Next Article in Journal
CLPP Depletion Causes Diplotene Arrest; Underlying Testis Mitochondrial Dysfunction Occurs with Accumulation of Perrault Proteins ERAL1, PEO1, and HARS2
Next Article in Special Issue
The Importance of Stem Cells Isolated from Human Dental Pulp and Exfoliated Deciduous Teeth as Therapeutic Approach in Nervous System Pathologies
Previous Article in Journal
The Multiple Roles of Periostin in Non-Neoplastic Disease
Previous Article in Special Issue
Therapeutic Potential of Mesenchymal Stem Cells in the Treatment of Epilepsy and Their Interaction with Antiseizure Medications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bone Tissue and the Nervous System: What Do They Have in Common?

by
Arianna Minoia
1,
Luca Dalle Carbonare
1,*,
Jens Christian Schwamborn
2,
Silvia Bolognin
2 and
Maria Teresa Valenti
3
1
Department of Medicine, University of Verona, 37100 Verona, Italy
2
Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 4365 Belvaux, Luxembourg
3
Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy
*
Author to whom correspondence should be addressed.
Cells 2023, 12(1), 51; https://doi.org/10.3390/cells12010051
Submission received: 29 September 2022 / Revised: 12 December 2022 / Accepted: 16 December 2022 / Published: 22 December 2022
(This article belongs to the Special Issue Mesenchymal Stem Cells and Their Role in Neurodegenerative Diseases)

Abstract

:
Degenerative diseases affecting bone tissues and the brain represent important problems with high socio-economic impact. Certain bone diseases, such as osteoporosis, are considered risk factors for the progression of neurological disorders. Often, patients with neurodegenerative diseases have bone fractures or reduced mobility linked to osteoarthritis. The bone is a dynamic tissue involved not only in movement but also in the maintenance of mineral metabolism. Bone is also associated with the generation of both hematopoietic stem cells (HSCs), and thus the generation of the immune system, and mesenchymal stem cells (MSCs). Bone marrow is a lymphoid organ and contains MSCs and HSCs, both of which are involved in brain health via the production of cytokines with endocrine functions. Hence, it seems clear that bone is involved in the regulation of the neuronal system and vice versa. This review summarizes the recent knowledge on the interactions between the nervous system and bone and highlights the importance of the interaction between nerve and bone cells. In addition, experimental models that study the interaction between nerve and skeletal cells are discussed, and innovative models are suggested to better evaluate the molecular interactions between these two cell types.

1. Introduction

Few studies have thoroughly analyzed the connections between the nervous system and bone. However, there is a relationship between these two systems, and some pathologies indicate the association between bone tissues and the nervous system. If we consider the two systems from an evolutionary perspective, it is apparent that the development of one influenced the development of the other. The modern human differs from the Neanderthal human not so much in the size of the brain as in the shape of the head [1]. The globularity of the modern human head is the result of a specific differentiation process after birth, at the stage where the brain determines the shape of the skull [2]. Therefore, it has been suggested that the globular shape of the skull of modern humans is not limited to a morphological change but is associated with neurofunctional processes [3].
Alterations in the balance of bone metabolism lead to a greater risk of fractures and an increase in osteoporosis [4]. The major hormonal variables that control bone metabolism are those that react to simultaneously perceived paracrine, autocrine and mechanical inputs [5]. Peripheral nerves regulate bone remodeling and recently it was demonstrated that sensory/motor nerve injury causes osteoporosis [6]. In addition, there is a relationship between osteoporosis and psychological stress, regulated by the hypothalamic–pituitary–adrenal (HPA) axis, glucocorticoid levels and a reduced response to factors that increase bone mass [7,8]. Chronic stress, through the production of inflammatory factors, promotes osteoclast differentiation as well as osteoblastic apoptosis [7], and it has been reported that anxiety levels can be considered a prognostic risk of fractures in postmenopausal women [8]. Importantly, although psychological stress and osteoporosis are distinct conditions, some factors such as glucocorticoids, catecholamines and insulin-like growth factors are involved in both alterations [9]. Osteoporotic fractures can be associated with spinal cord injuries. This lesion causes severe disability, compromising quality of life. After a spinal cord injury, the bone loss observed can be as high as 40% and the fractures can have devastating consequences [10]. Disuse following a spinal cord injury promotes increased levels of sclerostin, produced by osteocytes, reducing bone formation and inducing bone resorption through osteoclastic activation [11].
It has been reported that Alzheimer’s disease (AD) patients show lower bone mineral density and these patients often have fractures [12]. As osteoporosis has been shown to correlate with cognitive impairment, this condition is considered a risk factor for the development of AD [13]. In addition, Parkinson’s disease (PD) patients can be affected by Pisa syndrome due to trunk scoliosis, leading to further complications of disability [14]. Patients with AD and PD suffer from bone problems and osteoarthritis, and they have limited movement.
In addition, osteoporosis or skeletal abnormalities produce molecules capable of promoting neurodegenerative progression [15]. In a previous meta-analysis, the authors found that PD patients have a significantly increased risk of osteoporosis and osteopenia, and that female patients are more severely affected than male patients [16]. This gender difference in osteoporosis can be explained by the important role of endocrine and nutritional factors. From another point of view, it is known that bone mineral density is negatively affected by several neurotoxic metals, such as cadmium, lead, aluminum and arsenic, and these metals are implicated as well in neurodegenerative disorders [17].
Another important relationship between bone disease and neurodegenerative diseases is vitamin D levels. Hypovitaminosis D is associated with decreased bone mineral density and an increased risk of fractures [18], and accumulating evidence shows that vitamin D is essential for proper brain development, maturation and functions and neural network development [19]. Vitamin D deficiency seems to be related to various neurological conditions, such as MS, PD and AD, and vitamin D supplements resulted in no significant benefits for improving motor function for patients with PD [19,20].
In addition, vitamin D deficiency is associated with an increased risk of falls [21], worsening these events in patients with neurodegenerative diseases and further increasing the risk of fractures.
Cognitive alterations and motor disabilities are present in CDKL5 deficiency disorder (CDD). Patients with this disorder, a rare condition of X-linked impaired neurogenesis, have seizures and cognitive impairment and are characterized by microcephaly and scoliosis [22,23]. Interestingly, the Cdkl5 mutant zebrafish has been shown to be characterized by skeletal and neuronal disorders [24]. It has been suggested that CDKL5 plays an important role in bone metabolism and hypermethylation has been observed in osteoporotic patients with this gene [25].
On the basis of these reported findings and to understand the relationship between the two systems, it seems appropriate to evaluate the biology of the cells from which these systems derive and, in particular, the molecular crosstalk occurring between bone and nervous cells both at the physiological and pathological levels. The aim of this review is, therefore, to describe the “intersection” between the bone and the nervous system through the knowledge of related cellular and molecular systems. Finally, the intent of this review is also to provide suggestions and experimental models to identify therapeutic approaches/intervention tools to counteract some of the degenerative diseases that currently affect many subjects.

2. Search Strategy

Studies on the bone and nervous system were selected by consulting public databases. In particular, we identified 397 full articles from the following databases: PubMed, Web of Science and Scopus. The following keywords were used to search the titles and abstracts in all databases: bone, osteoblasts, stem cells, brain, nervous, diseases, molecule delivery in vitro experiments and in vivo experiments, with the Boolean operators (AND/OR/NOT).
Then, we removed duplicates and chose the article abstracts based on their agreement with our review topic. We removed several reviews and papers that were not recent. Thus, 226 papers were included in this review and are cited in the references.

3. Bone Marrow Mesenchymal Stem Cells (BM-MSCs)

The bone is a dynamic tissue that consists mainly of skeletal cells and bone marrow (BM) and has various roles, such as mineral metabolism and the generation of immune cells, hematopoietic stem cells and mesenchymal stem cells.
It has been reported that non-hematopoietic stem cells present in BM not only provide support to the microenvironment of hematopoietic stem cells, but also, thanks to their multipotency, are capable of differentiating into osteoblasts, chondroblasts and adipocytes [26].
MSCs represent a small population (from −0.001 to 0.01%) of the total nucleated cells. MSCs or MSC-like adult stem cells could be found and isolated from different mature tissues, such as adipose tissue, umbilical cord, amniotic fluid and peripheral blood [27]. The lineage commitment is promoted by specific molecular regulators of lineage. Chondrogenesis is regulated by interconnected molecular and cellular processes during embryogenesis. This is a multi-step process involving the recruitment, migration, proliferation and differentiation of mesenchymal cells. Furthermore, cellular interactions with the surrounding matrix and growth factors that modulate several transcription pathways regulate this process. The activation of cellular signaling, such as the Hedgehog signaling or Wnt signaling pathways, has been shown to play crucial roles in the development of cartilage tissue [28]. It has been reported that the Wnt/β-catenin pathway activates the expression of RUNX2, the master gene of osteogenic differentiation [29]. Yap and Taz, interacting with β-catenin, promote osteogenesis and inhibit chondrogenesis in neural crest cells [30]. The RUNX2 gene is not only the master gene of osteogenesis but is also associated with the regulation of neuronal processes. The RUNX2 gene is implicated in cleidocranial dysplasia [31] and acromegaly [32]; it controls the closure of cranial sutures [33] and is involved in the globularization of the skull [34,35]. In addition, RUNX2 participates in the development of the hippocampus [36,37] and the thalamus [38]. Mutations in RUNX2 are associated with mental disorders [39,40]. Therefore, RUNX2, whose expression plays an important role in the brain development (thalamus, hypothalamus and hippocampus) [38,41] is considered a candidate gene for serious mental illnesses, such as schizophrenia and bipolarity [37,39,40,42]. Moreover, RUNX2 transcriptionally activates osteocalcin and osteopontin [43], which, in addition to being important proteins of the bone matrix, are also involved in the organization of the brain [44].
Microglia, which are brain-specific macrophages, are another group of BM-derived cells that are in charge of the immunological defense of the brain via innate immunity mechanisms [45]. Some studies suggest that BM-derived hematopoietic cells infiltrate the brain and are subsequently able to differentiate into microglia, acquiring the ability to enter the CNS while keeping the blood–brain barrier (BBB) intact, thus colonizing the CNS especially in certain neurodegenerative diseases [46].
A fundamental role in this part is played by CCR2 (C-C chemokine receptor type 2) and CCR5 (C-C chemokine receptor type 5), chemokine receptors involved in the migration of microglial cells that have been shown to guide microglia derived from BM through the BBB, causing their accumulation in the brain parenchyma [45]. A reduced expression of CCR2 also leads to a decrease in BM-derived microglial cells within the brain and an increase in amyloid-β peptide levels [47].

