clin Psycho. 2019;15:399‐423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Bagit A, Hayward GC, MacPherson REK. Exercise and estrogen: common pathways in Alzheimer's disease pathology. Am J Physiol‐Endoc M. 2021;321:E164‐E168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Manolagas SC. From estrogen‐centric to aging and oxidative stress: a revised perspective of the pathogenesis of osteoporosis. Endocr Rev. 2010;31:266‐300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Jilka RL, Hangoc G, Girasole G, et al. Increased osteoclast development after estrogen loss: mediation by interleukin‐6. Science. 1992;257:88‐91. [DOI] [PubMed] [Google Scholar]
- 92. Kousteni S, Bellido T, Plotkin LI, et al. Nongenotropic, sex‐nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell. 2001;104:719‐730. [PubMed] [Google Scholar]
- 93. Fischer V, Haffner‐Luntzer M. Interaction between bone and immune cells: implications for postmenopausal osteoporosis. Semin Cell Dev Biol. 2022;123:14‐21. [DOI] [PubMed] [Google Scholar]
- 94. Cheng CH, Chen LR, Chen KH. Osteoporosis due to hormone imbalance: an overview of the effects of estrogen deficiency and glucocorticoid overuse on bone turnover. Int J Mol Sci. 2022;23:1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Pang H, Chen S, Klyne DM, et al. Low back pain and osteoarthritis pain: a perspective of estrogen. Bone Res. 2023;11:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Short RA, Bowen RL, O'Brien PC, Graff‐Radford NR. Elevated gonadotropin levels in patients with Alzheimer disease. Mayo Clin Proc. 2001;76:906‐909. [DOI] [PubMed] [Google Scholar]
- 97. Xiong J, Kang SS, Wang ZH. FSH blockade improves cognition in mice with Alzheimer's disease. Nature. 2022;603:470‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Zaidi M, Lizneva D, Kim SM, et al. FSH, bone mass, body fat, and biological aging. Endocrinology. 2018;159:3503‐3514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Murphy E, Glüer CC, Reid DM, et al. Thyroid function within the upper normal range is associated with reduced bone mineral density and an increased risk of nonvertebral fractures in healthy euthyroid postmenopausal women. J Clin Endocr Metab. 2010;95:3173‐3181. [DOI] [PubMed] [Google Scholar]
- 100. Daya NR, Fretz A, Martin SS, et al. Association between subclinical thyroid dysfunction and fracture risk. Jama Netw Open. 2022;5:e2240823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Zaidi M, Yuen T, Kim SM. Pituitary crosstalk with bone, adipose tissue and brain. Nat Rev Endocrinol. 2023;19:708‐721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Abe E, Marians RC, Yu W, et al. TSH is a negative regulator of skeletal remodeling. Cell. 2003;115:151‐162. [DOI] [PubMed] [Google Scholar]
- 103. Wang YY, Sheng Q, Hou X, et al. Thyrotropin and Alzheimer's disease risk in the elderly: a systematic review and meta‐analysis. Mol Neurobiol. 2016;53:1229‐1236. [DOI] [PubMed] [Google Scholar]
- 104. Marouli E, Yusuf L, Kjaergaard AD, et al. Thyroid function and the risk of Alzheimer's disease: a mendelian randomization study. Thyroid. 2021;31:1794‐1799. [DOI] [PubMed] [Google Scholar]
- 105. Tan ZS, Beiser A, Vasan RS, et al. Thyroid function and the risk of Alzheimer disease: the Framingham Study. Arch Intern Med. 2008;168:1514‐1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Salehipour A, Dolatshahi M, Haghshomar M, Amin J. The role of thyroid dysfunction in Alzheimer's disease: a systematic review and meta‐analysis. Jpad‐J Prev Alzheim. 2023;10:276‐286. [DOI] [PubMed] [Google Scholar]
- 107. Ahmad MH, Fatima M, Mondal AC. Role of hypothalamic‐pituitary‐adrenal axis, hypothalamic‐pituitary‐gonadal axis and insulin signaling in the pathophysiology of Alzheimer's disease. Neuropsychobiology. 2018;77:197‐205. [DOI] [PubMed] [Google Scholar]
- 108. Hebda‐Bauer EK, Simmons TA, Sugg A, et al. 3xTg‐AD mice exhibit an activated central stress axis during early‐stage pathology. J Alzheimers Dis. 2013;33:407‐422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Sotiropoulos I, Catania C, Riedemann T, et al. Glucocorticoids trigger Alzheimer disease‐like pathobiochemistry in rat neuronal cells expressing human tau. J Neurochem. 2008;107:385‐397. [DOI] [PubMed] [Google Scholar]
- 110. Kang JE, Cirrito JR, Dong H, Csernansky JG, Holtzman DM. Acute stress increases interstitial fluid amyloid‐β via corticotropin‐releasing factor and neuronal activity. P Natl Acad Sci USA. 2007;104:10673‐10678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Chotiyarnwong P, McCloskey EV. Pathogenesis of glucocorticoid‐induced osteoporosis and options for treatment. Nat Rev Endocrinol. 2020;16:437‐447. [DOI] [PubMed] [Google Scholar]
- 112. Liu P, Gao YS, Luo PB, et al. Glucocorticoid‐induced expansion of classical monocytes contributes to bone loss. Exp Mol Med. 2022;54:765‐776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Du X, Pang TY. Is dysregulation of the HPA‐axis a core pathophysiology mediating co‐morbid depression in neurodegenerative diseases? Front Psychiatry. 2015;6:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Swaab DF, Bao AM, Lucassen PJ. The stress system in the human brain in depression and neurodegeneration. Ageing Res Rev. 2005;4:141‐194. [DOI] [PubMed] [Google Scholar]
- 115. Caruso A, Nicoletti F, Mango D, Saidi A, Orlando R, Scaccianoce S. Stress as risk factor for Alzheimer's disease. Pharmacol Res. 2018;132:130‐134. [DOI] [PubMed] [Google Scholar]
- 116. Isales CM, Zaidi M, Blair HC. ACTH is a novel regulator of bone mass. Ann Ny Acad Sci. 2010;1192:110‐116. [DOI] [PubMed] [Google Scholar]
- 117. Chen QC, Zhang Y. The role of NPY in the regulation of bone metabolism. Front Endocrinol. 2022;13:833485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Sunderland T, Berrettini WH, Molchan SE, et al. Reduced cerebrospinal fluid dynorphin A1‐8 in Alzheimer's disease. Biol Psychiatry. 1991;30:81‐87. [DOI] [PubMed] [Google Scholar]
- 119. Koide S, Onishi H, Hashimoto H, Kai T, Yamagami S. Plasma neuropeptide Y is reduced in patients with Alzheimer's disease. Neurosci Lett. 1995;198:149‐151. [DOI] [PubMed] [Google Scholar]
- 120. Proto C, Romualdi D, Cento RM, et al. Plasma levels of neuropeptides in Alzheimer's disease. Gynecol Endocrinol. 2006;22:213‐218. [DOI] [PubMed] [Google Scholar]
