Next Article in Journal
Involvement of Orotic Acid in Mitochondrial Activity of Ovarian Granulosa Cells and Oocyte Meiotic Maturation
Previous Article in Journal
Reactions of Lanthanide Ions with Glycolic Acid or Tartaric Acid in the Presence of Spermine: Potentiometric and Spectroscopic Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Masticatory Force in Modulating Jawbone Immunity and Bone Homeostasis: A Review

1
Department of Orthodontics (WangFuJing Campus), School of Stomatology, Capital Medical University, Scylla alley No. 11, Beijing 100069, China
2
Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(10), 4478; https://doi.org/10.3390/ijms26104478
Submission received: 31 March 2025 / Revised: 4 May 2025 / Accepted: 6 May 2025 / Published: 8 May 2025
(This article belongs to the Section Molecular Immunology)

Abstract

:
Mastication exerts a significant influence on both the structural and immunological environment of the jawbone. The mechanical stress generated during chewing initiates bone remodeling through the coordinated activities of osteoclasts and osteoblasts, with these processes being modulated by immune cell responses. This review summarizes the interaction between masticatory forces and jawbone immunity, focusing on key mechanisms such as mechanotransduction in osteocytes, macrophage polarization, and the activation of T cells. The review also delves into the role of the receptor activator of nuclear factor κ-B ligand (RANKL), receptor activator of nuclear factor κ-B (RANK), and osteoprotegerin (OPG) signaling pathway, highlighting its critical function in bone resorption and immune regulation. Additionally, the review summarizes how masticatory forces modulate the immune response through changes in immune cells, particularly focusing on cytokines, and the involvement of hormonal and molecular pathways. These findings provide valuable insights into the complex interplay between mechanical forces and immune cells, with implications for bone health.

1. Introduction

Mastication, the process of chewing, plays a crucial role in maintaining not only the functional integrity of the oral cavity but also the structural health of the jawbone. The mechanical forces generated during chewing are essential for stimulating bone remodeling, a dynamic process that balances bone formation and resorption [1]. Beyond its biomechanical impact, mastication has significant effects on the immune environment of the jawbone. The jawbone is a unique site where immune cells are constantly exposed to mechanical stress and microbial challenges from the oral cavity, creating a complex interplay between bone cells and the immune system [2,3].
At the cellular level, osteocytes [4], osteoblasts [5], and osteoclasts [6] are key players in bone remodeling, responding to mechanical stimuli through mechanotransduction pathways [4,7,8,9,10]. These bone cells interact closely with immune cells, such as macrophages and T cells [11,12], in a bidirectional manner. Immune cells produce cytokines and signaling molecules that influence bone cell activity, while bone cells, in turn, modulate immune responses by secreting factors that affect immune cell function. Central to this dynamic interaction is the receptor activator of nuclear factor κ-B ligand (RANKL), receptor activator of nuclear factor κ-B (RANK), and osteoprotegerin (OPG) signaling axis, which not only governs osteoclast differentiation and activity but also links mechanical forces with immune modulation and bone resorption, illustrating the reciprocal influence between the skeletal and immune systems [13,14].
Understanding the relationship between mastication and the immune environment of the jawbone is critical, as this interaction affects bone health. This review aims to explore the mechanisms through which masticatory forces influence the immune response and bone remodeling, highlighting the roles of key cells and signaling pathways involved in this intricate process. By delving into these mechanisms, we can gain valuable insights into potential therapeutic strategies for improving jawbone health.

2. Mechanisms of Bone Homeostasis in the Jawbone

Bone homeostasis in the jawbone is a complex physiological process regulated by the interaction of various cell types, including bone cells and immune cells [3,15,16]. Unlike other skeletal structures, the jawbone is continuously exposed to mechanical stress and microbial challenges during mastication, requiring a finely tuned balance between bone formation and resorption. In addition to the activity of bone cells, the unique immune microenvironment of the jawbone, characterized by a higher concentration of immune cells, plays a critical role in maintaining homeostasis [2]. This coordinated response to mechanical forces and microbial threats ensures the dynamic regulation of bone remodeling and immune regulation within the jawbone.

2.1. Bone Cells in Jawbone Homeostasis

Bone remodeling in the jawbone is a finely tuned process involving the coordinated actions of various bone cells. These cells work together to regulate bone formation and resorption, maintaining skeletal balance.
Mesenchymal cells, such as those in the periodontal ligament, are key regulators of bone remodeling. They are major sources of RANKL, especially during inflammation, driving osteoclast formation and promoting bone resorption [15,17,18]. The RANKL/RANK/OPG axis plays a critical role here: RANKL secreted by mesenchymal cells binds to RANK on osteoclast precursors, promoting osteoclast differentiation and activity. OPG, which acts as a decoy receptor, helps inhibit this process, thus preserving bone mass during non-inflammatory conditions [19]. The balance between RANKL and OPG production by mesenchymal cells is crucial in regulating bone resorption and preventing pathological bone loss [20].
Osteocytes play a central role in bone metabolism by sensing mechanical stress and coordinating the activities of osteoblasts and osteoclasts to maintain homeostasis [4]. They are a significant source of RANKL, especially in response to mechanical unloading or microdamage [15], which drives osteoclast activity. Conversely, osteocytes also produce OPG, which acts as a decoy receptor for RANKL, inhibiting osteoclastogenesis and protecting against excessive bone resorption. The RANKL/OPG balance maintained by osteocytes is essential for regulating bone turnover and adapting to mechanical forces.
Osteoblasts are specialized cells essential for bone formation. They produce the bone matrix, which is primarily composed of collagen, and regulate the mineralization process [5]. Osteoblasts also express RANKL, a key factor in activating osteoclasts, the cells responsible for bone resorption. In inflammatory conditions, RANKL expression by osteoblasts increases, leading to heightened osteoclast activity and accelerated bone resorption [21]. To counterbalance this, osteoblasts produce OPG, which helps reduce bone resorption by inhibiting RANKL’s effects on osteoclasts [22].
Osteoclasts are the key cells responsible for bone resorption, breaking down bone tissue as part of the ongoing bone remodeling process [16]. Their activity is carefully controlled by the RANKL/RANK/OPG signaling pathway [23]. Inflammatory cytokines like tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 upregulate RANKL expression, which in turn promotes the formation and activation of osteoclasts, leading to increased bone resorption [14,24,25]. Periodontal ligament stem cells (PDLSCs) play a pivotal role in regulating osteoclast activity through multiple mechanisms, including the secretion of extracellular vesicles (EVs) [26] and apoptotic bodies (ABs) [27], as well as direct cellular interactions. EVs can carry mRNA, proteins, and enzymes that influence osteoclast differentiation and function [28]. ABs contain microRNAs like miR-223-3p, which target integrin beta 1 (Itgb1) to inhibit osteoclast activity and reduce bone resorption [27]. In inflammatory conditions such as periodontitis, PDLSC-derived EVs are enriched with RANKL and TNF-α, promoting osteoclast differentiation and activation [29]. Additionally, PDLSCs regulate osteoclast function through membrane-derived lipid mediators, which are essential for osteoclast morphology and activity [30].

2.2. Immune Cells in Jawbone Homeostasis

The jawbone, particularly the alveolar bone, possesses a unique immune microenvironment compared to other bones. This microenvironment is characterized by a higher concentration of mature immune cells that play a crucial role in responding to the constant mechanical and microbial challenges that arise during mastication [2]. These immune responses are essential for the process of bone remodeling, which involves balancing bone resorption and bone formation to adapt to the forces generated by chewing [3].
T cells are pivotal in regulating both bone resorption and regeneration, with different subsets playing distinct roles. RANK, the receptor for RANKL, is expressed not only on osteoclast precursors responsible for bone resorption but also on several immune cells, including T cells [31]. The RANKL/RANK signaling pathway affects the immune system by modulating the development and function of these immune cells. When RANKL binds to RANK on dendritic cells, it enhances their survival and ability to present antigens, which, in turn, activates T cells [32]. In bone metabolism, T helper 1 (Th1) cells produce cytokines such as IL-12 and Interferon (IFN)-γ, which inhibit osteoclast formation by suppressing RANKL and TNF-α pathways, both of which are essential for osteoclast development and activity [33]. On the other hand, T helper 17 (Th17) cells, a subset of CD4+ T cells, promote bone resorption by producing IL-17A, IL-17F, and other cytokines like IL-21 and IL-22 [34]. IL-17 increases RANKL expression on osteoblasts and other bone cells, thereby accelerating osteoclast formation, especially in inflammatory conditions [35,36]. Knocking out IL-17A and IL-17F has been shown to reduce bone loss, underscoring their role in driving inflammation-induced osteolysis [37].
In contrast, regulatory T cells (Tregs) help protect bone by inhibiting excessive immune activity. Through their production of IL-10 and transforming growth factor (TGF)-β, Tregs suppress Th17 cells and reduce RANKL expression, limiting osteoclast formation and bone resorption [11,38,39]. The balance between Th17 and Tregs is influenced by IL-6, which determines whether naive CD4+ T cells develop into pro-resorptive Th17 cells or bone-protective Tregs [40].
γδ T cells, a unique subset of T cells, are distinguished by their specific T-cell receptor (TCR), composed of γ and δ chains. Their role in bone biology is complex, as they produce IL-17A, a cytokine whose effects vary depending on the immune response phase and the surrounding microenvironment [41]. In conditions of chronic inflammation, IL-17A is linked to increased bone resorption. This occurs through the upregulation of RANKL expression on osteoblasts and stromal cells, which enhances osteoclast activation and differentiation, leading to bone breakdown. However, during the early stages of bone healing, γδ T cells’ production of IL-17A supports osteoblast proliferation and activity, promoting bone formation [35].
While certain B cell subsets contribute to bone loss in inflammatory conditions, other B cells can produce anti-inflammatory cytokines like IL-10, which help reduce inflammation and protect bone tissue [11,42,43]. For example, activated B cells in the jawbone significantly contribute to alveolar bone resorption, particularly in response to oral pathogen infections, by producing RANKL [44]. This was demonstrated in a mouse model of periodontitis, where B cell-specific conditional knockout mice revealed the essential role of RANKL-expressing effector B cells in pathogen-induced bone loss [44]. Additionally, B cells interact with osteoclasts and osteoblasts, influencing osteoclast differentiation and activity through the secretion of cytokines such as TNF and C-C chemokine motif ligand (CCL) 3 [45], further contributing to bone loss in inflammatory conditions. On the other hand, regulatory B cells (Bregs) play a crucial role in bone healing by modulating immune responses, particularly by suppressing proinflammatory cytokines and promoting the activity of Tregs [46], thereby helping to reduce inflammation and protect bone tissue.
Macrophages play a crucial role in bone remodeling, with pro-inflammatory macrophages (M1) promoting bone resorption by producing cytokines such as TNF-α, IL-1β, and IL-6, while anti-inflammatory (M2) macrophages support bone regeneration through the secretion of cytokines like TGF-β and IL-10 [11]. The switch from M1 to M2 macrophages is influenced by various factors, including Tregs, which promote the tissue-repairing M2 phenotype [47]. Jaw periosteal cells (JPCs) have been shown to further influence macrophage polarization [48]. In coculture systems, JPCs can induce a shift from M1 to M2 macrophages, promoting an anti-inflammatory environment through paracrine signaling, thereby supporting bone regeneration and tissue repair.
Neutrophils contribute to both bone destruction and repair [49]. During inflammation, they exacerbate bone damage by promoting osteoclast activity through the formation of neutrophil extracellular traps (NETs) and the production of RANKL-inducing cytokines like oncostatin M (OSM) [50]. NETs, formed when neutrophils release their chromatin and antimicrobial proteins into the extracellular space, serve to trap pathogens. However, these NETs also contribute to bone resorption by releasing pro-inflammatory cytokines, such as RANKL, and proteases like elastase, which activate osteoclasts. The formation of NETs triggers the production of reactive oxygen species (ROS), which activate key inflammatory signaling pathways like NF-κB, MAPK, and Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3). These pathways, in turn, amplify inflammation, promoting osteoclast differentiation, maturation, and bone resorption. Through the RANKL/RANK signaling axis, NETs enhance osteoclastogenesis by binding RANKL to the RANK receptor on osteoclast precursor cells, leading to increased bone degradation. However, neutrophils are also essential for initiating bone repair during the early stages of the inflammatory response. Additionally, neutrophils are influenced by RANKL, which stimulates their degranulation and migration [32], both essential for immune defense.
Dendritic cells (DCs), along with Langerhans cells, play a crucial role in regulating local immune responses and maintaining bone homeostasis [51]. Through the RANKL/RANK/OPG axis, RANKL enhances the survival and antigen presentation abilities of DCs [31,52], which can influence bone remodeling by activating the immune system. In the jawbone, DCs are particularly important in recognizing bacterial antigens and modulating immune responses [53], essential for maintaining oral health and preventing conditions such as periodontal disease. Additionally, research shows that JPCs can inhibit the maturation of DCs [54], a mechanism that could be beneficial in reducing excessive immune responses during tissue engineering and regenerative procedures.
Innate lymphoid cells (ILCs) play a significant role in both inflammation and tissue repair [55]. In the jawbone, ILC2s promote anti-inflammatory responses by secreting IL-5 and IL-13, which support tissue repair and activate M2 macrophages. They also help maintain the balance between bone resorption and formation [47,56]. Conversely, ILC3s produce IL-17 and IL-22 [57], which contribute to both protective immunity and inflammatory bone resorption.
Moreover, RANKL plays a role beyond immune cell interactions by contributing to the formation and maintenance of lymph nodes, which are central to immune system function [58]. This process is essential for the organization and operation of the immune response, as lymph nodes act as hubs for immune cell activation and interaction.