4. Crosstalk between Bone and Neural Cells

Many neurotransmitters affect the metabolism of bone cells, and there is evidence that the central nervous system, which controls bone tissue directly via efferent neural connections, plays a significant role in maintaining the homeostasis of bone metabolism. [48]. In particular, the skeleton is innervated by a complex peripheral nervous system. The peripheral nervous system, by infiltrating the bones, regulates skeletal homeostasis by controlling bone metabolism and stem cell activity and secreting neurofactors [49].
Despite the observed relationship between the bone and neuronal system, few studies have investigated the presence of pathways common to bone and neuronal cells. One of the main and important pathways that links bone metabolism and the brain is the Wnt/β-catenin pathway. The Wnt pathway is regulated through the activation and inactivation of non-canonical and canonical signals during brain development [50].
The homeostasis of bone tissue is fundamentally regulated by the Wnt/β-catenin pathway. When Wnt is secreted, it can connect to the Frizzled and Lrp5/6 receptors, causing β-catenin to accumulate in the cytoplasm and move into the nucleus to control gene expression [51]. Additionally, it has been demonstrated that parkin, an E3 ubiquitin ligase involved in neurodegenerative diseases [52], can control the differentiation of BMSCs into osteogenic lineages via modifying β-catenin signaling and the autophagy process. In fact, the overexpression of parkin could induce β-catenin expression and the autophagy process through the expression of specific osteo-markers [53]. Leptin has been shown to be involved in modulating bone mass by acting on the hypothalamus and altering the sympathetic system. In particular, the activity of leptin induces the release of noradrenaline, which, in turn, activates the beta-adrenergic receptors expressed by osteoblasts [54,55]. It has been reported that neuromedin, through hypothalamic signals, regulates bone mass by inducing the expression of osteoblastic genes [56]. Recently, particular interest has been shown to the evaluation of the microenvironment inside the bone, in particular the neural component, and the regulatory role of the peripheral neural system on bone metabolism has been reported [57,58]. Sensory nerves are present in both cortical and trabecular bone motor nerves [58]. The intraosseous motor nerves are divided into adrenergic and cholinergic and communicate with bone cells with the aim of regulating bone metabolism through neurotransmitters. Peptidergic neurons were observed at the level of the periosteum and mineralization sites, in contact with osteoblasts, osteoclasts, stem cells and hematopoietic and endothelial cells [59]. Sympathetic nerve fibers, at the level of osteoblasts and osteoclasts, release neurotransmitters and neuropeptides capable of regulating bone cells and, thus, skeletal metabolism [60]. Norepinephrine (NE), the principal neurotransmitter of the sympathetic nervous system found in bone, inhibits bone formation. 𝛽-adrenergic receptors (β-Ars) are present in osteoblasts and osteoclasts with the function of regulating bone metabolism [61,62,63]. This regulation takes place through noradrenaline, which activates β-ARs present in osteoblasts and reduces bone formation. In particular, reduced bone formation is a consequence of the expression of RANKL and interleukins (IL)-6 and IL-11, which in turn promote osteoclast differentiation and maturation [63,64]. β-ARs can also directly regulate osteoclastogenesis by promoting the formation of reactive oxygen species [65]. In addition, the differentiation of mesenchymal cells can also be regulated by β-Ars via the cAMP/PKA pathways [58]. Sometimes, β1-ARs and β2-ARs regulate bone remodeling in the opposite way by exerting anabolic and catabolic activity, respectively [58].
The nicotinic acetylcholine receptors (nAChRs) or muscarinic acetylcholine receptors bind to the acetylcholine (ACh) that is released by cholinergic neurons (mAChRs). It has been reported that ACh regulates bone metabolism through these receptors, also expressed by bone cells [66,67,68]. Another factor associated with bone remodeling is the calcitonin-gene-related peptide (CGRP), which stimulates osteogenic differentiation by the upregulation of transcription factor-4 (ATF4) and osteocalcin and reduces osteoclastogenesis [69,70,71]. Furthermore, CGRP is involved in the bone’s ability to adapt to mechanical stresses, such as compression [72]. In addition to being crucial for the growth, survival and differentiation of nerve cells, neurotrophins, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), glial-cell-line-derived neurotrophic factor (GDNF), neurotrophin 3 (NT-3) and neurotrophin 4/5 (NT 4/5) are also involved in bone metabolism and are essential for the communication between peripheral nerves and the bone [58].
Bone cells, by releasing osteokines such as osteocalcin and lipocalin-2 that are able to pass through the BBB, communicate with the brain. Osteocalcin, accumulating in the brainstem, can affects neurotransmitter synthesis as well as cellular signaling, thus influencing age-related memory loss [73]. About 50% of circulating lipocalin-2 is produced by osteoblasts. In the hippocampus, an increase in LCN2 has been observed following inflammation, and the administration of LCN2 in the CNS has also been shown to reduce food intake and body weight [74]. The fibroblast growth factor-23, produced by osteoblasts and osteocytes, has been also found in the hypothalamus, hippocampus and cortex, and an alteration of cognitive functions in the FGF23 knockout mouse has been observed [75]. RANKL, detected in the hypothalamus, controls body temperature in females [76]. In addition, it has been shown that the treatment with anti-RANKL reduces stress and depression in mice with chronic social defects [77]. Osteopontin (OPN), osteoprotegerin (OPG), Sclerostin and Dickkopf-124 are other osteokines that can impact neuronal cells [78,79,80].
In neural cells, the expression of SATB2, a DNA-binding protein involved in the development of the brain, depends on both Bone Morphogenic Proteins (BMP) and SHH [81] and downregulates HOXA2, a target gene of the language regulator FOXP2 [82,83]. HOX2A plays an important role in both brain and bone generation [84,85], and it is an important gene for skeletal morphology [83,86,87]. Interestingly, the activation of HOXA2 in the neural crest reduces the expression of BMP inhibitors, causing craniofacial and also cerebral alterations [88]. On the other hand, FOXP2 is involved in both bone formation and neuronal stem cell commitment during corticogenesis [89,90]. HES1, functionally related to RUNX2, plays an important role during neurogenesis by regulating the Slit/Robo pathway [91] as well by affecting GABAergic and dopaminergic neuron formation [92]. In particular, Hes1 silencing induces GABAergic neuron differentiation in BM-MSCs [92] and is involved in osteoarthritis diseases [93].

5. Experimental Models for the Study of Skeletal and Neuronal Cells

5.1. In Vitro Models

In order to analyze and study the crosstalk between neuronal cells and bone metabolism, both in vitro and animal models have been used [58,94]. Regarding the study of bone metabolism, it was found that the use of in vitro models in 2D cell cultures are effective for functional and gene expression studies as well as for protein analysis [95] (Table 1). However, to fully capture the complexity of human tissues and the interplay between several cell lines, more sophisticated 3D models are necessary. Numerous papers have addressed the differences between cells cultured in 2D vs 3D models, showing that cell morphology, proliferation and differentiation are closer to the physiological situation in 3D [96,97]. Three-dimensional models are useful for detailed research of stem cell behavior, drug development, disease modeling and genetic screening. In fact, 3D models can greatly multiply tissue-specific stem cells and their differentiated cells from incredibly small amounts of starting material [98]. In the AD context, it has been shown that iPSC-derived neurons in 3D can be used for phenotypic assessment [99]. It has been demonstrated that the 3D framework provided by Matrigel, as well as other hydrogels including collagen and alginate gels, expedites the formation of neural networks. More crucially, the matrix’s support enables vertical development, which is entirely impossible in 2D cultures and leads to unfavorable apical–basal polarity [100].
Additionally, the 3D environment has the ability to elicit mechanical cues that can be translated into biochemical signals that are underrepresented in traditional 2D cultures [101]. It has also been shown that 3D set-ups can be compatible with multiplexing and automated screening procedures [102]. Furthermore, it has been shown that the use of neural progenitor cells with midbrain floor plate identity can be a source for the development of midbrain specific organoids. These midbrain organoids can be used to study neurodegenerative diseases such as PD. As an example, patient-specific organoids with a mutation in the PD-associated gene LRRK2 recapitulate key pathological processes, which are also seen in the actual patient’s brain [103].
Bone organoids are 3D self-renewing and self-organized micro-bone tissues with biomimetic spatial features [100]. They are composed of directionally differentiated stem cells, such as bone stem cells and embryonic stem cells, or progenitor cells, such as osteoblast and/or osteoclast. Bone organoids need biocompatible materials, such as Matrigel or alternative synthesized hydrogels, applied as a support to the self-organization of the bone organoids [104]. In contrast to other 3D cell culture models (such as spheroids), bone organoids develop from tissues specific to humans and self-organize into tissues resembling organs [105].
Conversely, spheroids are densely packed cell aggregates with irregular distributions. Due to inappropriate cell types and excessive human intervention, typical bone tissue creation techniques still fall short of accurately simulating the physiological microenvironment [106].
The osteoblast/osteocyte population produced from ESCs (embryonic stem cells) exhibits excellent potential to produce bone organoids. It has been demonstrated that 3D culture conditions gradually improve the stemness, proliferation and differentiation of MSCs and that anti-inflammatory and anti-apoptotic capability also occurred [107]. As typical ECM-produced materials, Matrigel and tissue-specific extracellular matrix have been shown to facilitate successful organoid growth [108].
Matrigel, which contributes to the ECM’s chemistry, offers plentiful collagen supports and physiological properties that are biocompatible for the development of organoids [108]. However, Matrigel is expensive; it has an unclear chemical makeup and the batch-to-batch variability leads to reproducibility issues. Biomaterials, both natural and artificial, have been used to increase the reproducibility of the experiments [109]. Thus, the organoids have been cultured using collagen, gelatin, alginate, fibrin and hyaluronic acid (HA). The two earliest synthetic polymers that are still often utilized in organoid research are polyethylene glycol (PEG) and polyisocyanopeptide (PIC) [110,111,112,113].

5.2. In Vivo Models

Zebrafish and mouse are generally used as in vivo models for studying degenerative diseases. The main advantage of using zebrafish as a model for brain–bone crosstalk is the fact that zebrafish and mammals share many skeletal development and biological phenomena as well as overall inventory of bone types [114]. As in the case of mammals, teleosts undergo skeletal histogenesis, which involves the differentiation of mesenchymal stem cells into chondroblasts and osteoblasts that produce the collagen extracellular matrix [115]. In both mammals and fish, skeletal cells differentiate through a complex interplay between intracellular molecular pathways and chemicals that regulate the timing, position and process [116]. Additionally, Parkin KO results in a moderate loss (20%) of dopaminergic neurons and decreased mitochondrial complex I activity in the zebrafish model [117]. In fact, zebrafish is also used as an experimental model for PD, and Parkin, PINK1 and LRKK2 are examples of related protein homologs found in zebrafish. Although fish have three synuclein genes, no homologs of human synuclein have been found [118].
Furthermore, mouse models are frequently used to study age-related diseases, such as bone disorder and neurodegenerative diseases [119]. The results of research conducted using murine models frequently challenge conventional wisdom regarding normal and pathological osseous remodeling. To discriminate among outcomes that are therapeutically meaningful, one must fully comprehend the distinctions between the physiology of human and murine bones. The evidence to date suggests that the characteristics and timing of osseous loss related to age in mice are quite similar to those of human beings [120]. Mouse models clearly demonstrate a rise in cortical porosity with age, which may provide information on the state of human beings [120]. For neurodegenerative pathologies, in particular for PD, mouse models have been extensively employed because the genetic alteration of these animals is relatively simple and common. The lifespan of mice, which is only two years, makes them a poor model for diseases such as PD, which can take five to six decades or longer to manifest in humans [121]. Therefore, with autosomal dominant disorders, the overall treatment is on self-expression in an effort to shorten the time that the disease takes to appear in mice. Results in transgenic models have been validated using human post-mortem tissue [122]. Even though post-mortem analyses of humans typically focus on diseases at an advanced stage, the early signs of disease persist and are the gold standard for evaluating how well animal models can mimic actual pathogens [121]. To understand the first anomalies in the DA nigrostriatal system that have been discovered as result of these mutations, it may be helpful to study the current knockdown parkin, PINK1 and DJ-1 [123]. Similarly, the current transgenic LRRK2 models may be very helpful for researching the developmental disorders in the DA nigrostriatal system. Some transgenic α-synuclein animal models also have increasing sensory abnormalities caused by dopaminergic dysfunction [124]. Therefore, finding a model organism capable of satisfying the needs necessary to investigate the crosstalk between brain and bone in the best possible way is a challenge. Future therapeutic strategies must consider the intensive and dynamic brain–bone interaction as well as the genetic and neuropsychological comorbidities of the affected patients. A strategy that takes this into account could be the possibility of studying bone–brain crosstalk using bone and brain organoids in on-chip approaches, which may call for extra monitoring or even tailored treatment plans. At the moment, however, these approaches are not particularly used, and these tools are used to study either the bone or the neuronal system or both simultaneously.
Table 1. Several in vitro and in vivo models used in skeletal and neuronal studies.
Table 1. Several in vitro and in vivo models used in skeletal and neuronal studies.
2D In Vitro Models3D In Vitro ModelsIn Vivo Models
CELL CULTURE
Cell culture is effective for functional and gene expression studies, as well as for protein analysis. Main cells used:
SH-SY5Y, PBMS, MSCs, MLO-Y4, MLC3T3-E1, MNC and RAW264 [95,125].

Three-dimensional cell culture is useful for detailed research of stem cell behavior, drug development, disease modeling and genetic screening.
  • Three-dimensional neural cell culture [65,99,103];
  • Three-dimensional bone cell culture [100,126,127];
  • Bone spheroids [105,128];
  • Neural spheroids [129,130];
  • On-chip organoid for brain or neural models [131,132,133,134];
  • On-chip organoid for bone models [135,136]

 
 
 
Zebrafish and mammals share many skeletal development and biological phenomena.
  • Zebrafish for bone studies [24,114,116,137];
  • Zebrafish for neural studies [24,104,117,118];
  • Zebrafish model for bone and brain crosstalk [24,138];

  • Murine model for bone and brain crosstalk [72,139,140].