- 121. Martignoni E, Blandini F, Petraglia F, Pacchetti C, Bono G, Nappi G. Cerebrospinal fluid norepinephrine, 3‐methoxy‐4‐hydroxyphenylglycol and neuropeptide Y levels in Parkinson's disease, multiple system atrophy and dementia of the Alzheimer type. J Neural Transm Park Dis Dement Sect. 1992;4:191‐205. [DOI] [PubMed] [Google Scholar]
- 122. Diez M, Koistinaho J, Kahn K, Games D, Hökfelt T. Neuropeptides in hippocampus and cortex in transgenic mice overexpressing V717F beta‐amyloid precursor protein–initial observations. Neuroscience. 2000;100:259‐286. [DOI] [PubMed] [Google Scholar]
- 123. Diez M, Danner S, Frey P, et al. Neuropeptide alterations in the hippocampal formation and cortex of transgenic mice overexpressing β‐amyloid precursor protein (APP) with the Swedish double mutation (APP23). Neurobiol Dis. 2003;14:579‐594. [DOI] [PubMed] [Google Scholar]
- 124. Ramos B, Baglietto‐Vargas D, del Rio JC, et al. Early neuropathology of somatostatin/NPY GABAergic cells in the hippocampus of a PS1xAPP transgenic model of Alzheimer's disease. Neurobiol Aging. 2006;27:1658‐1672. [DOI] [PubMed] [Google Scholar]
- 125. Krezymon A, Richetin K, Halley H, et al. Modifications of hippocampal circuits and early disruption of adult neurogenesis in the Tg2576 mouse model of Alzheimer's disease. Plos One. 2013;8:e76497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Croce N, Gelfo F, Ciotti MT, et al. NPY modulates miR‐30a‐5p and BDNF in opposite direction in an in vitro model of Alzheimer disease: a possible role in neuroprotection? Mol Cell Biochem. 2013;376:189‐915. [DOI] [PubMed] [Google Scholar]
- 127. Croce N, Ciotti MT, Gelfo F, et al. Neuropeptide Y protects rat cortical neurons against β‐amyloid toxicity and re‐establishes synthesis and release of nerve growth factor. Acs Chem Neurosci. 2012;3:312‐318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Angelucci F, Gelfo F, Fiore M, et al. The effect of neuropeptide Y on cell survival and neurotrophin expression in in‐vitro models of Alzheimer's disease. Can J Physiol Pharm. 2014;92:621‐630. [DOI] [PubMed] [Google Scholar]
- 129. Spencer B, Potkar R, Metcalf J, et al. Systemic central nervous system (CNS)‐targeted delivery of neuropeptide Y (NPY) reduces neurodegeneration and increases neural precursor cell proliferation in a mouse model of Alzheimer disease. J Biol Chem. 2016;291:1905‐1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Li CR, Wu XJ, Liu S, Zhao Y, Zhu J, Liu KD. Roles of neuropeptide Y in neurodegenerative and neuroimmune diseases. Front Neurosci‐Switz. 2019;13:869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Duarte‐Neves J, de Almeida LP, Cavadas C. Neuropeptide Y (NPY) as a therapeutic target for neurodegenerative diseases. Neurobiol Dis. 2016;95:210‐224. [DOI] [PubMed] [Google Scholar]
- 132. Ishii M, Hiller AJ, Pham L, McGuire MJ, Iadecola C, Wang G. Amyloid‐beta modulates low‐threshold activated voltage‐gated L‐type calcium channels of arcuate neuropeptide Y neurons leading to calcium dysregulation and hypothalamic dysfunction. J Neurosci. 2019;39:8816‐8825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Wang FS, Lian WS, Weng WT, et al. Neuropeptide Y mediates glucocorticoid‐induced osteoporosis and marrow adiposity in mice. Osteoporosis Int. 2016;27:2777‐2789. [DOI] [PubMed] [Google Scholar]
- 134. Zhang Y, Chen CY, Liu YW, et al. Neuronal induction of bone‐fat imbalance through osteocyte neuropeptide Y. Adv Sci. 2021;8:e2100808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Chen ZJ, Lv MY, Liang J, et al. Neuropeptide Y‐mediated gut microbiota alterations aggravate postmenopausal osteoporosis. Adv Sci. 2023;10:e2303015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Lv X, Gao F, Li TP, et al. Skeleton interoception regulates bone and fat metabolism through hypothalamic neuroendocrine NPY. Elife. 2021;10:e70324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Xie WC, Li F, Han Y, et al. Neuropeptide Y1 receptor antagonist promotes osteoporosis and microdamage repair and enhances osteogenic differentiation of bone marrow stem cells via cAMP/PKA/CREB pathway. Aging‐Us. 2020;12:8120‐8136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Liu S, Jin D, Wu JQ, et al. Neuropeptide Y stimulates osteoblastic differentiation and VEGF expression of bone marrow mesenchymal stem cells related to canonical Wnt signaling activating in vitro. Neuropeptides. 2016;56:105‐113. [DOI] [PubMed] [Google Scholar]
- 139. Dong PH, Gu XC, Zhu GL, Li M, Ma B, Zi Y. Melatonin induces osteoblastic differentiation of mesenchymal stem cells and promotes fracture healing in a rat model of femoral fracture via neuropeptide Y/Neuropeptide Y Receptor Y1 Signaling. Pharmacology. 2018;102:272‐280. [DOI] [PubMed] [Google Scholar]
- 140. Park MH, Kim N, Jin HK, Bae JS. Neuropeptide Y‐based recombinant peptides ameliorate bone loss in mice by regulating hematopoietic stem/progenitor cell mobilization. Bmb Rep. 2017;50:138‐143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Abbara A, Clarke SA, Dhillo WS. Clinical potential of kisspeptin in reproductive health. Trends Mol Med. 2021;27:807‐823. [DOI] [PubMed] [Google Scholar]
- 142. Milton NGN, Chilumuri A, Rocha‐Ferreira E, Nercessian AN, Ashioti M. Kisspeptin prevention of amyloid‐β peptide neurotoxicity. Acs Chem Neurosci. 2012;3:706‐719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Mills EG, Yang L, Nielsen MF, Kassem M, Dhillo WS, Comninos AN. The relationship between bone and reproductive hormones beyond estrogens and androgens. Endocr Rev. 2021;42:691‐719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Comninos AN, Hansen MS, Courtney A, et al. Acute effects of kisspeptin administration on bone metabolism in healthy men. J Clin Endocr Metab. 2022;107:1529‐1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Herber CB, Krause WC, Wang LP, et al. Estrogen signaling in arcuate neurons suppresses a sex‐dependent female circuit promoting dense strong bones. Nat Commun. 2019;10:163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Mach DB, Rogers SD, Sabino MC, et al. Origins of skeletal pain: sensory and sympathetic innervation of the mouse femur. Neuroscience. 2002;113:155‐166. [DOI] [PubMed] [Google Scholar]
- 147. Affoo RH, Foley N, Rosenbek J, Kevin Shoemaker J, Martin RE. Swallowing dysfunction and autonomic nervous system dysfunction in Alzheimer's disease: a scoping review of the evidence. J Am Geriatr Soc. 2013;61:2203‐2213. [DOI] [PubMed] [Google Scholar]