2.3. Mechanisms of Mechanical Stress in Bone Homeostasis

Mechanical stress is a key regulator of bone remodeling, particularly in response to masticatory forces. One of the primary mechanisms involved is fluid shear stress (FSS), which arises from the compressive forces applied to the lacunar–canalicular system during mastication. This shear stress affects osteocytes, which are embedded within the bone matrix and play a pivotal role in mechanotransduction. The compression of the lacunar–canalicular system leads to the activation of intracellular signaling pathways that modulate bone resorption and formation [59].
Mechanoreceptors such as Piezo1 and Piezo2 play a significant role in the response to mechanical loading. These ion channels are activated by tension in the plasma membrane and are crucial for detecting mechanical deformation in cells. Upon activation, Piezo receptors initiate intracellular signaling cascades that influence both bone and immune cell activity, affecting bone remodeling and immune responses [60]. The presence of Piezo1 and Piezo2 mechanoreceptors in various bone and immune cells highlights their essential role in translating mechanical stimuli into biological responses that regulate bone homeostasis and immune regulation [61].
Furthermore, the cytoskeleton plays a crucial role in translating mechanical forces into cellular responses. When cells are subjected to mechanical loading, deformation of the cell membrane occurs, which in turn impacts the structure and function of the cytoskeleton. Focal adhesions and integrins within the cytoskeleton serve as mechanical signal transducers, converting physical forces into biochemical signals. This process is essential for the activation of various signaling pathways, such as the RANKL/RANK/OPG pathway, which regulates osteoclast differentiation and bone resorption [62].
While masticatory forces contribute to the regulation of bone homeostasis, excessive masticatory forces have been shown to cause significant mechanical damage to the jawbone. Overloading the jaw during chewing generates excessive stress on the bone tissue, leading to structural damage such as fractures, bone resorption, or even bone degradation over time [63]. Osteocytes, which are embedded within the bone matrix, are particularly sensitive to such excessive mechanical forces. When masticatory forces exceed the bone’s tolerance, osteocytes detect this damage and activate the bone remodeling process. Osteoclasts are recruited to resorb damaged bone, while osteoblasts are stimulated to generate new bone, repairing and restoring the structural integrity of the jawbone [64].

3. The Impact of Masticatory Forces on Immune Cells in the Jawbone

Masticatory forces not only affect the structural integrity of the jawbone but also play a critical role in influencing immune cell activity, for both innate immune cells and adaptive immune cells [65,66,67] (Figure 1). These forces cause mechanical damage to the gingival barrier, which leads to the release of IL-6 from epithelial cells, fibroblasts, osteoblasts, and skeletal muscle fibers [68]. This IL-6 release promotes the accumulation and function of Th17 cells, which are integral to the immune response in the oral cavity [12]. Additionally, the stiffness of the surrounding environment influences T cell activation and proliferation. Studies using engineered 3D hydrogels have shown that T cells in stiffer environments exhibit greater activation and higher production of cytokines such as IL-2, IFN-γ, and TNF-α compared to softer environments [69].
Mechanical forces also directly activate T cells. Studies using synthetic nanomotors have shown that mechanical forces can open cellular Ca2+ channels, triggering T cell activation [70]. This suggests that mechanical stress from mastication could similarly activate T cells in the jawbone, contributing to bone remodeling. Mechanical stress further enhances the expression of T cell-associated Th1 cytokines such as TNF-α and IFN-γ within periodontal tissues, promoting osteoclast activation and facilitating alveolar bone remodeling [65]. Inhibiting TNF-α has been shown to reduce the stimulatory effects of T cells, further underscoring the role of these cytokines in T cell-mediated bone remodeling.
In orthodontic tooth movement (OTM), mechanical forces increase the ratio of CD4+/CD3+ T cells in the alveolar bone, correlating with elevated expression of Th1 cytokines like TNF-α and IFN-γ, which promote bone resorption [64]. Similarly, γδ T cells produce IL-17A, which recruits monocytes and neutrophils, driving osteoclast activation [67]. Since both orthodontic and masticatory forces apply mechanical stress to the jawbone, mastication likely influences T cell activity in a similar way.
Macrophages, which are crucial for bone remodeling, respond to mechanical forces such as those generated during chewing by regulating the activity of osteoclasts and osteoblasts [66]. These mechanical forces influence macrophage polarization, shifting them between M1 and M2 states. Masticatory forces, akin to mechanical stretching, promote the polarization of M2 macrophages, which are essential for bone regeneration [71]. Mechanical stimuli, such as shear stress, compression, and tensile forces, are detected by macrophages through mechanosensors like integrins, ion channels, and other mechanoreceptors. These mechanosensitive molecules translate mechanical signals into biochemical signals, activating intracellular pathways that dictate macrophage function. When macrophages are exposed to mechanical forces, they initiate various signaling cascades. For example, shear stress activates the ERK1/2 signaling pathway in PDLSCs, which enhances the secretion of immunomodulatory molecules such as TGF-β1 and IL-10 [72,73,74,75]. These molecules promote the M2 polarization of macrophages. Additionally, integrins play a central role in mechanosensing by linking the extracellular matrix to the intracellular cytoskeleton. The activation of integrins in response to mechanical forces leads to the activation of downstream signaling pathways, including the NF-κB and MAPK pathways, which are critical in modulating the immune response and macrophage polarization. The mechanical forces also affect macrophage polarization indirectly through the release of extracellular vesicles, such as exosomes, from activated macrophages. These exosomes contain bioactive molecules, including ubiquitin carboxyl-terminal hydrolase isozyme L3 (UCHL3) [75], that further promote osteogenesis and contribute to the regeneration of alveolar bone. Furthermore, the mechanical environment influences macrophage behavior through the modulation of the extracellular matrix (ECM). In response to mechanical stress, the ECM undergoes remodeling, which in turn affects macrophage function. This remodeling process can provide physical cues that guide macrophage differentiation toward the M2 phenotype, further supporting tissue regeneration and bone homeostasis. M2 macrophages enhance osteogenesis in bone marrow mesenchymal stem cells (BMSCs) by producing anti-inflammatory factors like IL-10 and TGF-β1 [72,73,74,75]. Additionally, chewing stimulates the release of exosomes from activated macrophages, which contain factors such as ubiquitin carboxyl-terminal hydrolase isozyme L3 (UCHL3) [75,76]. These exosomes further promote osteogenesis in BMSCs and contribute to alveolar bone formation. This process demonstrates how macrophages, through both direct cytokine production and exosome-mediated signaling, respond to mechanical stress to support bone regeneration and maintain bone health. M2 macrophages also contribute to angiogenesis by increasing the expression of growth factors such as vascular endothelial growth factor (Vegfa) and placental growth factor (Pigf), promoting effective bone remodeling [73].
The Piezo1 ion channel, a key mechanosensor in macrophages, is upregulated by mechanical forces such as mastication, activating the protein kinase B (AKT)/glycogen synthase kinase 3 β (GSK3β) signaling pathway, which promotes macrophage proliferation and supports bone regeneration [72]. During orthodontic tooth movement, compressive forces also activate the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, driving osteoclast differentiation and alveolar bone resorption [77]. Given that chewing exerts similar mechanical stress on the jawbone, it is likely that mastication influences these pathways in a similar manner.
Finally, interactions between immune cells, particularly monocytes and macrophages, and MSCs are more pronounced in the jawbone compared to in long bones. This unique immune microenvironment underscores the complex communication between immune cells and bone cells in response to mechanical forces [2].

4. The Regulatory Effects of Masticatory Forces on Bone Cells in the Jawbone

Masticatory forces play a critical role in regulating bone remodeling by activating mechanotransduction pathways in osteocytes, osteoblasts, and osteoclasts. These mechanical signals influence both bone formation and resorption, ensuring the jawbone adapts to the functional demands of chewing while maintaining structural integrity (Figure 1).