6. Bone and Neuronal Cells in Degenerative Diseases

With increasing population aging, there is also an increase in neurodegenerative diseases and osteoporosis. Many data confirm the correlation between bone and neurodegenerative diseases [13,15,141].
Alzheimer’s disease and Parkinson’s disease are among the most common forms of neurodegenerative diseases.
Alzheimer’s disease is characterized by memory loss and cognitive decline. Even if there are common neuropathological features, the molecular mechanisms involved in the genetic susceptibility and progression of AD remain to be elucidated. In AD, bone mass reduction, characterized by the increase in inflammatory markers, can be considered a risk factor [13]. Recently, it has been reported that astrocyte alterations and an imbalance in calcium levels link osteoporosis and AD [142]. Moreover, AD causes beta amyloid accumulation in the bone, promoting RANKL-induced osteoclastic activation and thus favoring bone resorption [143]. In addition, by using AD mouse models, it has been demonstrated that lipocalin-2, produced by osteoblasts and regulated by miRNA-96-5p/Foxo1, promotes AD [144]. By performing single-cell RNA sequencing analyses, the age-associated transcriptomic profile in the cells of CNS such as microglia [145] has been recently reported and it has been demonstrated that beta amyloid, a pathological biomarker of AD, is able to modulate these cells [146]. In the PSEN1 p.G378E mutated-AD mouse model, the involvement of the receptor TYROBP in disease progression has been demonstrated [147]. Interestingly, the protein TYROBP is expressed in different cells, such as microglia and osteoclasts, two cells sharing a myeloid origin. Signaling pathways, such as TREM2/TYROBP, CSF1 and CCR5, are involved in AD and osteoporosis [148,149]. Moreover, skeletal fragility due to osteoclast activation is associated with increased Aβ42 levels in the brain and has been reported in an AD mouse model [140]. During aging, increased inflammatory processes cause both bone loss (by activating osteoclast activity) and AD by increasing the levels of the protease inhibitor neuronal α2-macroglobulin involved in plaque formation [150].
PD is characterized by the degeneration of the neurons of the midbrain nigrostriatal dopaminergic (DA) system that regulate not only neuronal functions, but also bone metabolism, and the Wnt/β-catenin pathway plays a crucial role in the development of many aspects of DA neuron development [151]. PD represents the second most common age-associated neurodegenerative disorder affecting the nervous system after AD and it is clinically characterized as a movement disorder. Often, patients affected by this pathology are more prone than age-matched controls to fractures and joint and bone problems due to reduced mobility, postural instability and neurological impairment [152].
There are recent results on the interaction of Parkin and β-catenin; elevated levels of sum-active (dephosphorylated) β-catenin were found in mice lacking the protein parkin [153]. Its increase in Wnt/β-catenin signaling suggests that reduced β-catenin degradation may result in DA neurons being lost as they attempt to re-enter the cell cycle. This latter finding contrasts with the active role of Wnt/β-catenin signaling during the development of midbrain DA neurons and stem cells, suggesting that pathological adult DA neurons may require less Wnt/β-catenin signaling, while DA precursors may benefit from enhanced Wnt activation/β-catenin signaling [154,155].
Moreover, parkin overexpression accelerates bone healing in tibial fracture model markers [53]. Additionally, parkin, which was found to be mutated in PD patients, caused impaired mitophagy with an accumulation of damaged mitochondria, causing the loss of DA neurons with age [151]. Furthermore, NR2F1, known as nuclear receptor 2 families 1 and being part of the Human Hormone Nuclear Receptor (hHNR) family, is upregulated during osteogenesis and plays a pivotal role during neurogenesis [156]. In particular, the NR2F1 transcript was recently reported to be considerably downregulated in dopaminergic neurons and midbrain organoids generated from PD patients carrying the LRRK2-G2019S mutation compared to the healthy controls [157] (Figure 1).
In addition to AD or PD, other neurodegenerative diseases are associated with bones. Multiple sclerosis and osteoporosis affect postmenopausal women and it seems that these two pathologies share pathogenetic modalities. Bisson et al. compared BMD in individuals with or without multiple sclerosis [158]. The authors observed a lower BMD and an increased prevalence of osteoporosis in individuals affected by multiple sclerosis compared to age- and sex-matched controls, suggesting that, in this population, the bone component needs to be evaluated in order to adopt a correct therapy.
Mutations in AUTS2, a gene involved in neurodevelopment as well as in several neurological disorders [159], cause cognitive impairments [160] that are often associated with skeletal anomalies [161]. Many AUTS2-associated proteins, which play an important role at the neuronal level and are associated with pathologies such as ASD and cognitive alterations, interact directly with the osteogenic master gene RUNX2 [35]. However, RUNX2 interacts with several candidate genes for autism, such as SMURF1 [162], involved in the control of axonogenesis [163,164]. SATB2, associated with ASD, cognitive disability and craniofacial alterations [165], is an important gene for osteoblastic differentiation and directly interacts with RUNX2 [166,167,168].
RUNX2, through FOXO1, interacts with DYRK1A [169], a gene located on the Down syndrome region of chromosome 21, is associated with facial and cognitive alterations [170,171] and acts as an inhibitor in the process of osteoclastogenesis [172]. In addition, DYRK1A phosphorylates SIRT1, a protein that controls neuronal and osteogenic differentiation [35]. In fact, SIRT1 upregulates and deacetylates RUNX2 and acts on β-catenin and FoxO expression in osteoblast progenitors, thereby promoting osteoblastic differentiation [35,173,174]. SIRT1 is also capable of inducing the neuronal differentiation of bone marrow mesenchymal stem cells under the activation of resveratrol [175]. AKT1, functionally related to RUNX2 [35], is involved in the regulation of the bone remodeling, a process performed by osteoblasts, cells with bone-forming activity, and osteoclasts, cells with bone resorption activity. Mutations in Akt1 and Akt2 in mice compromise bone formation [176,177]. However, AKT1 is also involved in growth-factor-induced neuronal survival [178].
Importantly, physical and cognitive alterations following traumatic brain injury (TBI) cause death or disability. TBI induces inflammation and the upregulation of leptin levels for the alteration of the blood–brain barrier or following the dysfunction of the hypothalamus–pituitary–adrenal axis. The increase in leptin levels causes the release of neurofactors, which in turn affects bone formation [179]. Additionally, heterotropic ossification is associated with TBI [180]. In particular, the reduced activity of PHD2 (prolyl hydroxylase domain proteins) due to a hypoxic environment as a consequence of TBI promotes angiogenesis; this, in turn, promotes chondrocyte hypertrophy in soft tissues, thus leading to heterotopic ossification.

7. Bone-Marrow-Derived Cells Influence Neurogenesis

The decline of stem cells due to aging is associated with tissue dysfunctions and reduced molecular repair.
During aging, a reduced ability of somatic stem cells to differentiate into cells fundamental for brain repair is observed. By using a mouse model, a reduced number of activated neuronal stem cells associated with aging has been observed [181]. An infiltration of immune cells into the stem cell niche is also observed during aging. The presence of immune cells, such as microglia and senescent cells, causes an inflammatory environment that surrounds the stem cell niche [182]. Thus, the modifications of the stem cell niche during aging change the spatial localization of these cells, which causes alterations between stem cells and supporting cells. In particular, the ECM produced by microglia, following inflammatory cytokine activation, causes the imbalance between oligodendrocytes and astrocytes during aging [183]. Alterations in function as well of cognition are associated with the loss of neurons and synapses in AD patients and a reduction in dopaminergic neurons in the substantia nigra in PD patients has been observed [184]. Therefore, MSCs have been considered in the attempts to counteract different neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases, amyotrophic lateral sclerosis (ALS), spinal cord injury or traumatic brain injury.
To this purpose, different studies performed in mice reported the beneficial administration effects of stem cells [185,186,187].
It has been reported that bone marrow stem cells (BM-SCs) are able to infiltrate the brain. The migration of BM-SCs into the CNS has been observed in rodents and humans. In particular, BM hematopoietic stem cells injected in rodents are able to colonize the CNS and differentiate into non-neuronal cells as well as probably even neuronal cells [185,186,187]. In AD mouse models, MSC injection was observed to counteract cognitive decline by promoting the regeneration process by releasing extracellular vesicles and modulating neuroinflammation [188,189].
Human BM mesenchymal stem cells (BMMSCs) injected intravenously in an Alzheimer’s disease mice model were able to pass the BBB and accumulate in the hippocampus [190]. Interestingly, the authors observed that the hBM-MSC injection reduced cerebral amyloid βeta levels. In addition, several enzymes and cerebral cytokines were also modulated, probably as consequence of the hMSC presence [190]. Thus, the immunomodulatory effects of BM-MSCs could play a crucial part in the management of AD by modulating the activity of microglia and astrocytes as well as by targeting transcription factor expression and levels of cytokines involved in neuroinflammation [191].
The injection of human (h)BM-MSCs has been reported to promote the maturation of murine stem cells in the neuronal lineage via neuronal growth factor secretion [192,193]. The transplantation of hBM-MSCs into a mouse model of autism promoted an improvement in cognitive abilities, suggesting the importance of neurogenesis induced by hBM-MSC transplantation [194]. The ability of transplanted hBM-MSCs to secrete BDNF has also been evidenced in rat and mouse models of PD with cerebral ischemia [195,196,197]. hBM-MSC transplantation is also able to stimulate EGFR (epidermal growth factor receptor) expression by enhancing neurogenesis in a PD model [198]. It has been demonstrated that BM-MSC transplantation also induces the upregulation of the neuronal precursor nestin [199]. In a mouse AD model, the injection of bone-marrow-derived microglia-like cells counteracts amyloid deposition, ameliorating cognitive capacity [139].
Vesicles originated from BM/MSCs can deliver lipids and proteins as well as post-transcriptional regulators, such as non-coding RNAs, thus affecting important cellular processes [200,201]. Thus, microvesicles or exosomes produced by BM-MSCs reduce cognitive alterations due to traumatic brain injury or PD [202,203,204,205]. It has been reported that exosomes originating from MSCs can deliver molecules, such as miRNAs promoting synaptic remodeling or inhibiting neuronal apoptosis and promoting neuronal recovery in pathological models [201,202]. Exosomes produced by MSCs can deliver miR 133b in neuronal cells of a stroke rat model, thus promoting cellular regeneration in the ischemic tissue [205]. Increased levels of miR 146a are found in peripheral blood and cerebral fluids of the spine of patients with AD and amyotrophic lateral sclerosis, and it has been reported that this miR promotes remyelination by preventing NF-kB pathway activation [205,206]. Therefore, Kubota et al. reported that mir146a delivered by exosomes produced in MSCs is involved in the prevention of cognitive decline in a diabetic rat model [207]. Many studies, therefore, report the positive effect of MSCs on the protection of neuronal cells. However, the quality of stem cells is essential for carrying out effective neuroprotective functions. Senescent MSCs are associated with the development of skeletal diseases, such as osteoporosis, and neurodegenerative diseases, such as AD and PD [203].
MSCs as well as molecules produced by MSCs have been utilized to combat neurodegenerative illnesses, including AD, PD, ALS or ischemic stroke [208,209]. Huntington’s disease has been also treated with SCs [209]. In particular, the use of extracellular vesicles (EVs) from MSCs appears very promising.
Thus, the beneficial effects of MSC transplants, such as improving cognitive functions and survival, have been observed in many experimental models. The possibility of manipulating MSCs to induce their neuronal differentiation has suggested the use of MSCs to replace damaged neuronal tissues. Many researchers have performed specific protocols to differentiate hMSCs into neuronal cells that are able to produce and secrete neural factors such as dopamine or acetylcholine [210,211,212]. However, the exact molecular mechanisms underlying the role of MSCs in modulating neuronal cells are not yet fully understood. Some challenges involving the use of stem cells have not yet been resolved. In particular, stem cells can be influenced by the microenvironment, having negative consequences for correct engraftment and differentiation [5].
More studies are therefore needed, especially for traumatic brain injuries, so that stem cells can be used successfully as a therapeutic tool.

8. Conclusions and New Perspectives

The bone and brain are closely associated and alterations in one system are reflected in the other. In recent years, following the increase in the aging of the population, it has been observed that bone and neurodegenerative pathologies are associated. The central and peripheral neural systems are key players in the remodeling of bone. Several sensory neurotransmitters can modulate aging and osteogenic differentiation with important repercussions on skeletal pathologies. Both microglia and osteoclast cells share the same hematopoietic precursor. Pathologies, such as osteoporosis, arthrosis and bone alterations due to oncological problems, involve the crosstalk between bone and nerves. Based on the observations that highlight the importance of the crosstalk between the brain and bone, the possibility of the early treatment of bone disorders and the consequent inflammatory state appears useful to delay or reduce the severity of neurodegenerative pathologies.
Therefore, the observation of bidirectional exchanges between bone and nerves can be explored to identify new therapeutic tools. Although many studies have been performed to understand the pathogenesis of neurodegenerative diseases, many molecular mechanisms involved in the susceptibility and progression of these diseases remain to be elucidated. For this purpose, in vitro models, such as 3D models, could be useful. However, at present, experimental models are not yet perfectly complete for the study of the bone–brain crosstalk, as those currently available are aimed at studying one system or the other. In addition, at present brain on a chip is considered a dream [213].
Several studies report the beneficial effects of stem cells in neurodegenerative diseases, especially through the release of molecules in micro-vesicles or exosomes from MSCs. These cells, which are able to pass through the blood–brain barrier, could restore neuronal cell alterations due to their ability to differentiate into neuron-like cells or through fusion processes with endogenous cells. Even if stem cell therapies applied in animal models have shown successful results, many clinical applications or trials in patients with neurodegenerative diseases have not produced exciting results.
Interestingly, nanoparticles (NPs) have been proposed for the delivery of drugs or biological macromolecules for CNS application due to their potential to cross the blood–brain barrier. Recently, preliminary pharmacodynamic analyses have been performed for the treatment of AD by applying nanoparticles containing memantine (an N-methyl-D-aspartate antagonist), a molecule used to treat severe cases of AD [214].
Thus, nanoformulations, targeting tissue-specific markers, could be considered promising therapeutics tools for different diseases, including neurodegenerative disorders.

Author Contributions

Conceptualization: L.D.C. and M.T.V.; investigation: A.M. and M.T.V.; supervision: S.B. and J.C.S.; writing—original draft: A.M., L.D.C., M.T.V. and S.B.; writing—review and editing: L.D.C., M.T.V., S.B. and J.C.S. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by FUR-Dep of Medicine, University of Verona (LDC).

Conflicts of Interest

The authors declare that there are no conflict of interest.