- 148. Ma C, Zhang Y, Cao Y, et al. Autonomic neural regulation in mediating the brain‐bone axis: mechanisms and implications for regeneration under psychological stress. Qjm‐Int J Med. 2024;117:95‐108. [DOI] [PubMed] [Google Scholar]
- 149. Bartus RT, Dean RL 3rd, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217:408‐414. [DOI] [PubMed] [Google Scholar]
- 150. Hampel H, Mesulam MM, Cuello AC, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer's disease. Brain. 2018;141:1917‐1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Ma YY, Li XX, Fu J, et al. Acetylcholine affects osteocytic MLO‐Y4 cells via acetylcholine receptors. Mol Cell Endocrinol. 2014;384:155‐164. [DOI] [PubMed] [Google Scholar]
- 152. Kliemann K, Kneffel M, Bergen I, et al. Quantitative analyses of bone composition in acetylcholine receptor M3R and alpha7 knockout mice. Life Sci. 2012;91:997‐1002. [DOI] [PubMed] [Google Scholar]
- 153. Shi Y, Oury F, Yadav VK, et al. Signaling through the M(3) muscarinic receptor favors bone mass accrual by decreasing sympathetic activity. Cell Metab. 2010;11:231‐238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Baumann L, Kauschke V, Vikman A, et al. Deletion of nicotinic acetylcholine receptor alpha9 in mice resulted in altered bone structure. Bone. 2019;120:285‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Bajayo A, Bar A, Denes A, et al. Skeletal parasympathetic innervation communicates central IL‐1 signals regulating bone mass accrual. P Natl Acad Sci USA. 2012;109:15455‐15460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Mandl P, Hayer S, Karonitsch T, et al. Nicotinic acetylcholine receptors modulate osteoclastogenesis. Arthritis Res Ther. 2016;18:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Elefteriou F. Impact of the autonomic nervous system on the skeleton. Physiol Rev. 2018;98:1083‐1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Kondo H, Nifuji A, Takeda S, et al. Unloading induces osteoblastic cell suppression and osteoclastic cell activation to lead to bone loss via sympathetic nervous system. J Biol Chem. 2005;280:30192‐30200. [DOI] [PubMed] [Google Scholar]
- 159. Dimitri P, Rosen C. The central nervous system and bone metabolism: an evolving story. Calcified Tissue Int. 2017;100:476‐485. [DOI] [PubMed] [Google Scholar]
- 160. Culibrk RA, Arabiyat AS, DeKalb CA, Hahn MS. Modeling sympathetic hyperactivity in Alzheimer's related bone loss. J Alzheimers Dis. 2021;84:647‐658. [DOI] [PubMed] [Google Scholar]
- 161. Hu CH, Sui BD, Liu J, Dang L, et al. Sympathetic neurostress drives osteoblastic exosomal MiR‐21 transfer to disrupt bone homeostasis and promote osteopenia. Small Methods. 2022;6:e2100763. [DOI] [PubMed] [Google Scholar]
- 162. Sun Y, Zhang HW, Zhang X, et al. Promotion of astrocyte‐neuron glutamate‐glutamine shuttle by SCFA contributes to the alleviation of Alzheimer's disease. Redox Biol. 2023;62:102690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Zott B, Simon MM, Hong W, et al. A vicious cycle of β amyloid‐dependent neuronal hyperactivation. Science. 2019;365:559‐565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Wood H. Glutamate perpetuates amyloid‐β‐dependent neuronal hyperactivity. Nat Rev Neurol. 2019;15:558‐559. [DOI] [PubMed] [Google Scholar]
- 165. Cox MF, Hascup ER, Bartke A, Hascup KN. Defining the role of glutamate in aging and Alzheimer's disease. Front Aging. 2022;3:929474. Friend or Foe? [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Hogan‐Cann AD, Anderson CM. Physiological roles of non‐neuronal NMDA receptors. Trends Pharmacol Sci. 2016;37:750‐767. [DOI] [PubMed] [Google Scholar]
- 167. Li JL, Zhao L, Cui B, Deng LF, Ning G, Liu JM. Multiple signaling pathways involved in stimulation of osteoblast differentiation by N‐methyl‐D‐aspartate receptors activation in vitro. Acta Pharmacol Sin. 2011;32:895‐903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Rivera‐Villaseñor A, Higinio‐Rodríguez F, Nava‐Gómez L, et al. NMDA receptor hypofunction in the aging‐associated malfunction of peripheral tissue. Front Physiol. 2021;12:687121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Xia W, Xie J, Cai ZQ, et al. Damaged brain accelerates bone healing by releasing small extracellular vesicles that target osteoprogenitors. Nat Commun. 2021;12:6043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Liu XX, Chen CY, Jiang YL, et al. Brain‐derived extracellular vesicles promote bone‐fat imbalance in Alzheimer's disease. Int J Biol Sci. 2023;19:2409‐2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Karsenty G. Osteocalcin: a multifaceted bone‐derived hormone. Annu Rev Nutr. 2023;43:55‐71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Razny U, Fedak D, Kiec‐Wilk B, et al. Carboxylated and undercarboxylated osteocalcin in metabolic complications of human obesity and prediabetes. Diabetes‐Metab Res. 2017;33:e2862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Tacey A, Smith C, Woessner MN, et al. Undercarboxylated osteocalcin is associated with vascular function in female older adults but does not influence vascular function in male rabbit carotid artery. Plos One. 2020;15:e0242774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Puig J, Blasco G, Daunis‐i‐Estadella J, et al. Lower serum osteocalcin concentrations are associated with brain microstructural changes and worse cognitive performance. Clin Endocrinol. 2016;84:756‐763. [DOI] [PubMed] [Google Scholar]