4.1. Mechanotransduction and Immune Response in Osteocytes

Mastication plays a critical role in regulating bone formation and resorption through its influence on osteocyte activity, which is key to maintaining jawbone health. Osteocytes, the most abundant bone cells, are crucial for mechanosensation—a process where these cells detect mechanical forces such as those produced by chewing [7,8,9]. Upon detecting mechanical loading, osteocytes activate gene expression and produce proteins that contribute to bone remodeling, either promoting bone formation or triggering bone resorption [78].
The mechanotransduction process in osteocytes is triggered by mechanical stress, leading to plasma membrane disruptions (PMDs), activation of integrin-based structures, and involvement of the lacunar–canalicular system (LCS). Studies by Yu Kanglun et al. demonstrate that mechanical loading disrupts the plasma membranes of osteocytes, which initiates calcium signaling and other pathways that promote bone adaptation [10]. This response is further supported by specialized structures like β3 integrin sites on osteocyte processes, which are particularly sensitive to mechanical strain, allowing osteocytes to effectively sense and respond to these forces [79]. Mastication also exerts mechanical forces such as compression, stretching, and bending on the jawbone, creating FSS in the intraosseous fluid within the LCS [80,81,82]. This process is crucial for osteocytes to sense mechanical load. Osteocytes, connected through dendrites to form a complete network within the bone matrix, detect FSS and other stimuli, including matrix stiffness and hydrostatic pressure [8,83,84]. These mechanical signals are transmitted via protein secretion and cell dendrites [85], contributing to bone remodeling. Further computational research suggests that the poroelastic properties of bone cells significantly affect their response to compressive loads, influencing both strain distribution and fluid flow within the bone matrix [86].
Kawakami et al.’s studies revealed that masticatory forces affect osteocyte micromorphology and bone lacunae characteristics. For example, rats fed a solid diet developed more complex osteocyte connectivity and larger bone lacunae compared to those on a powdered diet [85]. This indicates that chewing increases the structural complexity of osteocytes and enhances bone lacunae expansion. Additionally, Dentin matrix protein (DMP)-1, an important marker for osteocyte activity and bone calcification, showed higher localization in the solid feed group, suggesting that mastication significantly influences bone calcification [85].
The Wingless/Int-1 (Wnt)/β-catenin and Yes-associated protein (YAP)/Transcriptional co-activator with PDZ-binding motif (TAZ) pathways are both pivotal in the regulation of bone remodeling and mechanotransduction in osteocytes under mechanical loading. The Wnt/β-catenin pathway is central to maintaining bone mass, as its activation promotes osteoblastogenesis [87]. However, this pathway can be impaired by factors such as estrogen deficiency, as observed in ovariectomized mouse models [88]. Similarly, the YAP/TAZ pathway is activated by mechanical loading and regulates the expression of mechanosensitive genes and chemokines, which are essential for bone remodeling [89].
Further research has shown that increased masticatory activity, such as chewing a hard diet, suppresses sclerostin, a protein that inhibits bone formation, and induces the expression of Insulin-like growth factor (IGF)-1 in osteocytes [90]. IGF-1 receptor expression is also upregulated in chondroblasts due to enhanced muscle activity during mastication [91], which leads to thickening of the articular cartilage and promotes jawbone development [90]. Mechanical loading also stimulates the production of IL-6 in osteocytes, which activates JAK/STAT3 and extracellular signal-regulated kinase (ERK) signaling pathways. These pathways promote bone formation while inhibiting bone resorption [92,93].
Mastication further influences the immune environment of the jawbone by upregulating chemokines like C-X-C motif chemokine ligand (CXCL) 1 and CXCL2 in response to mechanical forces [94]. These chemokines promote the proliferation of bone cells, while other signaling molecules, such as macrophage colony-stimulating factor (M-CSF), CXCL1, CXCL2, CXCL3, CXCL9, and CXCL10, are expressed through the YAP/TAZ pathway, playing a significant role in osteocyte mechanotransduction and overall bone health [89]. Additionally, mechanical loading stimulates osteocytes to release nitric oxide (NO), prostaglandins (PGE2), and adenosine triphosphate (ATP), increasing the expression of osteogenic markers such as osterix (OSX) and alkaline phosphatase (ALP), which enhance osteoblast differentiation and bone mineralization [95,96].
Studies by Lohberger et al. show that cyclic mechanical stimulation, similar to that experienced during chewing, increases the expression of osteogenesis-specific markers, including type-I collagen and bone morphogenetic protein-2 in human intraoral mesenchymal stromal and progenitor cells [97]. This highlights the direct impact of mechanical forces, like mastication, on bone cell function and development.

4.2. Mechanotransduction and Immune Response in Osteoblastes

Mechanical stress significantly influences jawbone metabolism by affecting the activities of osteoblasts, the cells responsible for bone formation, and osteoclasts, which are involved in bone resorption. Specifically, mechanical stress enhances osteoblast activity through pathways involving key proteins such as glucose transporter (Glut) 1, sirtuin (SIRT) 1, and the transcription factor Runt-related transcription factor (Runx) 2, leading to increased bone formation [98,99].
Increased masticatory force activates osteocytes, the bone cells that act as mechanosensors. These activated osteocytes modify the expression of signaling molecules to promote the formation of osteoblasts. Specifically, they upregulate IGF-1 and suppress sclerostin, thereby enhancing the differentiation of osteoblasts from precursor cells [90].
Intermittent compressive force also stimulates osteoblast differentiation through the Wnt/β-catenin signaling pathway. This process is mediated by the release of ATP from the cells, which increases the expression of genes involved in bone formation and promotes mineralization [96]. Additionally, mechanical stress activates enzymes called matrix metalloproteinases (MMPs), particularly MMP-2, MMP-13, and MT1-MMP [100]. These enzymes are essential for osteoblast differentiation, facilitating the expression of important bone formation markers and contributing to the mineralization of the bone matrix.
Masticatory force also influences osteoblastic activity in the jaw by altering the expression of key signaling molecules such as RANKL and OPG. Mechanical stress shifts the balance toward increased RANKL expression, promoting bone remodeling through the formation of osteoclasts in a process known as osteoclastogenesis. This leads to bone resorption by osteoclasts [101], followed by new bone formation by osteoblasts [102]. Furthermore, mechanical stress enhances RANKL expression via the p38 MAPK pathway, underscoring the essential role of chewing forces in regulating osteoblast activity and maintaining bone health in the jaw [103,104].
Osteoblasts respond differently depending on the magnitude of compressive stress applied. At optimal stress levels, osteoblast differentiation is enhanced, evidenced by increased levels of bone formation markers such as Runx2 and ALP. However, excessive stress can inhibit the ability of osteoblasts to regulate the formation of osteoclasts [105].

4.3. Mechanotransduction and Immune Response in Osteoclastes

Mechanical forces, such as masticatory force, regulate osteoclast activity through a signaling pathway mediated by cementocytes. Under force loading, cementocytes facilitate osteoclastogenesis via the sphingosine-1-phosphate (S1P)/sphingosine-1-phosphate receptor (S1PR) 1/ras-related C3 botulinum toxin substrate 1 (Rac1) axis [106,107,108,109], where increased compression leads to higher synthesis and release of S1P, amplifying RANKL production and promoting osteoclast differentiation.
Moreover, masticatory force also exerts significant influence on osteoclast differentiation and macrophage activity through the actions of PDLSCs [110,111,112,113]. Under mechanical force, PDLSCs secrete exosomes that modulate immune responses, notably by suppressing IL-1β production through inhibition of the NF-κB signaling pathway in macrophages [112]. This mechanical stimulation also alters the exosomal proteome, increasing levels of annexin A3 (ANXA3), which enhances exosome uptake and activates the ERK pathway, promoting osteoclast differentiation [114]. Furthermore, force-induced production of hydrogen sulfide (H2S) by PDLSCs is associated with the secretion of monocyte chemoattractant protein (MCP)-1 and regulation of the RANKL/OPG system, both of which are critical for macrophage migration and osteoclast differentiation, key processes in bone remodeling [115,116,117].
In addition, masticatory force enhances osteoclastogenesis by bone marrow macrophages (BMMs) through the activation of colony-stimulating factor 1 receptor (CSF1R) signaling [118]. This process promotes osteoclast differentiation induced by TNF-α in the presence of M-CSF, independent of RANKL [119,120]. Notably, this highlights the crucial role of compressive force in driving osteoclastogenesis, even in the absence of mechanosensitive cells such as osteoblasts and fibroblasts.
While mechanical forces can promote osteoclastogenesis through enhanced RANKL production, other studies have demonstrated that increased masticatory force can actually diminish osteoclast activity by modulating the RANKL/OPG ratio. Specifically, increased masticatory force influenced the expression of essential proteins within the alveolar bone. Notably, the experimental group exhibited a substantial rise in OPG and mechano-growth factor (MGF) levels, alongside a marked reduction in RANKL. This led to a decreased RANKL/OPG ratio, diminishing osteoclast activity, which can help in maintaining or increasing bone density and is advantageous for bone health [121].

5. Broader Impacts of Masticatory Forces on Jawbone Immunity and Remodeling

Recent studies have revealed that masticatory forces influence the jawbone’s immune environment and bone remodeling not only through direct mechanical stimuli but also through hormonal interactions, non-coding RNA regulation, and muscle-bone communication (Figure 2). These mechanisms highlight the complexity of how chewing affects overall jawbone health, extending beyond basic bone cell activity to include broader systemic effects.

5.1. Hormonal Modulation of Jawbone Immunity by Masticatory Forces

Recent studies have shown that chewing affects immune function by altering hormone levels [122]. During fasting, leptin, a hormone that regulates immune cell activity [123], typically decreases. Meanwhile, corticosterone, a stress hormone known to suppress immune cell growth and induce cell death [124], increases. Research by Yang et al. found that chewing stimulation in fasting mice reduced the rise in corticosterone levels without affecting the decline in leptin. This suggests that chewing can mitigate stress-related immune suppression. Furthermore, chewing was observed to enhance antibody production after immunization, indicating that it may strengthen immune responses under fasting conditions by counteracting the effects of stress hormones [125].

5.2. Influence of Masticatory Forces on Jawbone Immunity Through Non-Coding RNA Regulation

Mechanical stress from chewing has been shown to influence bone remodeling by regulating small RNA molecules that control gene expression. MicroRNAs (miRNAs) are small, non-coding RNA molecules that regulate genes after transcription by binding to target messenger RNAs, leading to their degradation or inhibition of translation. They play critical roles in various biological processes, including bone remodeling [1].
Under compressive forces, specific miRNAs are either increased or decreased to mediate bone cell responses. For example, miR-494-3p levels increase under compressive stress and inhibit the proliferation of osteoblasts by targeting key genes essential for bone differentiation, such as FGFR2 and ROCK1 [126]. Similarly, miR-29 regulates components of the extracellular matrix, affecting bone matrix formation [127]. These miRNAs help balance the activity between osteoblasts and osteoclasts, driving bone resorption and remodeling in response to mechanical forces.
Mechanical stress also affects bone remodeling through long non-coding RNAs (lncRNAs), which are longer RNA molecules that do not code for proteins but regulate gene expression [128]. In response to mechanical stimuli like compressive forces, the lncRNA H19 is upregulated and plays a crucial role in the differentiation of mesenchymal stem cells into bone cells [129]. H19 primarily influences the Notch signaling pathway, essential for bone development and remodeling. Any imbalance in H19 expression can disrupt normal bone formation, highlighting the important regulatory function of lncRNAs in maintaining proper bone remodeling dynamics.
These findings underscore the complexity of how mechanical stress from chewing influences the immunological environment of the jawbone. By regulating non-coding RNAs like miRNAs and lncRNAs, mechanical forces modulate gene expression, affecting bone remodeling and immune responses in the jaw.