References

  1. Bruner, E. Geometric morphometrics and paleoneurology: Brain shape evolution in the genus Homo. J. Hum. Evol. 2004, 47, 279–303. [Google Scholar] [CrossRef] [PubMed]
  2. Gunz, P.; Neubauer, S.; Maureille, B.; Hublin, J.-J. Brain development after birth differs between Neanderthals and modern humans. Curr. Biol. 2010, 20, R921–R922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Boeckx, C.A.; Benítez-Burraco, A. The shape of the human language-ready brain. Front. Psychol. 2014, 5, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Valenti, M.T.; Dalle Carbonare, L.; Mottes, M. Osteogenic differentiation in healthy and pathological conditions. Int. J. Mol. Sci. 2016, 18, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Adugna, D.G.; Aragie, H.; Kibret, A.A.; Belay, D.G. Therapeutic Application of Stem Cells in the Repair of Traumatic Brain Injury. Stem Cells Cloning Adv. Appl. 2022, 15, 53. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, Y.; Zhou, J.; Liang, C.; Xiao, Q.; Chen, Y.; Yu, B. Effects of highly selective sensory/motor nerve injury on bone metabolism and bone remodeling in rats. J. Musculoskelet. Neuronal Interact. 2022, 22, 524–535. [Google Scholar]
  7. Eastell, R.; O’Neill, T.W.; Hofbauer, L.C.; Langdahl, B.; Reid, I.R.; Gold, D.T.; Cummings, S.R. Postmenopausal osteoporosis. Nat. Rev. Dis. Prim. 2016, 2, 16069. [Google Scholar] [CrossRef]
  8. Catalano, A.; Martino, G.; Bellone, F.; Gaudio, A.; Lasco, C.; Langher, V.; Lasco, A.; Morabito, N. Anxiety levels predict fracture risk in postmenopausal women assessed for osteoporosis. Menopause 2018, 25, 1110–1115. [Google Scholar] [CrossRef]
  9. Kelly, R.R.; McDonald, L.T.; Jensen, N.R.; Sidles, S.J.; LaRue, A.C. Impacts of psychological stress on osteoporosis: Clinical implications and treatment interactions. Front. Psychiatry 2019, 10, 200. [Google Scholar] [CrossRef]
  10. Haider, I.T.; Lobos, S.M.; Simonian, N.; Schnitzer, T.J.; Edwards, W.B. Bone fragility after spinal cord injury: Reductions in stiffness and bone mineral at the distal femur and proximal tibia as a function of time. Osteoporos. Int. 2018, 29, 2703–2715. [Google Scholar] [CrossRef]
  11. Battaglino, R.A.; Lazzari, A.A.; Garshick, E.; Morse, L.R. Spinal cord injury-induced osteoporosis: Pathogenesis and emerging therapies. Curr. Osteoporos. Rep. 2012, 10, 278–285. [Google Scholar] [CrossRef]
  12. Malochet-Guinamand, S.; Durif, F.; Thomas, T. Parkinson’s disease: A risk factor for osteoporosis. Jt. Bone Spine 2015, 82, 406–410. [Google Scholar] [CrossRef]
  13. Kang, H.G.; Park, H.Y.; Ryu, H.U.; Suk, S.-H. Bone mineral loss and cognitive impairment: The PRESENT project. Medicine 2018, 97, e12755. [Google Scholar] [CrossRef]
  14. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef]
  15. Yuan, J.; Meloni, B.P.; Shi, T.; Bonser, A.; Papadimitriou, J.M.; Mastaglia, F.L.; Zhang, C.; Zheng, M.; Gao, J. The potential influence of bone-derived modulators on the progression of Alzheimer’s disease. J. Alzheimer’s Dis. 2019, 69, 59–70. [Google Scholar] [CrossRef]
  16. Torsney, K.M.; Noyce, A.J.; Doherty, K.M.; Bestwick, J.P.; Dobson, R.; Lees, A.J. Bone health in Parkinson’s disease: A systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2014, 85, 1159–1166. [Google Scholar] [CrossRef] [Green Version]
  17. Huat, T.J.; Camats-Perna, J.; Newcombe, E.A.; Valmas, N.; Kitazawa, M.; Medeiros, R. Metal toxicity links to Alzheimer’s disease and neuroinflammation. J. Mol. Biol. 2019, 431, 1843–1868. [Google Scholar] [CrossRef]
  18. Dalle Carbonare, L.; Valenti, M.T.; Del Forno, F.; Caneva, E.; Pietrobelli, A. Vitamin D: Daily vs. monthly use in children and elderly—What is going on? Nutrients 2017, 9, 652. [Google Scholar] [CrossRef] [Green Version]
  19. Moretti, R.; Morelli, M.E.; Caruso, P. Vitamin D in neurological diseases: A rationale for a pathogenic impact. Int. J. Mol. Sci. 2018, 19, 2245. [Google Scholar] [CrossRef] [Green Version]
  20. Zhou, Z.; Zhou, R.; Zhang, Z.; Li, K. The association between vitamin D status, vitamin D supplementation, sunlight exposure, and Parkinson’s disease: A systematic review and meta-analysis. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 666. [Google Scholar] [CrossRef]
  21. Wu, H.; Pang, Q. The effect of vitamin D and calcium supplementation on falls in older adults. Der Orthopäde 2017, 46, 729–736. [Google Scholar] [CrossRef] [PubMed]
  22. Bahi-Buisson, N.; Nectoux, J.; Rosas-Vargas, H.; Milh, M.; Boddaert, N.; Girard, B.; Cances, C.; Ville, D.; Afenjar, A.; Rio, M. Key clinical features to identify girls with CDKL5 mutations. Brain 2008, 131, 2647–2661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Fehr, S.; Wilson, M.; Downs, J.; Williams, S.; Murgia, A.; Sartori, S.; Vecchi, M.; Ho, G.; Polli, R.; Psoni, S. The CDKL5 disorder is an independent clinical entity associated with early-onset encephalopathy. Eur. J. Hum. Genet. 2013, 21, 266–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Varela, T.; Varela, D.; Martins, G.; Conceição, N.; Cancela, M.L. Cdkl5 mutant zebrafish shows skeletal and neuronal alterations mimicking human CDKL5 deficiency disorder. Sci. Rep. 2022, 12, 9325. [Google Scholar] [CrossRef] [PubMed]
  25. Cheishvili, D.; Parashar, S.; Mahmood, N.; Arakelian, A.; Kremer, R.; Goltzman, D.; Szyf, M.; Rabbani, S.A. Identification of an epigenetic signature of osteoporosis in blood DNA of postmenopausal women. J. Bone Miner. Res. 2018, 33, 1980–1989. [Google Scholar] [CrossRef] [Green Version]
  26. Pontikoglou, C.; Deschaseaux, F.; Sensebé, L.; Papadaki, H.A. Bone marrow mesenchymal stem cells: Biological properties and their role in hematopoiesis and hematopoietic stem cell transplantation. Stem Cell Rev. Rep. 2011, 7, 569–589. [Google Scholar] [CrossRef]
  27. Hwang, N.S.; Zhang, C.; Hwang, Y.S.; Varghese, S. Mesenchymal stem cell differentiation and roles in regenerative medicine. Wiley Interdiscip. Rev. Syst. Biol. Med. 2009, 1, 97–106. [Google Scholar] [CrossRef]
  28. Deng, Q.; Li, P.; Che, M.; Liu, J.; Biswas, S.; Ma, G.; He, L.; Wei, Z.; Zhang, Z.; Yang, Y. Activation of hedgehog signaling in mesenchymal stem cells induces cartilage and bone tumor formation via Wnt/β-Catenin. Elife 2019, 8, e50208. [Google Scholar] [CrossRef]
  29. Vega, O.A.; Lucero, C.M.; Araya, H.F.; Jerez, S.; Tapia, J.C.; Antonelli, M.; Salazar-Onfray, F.; Las Heras, F.; Thaler, R.; Riester, S.M. Wnt/β-catenin signaling activates expression of the bone-related transcription factor RUNX2 in select human osteosarcoma cell types. J. Cell. Biochem. 2017, 118, 3662–3674. [Google Scholar] [CrossRef]
  30. Zhao, X.; Tang, L.; Le, T.P.; Nguyen, B.H.; Chen, W.; Zheng, M.; Yamaguchi, H.; Dawson, B.; You, S.; Martinez-Traverso, I.M. Yap and Taz promote osteogenesis and prevent chondrogenesis in neural crest cells in vitro and in vivo. Sci. Signal. 2022, 15, eabn9009. [Google Scholar] [CrossRef]
  31. Dalle Carbonare, L.; Antoniazzi, F.; Gandini, A.; Orsi, S.; Bertacco, J.; Li Vigni, V.; Minoia, A.; Griggio, F.; Perduca, M.; Mottes, M. Two Novel C-Terminus RUNX2 Mutations in Two Cleidocranial Dysplasia (CCD) Patients Impairing p53 Expression. Int. J. Mol. Sci. 2021, 22, 10336. [Google Scholar] [CrossRef]
  32. Valenti, M.T.; Mottes, M.; Cheri, S.; Deiana, M.; Micheletti, V.; Cosaro, E.; Davì, M.V.; Francia, G.; Dalle Carbonare, L. Runx2 overexpression compromises bone quality in acromegalic patients. Endocr.-Relat. Cancer 2018, 25, 269–277. [Google Scholar] [CrossRef]
  33. Stein, G.S.; Lian, J.B.; Van Wijnen, A.J.; Stein, J.L.; Montecino, M.; Javed, A.; Zaidi, S.K.; Young, D.W.; Choi, J.-Y.; Pockwinse, S.M. Runx2 control of organization, assembly and activity of the regulatory machinery for skeletal gene expression. Oncogene 2004, 23, 4315–4329. [Google Scholar] [CrossRef] [Green Version]
  34. Depew, M.J.; Liu, J.K.; Long, J.E.; Presley, R.; Meneses, J.J.; Pedersen, R.A.; Rubenstein, J. Dlx5 regulates regional development of the branchial arches and sensory capsules. Development 1999, 126, 3831–3846. [Google Scholar] [CrossRef]
  35. Boeckx, C.; Benítez-Burraco, A. Osteogenesis and neurogenesis: A robust link also for language evolution. Front. Cell. Neurosci. 2015, 9, 291. [Google Scholar] [CrossRef] [Green Version]
  36. Pleasure, S.J.; Anderson, S.; Hevner, R.; Bagri, A.; Marin, O.; Lowenstein, D.H.; Rubenstein, J.L. Cell migration from the ganglionic eminences is required for the development of hippocampal GABAergic interneurons. Neuron 2000, 28, 727–740. [Google Scholar] [CrossRef]
  37. Benes, F.M.; Lim, B.; Matzilevich, D.; Walsh, J.P.; Subburaju, S.; Minns, M. Regulation of the GABA cell phenotype in hippocampus of schizophrenics and bipolars. Proc. Natl. Acad. Sci. USA 2007, 104, 10164–10169. [Google Scholar] [CrossRef] [Green Version]
  38. Reale, M.E.; Webb, I.C.; Wang, X.; Baltazar, R.M.; Coolen, L.M.; Lehman, M.N. The transcription factor Runx2 is under circadian control in the suprachiasmatic nucleus and functions in the control of rhythmic behavior. PLoS ONE 2013, 8, e54317. [Google Scholar] [CrossRef] [Green Version]
  39. Talkowski, M.E.; Rosenfeld, J.A.; Blumenthal, I.; Pillalamarri, V.; Chiang, C.; Heilbut, A.; Ernst, C.; Hanscom, C.; Rossin, E.; Lindgren, A.M. Sequencing chromosomal abnormalities reveals neurodevelopmental loci that confer risk across diagnostic boundaries. Cell 2012, 149, 525–537. [Google Scholar] [CrossRef] [Green Version]
  40. Ruzicka, W.B.; Subburaju, S.; Benes, F.M. Circuit-and diagnosis-specific DNA methylation changes at γ-aminobutyric acid–related genes in postmortem human hippocampus in schizophrenia and bipolar disorder. JAMA Psychiatry 2015, 72, 541–551. [Google Scholar] [CrossRef]
  41. Jeong, J.H.; Jin, J.S.; Kim, H.N.; Kang, S.M.; Liu, J.C.; Lengner, C.J.; Otto, F.; Mundlos, S.; Stein, J.L.; Van Wijnen, A.J. Expression of Runx2 transcription factor in non-skeletal tissues, sperm and brain. J. Cell. Physiol. 2008, 217, 511–517. [Google Scholar] [CrossRef] [PubMed]
  42. Subburaju, S.; Benes, F.M. Induction of the GABA cell phenotype: An in vitro model for studying neurodevelopmental disorders. PLoS ONE 2012, 7, e33352. [Google Scholar] [CrossRef] [PubMed]
  43. Valenti, M.T.; Serafini, P.; Innamorati, G.; Gili, A.; Cheri, S.; Bassi, C.; Dalle Carbonare, L. Runx2 expression: A mesenchymal stem marker for cancer. Oncol. Lett. 2016, 12, 4167–4172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Schroeter, M.; Zickler, P.; Denhardt, D.T.; Hartung, H.-P.; Jander, S. Increased thalamic neurodegeneration following ischaemic cortical stroke in osteopontin-deficient mice. Brain 2006, 129, 1426–1437. [Google Scholar] [CrossRef] [PubMed]
  45. Soulet, D.; Rivest, S. Bone-marrow-derived microglia: Myth or reality? Curr. Opin. Pharmacol. 2008, 8, 508–518. [Google Scholar] [CrossRef]