- 176. Liu W, Hu Q, Zhang F, Shi K, Wu J. Investigation of the causal relationship between osteocalcin and dementia: a Mendelian randomization study. Heliyon. 2023;9:e21073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Oury F, Khrimian L, Denny CA, et al. Maternal and offspring pools of osteocalcin influence brain development and functions. Cell. 2013;155:228‐241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Khrimian L, Obri A, Ramos‐Brossier M, et al. Gpr158 mediates osteocalcin's regulation of cognition. J Exp Med. 2017;214:2859‐2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Kosmidis S, Polyzos A, Harvey L, et al. RbAp48 protein is a critical component of GPR158/OCN signaling and ameliorates age‐related memory loss. Cell Rep. 2018;25. 959‐973.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Shan C, Zhang D, Ma DN, et al. Osteocalcin ameliorates cognitive dysfunctions in a mouse model of Alzheimer's Disease by reducing amyloid β burden and upregulating glycolysis in neuroglia. Cell Death Discov. 2023;9:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Song CH, Orlandi C, Sutton LP, Martemyanov KA. The signaling proteins GPR158 and RGS7 modulate excitability of L2/3 pyramidal neurons and control A‐type potassium channel in the prelimbic cortex. J Biol Chem. 2019;294:13145‐13157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Qian ZJ, Li HC, Yang HY, et al. Osteocalcin attenuates oligodendrocyte differentiation and myelination via GPR37 signaling in the mouse brain. Sci Adv. 2021;7:eabi5811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Chen JF, Liu K, Hu B, et al. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer's disease. Neuron. 2021;109:2292‐2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Depp C, Sun T, Sasmita AO, et al. Myelin dysfunction drives amyloid‐β deposition in models of Alzheimer's disease. Nature. 2023;618:349‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Al Jaberi S, Cohen A, Abdulrazzaq YM, Ojha S, Bastaki S, Adeghate EA. Lipocalin‐2: structure, function, distribution and role in metabolic disorders. Biomed Pharmacother. 2021;142:112002. [DOI] [PubMed] [Google Scholar]
- 186. Costa D, Lazzarini E, Canciani B, et al. Altered bone development and turnover in transgenic mice over‐expressing lipocalin‐2 in bone. J Cell Physiol. 2013;228:2210‐2221. [DOI] [PubMed] [Google Scholar]
- 187. Mosialou I, Shikhel S, Liu JM, et al. MC4R‐dependent suppression of appetite by bone‐derived lipocalin 2. Nature. 2017;543:385‐390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Wu BW, Guo JD, Wu MS, et al. Osteoblast‐derived lipocalin‐2 regulated by miRNA‐96‐5p/Foxo1 advances the progression of Alzheimer's disease. Epigenomics‐Uk. 2020;12:1501‐1513. [DOI] [PubMed] [Google Scholar]
- 189. Eruysal E, Ravdin L, Kamel H, Iadecola C, Ishii M. Plasma lipocalin‐2 levels in the preclinical stage of Alzheimer's disease. Alzheimers Dement (Amst). 2019;11:646‐653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Hermann P, Villar‐Piqué A, Schmitz M, et al. Plasma Lipocalin 2 in Alzheimer's disease: potential utility in the differential diagnosis and relationship with other biomarkers. Alzheimers Res Ther. 2022;14:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Ferreira AC, Dá Mesquita S, Sousa JC, et al. From the periphery to the brain: lipocalin‐2, a friend or foe? Prog Neurobiol. 2015;131:120‐136. [DOI] [PubMed] [Google Scholar]
- 192. Riminucci M, Collins MT, Fedarko NS, et al. FGF‐23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. J Clin Invest. 2003;112:683‐692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Millar SA, Anderson SI, O'Sullivan SE. Osteokines and the vasculature: a review of the effects of osteocalcin, fibroblast growth factor‐23 and lipocalin‐2. Peerj. 2019;7:e7139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Prud'homme GJ, Kurt M, Wang Q. Pathobiology of the klotho antiaging protein and therapeutic considerations. Front Aging. 2022;3:931331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. McGrath ER, Himali JJ, Levy D, et al. Circulating fibroblast growth factor 23 levels and incident dementia: the Framingham heart study. Plos One. 2019;14:e0213321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Li BS, Zhou M, Peng J, et al. Mechanism of the fibroblast growth factor 23/α‐Klotho axis in peripheral blood mononuclear cell inflammation in Alzheimer's disease. Immunol Invest. 2022;51:1471‐1484. [DOI] [PubMed] [Google Scholar]
- 197. Liu PD, Chen L, Bai XY, Karaplis A, Miao DS, Gu N. Impairment of spatial learning and memory in transgenic mice overexpressing human fibroblast growth factor‐23. Brain Res. 2011;1412:9‐17. [DOI] [PubMed] [Google Scholar]
- 198. Boyce BF, Xing LP. Biology of RANK, RANKL, and osteoprotegerin. Arthritis Res Ther. 2007;9(1):S1. Suppl. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Han YJ, You XL, Xing WH, Zhang Z, Zou WG. Paracrine and endocrine actions of bone‐the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res. 2018;6:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Walsh MC, Choi Y. Biology of the RANKL‐RANK‐OPG system in immunity, bone, and beyond. Front Immunol. 2014;5:511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Emanuele E, Peros E, Scioli GA, et al. Plasma osteoprotegerin as a biochemical marker for vascular dementia and Alzheimer's disease. Int J Mol Med. 2004;13:849‐853. [PubMed] [Google Scholar]
- 202. Luckhaus C, Mahabadi B, Grass‐Kapanke B, et al. Blood biomarkers of osteoporosis in mild cognitive impairment and Alzheimer's disease. J Neural Transm. 2009;116:905‐911. [DOI] [PubMed] [Google Scholar]
- 203. Jefferson AL, Massaro JM, Wolf PA, et al. Inflammatory biomarkers are associated with total brain volume—The Framingham Heart Study. Neurology. 2007;68:1032‐1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Glasnović A, O'Mara N, Kovačić N, Grčević D, Gajović S. RANK/RANKL/OPG signaling in the brain: a systematic review of the literature. Front Neurol. 2020;11:590480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Icer MA, Gezmen‐Karadag M. The multiple functions and mechanisms of osteopontin. Clin Biochem. 2018;59:17‐24. [DOI] [PubMed] [Google Scholar]
- 206. Singh A, Gill G, Kaur H, Amhmed M, Jakhu H. Role of osteopontin in bone remodeling and orthodontic tooth movement: a review. Prog Orthod. 2018;19:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Fodor D, Bondor C, Albu A, Simon SP, Craciun A, Muntean L. The value of osteopontin in the assessment of bone mineral density status in postmenopausal women. J Invest Med. 2013;61:15‐21. [DOI] [PubMed] [Google Scholar]
- 208. Wei QS, Huang L, Tan X, Chen ZQ, Chen SM, Deng WM. Serum osteopontin levels in relation to bone mineral density and bone turnover markers in postmenopausal women. Scand J Clin Lab Inv. 2016;76:33‐39. [DOI] [PubMed] [Google Scholar]
- 209. Comi C, Carecchio M, Chiocchetti A, et al. Osteopontin is increased in the cerebrospinal fluid of patients with Alzheimer's disease and its levels correlate with cognitive decline. J Alzheimers Dis. 2010;19:1143‐1148. [DOI] [PubMed] [Google Scholar]