5.3. Muscle–Bone Interaction Mediated by Masticatory Forces

Over the past decade, the concept of muscle–bone crosstalk has gained significant attention. This interaction encompasses not only the mechanical forces generated during muscle contraction but also biochemical signals mediated by soluble molecules [130]. The masseter muscle, a primary muscle involved in chewing, has been identified as both a source of mechanical force affecting the jawbone’s immune environment through mechanotransduction and as a secretory organ influencing mandibular bone osteoimmunology [131,132].
In the mandible, this interaction is particularly crucial due to the close anatomical relationship between the masseter muscle and the jawbone. Muscle-derived signaling molecules, known as myokines, play a key role in communication between skeletal muscle and bone. These myokines include myostatin, brain-derived neurotrophic factor (BDNF), irisin, IGF, fibroblast growth factor (FGF)-2, β-aminoisobutyric acid (BAIBA), and several interleukins such as IL-6, IL-7, IL-8, and IL-15 [133,134,135,136]. For example, myostatin directly influences bone cell behavior by promoting the formation of osteoclasts, the cells responsible for bone resorption, through RANKL signaling. It also inhibits the differentiation of osteoblasts, the cells responsible for bone formation, by altering the content of exosomes released from osteocytes, which are mature bone cells [137,138]. Additionally, IGF-1, produced in skeletal muscle, is significantly increased following exercise [139]. This increase enhances muscle–bone interaction and affects the expression of osteoclasts and osteoblasts [140].
Conversely, bone-derived signaling molecules, known as osteokines, contribute to this bidirectional communication. Osteokines such as RANKL, osteocalcin, sclerostin, PGE2, TGF-β, Wnt3a, and FGF-23 play roles in regulating both local bone processes and systemic physiological functions. These molecules further emphasize the complexity of muscle–bone interactions and their impact on overall skeletal health [141,142,143,144].

6. Conclusions

In summary, the intricate interplay between mastication and the immune environment of the jawbone represents a critical focus within the field of osteoimmunology. This review underscores the complex mechanisms through which masticatory forces drive bone remodeling and modulate immune responses, with particular emphasis on the roles of key bone cells and immune cells. Central to this process is the RANKL/RANK/OPG signaling pathway, which regulates the balance between bone formation and resorption in response to mechanical stress. Moreover, masticatory forces exert systemic effects, influencing immune regulation through hormonal pathways and the modulation of non-coding RNAs. A deeper understanding of these interactions enhances our comprehension of jawbone health and offers potential avenues for the development of targeted therapies for conditions such as periodontal disease.
However, despite the insights provided, several challenges remain, such as the detrimental effects of excessive masticatory forces, which can lead to bone damage and inflammation. Addressing these issues requires further exploration into the specific molecular pathways involved and the identification of effective preventive or therapeutic interventions. For instance, developing mechanical force-modulating devices or exploring pharmacological approaches that target key signaling molecules within the RANKL/RANK/OPG pathway could offer solutions to mitigate excessive bone resorption or enhance bone formation. Additionally, the role of non-coding RNAs in immune modulation presents a promising area for the development of novel treatments. By advancing research in these areas, we may uncover strategies for preserving bone integrity and immune homeostasis in the jaw, contributing to better management of periodontal disease and other related conditions.

Author Contributions

Conceptualization, Y.L. and L.G.; Literature Collection, Y.S.; Writing—Original Draft Preparation, Y.S.; Writing—Review & Editing, Y.L. and L.G.; Supervision, Y.J.; Funding Acquisition, Y.L. and L.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Key R&D Program of China (Grant NO. 2022YFC2504200), the National Nature Science Foundation of China (82470936, 82201053), Young Clinical Research Fund of the Chinese Stomatological Association (CSA-SIS2022-11), the Beijing Municipal Administration of Hospitals Incubating Program (PX2023054), the Beijing Stomatological Hospital, Capital Medical University Young Scientist Program (YSP202105), Beijing Municipal Administration of Hospitals’ Youth Programme (QML20231506).

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

RANKLreceptor activator of nuclear factor κ-B ligand
RANKreceptor activator of nuclear factor κ-B
OPGosteoprotegerin
TNFtumor necrosis factor
ILinterleukin
PDLSCsperiodontal ligament stem cells
EVsextracellular vesicles
ABsapoptotic bodies
Itgb1integrin beta 1
Th1T helper 1 cells
IFNinterferon
Th17T helper 17 cells
Tregsregulatory T cells
TGFtransforming growth factor
TCRT-cell receptor
CCLC-C chemokine motif ligand
M1pro-inflammatory macrophages
M2anti-inflammatory macrophages
JPCsjaw periosteal cells
NETsneutrophil extracellular traps
ROSreactive oxygen species
OSMoncostatin M
DCsdendritic cells
ILCsinnate lymphoid cells
OTMorthodontic tooth movement
BMSCsbone marrow mesenchymal stem cells
UCHL3ubiquitin carboxyl-terminal hydrolase isozyme L3
Vegfavascular endothelial growth factor
Pigfplacental growth factor
AKTprotein kinase B
GSK3βglycogen synthase kinase 3 β
NLRP3NOD-like receptor family pyrin domain containing 3
PMDplasma membrane disruptions
LCSlacunar-canalicular system
FSSfluid shear stress
DMPdentin matrix protein
WntWingless/Int-1
YAPyes-associated protein
TAZtranscriptional co-activator with PDZ-binding motif
IGFinsulin-like growth factor
JAKjanus kinase
STAT3signal transducer and activator of transcription 3
ERKextracellular signal-regulated kinase
CXCLC-X-C motif chemokine ligand
M-CSFmacrophage colony-stimulating factor
NOnitric oxide
PGE2prostaglandins
ATPadenosine triphosphate
OSXosterix
ALPalkaline phosphatase
Glutglucose transporter
SIRTsirtuin
Runxrunt-related transcription factor
S1Psphingosine-1-phosphate
MMPsmatrix metalloproteinases
Rac1ras-related C3 botulinum toxin substrate 1
ANXA3annexin A3
H2Shydrogen sulfide
MCPmonocyte chemoattractant protein
BMMsbone marrow macrophages
CSF1Rcolony-stimulating factor 1 receptor
MGFmechano-growth factor
miRNAsmicroRNAs
lncRNAslong non-coding RNAs
BDNFbrain-derived neurotrophic factor
FGFFibroblast growth factor
BAIBAβ-aminoisobutyric acid