  46. Cartier, N.; Lewis, C.-A.; Zhang, R.; Rossi, F. The role of microglia in human disease: Therapeutic tool or target? Acta Neuropathol. 2014, 128, 363–380. [Google Scholar] [CrossRef] [Green Version]
  47. El Khoury, J.; Luster, A.D. Mechanisms of microglia accumulation in Alzheimer’s disease: Therapeutic implications. Trends Pharmacol. Sci. 2008, 29, 626–632. [Google Scholar] [CrossRef]
  48. Huang, S.; Li, Z.; Liu, Y.; Gao, D.; Zhang, X.; Hao, J.; Yang, F. Neural regulation of bone remodeling: Identifying novel neural molecules and pathways between brain and bone. J. Cell. Physiol. 2019, 234, 5466–5477. [Google Scholar] [CrossRef]
  49. Rajpar, I.; Tomlinson, R.E. Function of peripheral nerves in the development and healing of tendon and bone. Proc. Semin. Cell Dev. Biol. 2022, 123, 48–56. [Google Scholar] [CrossRef]
  50. Freese, J.L.; Pino, D.; Pleasure, S.J. Wnt signaling in development and disease. Neurobiol. Dis. 2010, 38, 148–153. [Google Scholar] [CrossRef] [Green Version]
  51. Kele-Olovsson, J.M. Regulation Of Midbrain Dopaminergic Neuron Development by Wnts, Sfrps And bHLH Proteins; Karolinska Institutet (Sweden): Solna, Sweden, 2007. [Google Scholar]
  52. Kane, L.A.; Lazarou, M.; Fogel, A.I.; Li, Y.; Yamano, K.; Sarraf, S.A.; Banerjee, S.; Youle, R.J. PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase activity. J. Cell Biol. 2014, 205, 143–153. [Google Scholar] [CrossRef]
  53. Zhang, W.; Hou, W.; Chen, M.; Chen, E.; Xue, D.; Ye, C.; Li, W.; Pan, Z. Upregulation of parkin accelerates osteoblastic differentiation of bone marrow-derived mesenchymal stem cells and bone regeneration by enhancing autophagy and β-Catenin signaling. Front. Cell Dev. Biol. 2020, 907, 576104. [Google Scholar] [CrossRef]
  54. Takeda, S.; Elefteriou, F.; Levasseur, R.; Liu, X.; Zhao, L.; Parker, K.L.; Armstrong, D.; Ducy, P.; Karsenty, G. Leptin regulates bone formation via the sympathetic nervous system. Cell 2002, 111, 305–317. [Google Scholar] [CrossRef] [Green Version]
  55. Elefteriou, F.; Ahn, J.D.; Takeda, S.; Starbuck, M.; Yang, X.; Liu, X.; Kondo, H.; Richards, W.G.; Bannon, T.W.; Noda, M. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature 2005, 434, 514–520. [Google Scholar] [CrossRef]
  56. Sato, S.; Hanada, R.; Kimura, A.; Abe, T.; Matsumoto, T.; Iwasaki, M.; Inose, H.; Ida, T.; Mieda, M.; Takeuchi, Y. Central control of bone remodeling by neuromedin U. Nat. Med. 2007, 13, 1234–1240. [Google Scholar] [CrossRef]
  57. Hohmann, E.L.; Elde, R.P.; Rysavy, J.A.; Einzig, S.; Gebhard, R.L. Innervation of periosteum and bone by sympathetic vasoactive intestinal peptide-containing nerve fibers. Science 1986, 232, 868–871. [Google Scholar] [CrossRef]
  58. Wan, Q.Q.; Qin, W.P.; Ma, Y.X.; Shen, M.J.; Li, J.; Zhang, Z.B.; Chen, J.H.; Tay, F.R.; Niu, L.N.; Jiao, K. Crosstalk between bone and nerves within bone. Adv. Sci. 2021, 8, 2003390. [Google Scholar] [CrossRef]
  59. Zhu, Y.; Ma, Y.; Elefteriou, F. Cortical bone is an extraneuronal site of norepinephrine uptake in adult mice. Bone Rep. 2018, 9, 188–198. [Google Scholar] [CrossRef]
  60. Robles, H.; Park, S.; Joens, M.S.; Fitzpatrick, J.A.; Craft, C.S.; Scheller, E.L. Characterization of the bone marrow adipocyte niche with three-dimensional electron microscopy. Bone 2019, 118, 89–98. [Google Scholar] [CrossRef]
  61. Mulcrone, P.L.; Campbell, J.P.; Clément-Demange, L.; Anbinder, A.L.; Merkel, A.R.; Brekken, R.A.; Sterling, J.A.; Elefteriou, F. Skeletal colonization by breast cancer cells is stimulated by an osteoblast and β2AR-dependent neo-angiogenic switch. J. Bone Miner. Res. 2017, 32, 1442–1454. [Google Scholar] [CrossRef]
  62. Hirai, T.; Tanaka, K.; Togari, A. β-adrenergic receptor signaling regulates Ptgs2 by driving circadian gene expression in osteoblasts. J. Cell Sci. 2014, 127, 3711–3719. [Google Scholar] [CrossRef] [PubMed]
  63. Yao, Q.; Liang, H.; Huang, B.; Xiang, L.; Wang, T.; Xiong, Y.; Yang, B.; Guo, Y.; Gong, P. Beta-adrenergic signaling affect osteoclastogenesis via osteocytic MLO-Y4 cells’ RANKL production. Biochem. Biophys. Res. Commun. 2017, 488, 634–640. [Google Scholar] [CrossRef] [PubMed]
  64. Ma, Y.; Nyman, J.S.; Tao, H.; Moss, H.H.; Yang, X.; Elefteriou, F. β2-Adrenergic receptor signaling in osteoblasts contributes to the catabolic effect of glucocorticoids on bone. Endocrinology 2011, 152, 1412–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Wu, H.; Song, Y.; Li, J.; Lei, X.; Zhang, S.; Gao, Y.; Cheng, P.; Liu, B.; Miao, S.; Bi, L. Blockade of adrenergic β-receptor activation through local delivery of propranolol from a 3D collagen/polyvinyl alcohol/hydroxyapatite scaffold promotes bone repair in vivo. Cell Prolif. 2020, 53, e12725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Bellier, J.-P.; Kimura, H. Peripheral type of choline acetyltransferase: Biological and evolutionary implications for novel mechanisms in cholinergic system. J. Chem. Neuroanat. 2011, 42, 225–235. [Google Scholar] [CrossRef]
  67. Bajayo, A.; Bar, A.; Denes, A.; Bachar, M.; Kram, V.; Attar-Namdar, M.; Zallone, A.; Kovács, K.J.; Yirmiya, R.; Bab, I. Skeletal parasympathetic innervation communicates central IL-1 signals regulating bone mass accrual. Proc. Natl. Acad. Sci. USA 2012, 109, 15455–15460. [Google Scholar] [CrossRef] [Green Version]
  68. Sato, T.; Abe, T.; Chida, D.; Nakamoto, N.; Hori, N.; Kokabu, S.; Sakata, Y.; Tomaru, Y.; Iwata, T.; Usui, M. Functional role of acetylcholine and the expression of cholinergic receptors and components in osteoblasts. FEBS Lett. 2010, 584, 817–824. [Google Scholar] [CrossRef] [Green Version]
  69. Brazill, J.M.; Beeve, A.T.; Craft, C.S.; Ivanusic, J.J.; Scheller, E.L. Nerves in bone: Evolving concepts in pain and anabolism. J. Bone Miner. Res. 2019, 34, 1393–1406. [Google Scholar] [CrossRef]
  70. Oostinga, D.; Steverink, J.G.; van Wijck, A.J.; Verlaan, J.-J. An understanding of bone pain: A narrative review. Bone 2020, 134, 115272. [Google Scholar] [CrossRef]
  71. Zhou, Y.; Zhang, H.; Zhang, G.; He, Y.; Zhang, P.; Sun, Z.; Gao, Y.; Tan, Y. Calcitonin gene-related peptide reduces Porphyromonas gingivalis LPS-induced TNF-α release and apoptosis in osteoblasts. Mol. Med. Rep. 2018, 17, 3246–3254. [Google Scholar]
  72. Heffner, M.A.; Genetos, D.C.; Christiansen, B.A. Bone adaptation to mechanical loading in a mouse model of reduced peripheral sensory nerve function. PLoS ONE 2017, 12, e0187354. [Google Scholar] [CrossRef] [Green Version]
  73. Kosmidis, S.; Polyzos, A.; Harvey, L.; Youssef, M.; Denny, C.A.; Dranovsky, A.; Kandel, E.R. RbAp48 protein is a critical component of GPR158/OCN signaling and ameliorates age-related memory loss. Cell Rep. 2018, 25, 959–973.e956. [Google Scholar] [CrossRef]
  74. Bhusal, A.; Rahman, M.H.; Lee, W.-H.; Bae, Y.C.; Lee, I.-K.; Suk, K. Paradoxical role of lipocalin-2 in metabolic disorders and neurological complications. Biochem. Pharmacol. 2019, 169, 113626. [Google Scholar] [CrossRef]
  75. Laszczyk, A.; Nettles, D.; Pollock, T.; Fox, S.; Garcia, M.; Wang, J.; Quarles, L.; King, G. FGF-23 deficiency impairs hippocampal-dependent cognitive function. eNeuro 2019, 6, e0469. [Google Scholar] [CrossRef]
  76. Hanada, R.; Leibbrandt, A.; Hanada, T.; Kitaoka, S.; Furuyashiki, T.; Fujihara, H.; Trichereau, J.; Paolino, M.; Qadri, F.; Plehm, R. Central control of fever and female body temperature by RANKL/RANK. Nature 2009, 462, 505–509. [Google Scholar] [CrossRef]
  77. Zhang, J.; Fujita, Y.; Chang, L.; Pu, Y.; Qu, Y.; Wang, S.; Hashimoto, K. Beneficial effects of anti-RANKL antibody in depression-like phenotype, inflammatory bone markers, and bone mineral density in male susceptible mice after chronic social defeat stress. Behav. Brain Res. 2020, 379, 112397. [Google Scholar] [CrossRef]
  78. Zhang, D.-D.; Cao, Y.; Mu, J.-Y.; Liu, Y.-M.; Gao, F.; Han, F.; Zhai, F.-F.; Zhou, L.-X.; Ni, J.; Yao, M. Inflammatory biomarkers and cerebral small vessel disease: A community-based cohort study (P3-3.001). AAN Enterprises 2022, 98, 1915. [Google Scholar] [CrossRef]
  79. Levey, A.I.; Qiu, D.; Zhao, L.; Hu, W.T.; Duong, D.M.; Higginbotham, L.; Dammer, E.B.; Seyfried, N.T.; Wingo, T.S.; Hales, C.M. A phase II study repurposing atomoxetine for neuroprotection in mild cognitive impairment. Brain 2022, 145, 1924–1938. [Google Scholar] [CrossRef]
  80. Ross, R.D.; Shah, R.C.; Leurgans, S.; Bottiglieri, T.; Wilson, R.S.; Sumner, D.R. Circulating Dkk1 and TRAIL are associated with cognitive decline in community-dwelling, older adults with cognitive concerns. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2018, 73, 1688. [Google Scholar] [CrossRef]
  81. Sheehan-Rooney, K.; Swartz, M.E.; Lovely, C.B.; Dixon, M.J.; Eberhart, J.K. Bmp and Shh signaling mediate the expression of satb2 in the pharyngeal arches. PLoS ONE 2013, 8, e59533. [Google Scholar] [CrossRef] [Green Version]
  82. Konopka, G.; Bomar, J.M.; Winden, K.; Coppola, G.; Jonsson, Z.O.; Gao, F.; Peng, S.; Preuss, T.M.; Wohlschlegel, J.A.; Geschwind, D.H. Human-specific transcriptional regulation of CNS development genes by FOXP2. Nature 2009, 462, 213–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ye, J.-H.; Xu, Y.-J.; Gao, J.; Yan, S.-G.; Zhao, J.; Tu, Q.; Zhang, J.; Duan, X.-J.; Sommer, C.A.; Mostoslavsky, G. Critical-size calvarial bone defects healing in a mouse model with silk scaffolds and SATB2-modified iPSCs. Biomaterials 2011, 32, 5065–5076. [Google Scholar] [PubMed]
  84. Miguez, A.; Ducret, S.; Di Meglio, T.; Parras, C.; Hmidan, H.; Haton, C.; Sekizar, S.; Mannioui, A.; Vidal, M.; Kerever, A. Opposing roles for Hoxa2 and Hoxb2 in hindbrain oligodendrocyte patterning. J. Neurosci. 2012, 32, 17172–17185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Geisen, M.J.; Meglio, T.D.; Pasqualetti, M.; Ducret, S.; Brunet, J.-F.; Chedotal, A.; Rijli, F.M. Hox paralog group 2 genes control the migration of mouse pontine neurons through slit-robo signaling. PLoS Biol. 2008, 6, e142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Dobreva, G.; Chahrour, M.; Dautzenberg, M.; Chirivella, L.; Kanzler, B.; Fariñas, I.; Karsenty, G.; Grosschedl, R. SATB2 is a multifunctional determinant of craniofacial patterning and osteoblast differentiation. Cell 2006, 125, 971–986. [Google Scholar] [CrossRef] [Green Version]
  87. Tavella, S.; Bobola, N. Expressing Hoxa2 across the entire endochondral skeleton alters the shape of the skeletal template in a spatially restricted fashion. Differentiation 2010, 79, 194–202. [Google Scholar] [CrossRef]
  88. Garcez, R.C.; Le Douarin, N.M.; Creuzet, S.E. Combinatorial activity of Six1-2-4 genes in cephalic neural crest cells controls craniofacial and brain development. Cell. Mol. Life Sci. 2014, 71, 2149–2164. [Google Scholar] [CrossRef]
  89. Zhao, H.; Zhou, W.; Yao, Z.; Wan, Y.; Cao, J.; Zhang, L.; Zhao, J.; Li, H.; Zhou, R.; Li, B. Foxp1/2/4 regulate endochondral ossification as a suppresser complex. Dev. Biol. 2015, 398, 242–254. [Google Scholar]