- 210. Sun Y, Yin XS, Guo H, Han RK, He RD, Chi LJ. Elevated osteopontin levels in mild cognitive impairment and Alzheimer's disease. Mediat Inflamm. 2013;2013:615745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Chai YL, Chong JR, Raquib AR, et al. Plasma osteopontin as a biomarker of Alzheimer's disease and vascular cognitive impairment. Sci Rep‐Uk. 2021;11:4010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Qiu Y, Shen X, Ravid O, et al. Definition of the contribution of an Osteopontin‐producing CD11c(+) microglial subset to Alzheimer's disease. Proc Natl Acad Sci U S A. 2023;120:e2218915120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Ataman B, Boulting GL, Harmin DA, et al. Evolution of Osteocrin as an activity‐regulated factor in the primate brain. Nature. 2016;539:242‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Thomas G, Moffatt P, Salois P, et al. Osteocrin, a novel bone‐specific secreted protein that modulates the osteoblast phenotype. J Biol Chem. 2003;278:50563‐50571. [DOI] [PubMed] [Google Scholar]
- 215. Watanabe‐Takano H, Ochi H, Chiba A, et al. Mechanical load regulates bone growth via periosteal Osteocrin. Cell Rep. 2021;36:109380. [DOI] [PubMed] [Google Scholar]
- 216. Kanai Y, Yasoda A, Mori KP, et al. Circulating osteocrin stimulates bone growth by limiting C‐type natriuretic peptide clearance. J Clin Invest. 2017;127:4136‐4147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Mazur CM, Castro Andrade CD, Tokavanich N, et al. Partial prevention of glucocorticoid‐induced osteocyte deterioration in young male mice with osteocrin gene therapy. iScience. 2022;25:105019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Wang JS, Kamath T, Mazur CM, et al. Control of osteocyte dendrite formation by Sp7 and its target gene osteocrin. Nat Commun. 2021;12:6271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Ataka K, Asakawa A, Iwai H, Kato I. Musclin prevents depression‐like behavior in male mice by activating urocortin 2 signaling in the hypothalamus. Front Endocrinol (Lausanne). 2023;14:1288282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Cho W, Oh H, Choi SW, et al. Musclin mitigates the attachment of HUVECs to THP‐1 monocytes in hyperlipidemic conditions through PPARalpha/HO‐1‐mediated attenuation of inflammation. Inflammation. 2024;47:1‐12. [DOI] [PubMed] [Google Scholar]
- 221. Delgado‐Calle J, Sato AY, Bellido T. Role and mechanism of action of sclerostin in bone. Bone. 2017;96:29‐37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Gao Y, Chen N, Fu Z, Zhang Q. Progress of Wnt signaling pathway in osteoporosis. Biomolecules. 2023;13:483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Chauhan A, Bhakhar MK, Goyal P. Serum levels of sclerostin in prediabetes and its correlation with bone mineral density. J Family Med Prim Care. 2023;12:2702‐2707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Starlinger J, Santol J, Kaiser G, Sarahrudi K. Close negative correlation of local and circulating Dickkopf‐1 and Sclerostin levels during human fracture healing. Sci Rep. 2024;14:6524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Yuan J, Pedrini S, Thota R, et al. Elevated plasma sclerostin is associated with high brain amyloid‐β load in cognitively normal older adults. NPJ Aging. 2023;9:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Shi T, Shen S, Shi Y, et al. Osteocyte‐derived sclerostin impairs cognitive function during ageing and Alzheimer's disease progression. Nat Metab. 2024;6:531‐549. [DOI] [PubMed] [Google Scholar]
- 227. Xu C, Di C. The BMP signaling and in vivo bone formation. Gene. 2005;357:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Bragdon B, Moseychuk O, Saldanha S, King D, Julian J, Nohe A. Bone morphogenetic proteins: a critical review. Cell Signal. 2011;23:609‐620. [DOI] [PubMed] [Google Scholar]
- 229. Nakatsu D, Kunishige R, Taguchi Y, et al. BMP4‐SMAD1/5/9‐RUNX2 pathway activation inhibits neurogenesis and oligodendrogenesis in Alzheimer's patients' iPSCs in senescence‐related conditions. Stem Cell Rep. 2023;18:688‐705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Li DB, Tang J, Xu HW, Fan XT, Bai Y, Yang L. Decreased hippocampal cell proliferation correlates with increased expression of BMP4 in the APPswe/PS1ΔE9 mouse model of Alzheimer's disease. Hippocampus. 2008;18:692‐698. [DOI] [PubMed] [Google Scholar]
- 231. Zhang XQ, Li J, Ma L, et al. BMP4 overexpression induces the upregulation of APP/Tau and memory deficits in Alzheimer's disease. Cell Death Discov. 2021;7:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Skauli N, Savchenko E, Ottersen OP, Roybon L, Amiry‐Moghaddam M. Canonical bone morphogenetic protein signaling regulates expression of aquaporin‐4 and its anchoring complex in mouse astrocytes. Front Cell Neurosci. 2022;16:878154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Sun L, Guo CN, Song Y, Sheng JH, Xiao SF, AsDN Initi. Blood BMP6 associated with cognitive performance and Alzheimer's disease diagnosis: a longitudinal study of elders. J Alzheimers Dis. 2022;88:641‐651. [DOI] [PubMed] [Google Scholar]
- 234. Cai Y, Wang Z, Liao B, Sun Z, Zhu P. Anti‐inflammatory and chondroprotective effects of platelet‐derived growth factor‐BB on osteoarthritis rat models. J Gerontol A Biol Sci Med Sci. 2023;78:51‐59. [DOI] [PubMed] [Google Scholar]
- 235. Zhu P, Wang Z, Sun Z, Liao B, Cai Y. Recombinant platelet‐derived growth factor‐BB alleviates osteoarthritis in a rat model by decreasing chondrocyte apoptosis in vitro and in vivo. J Cell Mol Med. 2021;25:7472‐7484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Santhanam L, Liu G, Jandu S, et al. Skeleton‐secreted PDGF‐BB mediates arterial stiffening. J Clin Invest. 2021;131:e147116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Zaidi M, Lizneva D, Yuen T. The role of PDGF‐BB in the bone‐vascular relationship during aging. J Clin Invest. 2021;131:e153644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Su W, Liu G, Liu X, et al. Angiogenesis stimulated by elevated PDGF‐BB in subchondral bone contributes to osteoarthritis development. JCI Insight. 2020;5:e135446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Xie H, Cui Z, Wang L, et al. PDGF‐BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis. Nat Med. 2014;20:1270‐1278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Wang JK, Fang CL, Noller K, et al. Bone‐derived PDGF‐BB drives brain vascular calcification in male mice. J Clin Invest. 2023;133:e168447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Liu G, Wang J, Wei Z, et al. Elevated PDGF‐BB from Bone Impairs Hippocampal Vasculature by Inducing PDGFRβ Shedding from Pericytes. Adv Sci (Weinh). 2023;10:e2206938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Xu Z, Rao YX, Huang YB, et al. Efficient strategies for microglia replacement in the central nervous system. Cell Rep. 2020;32:108041. [DOI] [PubMed] [Google Scholar]