References

  1. Wang, Y.; Jia, L.; Zheng, Y.; Li, W. Bone remodeling induced by mechanical forces is regulated by miRNAs. Biosci. Rep. 2018, 38, BSR20180448. [Google Scholar] [CrossRef] [PubMed]
  2. Lin, W.; Li, Q.; Zhang, D.; Zhang, X.; Qi, X.; Wang, Q.; Chen, Y.; Liu, C.; Li, H.; Zhang, S.; et al. Mapping the immune microenvironment for mandibular alveolar bone homeostasis at single-cell resolution. Bone Res. 2021, 9, 17. [Google Scholar] [CrossRef] [PubMed]
  3. Lin, W.-M.; Quan, Y. Latest research findings on immune microenvironment regulation in jawbone-related diseases. J. Sichuan Univ. (Med. Sci.) 2022, 53, 528–531. [Google Scholar]
  4. Bonewald, L.F. The amazing osteocyte. J. Bone Miner. Res. 2011, 26, 229–238. [Google Scholar] [CrossRef]
  5. Zarrer, J.; Taipaleenmäki, H. The osteoblast in regulation of tumor cell dormancy and bone metastasis. J. Bone Oncol. 2024, 45, 100597. [Google Scholar] [CrossRef]
  6. Del Fattore, A.; Teti, A.; Rucci, N. Bone cells and the mechanisms of bone remodelling. Front. Biosci. Elite 2012, 4, 2302–2321. [Google Scholar] [CrossRef]
  7. Lara-Castillo, N.; Kim-Weroha, N.A.; Kamel, M.A.; Javaheri, B.; Ellies, D.L.; Krumlauf, R.E.; Thiagarajan, G.; Johnson, M.L. In vivo mechanical loading rapidly activates β-catenin signaling in osteocytes through a prostaglandin mediated mechanism. Bone 2015, 76, 58–66. [Google Scholar] [CrossRef]
  8. Ethier, C.R.; Simmons, C.A. Introductory Biomechanics: From Cells to Organisms; Cambridge University Press: Cambridge, UK, 2007. [Google Scholar]
  9. Uda, Y.; Azab, E.; Sun, N.; Shi, C.; Pajevic, P.D. Osteocyte mechanobiology. Curr. Osteoporos. Rep. 2017, 15, 318–325. [Google Scholar] [CrossRef]
  10. Yu, K.; Sellman, D.P.; Bahraini, A.; Hagan, M.L.; Elsherbini, A.; Vanpelt, K.T.; Marshall, P.L.; Hamrick, M.W.; McNeil, A.; McNeil, P.L.; et al. Mechanical loading disrupts osteocyte plasma membranes which initiates mechanosensation events in bone. J. Orthop. Res. 2018, 36, 653–662. [Google Scholar] [CrossRef]
  11. Gruber, R. Osteoimmunology: Inflammatory osteolysis and regeneration of the alveolar bone. J. Clin. Periodontol. 2019, 46, 52–69. [Google Scholar] [CrossRef]
  12. Dutzan, N.; Abusleme, L.; Bridgeman, H.; Greenwell-Wild, T.; Zangerle-Murray, T.; Fife, M.E.; Bouladoux, N.; Linley, H.; Brenchley, L.; Wemyss, K.; et al. On-going mechanical damage from mastication drives homeostatic Th17 cell responses at the oral barrier. Immunity 2017, 46, 133–147. [Google Scholar] [CrossRef] [PubMed]
  13. O’Brien, C.A. Control of RANKL gene expression. Bone 2010, 46, 911–919. [Google Scholar] [CrossRef] [PubMed]
  14. Udagawa, N.; Koide, M.; Nakamura, M.; Nakamichi, Y.; Yamashita, T.; Uehara, S.; Kobayashi, Y.; Furuya, Y.; Yasuda, H.; Fukuda, C.; et al. Osteoclast differentiation by RANKL and OPG signaling pathways. J. Bone Miner. Metab. 2021, 39, 19–26. [Google Scholar] [CrossRef]
  15. Graves, D.T.; Alshabab, A.; Albiero, M.L.; Mattos, M.; Corrêa, J.D.; Chen, S.; Yang, Y. Osteocytes play an important role in experimental periodontitis in healthy and diabetic mice through expression of RANKL. J. Clin. Periodontol. 2018, 45, 285–292. [Google Scholar] [CrossRef]
  16. Eriksen, E.F. Cellular mechanisms of bone remodeling. Rev. Endocr. Metab. Disord. 2010, 11, 219–227. [Google Scholar] [CrossRef]
  17. Pacios, S.; Xiao, W.; Mattos, M.; Lim, J.; Tarapore, R.S.; Alsadun, S.; Yu, B.; Wang, C.Y.; Graves, D.T. Osteoblast lineage cells play an essential role in periodontal bone loss through activation of nuclear factor-kappa B. Sci. Rep. 2015, 5, 16694. [Google Scholar] [CrossRef]
  18. Yang, C.Y.; Jeon, H.H.; Alshabab, A.; Lee, Y.J.; Chung, C.H.; Graves, D.T. RANKL deletion in periodontal ligament and bone lining cells blocks orthodontic tooth movement. Int. J. Oral Sci. 2018, 10, 3. [Google Scholar] [CrossRef]
  19. Simonet, W.S.; Lacey, D.L.; Dunstan, C.R.; Kelley, M.C.; Chang, M.S.; Lüthy, R.; Nguyen, H.Q.; Wooden, S.; Bennett, L.; Boone, T.; et al. Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell 1997, 89, 309–319. [Google Scholar] [CrossRef]
  20. Kostenuik, P.J. Osteoprotegerin and RANKL regulate bone resorption, density, geometry and strength. Curr. Opin. Pharmacol. 2005, 5, 618–625. [Google Scholar] [CrossRef]
  21. Liang, X.; Hou, Y.; Han, L.; Yu, S.; Zhang, Y.; Cao, X.; Yan, J. ELMO1 regulates RANKL-stimulated differentiation and bone resorption of osteoclasts. Front. Cell Dev. Biol. 2021, 9, 702916. [Google Scholar] [CrossRef]
  22. Zhao, X.; Patil, S.; Xu, F.; Lin, X.; Qian, A. Role of biomolecules in osteoclasts and their therapeutic potential for osteoporosis. Biomolecules 2021, 11, 747. [Google Scholar] [CrossRef] [PubMed]
  23. Elson, A.; Anuj, A.; Barnea-Zohar, M.; Reuven, N. The origins and formation of bone-resorbing osteoclasts. Bone 2022, 164, 116538. [Google Scholar] [CrossRef] [PubMed]
  24. Jimi, E.; Akiyama, S.; Tsurukai, T.; Okahashi, N.; Kobayashi, K.; Udagawa, N.; Nishihara, T.; Takahashi, N.; Suda, T. Osteoclast differentiation factor acts as a multifunctional regulator in murine osteoclast differentiation and function. J. Immunol. 1999, 163, 434–442. [Google Scholar] [CrossRef] [PubMed]
  25. Lin, A.; Kitaura, H.; Ohori, F.; Noguchi, T.; Marahleh, A.; Ma, J.; Ren, J.; Miura, M.; Fan, Z.; Narita, K.; et al. (D-Ala2) GIP Inhibits Inflammatory Bone Resorption by Suppressing TNF-α and RANKL Expression and Directly Impeding Osteoclast Formation. Int. J. Mol. Sci. 2024, 25, 2555. [Google Scholar] [CrossRef]
  26. Liu, C.; Li, Y.; Han, G. Advances of mesenchymal stem cells released extracellular vesicles in periodontal bone remodeling. DNA Cell Biol. 2022, 41, 935–950. [Google Scholar] [CrossRef]
  27. Li, X.; Jiang, Y.; Liu, X.; Fu, J.; Du, J.; Luo, Z.; Xu, J.; Bhawal, U.K.; Liu, Y.; Guo, L.; et al. Mesenchymal stem cell-derived apoptotic bodies alleviate alveolar bone destruction by regulating osteoclast differentiation and function. Int. J. Oral Sci. 2023, 15, 51. [Google Scholar] [CrossRef]
  28. Muraca, M.; Cappariello, A. The role of extracellular vesicles (EVs) in the epigenetic regulation of bone metabolism and osteoporosis. Int. J. Mol. Sci. 2020, 21, 8682. [Google Scholar] [CrossRef]
  29. Dai, Z.; Zheng, W.; Li, S. Receptor activator of nuclear factor-κB ligand and tumor necrosis factor-α promotes osteoclast differentiation through the exosomes of inflammatory periodontal ligament stem cells. Hua Xi Kou Qiang Yi Xue Za Zhi = Huaxi Kouqiang Yixue Zazhi = West China J. Stomatol. 2022, 40, 377–385. [Google Scholar]
  30. Oikawa, T.; Kuroda, Y.; Matsuo, K. Regulation of osteoclasts by membrane-derived lipid mediators. Cell. Mol. Life Sci. 2013, 70, 3341–3353. [Google Scholar] [CrossRef]
  31. Li, B.; Wang, P.; Jiao, J.; Wei, H.; Xu, W.; Zhou, P. Roles of the RANKL–RANK Axis in Immunity—Implications for Pathogenesis and Treatment of Bone Metastasis. Front. Immunol. 2022, 13, 824117. [Google Scholar] [CrossRef]
  32. Carbone, F.; Crowe, L.A.; Roth, A.; Burger, F.; Lenglet, S.; Braunersreuther, V.; Brandt, K.J.; Quercioli, A.; Mach, F.; Vallée, J.P.; et al. Treatment with anti-RANKL antibody reduces infarct size and attenuates dysfunction impacting on neutrophil-mediated injury. J. Mol. Cell. Cardiol. 2016, 94, 82–94. [Google Scholar] [CrossRef] [PubMed]
  33. Kitaura, H.; Kimura, K.; Ishida, M.; Hakami, Z.; Saeed, J.; Sugisawa, H.; Kohara, H.; Yoshimatsu, M.; Takano-Yamamoto, T. The Role of Th1 Cytokines on Mechanical Loading-Induced Osteoclastogenesis and Bone Resorption. In Interface Oral Health Science 2014: Innovative Research on Biosis-Abiosis Intelligent Interface; Springer: Berlin/Heidelberg, Germany, 2015; pp. 269–279. [Google Scholar]
  34. Amatya, N.; Garg, A.V.; Gaffen, S.L. IL-17 signaling: The yin and the yang. Trends Immunol. 2017, 38, 310–322. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, H.J.; Seo, S.J.; Kim, J.Y.; Kim, Y.G.; Lee, Y. IL-17 promotes osteoblast differentiation, bone regeneration, and remodeling in mice. Biochem. Biophys. Res. Commun. 2020, 524, 1044–1050. [Google Scholar] [CrossRef] [PubMed]
  36. Rossini, M.; Viapiana, O.; Adami, S.; Idolazzi, L.; Fracassi, E.; Gatti, D. Focal bone involvement in inflammatory arthritis: The role of IL17. Rheumatol. Int. 2016, 36, 469–482. [Google Scholar] [CrossRef]
  37. Tsukasaki, M.; Komatsu, N.; Nagashima, K.; Nitta, T.; Pluemsakunthai, W.; Shukunami, C.; Iwakura, Y.; Nakashima, T.; Okamoto, K.; Takayanagi, H.; et al. Host defense against oral microbiota by bone-damaging T cells. Nat. Commun. 2018, 9, 701. [Google Scholar] [CrossRef]
  38. Glowacki, A.J.; Yoshizawa, S.; Jhunjhunwala, S.; Vieira, A.E.; Garlet, G.P.; Sfeir, C.; Little, S.R. Prevention of inflammation-mediated bone loss in murine and canine periodontal disease via recruitment of regulatory lymphocytes. Proc. Natl. Acad. Sci. USA 2013, 110, 18525–18530. [Google Scholar] [CrossRef]
  39. Park, J.Y.; Chung, H.; DiPalma, D.T.; Tai, X.; Park, J.H. Immune quiescence in the oral mucosa is maintained by a uniquely large population of highly activated Foxp3+ regulatory T cells. Mucosal Immunol. 2018, 11, 1092–1102. [Google Scholar] [CrossRef]
  40. Bettelli, E.; Carrier, Y.; Gao, W.; Korn, T.; Strom, T.B.; Oukka, M.; Weiner, H.L.; Kuchroo, V.K. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006, 441, 235–238. [Google Scholar] [CrossRef]