  90. MuhChyi, C.; Juliandi, B.; Matsuda, T.; Nakashima, K. Epigenetic regulation of neural stem cell fate during corticogenesis. Int. J. Dev. Neurosci. 2013, 31, 424–433. [Google Scholar] [CrossRef]
  91. Borrell, V.; Cárdenas, A.; Ciceri, G.; Galcerán, J.; Flames, N.; Pla, R.; Nóbrega-Pereira, S.; García-Frigola, C.; Peregrín, S.; Zhao, Z. Slit/Robo signaling modulates the proliferation of central nervous system progenitors. Neuron 2012, 76, 338–352. [Google Scholar] [CrossRef] [Green Version]
  92. Long, Q.; Qiu, B.; Wang, K.; Yang, J.; Jia, C.; Xin, W.; Wang, P.; Han, R.; Fei, Z.; Liu, W. Genetically engineered bone marrow mesenchymal stem cells improve functional outcome in a rat model of epilepsy. Brain Res. 2013, 1532, 1–13. [Google Scholar] [CrossRef]
  93. Sugita, S.; Hosaka, Y.; Okada, K.; Mori, D.; Yano, F.; Kobayashi, H.; Taniguchi, Y.; Mori, Y.; Okuma, T.; Chang, S.H. Transcription factor Hes1 modulates osteoarthritis development in cooperation with calcium/calmodulin-dependent protein kinase 2. Proc. Natl. Acad. Sci. USA 2015, 112, 3080–3085. [Google Scholar] [CrossRef] [Green Version]
  94. Otto, E.; Knapstein, P.-R.; Jahn, D.; Appelt, J.; Frosch, K.-H.; Tsitsilonis, S.; Keller, J. Crosstalk of brain and bone—Clinical observations and their molecular bases. Int. J. Mol. Sci. 2020, 21, 4946. [Google Scholar] [CrossRef]
  95. Owen, R.; Reilly, G.C. In vitro models of bone remodelling and associated disorders. Front. Bioeng. Biotechnol. 2018, 6, 134. [Google Scholar] [CrossRef] [Green Version]
  96. Blesa, J.S.; Przedborski, S. Parkinson’s disease: Animal models and dopaminergic cell vulnerability. Front. Neuroanat 2014, 8, 155. [Google Scholar] [CrossRef] [Green Version]
  97. Oliveros Anerillas, L.; Kingham, P.J.; Lammi, M.J.; Wiberg, M.; Kelk, P. Three-dimensional osteogenic differentiation of bone marrow mesenchymal stem cells promotes matrix metallopeptidase 13 (MMP13) expression in Type I collagen hydrogels. Int. J. Mol. Sci. 2021, 22, 13594. [Google Scholar] [CrossRef]
  98. Fatehullah, A.; Tan, S.H.; Barker, N. Organoids as an in vitro model of human development and disease. Nat. Cell Biol. 2016, 18, 246–254. [Google Scholar] [CrossRef] [Green Version]
  99. Choi, S.H.; Kim, Y.H.; Hebisch, M.; Sliwinski, C.; Lee, S.; D’Avanzo, C.; Chen, H.; Hooli, B.; Asselin, C.; Muffat, J. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 2014, 515, 274–278. [Google Scholar] [CrossRef] [Green Version]
  100. Tibbitt, M.W.; Anseth, K.S. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol. Bioeng. 2009, 103, 655–663. [Google Scholar] [CrossRef] [Green Version]
  101. Amelian, A.; Wasilewska, K.; Megias, D.; Winnicka, K. Application of standard cell cultures and 3D in vitro tissue models as an effective tool in drug design and development. Pharmacol. Rep. 2017, 69, 861–870. [Google Scholar] [CrossRef]
  102. Bolognin, S.; Fossépré, M.; Qing, X.; Jarazo, J.; Ščančar, J.; Moreno, E.L.; Nickels, S.L.; Wasner, K.; Ouzren, N.; Walter, J. 3D cultures of Parkinson’s disease-specific dopaminergic neurons for high content phenotyping and drug testing. Adv. Sci. 2019, 6, 1800927. [Google Scholar] [CrossRef] [PubMed]
  103. Oun, A.; Sabogal-Guaqueta, A.M.; Galuh, S.; Alexander, A.; Kortholt, A.; Dolga, A. The multifaceted role of LRRK2 in Parkinson’s disease: From human iPSC to organoids. Neurobiol. Dis. 2022, 173, 105837. [Google Scholar] [CrossRef] [PubMed]
  104. Fett, M.E.; Pilsl, A.; Paquet, D.; Van Bebber, F.; Haass, C.; Tatzelt, J.; Schmid, B.; Winklhofer, K.F. Parkin is protective against proteotoxic stress in a transgenic zebrafish model. PLoS ONE 2010, 5, e11783. [Google Scholar] [CrossRef] [PubMed]
  105. Baptista, L.S.; Kronemberger, G.S.; Côrtes, I.; Charelli, L.E.; Matsui, R.A.M.; Palhares, T.N.; Sohier, J.; Rossi, A.M.; Granjeiro, J.M. Adult stem cells spheroids to optimize cell colonization in scaffolds for cartilage and bone tissue engineering. Int. J. Mol. Sci. 2018, 19, 1285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Collins, M.N.; Ren, G.; Young, K.; Pina, S.; Reis, R.L.; Oliveira, J.M. Scaffold fabrication technologies and structure/function properties in bone tissue engineering. Adv. Funct. Mater. 2021, 31, 2010609. [Google Scholar] [CrossRef]
  107. Kargozar, S.; Mozafari, M.; Hamzehlou, S.; Brouki Milan, P.; Kim, H.-W.; Baino, F. Bone tissue engineering using human cells: A comprehensive review on recent trends, current prospects, and recommendations. Appl. Sci. 2019, 9, 174. [Google Scholar]
  108. Heo, J.H.; Kang, D.; Seo, S.J.; Jin, Y. Engineering the extracellular matrix for organoid culture. Int. J. Stem Cells 2022, 15, 60–69. [Google Scholar] [CrossRef]
  109. Chen, S.; Chen, X.; Geng, Z.; Su, J. The horizon of bone organoid: A perspective on construction and application. Bioact. Mater. 2022, 18, 15–25. [Google Scholar] [CrossRef]
  110. Ye, W.; Luo, C.; Li, C.; Huang, J.; Liu, F. Organoids to study immune functions, immunological diseases and immunotherapy. Cancer Lett. 2020, 477, 31–40. [Google Scholar]
  111. Grebenyuk, S.; Ranga, A. Engineering organoid vascularization. Front. Bioeng. Biotechnol. 2019, 7, 39. [Google Scholar] [CrossRef] [Green Version]
  112. Singh, A.; Nikkhah, M.; Annabi, N. Biomaterials, Cells, and Patho-physiology: Building Better Organoids and On-Chip Technologies. Biomaterials 2019, 198, 1–2. [Google Scholar] [CrossRef]
  113. Torisawa, Y.-s.; Spina, C.S.; Mammoto, T.; Mammoto, A.; Weaver, J.C.; Tat, T.; Collins, J.J.; Ingber, D.E. Bone marrow–on–a–chip replicates hematopoietic niche physiology in vitro. Nat. Methods 2014, 11, 663–669. [Google Scholar] [CrossRef] [Green Version]
  114. Tonelli, F.; Bek, J.W.; Besio, R.; De Clercq, A.; Leoni, L.; Salmon, P.; Coucke, P.J.; Willaert, A.; Forlino, A. Zebrafish: A resourceful vertebrate model to investigate skeletal disorders. Front. Endocrinol. 2020, 11, 489. [Google Scholar] [CrossRef]
  115. Finley, M.L.; Kidd, K.A.; Curry, R.A.; Lescord, G.L.; Clayden, M.G.; O’Driscoll, N.J. A comparison of mercury biomagnification through lacustrine food webs supporting brook trout (Salvelinus fontinalis) and other salmonid fishes. Front. Environ. Sci. 2016, 4, 23. [Google Scholar] [CrossRef] [Green Version]
  116. Bergen, D.J.; Kague, E.; Hammond, C.L. Zebrafish as an emerging model for osteoporosis: A primary testing platform for screening new osteo-active compounds. Front. Endocrinol. 2019, 10, 6. [Google Scholar] [CrossRef]
  117. Flinn, L.; Mortiboys, H.; Volkmann, K.; Köster, R.W.; Ingham, P.W.; Bandmann, O. Complex I deficiency and dopaminergic neuronal cell loss in parkin-deficient zebrafish (Danio rerio). Brain 2009, 132, 1613–1623. [Google Scholar] [CrossRef] [Green Version]
  118. Razali, K.; Othman, N.; Mohd Nasir, M.H.; Doolaanea, A.A.; Kumar, J.; Ibrahim, W.N.; Mohamed Ibrahim, N.; Mohamed, W.M. The Promise of the zebrafish model for Parkinson’s disease: Today’s science and tomorrow’s treatment. Front. Genet. 2021, 12, 655550. [Google Scholar] [CrossRef]
  119. Kõks, S.; Dogan, S.; Tuna, B.G.; González-Navarro, H.; Potter, P.; Vandenbroucke, R.E. Mouse models of ageing and their relevance to disease. Mech. Ageing Dev. 2016, 160, 41–53. [Google Scholar] [CrossRef]
  120. Jilka, R.L. The relevance of mouse models for investigating age-related bone loss in humans. J. Gerontol. Ser. A: Biomed. Sci. Med. Sci. 2013, 68, 1209–1217. [Google Scholar] [CrossRef]
  121. Klæstrup, I.H.; Just, M.K.; Holm, K.L.; Alstrup, A.K.O.; Romero-Ramos, M.; Borghammer, P.; Van Den Berge, N. Impact of aging on animal models of Parkinson’s disease. Front. Aging Neurosci. 2022, 14, 909273. [Google Scholar] [CrossRef]
  122. Moore, D.J.; Dawson, T.M. Value of genetic models in understanding the cause and mechanisms of Parkinson’s disease. Curr. Neurol. Neurosci. Rep. 2008, 8, 288–296. [Google Scholar] [PubMed]
  123. Van der Vlag, M.; Havekes, R.; Heckman, P.R. The contribution of Parkin, PINK1 and DJ-1 genes to selective neuronal degeneration in Parkinson’s disease. Eur. J. Neurosci. 2020, 52, 3256–3268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Chang, E.E.S.; Ho, P.W.-L.; Liu, H.-F.; Pang, S.Y.-Y.; Leung, C.-T.; Malki, Y.; Choi, Z.Y.-K.; Ramsden, D.B.; Ho, S.-L. LRRK2 mutant knock-in mouse models: Therapeutic relevance in Parkinson’s disease. Transl. Neurodegener. 2022, 11, 10. [Google Scholar] [PubMed]
  125. Yang, T.-X.; Zhu, Y.-F.; Wang, C.-C.; Yang, J.-Y.; Xue, C.-H.; Huang, Q.-R.; Wang, Y.-M.; Zhang, T.-T. Epa-enriched plasmalogen attenuates the cytotoxic effects of lps-stimulated microglia on the sh-sy5y neuronal cell line. Brain Res. Bull. 2022, 186, 143–152. [Google Scholar] [CrossRef] [PubMed]
  126. Brennan, M.Á.; Renaud, A.; Gamblin, A.-l.; D’arros, C.; Nedellec, S.; Trichet, V.; Layrolle, P. 3D cell culture and osteogenic differentiation of human bone marrow stromal cells plated onto jet-sprayed or electrospun micro-fiber scaffolds. Biomed. Mater. 2015, 10, 045019. [Google Scholar] [PubMed]
  127. Noroozi, R.; Shamekhi, M.A.; Mahmoudi, R.; Zolfagharian, A.; Asgari, F.; Mousavizadeh, A.; Bodaghi, M.; Hadi, A.; Haghighipour, N. In vitro static and dynamic cell culture study of novel bone scaffolds based on 3D-printed PLA and cell-laden alginate hydrogel. Biomed. Mater. 2022, 17, 045024. [Google Scholar] [CrossRef]
  128. Ohori-Morita, Y.; Niibe, K.; Limraksasin, P.; Nattasit, P.; Miao, X.; Yamada, M.; Mabuchi, Y.; Matsuzaki, Y.; Egusa, H. Novel Mesenchymal Stem Cell Spheroids with Enhanced Stem Cell Characteristics and Bone Regeneration Ability. Stem Cells Transl. Med. 2022, 11, 434–449. [Google Scholar]
  129. Yuan, P.; Zhang, M.; Tong, L.; Morse, T.M.; McDougal, R.A.; Ding, H.; Chan, D.; Cai, Y.; Grutzendler, J. PLD3 affects axonal spheroids and network defects in Alzheimer’s disease. Nature 2022, 612, 328–337. [Google Scholar]
  130. Rabadan, M.; De La Cruz, E.D.; Rao, S.B.; Chen, Y.; Gong, C.; Crabtree, G.; Xu, B.; Markx, S.; Gogos, J.A.; Yuste, R. An in vitro model of neuronal ensembles. Nat. Commun. 2022, 13, 3340. [Google Scholar] [CrossRef]
  131. Spitz, S.; Bolognin, S.; Brandauer, K.; Fuessl, J.; Schuller, P.; Schobesberger, S.; Jordan, C.; Schaedl, B.; Grillari, J.; Wanzenboeck, H.D. Development of a multi-sensor integrated midbrain organoid-on-a-chip platform for studying Parkinson’s disease. bioRxiv 2022. [Google Scholar] [CrossRef]
  132. Shin, N.; Kim, Y.; Ko, J.; Choi, S.W.; Hyung, S.; Lee, S.E.; Park, S.; Song, J.; Jeon, N.L.; Kang, K.S. Vascularization of iNSC spheroid in a 3D spheroid-on-a-chip platform enhances neural maturation. Biotechnol. Bioeng. 2022, 119, 566–574. [Google Scholar] [CrossRef]
  133. Brighi, C.; Cordella, F.; Chiriatti, L.; Soloperto, A.; Di Angelantonio, S. Retinal and brain organoids: Bridging the gap between in vivo physiology and in vitro micro-physiology for the study of alzheimer’s diseases. Front. Neurosci. 2020, 14, 655. [Google Scholar] [CrossRef]