- 243. Varvel NH, Grathwohl SA, Degenhardt K, et al. Replacement of brain‐resident myeloid cells does not alter cerebral amyloid‐β deposition in mouse models of Alzheimer's disease. J Exp Med. 2015;212:1803‐1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Zhou R, Wang L, Chen L, et al. Bone marrow‐derived GCA(+) immune cells drive Alzheimer's disease progression. Adv Sci (Weinh). 2023;10:e2303402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Arthur A, Gronthos S. Clinical application of bone marrow mesenchymal stem/stromal cells to repair skeletal tissue. Int J Mol Sci. 2020;21:9759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Jiang YH, Zhang P, Zhang X, Lv LW, Zhou YS. Advances in mesenchymal stem cell transplantation for the treatment of osteoporosis. Cell Proliferat. 2021;54:e12956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Qin C, Lu YL, Wang KW, et al. Transplantation of bone marrow mesenchymal stem cells improves cognitive deficits and alleviates neuropathology in animal models of Alzheimer's disease: a meta‐analytic review on potential mechanisms. Transl Neurodegener. 2020;9:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Bae JS, Jin HK, Lee JK, Richardson JC, Carter JE. Bone marrow‐derived mesenchymal stem cells contribute to the reduction of amyloid‐β deposits and the improvement of synaptic transmission in a mouse model of pre‐dementia Alzheimer's disease. Curr Alzheimer Res. 2013;10:524‐531. [PubMed] [Google Scholar]
- 249. Lee JK, Jin HK, Endo S, Schuchman EH, Carter JE, Bae JS. Intracerebral transplantation of bone marrow‐derived mesenchymal stem cells reduces amyloid‐beta deposition and rescues memory deficits in Alzheimer's disease mice by modulation of immune responses. Stem Cells. 2010;28:329‐343. [DOI] [PubMed] [Google Scholar]
- 250. Lee JK, Jin HK, Bae JS. Bone marrow‐derived mesenchymal stem cells reduce brain amyloid‐β deposition and accelerate the activation of microglia in an acutely induced Alzheimer's disease mouse model. Neurosci Lett. 2009;450:136‐141. [DOI] [PubMed] [Google Scholar]
- 251. Neves AF, Camargo C, Premer C, Hare JM, Baumel BS, Pinto M. Intravenous administration of mesenchymal stem cells reduces Tau phosphorylation and inflammation in the 3xTg‐AD mouse model of Alzheimer's disease. Exp Neurol. 2021;341:113706. [DOI] [PubMed] [Google Scholar]
- 252. Qin C, Li YN, Wang KW. Functional mechanism of bone marrow‐derived mesenchymal stem cells in the treatment of animal models with Alzheimer's disease: inhibition of neuroinflammation. J Inflamm Res. 2021;14:4761‐4775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Naaldijk Y, Jäger C, Fabian C, et al. Effect of systemic transplantation of bone marrow‐derived mesenchymal stem cells on neuropathology markers in APP/PS1 Alzheimer mice. Neuropath Appl Neuro. 2017;43:299‐314. [DOI] [PubMed] [Google Scholar]
- 254. Wei Y, Xie ZH, Bi JZ, Zhu ZY. Anti‐inflammatory effects of bone marrow mesenchymal stem cells on mice with Alzheimer's disease. Exp Ther Med. 2018;16:5015‐5020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Farahzadi R, Fathi E, Vietor I. Mesenchymal stem cells could be considered as a candidate for further studies in cell‐based therapy of Alzheimer's disease via targeting the signaling pathways. Acs Chem Neurosci. 2020;11:1424‐1435. [DOI] [PubMed] [Google Scholar]
- 256. Qin C, Bai L, Li YN, Wang KW. The functional mechanism of bone marrow‐derived mesenchymal stem cells in the treatment of animal models with Alzheimer's disease: crosstalk between autophagy and apoptosis. Stem Cell Res Ther. 2022;13:90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Herisson F, Frodermann V, Courties G, et al. Direct vascular channels connect skull bone marrow and the brain surface enabling myeloid cell migration. Nat Neurosci. 2018;21:1209‐1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Cugurra A, Mamuladze T, Rustenhoven J, et al. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma. Science. 2021;373:eabf7844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Pulous FE, Cruz‐Hernandez JC, Yang CB, et al. Cerebrospinal fluid can exit into the skull bone marrow and instruct cranial hematopoiesis in mice with bacterial meningitis. Nat Neurosci. 2022;25:567‐576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Tijms BM, Vromen EM, Mjaavatten O, et al. Cerebrospinal fluid proteomics in patients with Alzheimer's disease reveals five molecular subtypes with distinct genetic risk profiles. Nature Aging. 2024;4:33‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Liu J, Li DF, Wu XH, Dang L, Lu AP, Zhang G. Bone‐derived exosomes. Curr Opin Pharmacol. 2017;34:64‐69. [DOI] [PubMed] [Google Scholar]
- 262. Xie Y, Chen YY, Zhang LC, Ge W, Tang PF. The roles of bone‐derived exosomes and exosomal microRNAs in regulating bone remodelling. J Cell Mol Med. 2017;21:1033‐1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Jiang YL, Wang ZX, Liu XX, et al. The protective effects of osteocyte‐derived extracellular vesicles against Alzheimer's disease diminished with aging. Adv Sci. 2022;9:e2105316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Wang ZX, Luo ZW, Li FXZ, et al. Aged bone matrix‐derived extracellular vesicles as a messenger for calcification paradox. Nat Commun. 2022;13:1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Lin YF, Smith AV, Aspelund T, et al. Genetic overlap between vascular pathologies and Alzheimer's dementia and potential causal mechanisms. Alzheimers Dement. 2019;15:65‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Beukel TC van den, Wolters FJ, Siebert U, et al. Intracranial arteriosclerosis and the risk of dementia: a population‐based cohort study. Alzheimers Dement. 2024;20:869‐879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Elia CA, Tamborini M, Rasile M, et al. Intracerebral injection of extracellular vesicles from mesenchymal stem cells exerts reduced Aβ plaque burden in early stages of a preclinical model of Alzheimer's disease. Cells‐Basel. 2019;8:1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Cone AS, Yuan XG, Sun L, et al. Mesenchymal stem cell‐derived extracellular vesicles ameliorate Alzheimer's disease‐like phenotypes in a preclinical mouse model. Theranostics. 2021;11:8129‐8142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Wang SS, Jia JJ, Wang ZF. Mesenchymal stem cell‐derived extracellular vesicles suppresses iNOS expression and ameliorates neural impairment in Alzheimer's disease mice. J Alzheimers Dis. 2018;61:1005‐1013. [DOI] [PubMed] [Google Scholar]