  41. Ness-Schwickerath, K.J.; Morita, C.T. Regulation and function of IL-17A-and IL-22-producing γδ T cells. Cell. Mol. Life Sci. 2011, 68, 2371–2390. [Google Scholar] [CrossRef]
  42. Zhu, M.; Belkina, A.C.; DeFuria, J.; Carr, J.D.; Van Dyke, T.E.; Gyurko, R.; Nikolajczyk, B.S. B cells promote obesity-associated periodontitis and oral pathogen-associated inflammation. J. Leukoc. Biol. 2014, 96, 349–357. [Google Scholar] [CrossRef]
  43. Hu, F.; Liu, H.; Liu, X.; Zhang, X.; Xu, L.; Zhu, H.; Li, Y.; Shi, L.; Ren, L.; Zhang, J.; et al. Pathogenic conversion of regulatory B10 cells into osteoclast-priming cells in rheumatoid arthritis. J. Autoimmun. 2017, 76, 53–62. [Google Scholar] [CrossRef] [PubMed]
  44. Settem, R.P.; Honma, K.; Chinthamani, S.; Kawai, T.; Sharma, A. B-cell RANKL contributes to pathogen-induced alveolar bone loss in an experimental periodontitis mouse model. Front. Physiol. 2021, 12, 722859. [Google Scholar] [CrossRef] [PubMed]
  45. Sun, W.; Meednu, N.; Rosenberg, A.; Rangel-Moreno, J.; Wang, V.; Glanzman, J.; Owen, T.; Zhou, X.; Zhang, H.; Boyce, B.F.; et al. B cells inhibit bone formation in rheumatoid arthritis by suppressing osteoblast differentiation. Nat. Commun. 2018, 9, 5127. [Google Scholar] [CrossRef] [PubMed]
  46. Sapra, L.; Bhardwaj, A.; Mishra, P.K.; Garg, B.; Verma, B.; Mishra, G.C.; Srivastava, R.K. Regulatory B cells (Bregs) inhibit osteoclastogenesis and play a potential role in ameliorating ovariectomy-induced bone loss. Front. Immunol. 2021, 12, 691081. [Google Scholar] [CrossRef]
  47. Molofsky, A.B.; Nussbaum, J.C.; Liang, H.E.; Van Dyken, S.J.; Cheng, L.E.; Mohapatra, A.; Chawla, A.; Locksley, R.M. Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J. Exp. Med. 2013, 210, 535–549. [Google Scholar] [CrossRef]
  48. He, F.; Umrath, F.; von Ohle, C.; Reinert, S.; Alexander, D. Analysis of the influence of jaw periosteal cells on macrophages phenotype using an innovative horizontal coculture system. Biomedicines 2021, 9, 1753. [Google Scholar] [CrossRef]
  49. Ando, Y.; Tsukasaki, M.; Huynh, N.C.; Zang, S.; Yan, M.; Muro, R.; Nakamura, K.; Komagamine, M.; Komatsu, N.; Okamoto, K.; et al. The neutrophil–osteogenic cell axis promotes bone destruction in periodontitis. Int. J. Oral Sci. 2024, 16, 18. [Google Scholar] [CrossRef]
  50. Bruce, A.G.; Hoggatt, I.H.; Rose, T.M. Oncostatin M is a differentiation factor for myeloid leukemia cells. J. Immunol. 1992, 149, 1271–1275. [Google Scholar] [CrossRef]
  51. Arizon, M.; Nudel, I.; Segev, H.; Mizraji, G.; Elnekave, M.; Furmanov, K.; Eli-Berchoer, L.; Clausen, B.E.; Shapira, L.; Wilensky, A.; et al. Langerhans cells down-regulate inflammation-driven alveolar bone loss. Proc. Natl. Acad. Sci. USA 2012, 109, 7043–7048. [Google Scholar] [CrossRef]
  52. Ono, T.; Hayashi, M.; Sasaki, F.; Nakashima, T. RANKL biology: Bone metabolism, the immune system, and beyond. Inflamm. Regen. 2020, 40, 2. [Google Scholar] [CrossRef]
  53. Elsayed, R.; Kurago, Z.; Cutler, C.W.; Arce, R.M.; Gerber, J.; Celis, E.; Sultan, H.; Elashiry, M.; Meghil, M.; Sun, C.; et al. Role of dendritic cell-mediated immune response in oral homeostasis: A new mechanism of osteonecrosis of the jaw. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2020, 34, 2595. [Google Scholar] [CrossRef]
  54. Dai, J.; Umrath, F.; Reinert, S.; Alexander, D. Jaw periosteal cells seeded in beta-tricalcium phosphate inhibit dendritic cell maturation. Biomolecules 2020, 10, 887. [Google Scholar] [CrossRef] [PubMed]
  55. Wojno, E.D.T.; Artis, D. Innate lymphoid cells: Balancing immunity, inflammation, and tissue repair in the intestine. Cell Host Microbe 2012, 12, 445–457. [Google Scholar]
  56. Qin, X.; Hoda, M.N.; Susin, C.; Wheeler, J.N.; Marshall, B.; Perry, L.; Saad, N.; Yin, L.; Elsayed, R.; Elsalanty, M.; et al. Increased innate lymphoid cells in periodontal tissue of the murine model of periodontitis: The role of AMP-activated protein kinase and relevance for the human condition. Front. Immunol. 2017, 8, 922. [Google Scholar] [CrossRef] [PubMed]
  57. Bernink, J.H.; Ohne, Y.; Teunissen, M.B.; Wang, J.; Wu, J.; Krabbendam, L.; Guntermann, C.; Volckmann, R.; Koster, J.; van Tol, S.; et al. c-Kit-positive ILC2s exhibit an ILC3-like signature that may contribute to IL-17-mediated pathologies. Nat. Immunol. 2019, 20, 992–1003. [Google Scholar] [CrossRef]
  58. Zhang, Y.; Liang, J.; Liu, P.; Wang, Q.; Liu, L.; Zhao, H. The RANK/RANKL/OPG system and tumor bone metastasis: Potential mechanisms and therapeutic strategies. Front. Endocrinol. 2022, 13, 1063815. [Google Scholar] [CrossRef]
  59. Nile, M.; Folwaczny, M.; Wichelhaus, A.; Baumert, U.; Janjic Rankovic, M. Fluid flow shear stress and tissue remodeling—An orthodontic perspective: Evidence synthesis and differential gene expression network analysis. Front. Bioeng. Biotechnol. 2023, 11, 1256825. [Google Scholar] [CrossRef]
  60. Qin, L.; He, T.; Chen, S.; Yang, D.; Yi, W.; Cao, H.; Xiao, G. Roles of mechanosensitive channel Piezo1/2 proteins in skeleton and other tissues. Bone Res. 2021, 9, 44. [Google Scholar] [CrossRef]
  61. Zong, B.; Yu, F.; Zhang, X.; Pang, Y.; Zhao, W.; Sun, P.; Li, L. Mechanosensitive Piezo1 channel in physiology and pathophysiology of the central nervous system. Ageing Res. Rev. 2023, 90, 102026. [Google Scholar] [CrossRef]
  62. Gould, N.R.; Torre, O.M.; Leser, J.M.; Stains, J.P. The cytoskeleton and connected elements in bone cell mechano-transduction. Bone 2021, 149, 115971. [Google Scholar] [CrossRef]
  63. Wang, T.; Liu, X.; Li, J.; Yue, Y.; Li, J.; Wang, M.; Wei, N.; Hao, L. Mechanisms of mechanical force in periodontal homeostasis: A review. Front. Immunol. 2024, 15, 1438726. [Google Scholar] [CrossRef] [PubMed]
  64. Nakai, Y.; Praneetpong, N.; Ono, W.; Ono, N. Mechanisms of osteoclastogenesis in orthodontic tooth movement and orthodontically induced tooth root resorption. J. Bone Metab. 2023, 30, 297. [Google Scholar] [CrossRef] [PubMed]
  65. Chaushu, S.; Klein, Y.; Mandelboim, O.; Barenholz, Y.; Fleissig, O. Immune changes induced by orthodontic forces: A critical review. J. Dent. Res. 2022, 101, 11–20. [Google Scholar] [CrossRef]
  66. Lassus, J.; Salo, J.; Jiranek, W.A.; Santavirta, S.; Nevalainen, J.; Matucci-Cerinic, M.; Horák, P.; Konttinen, Y. Macrophage activation results in bone resorption. Clin. Orthop. Relat. Res. (1976–2007) 1998, 352, 7–15. [Google Scholar] [CrossRef]
  67. Pöllinger, B.; Junt, T.; Metzler, B.; Walker, U.A.; Tyndall, A.; Allard, C.; Bay, S.; Keller, R.; Raulf, F.; Di Padova, F.; et al. Th17 cells, not IL-17+ γδ T cells, drive arthritic bone destruction in mice and humans. J. Immunol. 2011, 186, 2602–2612. [Google Scholar] [CrossRef]
  68. Blanchard, F.; Duplomb, L.; Baud’huin, M.; Brounais, B. The dual role of IL-6-type cytokines on bone remodeling and bone tumors. Cytokine Growth Factor Rev. 2009, 20, 19–28. [Google Scholar] [CrossRef]
  69. Majedi, F.S.; Hasani-Sadrabadi, M.M.; Thauland, T.J.; Li, S.; Bouchard, L.S.; Butte, M.J. T-cell activation is modulated by the 3D mechanical microenvironment. Biomaterials 2020, 252, 120058. [Google Scholar] [CrossRef]
  70. Xie, D.; Fu, D.; Fu, S.; Chen, B.; He, W.; Wilson, D.A.; Peng, F. Mechanical activation of immune T cells via a water driven nanomotor. Adv. Healthc. Mater. 2022, 11, 2200042. [Google Scholar] [CrossRef]
  71. Chansaenroj, J.; Suwittayarak, R.; Egusa, H.; Samaranayake, L.P.; Osathanon, T. Mechanical force modulates inflammation and immunomodulation in periodontal ligament cells. Med. Rev. 2024, 4, 544–548. [Google Scholar] [CrossRef]
  72. Xu, H.; Guan, J.; Jin, Z.; Yin, C.; Wu, S.; Sun, W.; Zhang, H.; Yan, B. Mechanical force modulates macrophage proliferation via Piezo1-AKT-cyclin D1 axis. FASEB J. 2022, 36, e22423. [Google Scholar] [CrossRef]
  73. Liang, W.; Ding, P.; Qian, J.; Li, G.; Lu, E.; Zhao, Z. Polarized M2 macrophages induced by mechanical stretching modulate bone regeneration of the craniofacial suture for midfacial hypoplasia treatment. Cell Tissue Res. 2021, 386, 585–603. [Google Scholar] [CrossRef] [PubMed]
  74. Dong, L.; Song, Y.; Zhang, Y.; Zhao, W.; Wang, C.; Lin, H.; Al-ani, M.K.; Liu, W.; Xue, R.; Yang, L. Mechanical stretch induces osteogenesis through the alternative activation of macrophages. J. Cell. Physiol. 2021, 236, 6376–6390. [Google Scholar] [CrossRef] [PubMed]
  75. Cai, G.; Lu, Y.; Zhong, W.; Wang, T.; Li, Y.; Ruan, X.; Chen, H.; Sun, L.; Guan, Z.; Li, G.; et al. Piezo1-mediated M2 macrophage mechanotransduction enhances bone formation through secretion and activation of transforming growth factor-β1. Cell Prolif. 2023, 56, e13440. [Google Scholar] [CrossRef] [PubMed]
  76. Pu, P.; Wu, S.; Zhang, K.; Xu, H.; Guan, J.; Jin, Z.; Sun, W.; Zhang, H.; Yan, B. Mechanical force induces macrophage-derived exosomal UCHL3 promoting bone marrow mesenchymal stem cell osteogenesis by targeting SMAD1. J. Nanobiotechnology 2023, 21, 88. [Google Scholar] [CrossRef]
  77. Han, Y.; Yang, Q.; Huang, Y.; Gao, P.; Jia, L.; Zheng, Y.; Li, W. Compressive force regulates orthodontic tooth movement via activating the NLRP3 inflammasome. FASEB J. 2022, 36, e22627. [Google Scholar] [CrossRef]