  134. Gonzalez, C.; Armijo, E.; Bravo-Alegria, J.; Becerra-Calixto, A.; Mays, C.E.; Soto, C. Modeling amyloid beta and tau pathology in human cerebral organoids. Mol. Psychiatry 2018, 23, 2363–2374. [Google Scholar] [CrossRef]
  135. Zhang, Y.; Yu, T.; Ding, J.; Li, Z. Bone-on-a-chip platforms and integrated biosensors: Towards advanced in vitro bone models with real-time biosensing. Biosens. Bioelectron. 2022, 219, 114798. [Google Scholar] [CrossRef]
  136. Cha, C. Microfluidic Biotechnology for “Bone-on-a-Chip”. Biofabr. Orthop. Methods Tech. Appl. 2022, 1, 181–209. [Google Scholar]
  137. Gan, S.; Huang, Z.; Liu, N.; Su, R.; Xie, G.; Zhong, B.; Zhang, K.; Wang, S.; Hu, X.; Zhang, J. Micro RNA-140-5p impairs zebrafish embryonic bone development via targeting BMP-2. FEBS Lett. 2016, 590, 1438–1446. [Google Scholar] [CrossRef] [Green Version]
  138. Méndez-Martínez, L.; Guerrero-Peña, L.; Suárez-Bregua, P.; Naranjo, S.; Tena, J.J.; Rotllant, J. Neural Regulation of Bone Mineral Homeostasis in Fish: Functional and Transcriptional Characterization of pth4 Neurons. In Proceedings of the 6th International Symposium on Genomics in Aquaculture, Granada, Spain, 4–6 May 2022. [Google Scholar]
  139. Kawanishi, S.; Takata, K.; Itezono, S.; Nagayama, H.; Konoya, S.; Chisaki, Y.; Toda, Y.; Nakata, S.; Yano, Y.; Kitamura, Y. Bone-marrow-derived microglia-like cells ameliorate brain amyloid pathology and cognitive impairment in a mouse model of Alzheimer’s disease. J. Alzheimer’s Dis. 2018, 64, 563–585. [Google Scholar] [CrossRef]
  140. LLabre, J.E.; 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. [Google Scholar] [CrossRef]
  141. Cardoso, A.L.; Fernandes, A.; Aguilar-Pimentel, J.A.; de Angelis, M.H.; Guedes, J.R.; Brito, M.A.; Ortolano, S.; Pani, G.; Athanasopoulou, S.; Gonos, E.S. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res. Rev. 2018, 47, 214–277. [Google Scholar] [CrossRef]
  142. Tsai, Y.-L.; Yen, C.-T.; Wang, Y.-F. Astrocyte Dysregulation and Calcium Ion Imbalance May Link the Development of Osteoporosis and Alzheimer’s Disease. J. Alzheimer’s Dis. 2022, 88, 439–445. [Google Scholar] [CrossRef]
  143. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wu, B.-W.; Guo, J.-D.; Wu, M.-S.; Liu, Y.; Lu, M.; Zhou, Y.-H.; Han, H.-W. Osteoblast-derived lipocalin-2 regulated by miRNA-96-5p/Foxo1 advances the progression of Alzheimer’s disease. Epigenomics 2020, 12, 1501–1513. [Google Scholar] [CrossRef] [PubMed]
  145. Ximerakis, M.; Lipnick, S.L.; Innes, B.T.; Simmons, S.K.; Adiconis, X.; Dionne, D.; Mayweather, B.A.; Nguyen, L.; Niziolek, Z.; Ozek, C. Single-cell transcriptomic profiling of the aging mouse brain. Nat. Neurosci. 2019, 22, 1696–1708. [Google Scholar] [CrossRef] [PubMed]
  146. Zhong, L.; Wang, Z.; Wang, D.; Wang, Z.; Martens, Y.A.; Wu, L.; Xu, Y.; Wang, K.; Li, J.; Huang, R. Amyloid-beta modulates microglial responses by binding to the triggering receptor expressed on myeloid cells 2 (TREM2). Mol. Neurodegener. 2018, 13, 15. [Google Scholar] [CrossRef] [PubMed]
  147. Li, R.; Lv, Z.-Y.; Li, Y.-X.; Hao, Y.-L. Effects of TYROBP Deficiency on Neuroinflammation of a Alzheimer’s Disease Mouse Model Carrying a PSEN1 p. G378E Mutation. Chin. Med. Sci. J. Chung-Kuo I Hsueh K’o Hsueh Tsa Chih 2022. [Google Scholar] [CrossRef]
  148. Ulland, T.K.; Colonna, M. TREM2—A key player in microglial biology and Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 667–675. [Google Scholar] [CrossRef]
  149. Tsukasaki, M.; Takayanagi, H. Osteoimmunology: Evolving concepts in bone–immune interactions in health and disease. Nat. Rev. Immunol. 2019, 19, 626–642. [Google Scholar] [CrossRef]
  150. Varma, V.; Varma, S.; An, Y.; Hohman, T.; Seddighi, S.; Casanova, R.; Beri, A.; Dammer, E.; Seyfried, N.; Pletnikova, O. Alpha-2 macroglobulin in Alzheimer’s disease: A marker of neuronal injury through the RCAN1 pathway. Mol. Psychiatry 2017, 22, 13–23. [Google Scholar] [CrossRef]
  151. Xiong, L.; Pan, J.-X.; Guo, H.-h.; Mei, L.; Xiong, W.-C. Parkinson’s in the bone. Cell Biosci. 2021, 11, 190. [Google Scholar] [CrossRef]
  152. Allen, N.E.; Canning, C.G.; Almeida, L.R.S.; Bloem, B.R.; Keus, S.H.; Löfgren, N.; Nieuwboer, A.; Verheyden, G.S.; Yamato, T.P.; Sherrington, C. Interventions for preventing falls in Parkinson’s disease. Cochrane Database Syst. Rev. 2022, 6, CD011574. [Google Scholar]
  153. Berwick, D.C.; Harvey, K. The regulation and deregulation of Wnt signaling by PARK genes in health and disease. J. Mol. Cell Biol. 2014, 6, 3–12. [Google Scholar] [CrossRef] [Green Version]
  154. Inestrosa, N.C.; Arenas, E. Emerging roles of Wnts in the adult nervous system. Nat. Rev. Neurosci. 2010, 11, 77–86. [Google Scholar] [CrossRef]
  155. L’Episcopo, F.; Tirolo, C.; Testa, N.; Caniglia, S.; Morale, M.C.; Serapide, M.F.; Pluchino, S.; Marchetti, B. Wnt/β-catenin signaling is required to rescue midbrain dopaminergic progenitors and promote neurorepair in ageing mouse model of Parkinson’s disease. Stem Cells 2014, 32, 2147–2163. [Google Scholar] [CrossRef] [Green Version]
  156. Manikandan, M.; Abuelreich, S.; Elsafadi, M.; Alsalman, H.; Almalak, H.; Siyal, A.; Hashmi, J.A.; Aldahmash, A.; Kassem, M.; Alfayez, M. NR2F1 mediated down-regulation of osteoblast differentiation was rescued by bone morphogenetic protein-2 (BMP-2) in human MSC. Differentiation 2018, 104, 36–41. [Google Scholar] [CrossRef]
  157. Walter, J.; Bolognin, S.; Poovathingal, S.K.; Magni, S.; Gérard, D.; Antony, P.M.; Nickels, S.L.; Salamanca, L.; Berger, E.; Smits, L.M. The Parkinson’s-disease-associated mutation LRRK2-G2019S alters dopaminergic differentiation dynamics via NR2F1. Cell Rep. 2021, 37, 109864. [Google Scholar] [CrossRef]
  158. Bisson, E.J.; Finlayson, M.L.; Ekuma, O.; Leslie, W.D.; Marrie, R.A. Multiple sclerosis is associated with low bone mineral density and osteoporosis. Neurol. Clin. Pract. 2019, 9, 391–399. [Google Scholar] [CrossRef]
  159. Biel, A.; Castanza, A.S.; Rutherford, R.; Fair, S.R.; Chifamba, L.; Wester, J.C.; Hester, M.E.; Hevner, R.F. AUTS2 syndrome: Molecular mechanisms and model systems. Front. Mol. Neurosci. 2022, 15, 858582. [Google Scholar] [CrossRef]
  160. Eisenberg, E.; Levanon, E.Y. Human housekeeping genes, revisited. TRENDS Genet. 2013, 29, 569–574. [Google Scholar] [CrossRef]
  161. Beunders, G.; Voorhoeve, E.; Golzio, C.; Pardo, L.M.; Rosenfeld, J.A.; Talkowski, M.E.; Simonic, I.; Lionel, A.C.; Vergult, S.; Pyatt, R.E. Exonic deletions in AUTS2 cause a syndromic form of intellectual disability and suggest a critical role for the C terminus. Am. J. Hum. Genet. 2013, 92, 210–220. [Google Scholar] [CrossRef]
  162. De Rubeis, S.; He, X.; Goldberg, A.P.; Poultney, C.S.; Samocha, K.; Ercument Cicek, A.; Kou, Y.; Liu, L.; Fromer, M.; Walker, S. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 2014, 515, 209–215. [Google Scholar] [CrossRef] [Green Version]
  163. Cheng, P.-l.; Lu, H.; Shelly, M.; Gao, H.; Poo, M.-m. Phosphorylation of E3 ligase Smurf1 switches its substrate preference in support of axon development. Neuron 2011, 69, 231–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Benitez-Burraco, A.; Boeckx, C. Possible functional links among brain-and skull-related genes selected in modern humans. Front. Psychol. 2015, 6, 794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Liedén, A.; Kvarnung, M.; Nilssson, D.; Sahlin, E.; Lundberg, E.S. Intragenic duplication—A novel causative mechanism for SATB2-associated syndrome. Am. J. Med. Genet. Part A 2014, 164, 3083–3087. [Google Scholar] [CrossRef] [PubMed]
  166. Zhao, X.; Qu, Z.; Tickner, J.; Xu, J.; Dai, K.; Zhang, X. The role of SATB2 in skeletogenesis and human disease. Cytokine Growth Factor Rev. 2014, 25, 35–44. [Google Scholar] [CrossRef] [PubMed]
  167. Hassan, M.Q.; Gordon, J.A.; Beloti, M.M.; Croce, C.M.; Wijnen, A.J.v.; Stein, J.L.; Stein, G.S.; Lian, J.B. A network connecting Runx2, SATB2, and the miR-23a∼ 27a∼ 24-2 cluster regulates the osteoblast differentiation program. Proc. Natl. Acad. Sci. USA 2010, 107, 19879–19884. [Google Scholar] [CrossRef] [Green Version]
  168. Gong, Y.; Qian, Y.; Yang, F.; Wang, H.; Yu, Y. Lentiviral-mediated expression of SATB 2 promotes osteogenic differentiation of bone marrow stromal cells in vitro and in vivo. Eur. J. Oral Sci. 2014, 122, 190–197. [Google Scholar] [CrossRef]
  169. Huang, H.; Tindall, D.J. Dynamic FoxO transcription factors. J. Cell Sci. 2007, 120, 2479–2487. [Google Scholar] [CrossRef] [Green Version]
  170. Van Bon, B.; Hoischen, A.; Hehir-Kwa, J.; De Brouwer, A.; Ruivenkamp, C.; Gijsbers, A.; Marcelis, C.; De Leeuw, N.; Veltman, J.; Brunner, H. Intragenic deletion in DYRK1A leads to mental retardation and primary microcephaly. Clin. Genet. 2011, 79, 296–299. [Google Scholar] [CrossRef]
  171. Courcet, J.-B.; Faivre, L.; Malzac, P.; Masurel-Paulet, A.; Lopez, E.; Callier, P.; Lambert, L.; Lemesle, M.; Thevenon, J.; Gigot, N. The DYRK1A gene is a cause of syndromic intellectual disability with severe microcephaly and epilepsy. J. Med. Genet. 2012, 49, 731–736. [Google Scholar] [CrossRef]
  172. Lee, Y.; Ha, J.; Kim, H.J.; Kim, Y.-S.; Chang, E.-J.; Song, W.-J.; Kim, H.-H. Negative feedback Inhibition of NFATc1 by DYRK1A regulates bone homeostasis. J. Biol. Chem. 2009, 284, 33343–33351. [Google Scholar] [CrossRef] [Green Version]
  173. Shakibaei, M.; Shayan, P.; Busch, F.; Aldinger, C.; Buhrmann, C.; Lueders, C.; Mobasheri, A. Resveratrol mediated modulation of Sirt-1/Runx2 promotes osteogenic differentiation of mesenchymal stem cells: Potential role of Runx2 deacetylation. PLoS ONE 2012, 7, e35712. [Google Scholar] [CrossRef] [Green Version]
  174. Iyer, S.; Han, L.; Bartell, S.M.; Kim, H.-N.; Gubrij, I.; de Cabo, R.; O’Brien, C.A.; Manolagas, S.C.; Almeida, M. Sirtuin1 (Sirt1) promotes cortical bone formation by preventing β-catenin sequestration by FoxO transcription factors in osteoblast progenitors. J. Biol. Chem. 2014, 289, 24069–24078. [Google Scholar] [CrossRef] [Green Version]
  175. Joe, I.-S.; Jeong, S.-G.; Cho, G.-W. Resveratrol-induced SIRT1 activation promotes neuronal differentiation of human bone marrow mesenchymal stem cells. Neurosci. Lett. 2015, 584, 97–102. [Google Scholar] [CrossRef]
  176. Peng, X.-d.; Xu, P.-Z.; Chen, M.-L.; Hahn-Windgassen, A.; Skeen, J.; Jacobs, J.; Sundararajan, D.; Chen, W.S.; Crawford, S.E.; Coleman, K.G. Dwarfism, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. Genes Dev. 2003, 17, 1352–1365. [Google Scholar] [CrossRef] [Green Version]
  177. Mukherjee, A.; Larson, E.A.; Klein, R.F.; Rotwein, P. Distinct actions of akt1 on skeletal architecture and function. PLoS ONE 2014, 9, e93040. [Google Scholar] [CrossRef] [Green Version]
  178. Dudek, H.; Datta, S.R.; Franke, T.F.; Birnbaum, M.J.; Yao, R.; Cooper, G.M.; Segal, R.A.; Kaplan, D.R.; Greenberg, M.E. Regulation of neuronal survival by the serine-threonine protein kinase Akt. Science 1997, 275, 661–665. [Google Scholar] [CrossRef]