- 270. Liu S, Fan M, Xu JX, et al. Exosomes derived from bone‐marrow mesenchymal stem cells alleviate cognitive decline in AD‐like mice by improving BDNF‐related neuropathology. J Neuroinflamm. 2022;19:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Chang J, Wang Z, Tang E, et al. Inhibition of osteoblastic bone formation by nuclear factor‐κB. Nat Med. 2009;15:682‐689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Arra M, Swarnkar G, Ke K, et al. LDHA‐mediated ROS generation in chondrocytes is a potential therapeutic target for osteoarthritis. Nat Commun. 2020;11:3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Yu B, Wang CY. Osteoporosis and periodontal diseases—An update on their association and mechanistic links. Periodontol 2000. 2022;89:99‐113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Wang TT, He CQ. Pro‐inflammatory cytokines: the link between obesity and osteoarthritis. Cytokine Growth F R. 2018;44:38‐50. [DOI] [PubMed] [Google Scholar]
- 275. Kany S, Vollrath JT, Relja B. Cytokines in inflammatory disease. Int J Mol Sci. 2019;20:6008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Ishihara K, Hirano T. IL‐6 in autoimmune disease and chronic inflammatory proliferative disease. Cytokine Growth Factor Rev. 2002;13:357‐368. [DOI] [PubMed] [Google Scholar]
- 277. De Roover A, Escribano‐Nunez A, Monteagudo S, Lories R. Fundamentals of osteoarthritis: inflammatory mediators in osteoarthritis. Osteoarthr Cartilage. 2023;31:1303‐1311. [DOI] [PubMed] [Google Scholar]
- 278. Zhao YP, Li YH, Qu RZ, et al. Cortistatin binds to TNF‐α receptors and protects against osteoarthritis. Ebiomedicine. 2019;41:556‐570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Calsolaro V, Edison P. Neuroinflammation in Alzheimer's disease: current evidence and future directions. Alzheimers Dement. 2016;12:719‐732. [DOI] [PubMed] [Google Scholar]
- 280. Heneka MT, Carson MJ, El Khoury J, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015;14:388‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Li CH, Stebbins RC, Noppert GA, et al. Peripheral immune function and Alzheimer's disease: a living systematic review and critical appraisal. Mol Psychiatr. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiat. 2015;77:43‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Jin XZ, Beguerie JR, Zhang WY, et al. Circulating C reactive protein in osteoarthritis: a systematic review and meta‐analysis. Ann Rheum Dis. 2015;74:703‐710. [DOI] [PubMed] [Google Scholar]
- 284. Perruccio AV, Chandran V, Power JD, Kapoor M, Mahomed NN, Gandhi R. Systemic inflammation and painful joint burden in osteoarthritis: a matter of sex? Osteoarthritis Cartilage. 2017;25:53‐59. [DOI] [PubMed] [Google Scholar]
- 285. Di D, Zhou H, Cui Z, et al. Frailty phenotype as mediator between systemic inflammation and osteoporosis and fracture risks: a prospective study. J Cachexia Sarcopenia Muscle. 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Cunningham C, Campion S, Lunnon K, et al. Systemic inflammation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biol Psychiat. 2009;65:304‐312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Lopez‐Rodriguez AB, Hennessy E, et al. Acute systemic inflammation exacerbates neuroinflammation in Alzheimer's disease: iL‐1β drives amplified responses in primed astrocytes and neuronal network dysfunction. Alzheimers Dement. 2021;17:1735‐1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Walker KA, Le Page LM, Terrando N, Duggan MR, Heneka MT, Bettcher BM. The role of peripheral inflammatory insults in Alzheimer's disease: a review and research roadmap. Mol Neurodegener. 2023;18:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Kyrkanides S, Tallents RH, Miller JNH, et al. Osteoarthritis accelerates and exacerbates Alzheimer's disease pathology in mice. J Neuroinflamm. 2011;8:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Gupta DP, Lee YS, Choe Y, Kim KT, Song GJ, Hwang SC. Knee osteoarthritis accelerates amyloid beta deposition and neurodegeneration in a mouse model of Alzheimer's disease. Mol Brain. 2023;16:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Lee JW, Lee IH, Iimura T, Kong SW. Two macrophages, osteoclasts and microglia: from development to pleiotropy. Bone Res. 2021;9:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Gharpure M, Vyavahare S, Ahluwalia P, et al. Alterations in Alzheimer's disease microglia transcriptome might be involved in bone pathophysiology. Neurobiol Dis. 2024;191:106404. [DOI] [PubMed] [Google Scholar]
- 293. Lin Z, Shi G, Liao X, et al. Correlation between sedentary activity, physical activity and bone mineral density and fat in America: National Health and Nutrition Examination Survey, 2011‐2018. Sci Rep. 2023;13:10054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Raichlen DA, Aslan DH, Sayre MK, et al. Sedentary behavior and incident dementia among older adults. Jama. 2023;330:934‐940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295. Raffin J, Barreto PD, Le Traon AP, Vellas B, Aubertin‐Leheudre M, Rolland Y. Sedentary behavior and the biological hallmarks of aging. Ageing Res Rev. 2023;83:101807. [DOI] [PubMed] [Google Scholar]
- 296. Stanghelle B, Bentzen H, Giangregorio L, Pripp AH, Skelton DA, Bergland A. Effects of a resistance and balance exercise programme on physical fitness, health‐related quality of life and fear of falling in older women with osteoporosis and vertebral fracture: a randomized controlled trial (vol 45, pg 891, 2020). Osteoporosis Int. 2020;31:1187. [DOI] [PubMed] [Google Scholar]