  78. Kulkarni, R.N.; Bakker, A.D.; Everts, V.; Klein-Nulend, J. Mechanical loading prevents the stimulating effect of IL-1β on osteocyte-modulated osteoclastogenesis. Biochem. Biophys. Res. Commun. 2012, 420, 11–16. [Google Scholar] [CrossRef]
  79. Cabahug-Zuckerman, P.; Stout, R.F., Jr.; Majeska, R.J.; Thi, M.M.; Spray, D.C.; Weinbaum, S.; Schaffler, M.B. Potential role for a specialized β3 integrin-based structure on osteocyte processes in bone mechanosensation. J. Orthop. Res. 2018, 36, 642–652. [Google Scholar] [CrossRef]
  80. Weinbaum, S.; Cowin, S.C.; Zeng, Y. A model for the excitation of osteocytes by mechanical loading-induced bone fluid shear stresses. J. Biomech. 1994, 27, 339–360. [Google Scholar] [CrossRef]
  81. Klein-Nulend, J.; Bakker, A.D.; Bacabac, R.G.; Vatsa, A.; Weinbaum, S. Mechanosensation and transduction in osteocytes. Bone 2013, 54, 182–190. [Google Scholar] [CrossRef]
  82. Smit, T.H.; Huyghe, J.M.; Cowin, S.C. Estimation of the poroelastic parameters of cortical bone. J. Biomech. 2002, 35, 829–835. [Google Scholar] [CrossRef]
  83. Wang, L.; You, X.; Zhang, L.; Zhang, C.; Zou, W. Mechanical regulation of bone remodeling. Bone Res. 2022, 10, 16. [Google Scholar] [CrossRef] [PubMed]
  84. Moraes, C.; Sun, Y.; Simmons, C.A. (Micro) managing the mechanical microenvironment. Integr. Biol. 2011, 3, 959–971. [Google Scholar] [CrossRef] [PubMed]
  85. Kawakami, T.; Takise, S.; Fuchimoto, T.; Kawata, H. Effects of masticatory movement on cranial bone mass and micromorphology of osteocytes and osteoblasts in developing rats. Asia Pac. J. Clin. Nutr. 2009, 18, 96–104. [Google Scholar] [PubMed]
  86. Wang, L.; Dong, J.; Xian, C.J. Computational investigation on the biomechanical responses of the osteocytes to the compressive stimulus: A poroelastic model. BioMed Res. Int. 2018, 2018, 4071356. [Google Scholar] [CrossRef]
  87. Bonewald, L.F.; Johnson, M.L. Osteocytes, mechanosensing and Wnt signaling. Bone 2008, 42, 606–615. [Google Scholar] [CrossRef]
  88. Jackson, E.; Lara-Castillo, N.; Akhter, M.P.; Dallas, M.; Scott, J.M.; Ganesh, T.; Johnson, M.L. Osteocyte Wnt/β-catenin pathway activation upon mechanical loading is altered in ovariectomized mice. Bone Rep. 2021, 15, 101129. [Google Scholar] [CrossRef]
  89. Zarka, M.; Etienne, F.; Bourmaud, M.; Szondi, D.; Schwartz, J.M.; Kampmann, K.; Helary, C.; Rannou, F.; Haÿ, E.; Cohen-Solal, M. Mechanical loading activates the YAP/TAZ pathway and chemokine expression in the MLO-Y4 osteocyte-like cell line. Lab. Investig. 2021, 101, 1597–1604. [Google Scholar] [CrossRef]
  90. Inoue, M.; Ono, T.; Kameo, Y.; Sasaki, F.; Ono, T.; Adachi, T.; Nakashima, T. Forceful mastication activates osteocytes and builds a stout jawbone. Sci. Rep. 2019, 9, 4404. [Google Scholar] [CrossRef]
  91. Hichijo, N.; Kawai, N.; Mori, H.; Sano, R.; Ohnuki, Y.; Okumura, S.; Langenbach, G.E.; Tanaka, E. Effects of the masticatory demand on the rat mandibular development. J. Oral Rehabil. 2014, 41, 581–587. [Google Scholar] [CrossRef]
  92. Hao, Z.; Ma, Y.; Wu, J.; Li, X.; Chen, H.; Shen, J.; Wang, H. Osteocytes regulate osteoblast differentiation and osteoclast activity through Interleukin-6 under mechanical loading. RSC Adv. 2017, 7, 50200–50209. [Google Scholar] [CrossRef]
  93. Bonetto, A.; Aydogdu, T.; Kunzevitzky, N.; Guttridge, D.C.; Khuri, S.; Koniaris, L.G.; Zimmers, T.A. STAT3 activation in skeletal muscle links muscle wasting and the acute phase response in cancer cachexia. PLoS ONE 2011, 6, e22538. [Google Scholar] [CrossRef] [PubMed]
  94. Dwivedi, A.; Kiely, P.A.; Hoey, D.A. Mechanically stimulated osteocytes promote the proliferation and migration of breast cancer cells via a potential CXCL1/2 mechanism. Biochem. Biophys. Res. Commun. 2021, 534, 14–20. [Google Scholar] [CrossRef] [PubMed]
  95. Yan, Y.; Wang, L.; Ge, L.; Pathak, J.L. Osteocyte-mediated translation of mechanical stimuli to cellular signaling and its role in bone and non-bone-related clinical complications. Curr. Osteoporos. Rep. 2020, 18, 67–80. [Google Scholar] [CrossRef]
  96. Sindhavajiva, P.R.; Sastravaha, P.; Arksornnukit, M.; Pavasant, P. Intermittent compressive force induces human mandibular-derived osteoblast differentiation via WNT/β-catenin signaling. J. Cell. Biochem. 2018, 119, 3474–3485. [Google Scholar] [CrossRef] [PubMed]
  97. Lohberger, B.; Kaltenegger, H.; Stuendl, N.; Payer, M.; Rinner, B.; Leithner, A. Effect of cyclic mechanical stimulation on the expression of osteogenesis genes in human intraoral mesenchymal stromal and progenitor cells. BioMed Res. Int. 2014, 2014, 189516. [Google Scholar] [CrossRef]
  98. Liu, P.; Tu, J.; Wang, W.; Li, Z.; Li, Y.; Yu, X.; Zhang, Z. Effects of mechanical stress stimulation on function and expression mechanism of osteoblasts. Front. Bioeng. Biotechnol. 2022, 10, 830722. [Google Scholar] [CrossRef]
  99. Baumert, U.; Golan, I.; Becker, B.; Hrala, B.P.; Redlich, M.; Roos, H.A.; Palmon, A.; Reichenberg, E.; Müßig, D. Pressure simulation of orthodontic force in osteoblasts: A pilot study. Orthod. Craniofacial Res. 2004, 7, 3–9. [Google Scholar] [CrossRef]
  100. Barthelemi, S.; Robinet, J.; Garnotel, R.; Antonicelli, F.; Schittly, E.; Hornebeck, W.; Lorimier, S. Mechanical forces-induced human osteoblasts differentiation involves MMP-2/MMP-13/MT1-MMP proteolytic cascade. J. Cell. Biochem. 2012, 113, 760–772. [Google Scholar] [CrossRef]
  101. Sims, N.A.; Gooi, J.H. Bone remodeling: Multiple cellular interactions required for coupling of bone formation and resorption. In Seminars in Cell & Developmental Biology; Academic Press: Cambridge, MA, USA, 2008; Volume 19. [Google Scholar]
  102. Boyce, B.F.; Xing, L. Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch. Biochem. Biophys. 2008, 473, 139–146. [Google Scholar] [CrossRef]
  103. Yamamoto, K.; Yamamoto, T.; Ichioka, H.; Akamatsu, Y.; Oseko, F.; Mazda, O.; Imanishi, J.; Kanamura, N.; Kita, M. Effects of mechanical stress on cytokine production in mandible-derived osteoblasts. Oral Dis. 2011, 17, 712–719. [Google Scholar] [CrossRef]
  104. Yamamoto, K.; Yamamoto, T.; Kanamura, N.; Kita, M. Role of mechanical stress in mandible bone metabolism. Inflamm. Regen. 2012, 32, 119–123. [Google Scholar] [CrossRef]
  105. Vahidi, B. Simulation of mechanical modulation of an osteoblast cell due to fluid flow. In Proceedings of the 2018 25th National and 3rd International Iranian Conference on Biomedical Engineering (ICBME), Qom, Iran, 29–30 November 2018; IEEE: Piscataway, NJ, USA, 2018. [Google Scholar]
  106. Wang, H.; Li, T.; Wang, X.; Guan, Y.; Jiang, Y.; Chen, S.; Zou, S.; Duan, P. Mechanisms of sphingosine-1-phosphate (S1P) signaling on excessive stress-induced root resorption during orthodontic molar intrusion. Clin. Oral Investig. 2021, 26, 1003–1016. [Google Scholar] [CrossRef] [PubMed]
  107. Yu, H.; Sun, C.; Argraves, K.M. Periodontal inflammation and alveolar bone loss induced by A ggregatibacter actinomycetemcomitans is attenuated in sphingosine kinase 1-deficient mice. J. Periodontal Res. 2016, 51, 38–49. [Google Scholar] [CrossRef] [PubMed]
  108. Xiao, L.; Zhou, Y.; Zhu, L.; Yang, S.; Huang, R.; Shi, W.; Peng, B.; Xiao, Y. SPHK1-S1PR1-RANKL axis regulates the interactions between macrophages and BMSCs in inflammatory bone loss. J. Bone Miner. Res. 2018, 33, 1090–1104. [Google Scholar] [CrossRef]
  109. Wang, H.; Li, T.; Jiang, Y.; Chen, S.; Zou, S.; Bonewald, L.F.; Duan, P. Force-Loaded Cementocytes Regulate Osteoclastogenesis via S1P/S1PR1/Rac1 Axis. J. Dent. Res. 2023, 102, 1376–1386. [Google Scholar] [CrossRef]
  110. Kook, S.-H.; Jang, Y.-S.; Lee, J.-C. Human periodontal ligament fibroblasts stimulate osteoclastogenesis in response to compression force through TNF-α-mediated activation of CD4+ T cells. J. Cell. Biochem. 2011, 112, 2891–2901. [Google Scholar] [CrossRef]
  111. Huang, H.; Yang, R.; Zhou, Y.-H. Mechanobiology of periodontal ligament stem cells in orthodontic tooth movement. Stem Cells Int. 2018, 2018, 6531216. [Google Scholar] [CrossRef]
  112. Wang, Z.; Maruyama, K.; Sakisaka, Y.; Suzuki, S.; Tada, H.; Suto, M.; Saito, M.; Yamada, S.; Nemoto, E. Cyclic stretch force induces periodontal ligament cells to secrete exosomes that suppress IL-1β production through the inhibition of the NF-κB signaling pathway in macrophages. Front. Immunol. 2019, 10, 1310. [Google Scholar] [CrossRef]
  113. Bauernfeind, F.G.; Horvath, G.; Stutz, A.; Alnemri, E.S.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B.G.; Fitzgerald, K.A.; et al. Cutting edge: NF-κB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J. Immunol. 2009, 183, 787–791. [Google Scholar] [CrossRef]
  114. Huang, H.M.; Han, C.S.; Cui, S.J.; Zhou, Y.K.; Xin, T.Y.; Zhang, T.; Zhu, S.B.; Zhou, Y.H.; Yang, R.L. Mechanical force-promoted osteoclastic differentiation via periodontal ligament stem cell exosomal protein ANXA3. Stem Cell Rep. 2022, 17, 1842–1858. [Google Scholar] [CrossRef]
  115. Liu, Y.; Yang, R.; Liu, X.; Zhou, Y.; Qu, C.; Kikuiri, T.; Wang, S.; Zandi, E.; Du, J.; Ambudkar, I.S.; et al. Hydrogen sulfide maintains mesenchymal stem cell function and bone homeostasis via regulation of Ca2+ channel sulfhydration. Cell Stem Cell 2014, 15, 66–78. [Google Scholar] [CrossRef] [PubMed]