  179. Bajwa, N.M.; Kesavan, C.; Mohan, S. Long-term consequences of traumatic brain injury in bone metabolism. Front. Neurol. 2018, 9, 115. [Google Scholar] [CrossRef] [Green Version]
  180. Jodoin, M.; Rouleau, D.M.; Therrien, E.; Chauny, J.-M.; Sandman, E.; Larson-Dupuis, C.; Leduc, S.; Gosselin, N.; De Beaumont, L. Investigating the incidence and magnitude of heterotopic ossification with and without joints involvement in patients with a limb fracture and mild traumatic brain injury. Bone Rep. 2019, 11, 100222. [Google Scholar] [CrossRef]
  181. Kalamakis, G.; Brüne, D.; Ravichandran, S.; Bolz, J.; Fan, W.; Ziebell, F.; Stiehl, T.; Catalá-Martinez, F.; Kupke, J.; Zhao, S. Quiescence modulates stem cell maintenance and regenerative capacity in the aging brain. Cell 2019, 176, 1407–1419.e1414. [Google Scholar] [CrossRef]
  182. Brunet, A.; Goodell, M.A.; Rando, T.A. Ageing and rejuvenation of tissue stem cells and their niches. Nat. Rev. Mol. Cell Biol. 2022, 24, 45–62. [Google Scholar] [CrossRef]
  183. Baror, R.; Neumann, B.; Segel, M.; Chalut, K.J.; Fancy, S.P.; Schafer, D.P.; Franklin, R.J. Transforming growth factor-beta renders ageing microglia inhibitory to oligodendrocyte generation by CNS progenitors. Glia 2019, 67, 1374–1384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Desplats, P.; Lee, H.-J.; Bae, E.-J.; Patrick, C.; Rockenstein, E.; Crews, L.; Spencer, B.; Masliah, E.; Lee, S.-J. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of α-synuclein. Proc. Natl. Acad. Sci. USA 2009, 106, 13010–13015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Eglitis, M.A.; Mezey, É. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc. Natl. Acad. Sci. USA 1997, 94, 4080–4085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Brazelton, T.R.; Rossi, F.M.; Keshet, G.I.; Blau, H.M. From marrow to brain: Expression of neuronal phenotypes in adult mice. Science 2000, 290, 1775–1779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Mezey, E.; Chandross, K.J.; Harta, G.; Maki, R.A.; McKercher, S.R. Turning blood into brain: Cells bearing neuronal antigens generated in vivo from bone marrow. Science 2000, 290, 1779–1782. [Google Scholar] [CrossRef] [Green Version]
  188. Liu, X.-Y.; Yang, L.-P.; Zhao, L. Stem cell therapy for Alzheimer’s disease. World J. Stem Cells 2020, 12, 787. [Google Scholar] [CrossRef]
  189. Lee, H.J.; Lee, J.K.; Lee, H.; Carter, J.E.; Chang, J.W.; Oh, W.; Yang, Y.S.; Suh, J.-G.; Lee, B.-H.; Jin, H.K. Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer’s disease mouse model through modulation of neuroinflammation. Neurobiol. Aging 2012, 33, 588–602. [Google Scholar] [CrossRef]
  190. Harach, T.; Jammes, F.; Muller, C.; Duthilleul, N.; Cheatham, V.; Zufferey, V.; Cheatham, D.; Lukasheva, Y.A.; Lasser, T.; Bolmont, T. Administrations of human adult ischemia-tolerant mesenchymal stem cells and factors reduce amyloid beta pathology in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2017, 51, 83–96. [Google Scholar] [CrossRef]
  191. Zhang, L.; Dong, Z.-f.; Zhang, J.-y. Immunomodulatory role of mesenchymal stem cells in Alzheimer’s disease. Life Sci. 2020, 246, 117405. [Google Scholar] [CrossRef]
  192. Munoz, J.R.; Stoutenger, B.R.; Robinson, A.P.; Spees, J.L.; Prockop, D.J. Human stem/progenitor cells from bone marrow promote neurogenesis of endogenous neural stem cells in the hippocampus of mice. Proc. Natl. Acad. Sci. USA 2005, 102, 18171–18176, Erratum in Proc. Natl. Acad. Sci. USA 2006, 103, 2000–2002. [Google Scholar] [CrossRef] [Green Version]
  193. Kan, I.; Barhum, Y.; Melamed, E.; Offen, D. Mesenchymal stem cells stimulate endogenous neurogenesis in the subventricular zone of adult mice. Stem Cell Rev. Rep. 2011, 7, 404–412. [Google Scholar] [CrossRef]
  194. Segal-Gavish, H.; Karvat, G.; Barak, N.; Barzilay, R.; Ganz, J.; Edry, L.; Aharony, I.; Offen, D.; Kimchi, T. Mesenchymal stem cell transplantation promotes neurogenesis and ameliorates autism related behaviors in BTBR mice. Autism Res. 2016, 9, 17–32. [Google Scholar] [CrossRef]
  195. Cova, L.; Armentero, M.; Zennaro, E.; Calzarossa, C.; Bossolasco, P.; Busca, G.; Lambertenghi Deliliers, G.; Polli, E.; Nappi, G.; Silani, V.; et al. Multiple neurogenic and neurorescue effects of human mesenchymal stem cell after transplantation in an experimental model of Parkinson’s disease. Brain Res. 2010, 1311, 12–27. [Google Scholar] [CrossRef]
  196. Bao, X.; Wei, J.; Feng, M.; Lu, S.; Li, G.; Dou, W.; Ma, W.; Ma, S.; An, Y.; Qin, C. Transplantation of human bone marrow-derived mesenchymal stem cells promotes behavioral recovery and endogenous neurogenesis after cerebral ischemia in rats. Brain Res. 2011, 1367, 103–113. [Google Scholar] [CrossRef]
  197. Volkman, R.; Offen, D. Concise review: Mesenchymal stem cells in neurodegenerative diseases. Stem Cells 2017, 35, 1867–1880. [Google Scholar] [CrossRef] [Green Version]
  198. Park, H.-J.; Shin, J.Y.; Lee, B.R.; Kim, H.O.; Lee, P.H. Mesenchymal stem cells augment neurogenesis in the subventricular zone and enhance differentiation of neural precursor cells into dopaminergic neurons in the substantia nigra of a parkinsonian model. Cell Transplant. 2012, 21, 1629–1640. [Google Scholar] [CrossRef]
  199. Sanchez-Ramos, J.; Song, S.; Cardozo-Pelaez, F.; Hazzi, C.; Stedeford, T.; Willing, A.; Freeman, T.; Saporta, S.; Janssen, W.; Patel, N. Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp. Neurol. 2000, 164, 247–256. [Google Scholar] [CrossRef] [Green Version]
  200. Nakano, M.; Nagaishi, K.; Konari, N.; Saito, Y.; Chikenji, T.; Mizue, Y.; Fujimiya, M. Bone marrow-derived mesenchymal stem cells improve diabetes-induced cognitive impairment by exosome transfer into damaged neurons and astrocytes. Sci. Rep. 2016, 6, 24805. [Google Scholar] [CrossRef] [Green Version]
  201. Börger, V.; Bremer, M.; Ferrer-Tur, R.; Gockeln, L.; Stambouli, O.; Becic, A.; Giebel, B. Mesenchymal stem/stromal cell-derived extracellular vesicles and their potential as novel immunomodulatory therapeutic agents. Int. J. Mol. Sci. 2017, 18, 1450. [Google Scholar] [CrossRef]
  202. Xiong, Y.; Mahmood, A.; Chopp, M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen. Res. 2017, 12, 19. [Google Scholar] [CrossRef]
  203. Yang, Y.; Ye, Y.; Su, X.; He, J.; Bai, W.; He, X. MSCs-derived exosomes and neuroinflammation, neurogenesis and therapy of traumatic brain injury. Front. Cell. Neurosci. 2017, 11, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Yu, Z.; Ling, Z.; Lu, L.; Zhao, J.; Chen, X.; Xu, P.; Zou, X. Regulatory Roles of Bone in Neurodegenerative Diseases. Front. Aging Neurosci. 2020, 12, 610581. [Google Scholar] [CrossRef] [PubMed]
  205. Xin, H.; Li, Y.; Buller, B.; Katakowski, M.; Zhang, Y.; Wang, X.; Shang, X.; Zhang, Z.G.; Chopp, M. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 2012, 30, 1556–1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Zhang, J.; Zhang, Z.G.; Lu, M.; Wang, X.; Shang, X.; Elias, S.B.; Chopp, M. MiR-146a promotes remyelination in a cuprizone model of demyelinating injury. Neuroscience 2017, 348, 252–263. [Google Scholar] [CrossRef] [PubMed]
  207. Kubota, K.; Nakano, M.; Kobayashi, E.; Mizue, Y.; Chikenji, T.; Otani, M.; Nagaishi, K.; Fujimiya, M. An enriched environment prevents diabetes-induced cognitive impairment in rats by enhancing exosomal miR-146a secretion from endogenous bone marrow-derived mesenchymal stem cells. PLoS ONE 2018, 13, e0204252. [Google Scholar] [CrossRef] [Green Version]
  208. Yuyama, K.; Sun, H.; Mitsutake, S.; Igarashi, Y. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-β by microglia. J. Biol. Chem. 2012, 287, 10977–10989. [Google Scholar] [CrossRef] [Green Version]
  209. Vatsa, P.; Negi, R.; Ansari, U.; Khanna, V.; Pant, A. Insights of Extracellular Vesicles of Mesenchymal Stem Cells: A Prospective Cell-Free Regenerative Medicine for Neurodegenerative Disorders. Mol. Neurobiol. 2022, 59, 459–474. [Google Scholar] [CrossRef]
  210. Barzilay, R.; Kan, I.; Ben-Zur, T.; Bulvik, S.; Melamed, E.; Offen, D. Induction of human mesenchymal stem cells into dopamine-producing cells with different differentiation protocols. Stem Cells Dev. 2008, 17, 547–554. [Google Scholar] [CrossRef]
  211. Liang, J.; Wu, S.; Zhao, H.; Li, S.-l.; Liu, Z.-x.; Wu, J.; Zhou, L. Human umbilical cord mesenchymal stem cells derived from Wharton’s jelly differentiate into cholinergic-like neurons in vitro. Neurosci. Lett. 2013, 532, 59–63. [Google Scholar] [CrossRef]
  212. Liu, X.; Li, D.; Jiang, D.; Fang, Y. Acetylcholine secretion by motor neuron-like cells from umbilical cord mesenchymal stem cells. Neural Regen. Res. 2013, 8, 2086. [Google Scholar]
  213. Brofiga, M.; Massobrio, P. Brain-on-a-Chip: Dream or Reality? Front. Neurosci. 2022, 16, 837623. [Google Scholar] [CrossRef]
  214. Kaur, A.; Nigam, K.; Tyagi, A.; Dang, S. A Preliminary Pharmacodynamic Study for the Management of Alzheimer’s Disease Using Memantine-Loaded PLGA Nanoparticles. AAPS PharmSciTech 2022, 23, 298. [Google Scholar] [CrossRef]
Figure 1. Bone morphogenetic proteins (BMPs) and the master gene of the osteoblastic commitment, RUNX2, play an important role in osteogenic differentiation. Further, the secretion of neuronal growth factors promotes neuronal commitment. Wnt/β-catenin signaling is a common pathway of these two different lines. In particular, for osteoblastic commitment, an increase in Park2 stimulates the activation of β-catenin. The expression of NR2F1 is downregulated in the DA neuron of PD patients carrying the LRRK2-G2019S mutation.
Figure 1. Bone morphogenetic proteins (BMPs) and the master gene of the osteoblastic commitment, RUNX2, play an important role in osteogenic differentiation. Further, the secretion of neuronal growth factors promotes neuronal commitment. Wnt/β-catenin signaling is a common pathway of these two different lines. In particular, for osteoblastic commitment, an increase in Park2 stimulates the activation of β-catenin. The expression of NR2F1 is downregulated in the DA neuron of PD patients carrying the LRRK2-G2019S mutation.
Cells 12 00051 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Minoia, A.; Dalle Carbonare, L.; Schwamborn, J.C.; Bolognin, S.; Valenti, M.T. Bone Tissue and the Nervous System: What Do They Have in Common? Cells 2023, 12, 51. https://doi.org/10.3390/cells12010051

AMA Style

Minoia A, Dalle Carbonare L, Schwamborn JC, Bolognin S, Valenti MT. Bone Tissue and the Nervous System: What Do They Have in Common? Cells. 2023; 12(1):51. https://doi.org/10.3390/cells12010051

Chicago/Turabian Style

Minoia, Arianna, Luca Dalle Carbonare, Jens Christian Schwamborn, Silvia Bolognin, and Maria Teresa Valenti. 2023. "Bone Tissue and the Nervous System: What Do They Have in Common?" Cells 12, no. 1: 51. https://doi.org/10.3390/cells12010051

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

Article Metrics

Back to TopTop