- 297. Zhang SF, Huang XX, Zhao XY, et al. Effect of exercise on bone mineral density among patients with osteoporosis and osteopenia: a systematic review and network meta‐analysis. J Clin Nurs. 2022;31:2100‐2111. [DOI] [PubMed] [Google Scholar]
- 298. Alkhouli MF, Hung J, Squire M, et al. Exercise and resveratrol increase fracture resistance in the 3xTg‐AD mouse model of Alzheimer's disease. Bmc Complem Altern M. 2019;19:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. López‐Ortiz S, Valenzuela PL, Seisdedos MM, et al. Exercise interventions in Alzheimer's disease: a systematic review and meta‐analysis of randomized controlled trials. Ageing Res Rev. 2021;72:101479. [DOI] [PubMed] [Google Scholar]
- 300. Li L, Zhang W, Cheng SW, Cao DF, Parent M. Isoprenoids and related pharmacological interventions: potential application in Alzheimer's disease. Mol Neurobiol. 2012;46:64‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Fawcett JR, Bordayo EZ, Jackson K, et al. Inactivation of the human brain muscarinic acetylcholine receptor by oxidative damage catalyzed by a low molecular weight endogenous inhibitor from Alzheimer's brain is prevented by pyrophosphate analogs, bioflavonoids and other antioxidants. Brain Res. 2002;950:10‐20. [DOI] [PubMed] [Google Scholar]
- 302. Zameer S, Najmi AK, Vohora D, Akhtar M. Bisphosphonates: future perspective for neurological disorders. Pharmacol Rep. 2018;70:900‐907. [DOI] [PubMed] [Google Scholar]
- 303. Cibicková L, Palicka V, Cibicek N, et al. Differential effects of statins and alendronate on cholinesterases in serum and brain of rats. Physiol Res. 2007;56:765‐770. [DOI] [PubMed] [Google Scholar]
- 304. Lips P, van Schoor NM. The effect of vitamin D on bone and osteoporosis. Best Pract Res Clin Endocrinol Metab. 2011;25:585‐591. [DOI] [PubMed] [Google Scholar]
- 305. Gáll Z, Székely O. Role of vitamin D in cognitive dysfunction: new molecular concepts and discrepancies between animal and human findings. Nutrients. 2021;13:3672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306. Cui X, McGrath JJ, Burne THJ, Eyles DW. Vitamin D and schizophrenia: 20 years on. Mol Psychiatry. 2021;26:2708‐2720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Hendriks S, Ranson JM, Peetoom K, et al. Risk factors for young‐onset dementia in the UK biobank. Jama Neurol. 2024;81:188‐189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Chen LJ, Sha S, Stocker H, Brenner H, Schöttker B. The associations of serum vitamin D status and vitamin D supplements use with all‐cause dementia, Alzheimer's disease, and vascular dementia: a UK Biobank based prospective cohort study. Am J Clin Nutr. 2024;119:1052‐1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309. Di Somma C, Scarano E, Barrea L, et al. Vitamin D and neurological diseases: an endocrine view. Int J Mol Sci. 2017;18:2482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Lai RH, Hsu CC, Yu BH, et al. Vitamin D supplementation worsens Alzheimer's progression: animal model and human cohort studies. Aging Cell. 2022;21:e13670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Vieth R. Mistakes in terminology cause false conclusions: vitamin D does not increase the risk of dementia. Aging Cell. 2022;21:e13722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312. 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]
- 313. Wong D, Broberg DN, Doad J, et al. Effect of memantine treatment and combination with vitamin D supplementation on body composition in the APP/PS1 mouse model of Alzheimer's disease following chronic vitamin D deficiency. J Alzheimers Dis. 2021;81:375‐388. [DOI] [PubMed] [Google Scholar]
- 314. Morello M, Landel V, Lacassagne E, et al. Vitamin D improves neurogenesis and cognition in a mouse model of Alzheimer's disease. Mol Neurobiol. 2018;55:6463‐6479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315. Landel V, Millet P, Baranger K, Loriod B, Féron F. Vitamin D interacts with Esr1 and Igf1 to regulate molecular pathways relevant to Alzheimer's disease. Mol Neurodegener. 2016;11:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Breitner JCS, Haneuse SJPA, Walker R, et al. Risk of dementia and AD with prior exposure to NSAIDs in an elderly community‐based cohort. Neurology. 2009;72:1899‐1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317. Zhang CX, Wang Y, Wang DY, Zhang JD, Zhang FF. NSAID exposure and risk of Alzheimer's disease: an updated meta‐analysis from cohort studies. Front Aging Neurosci. 2018;10:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318. Pauls E, Bayod S, Mateo L, et al. Identification and drug‐induced reversion of molecular signatures of Alzheimer's disease onset and progression in App(NL‐G‐F), App(NL‐F), and 3xTg‐AD mouse models. Genome Med. 2021;13:168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319. Bennett DA, Whitmer RA. NSAID exposure and risk of Alzheimer disease: is timing everything? Neurology. 2009;72:1884‐1885. [DOI] [PubMed] [Google Scholar]
- 320. McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. New Engl J Med. 2014;370:412‐420. [DOI] [PubMed] [Google Scholar]
- 321. Lim SY, Bolster MB. Clinical utility of romosozumab in the management of osteoporosis: focus on patient selection and perspectives. Int J Womens Health. 2022;14:1733‐1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322. Markham A. Romosozumab: first global approval. Drugs. 2019;79:471‐476. [DOI] [PubMed] [Google Scholar]
- 323. Yoo Y, Neumayer G, Shibuya Y, Mader MMD, Wernig M. A cell therapy approach to restore microglial Trem2 function in a mouse model of Alzheimer's disease. Cell Stem Cell. 2023;30:1043‐1053. [DOI] [PubMed] [Google Scholar]
- 324. Zhou XC, Cao H, Guo JM, Yuan Y, Ni GX. Effects of BMSC‐derived EVs on bone metabolism. Pharmaceutics. 2022;14:1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325. Guo MT, Yin ZY, Chen FL, Lei P. Mesenchymal stem cell‐derived exosome: a promising alternative in the therapy of Alzheimer's disease. Alzheimers Res Ther. 2020;12:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326. Yin T, Liu Y, Ji WB, et al. Engineered mesenchymal stem cell‐derived extracellular vesicles: a state‐of‐the‐art multifunctional weapon against Alzheimer's disease. Theranostics. 2023;13:1264‐1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supporting Information