  116. Su, Y.; Liu, D.; Liu, Y.; Zhang, C.; Wang, J.; Wang, S. Physiologic levels of endogenous hydrogen sulfide maintain the proliferation and differentiation capacity of periodontal ligament stem cells. J. Periodontol. 2015, 86, 1276–1286. [Google Scholar] [CrossRef] [PubMed]
  117. Liu, F.; Wen, F.; He, D.; Liu, D.; Yang, R.; Wang, X.; Yan, Y.; Liu, Y.; Kou, X.; Zhou, Y. Force-induced H2S by PDLSCs modifies osteoclastic activity during tooth movement. J. Dent. Res. 2017, 96, 694–702. [Google Scholar] [CrossRef] [PubMed]
  118. Kim, N.; Kadono, Y.; Takami, M.; Lee, J.; Lee, S.H.; Okada, F.; Kim, J.H.; Kobayashi, T.; Odgren, P.R.; Nakano, H.; et al. Osteoclast differentiation independent of the TRANCE–RANK–TRAF6 axis. J. Exp. Med. 2005, 202, 589–595. [Google Scholar] [CrossRef]
  119. Kwak, H.B.; Jin, H.M.; Ha, H.; Kang, M.J.; Lee, S.B.; Kim, H.H.; Lee, Z.H. Tumor necrosis factor-α induces differentiation of human peripheral blood mononuclear cells into osteoclasts through the induction of p21 (WAF1/Cip1). Biochem. Biophys. Res. Commun. 2005, 330, 1080–1086. [Google Scholar] [CrossRef]
  120. Cho, E.S.; Lee, K.S.; Son, Y.O.; Jang, Y.S.; Lee, S.Y.; Kwak, S.Y.; Yang, Y.M.; Park, S.M.; Lee, J.C. Compressive mechanical force augments osteoclastogenesis by bone marrow macrophages through activation of c-Fms-mediated signaling. J. Cell. Biochem. 2010, 111, 1260–1269. [Google Scholar] [CrossRef]
  121. Ma, Z.; Li, S.; Sun, Y. Effect of enhanced masticatory force on OPG, RANKL and MGF in alveolar bone of ovariectomized rats. J. Appl. Oral Sci. 2020, 28, e20190409. [Google Scholar] [CrossRef]
  122. Tahara, Y.; Sakurai, K.; Ando, T. Influence of chewing and clenching on salivary cortisol levels as an indicator of stress. J. Prosthodont. 2007, 16, 129–135. [Google Scholar] [CrossRef]
  123. Abella, V.; Scotece, M.; Conde, J.; Pino, J.; Gonzalez-Gay, M.A.; Gomez-Reino, J.J.; Mera, A.; Lago, F.; Gómez, R.; Gualillo, O. Leptin in the interplay of inflammation, metabolism and immune system disorders. Nat. Rev. Rheumatol. 2017, 13, 100–109. [Google Scholar] [CrossRef]
  124. DeVries, A.C.; Gerber, J.M.; Richardson, H.N.; Moffatt, C.A.; Demas, G.E.; Taymans, S.E.; Nelson, R.J. Stress affects corticosteroid and immunoglobulin concentrations in male house mice (Mus musculus) and prairie voles (Microtus ochrogaster). Comp. Biochem. Physiol. Part A Physiol. 1997, 118, 655–663. [Google Scholar] [CrossRef]
  125. Yang, S.; Park, Y.; Choi, T. Effects of mastication on antibody production under fasting conditions in mice. Int. J. Med. Sci. 2023, 20, 232. [Google Scholar] [CrossRef] [PubMed]
  126. Iwawaki, Y.; Mizusawa, N.; Iwata, T.; Higaki, N.; Goto, T.; Watanabe, M.; Tomotake, Y.; Ichikawa, T.; Yoshimoto, K. MiR-494-3p induced by compressive force inhibits cell proliferation in MC3T3-E1 cells. J. Biosci. Bioeng. 2015, 120, 456–462. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, Y.; Mohammed, A.; Oubaidin, M.; Evans, C.A.; Zhou, X.; Luan, X.; Diekwisch, T.G.; Atsawasuwan, P. Cyclic stretch and compression forces alter microRNA-29 expression of human periodontal ligament cells. Gene 2015, 566, 13–17. [Google Scholar] [CrossRef] [PubMed]
  128. Quinodoz, S.; Guttman, M. Long noncoding RNAs: An emerging link between gene regulation and nuclear organization. Trends Cell Biol. 2014, 24, 651–663. [Google Scholar] [CrossRef]
  129. Liao, J.; Yu, X.; Hu, X.; Fan, J.; Wang, J.; Zhang, Z.; Zhao, C.; Zeng, Z.; Shu, Y.; Zhang, R.; et al. lncRNA H19 mediates BMP9-induced osteogenic differentiation of mesenchymal stem cells (MSCs) through Notch signaling. Oncotarget 2017, 8, 53581. [Google Scholar] [CrossRef]
  130. Buvinic, S.; Balanta-Melo, J.; Kupczik, K.; Vásquez, W.; Beato, C.; Toro-Ibacache, V. Muscle-bone crosstalk in the masticatory system: From biomechanical to molecular interactions. Front. Endocrinol. 2021, 11, 606947. [Google Scholar] [CrossRef]
  131. Maurel, D.B.; Jähn, K.; Lara-Castillo, N. Muscle–bone crosstalk: Emerging opportunities for novel therapeutic approaches to treat musculoskeletal pathologies. Biomedicines 2017, 5, 62. [Google Scholar] [CrossRef]
  132. Brotto, M.; Johnson, M.L. Endocrine crosstalk between muscle and bone. Curr. Osteoporos. Rep. 2014, 12, 135–141. [Google Scholar] [CrossRef]
  133. Brotto, M.; Bonewald, L. Bone and muscle: Interactions beyond mechanical. Bone 2015, 80, 109–114. [Google Scholar] [CrossRef]
  134. Kitase, Y.; Vallejo, J.A.; Gutheil, W.; Vemula, H.; Jähn, K.; Yi, J.; Zhou, J.; Brotto, M.; Bonewald, L.F. β-aminoisobutyric acid, l-BAIBA, is a muscle-derived osteocyte survival factor. Cell Rep. 2018, 22, 1531–1544. [Google Scholar] [CrossRef]
  135. Giudice, J.; Taylor, J.M. Muscle as a paracrine and endocrine organ. Curr. Opin. Pharmacol. 2017, 34, 49–55. [Google Scholar] [CrossRef]
  136. Bonewald, L. Use it or lose it to age: A review of bone and muscle communication. Bone 2019, 120, 212–218. [Google Scholar] [CrossRef] [PubMed]
  137. Dankbar, B.; Fennen, M.; Brunert, D.; Hayer, S.; Frank, S.; Wehmeyer, C.; Beckmann, D.; Paruzel, P.; Bertrand, J.; Redlich, K.; et al. Myostatin is a direct regulator of osteoclast differentiation and its inhibition reduces inflammatory joint destruction in mice. Nat. Med. 2015, 21, 1085–1090. [Google Scholar] [CrossRef] [PubMed]
  138. Qin, Y.; Peng, Y.; Zhao, W.; Pan, J.; Ksiezak-Reding, H.; Cardozo, C.; Wu, Y.; Pajevic, P.D.; Bonewald, L.F.; Bauman, W.A.; et al. Myostatin inhibits osteoblastic differentiation by suppressing osteocyte-derived exosomal microRNA-218: A novel mechanism in muscle-bone communication. J. Biol. Chem. 2017, 292, 11021–11033. [Google Scholar] [CrossRef] [PubMed]
  139. Goldspink, G. Mechanical signals, IGF-I gene splicing, and muscle adaptation. Physiology 2005, 20, 232–238. [Google Scholar] [CrossRef]
  140. Tokimasa, C.; Kawata, T.; Fujita, T.; Kaku, M.; Kohno, S.; Tsutsui, K.; Tenjou, K.; Ohtani, J.; Motokawa, M.; Tanne, K. Effects of insulin-like growth factor-I on the expression of osteoclasts and osteoblasts in the nasopremaxillary suture under different masticatory loading conditions in growing mice. Arch. Oral Biol. 2003, 48, 31–38. [Google Scholar] [CrossRef]
  141. Chowdhury, S.; Schulz, L.; Palmisano, B.; Singh, P.; Berger, J.M.; Yadav, V.K.; Mera, P.; Ellingsgaard, H.; Hidalgo, J.; Brüning, J.; et al. Muscle-derived interleukin 6 increases exercise capacity by signaling in osteoblasts. J. Clin. Investig. 2020, 130, 2888–2902. [Google Scholar] [CrossRef]
  142. Mo, C.; Romero-Suarez, S.; Bonewald, L.; Johnson, M.; Brotto, M. Prostaglandin E2: From clinical applications to its potential role in bone-muscle crosstalk and myogenic differentiation. Recent Pat. Biotechnol. 2012, 6, 223–229. [Google Scholar] [CrossRef]
  143. Bonnet, N.; Bourgoin, L.; Biver, E.; Douni, E.; Ferrari, S. RANKL inhibition improves muscle strength and insulin sensitivity and restores bone mass. J. Clin. Investig. 2023, 129, 3214–3223. [Google Scholar] [CrossRef]
  144. Huang, J.; Romero Suarez, S.; Lara, N.; Mo, C.; Kaja, S.; Brotto, L.; Dallas, S.L.; Johnson, M.L.; Jähn, K.; Bonewald, L.F.; et al. Crosstalk between MLO Y4 osteocytes and C2C12 muscle cells is mediated by the Wnt/β catenin pathway. J. Bone Miner. Res. Plus 2017, 1, 86–100. [Google Scholar] [CrossRef]
Figure 1. The mandibular immune system and bone cells exhibit a coordinated response under the impact of masticatory forces. Fibroblasts, osteoblasts, and osteocytes secrete IL-6, which leads to subsequent effects. Mechanical forces, such as pressure and stretching, regulate the transition between M1 and M2 macrophages, influencing angiogenesis and osteogenesis. T cells modulate osteoclast activity by regulating the secretion of Rankl. Additionally, the secretions of bone cells affect the differentiation of MSCs and the production of preosteoclasts. M1: M1 macrophages, M2: M2 macrophages, MSC: mesenchymal stem cell, HSC: hematopoietic stem cell.
Figure 1. The mandibular immune system and bone cells exhibit a coordinated response under the impact of masticatory forces. Fibroblasts, osteoblasts, and osteocytes secrete IL-6, which leads to subsequent effects. Mechanical forces, such as pressure and stretching, regulate the transition between M1 and M2 macrophages, influencing angiogenesis and osteogenesis. T cells modulate osteoclast activity by regulating the secretion of Rankl. Additionally, the secretions of bone cells affect the differentiation of MSCs and the production of preosteoclasts. M1: M1 macrophages, M2: M2 macrophages, MSC: mesenchymal stem cell, HSC: hematopoietic stem cell.
Ijms 26 04478 g001
Figure 2. Masticatory forces influence jawbone immunity and bone balance through other pathways. Masticatory forces can influence jawbone immunity and bone balance by (a) regulating hormone levels, (b) affecting non-coding RNA, and (c) modulating muscle-bone interaction.
Figure 2. Masticatory forces influence jawbone immunity and bone balance through other pathways. Masticatory forces can influence jawbone immunity and bone balance by (a) regulating hormone levels, (b) affecting non-coding RNA, and (c) modulating muscle-bone interaction.
Ijms 26 04478 g002
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

Song, Y.; Jiao, Y.; Liu, Y.; Guo, L. Role of Masticatory Force in Modulating Jawbone Immunity and Bone Homeostasis: A Review. Int. J. Mol. Sci. 2025, 26, 4478. https://doi.org/10.3390/ijms26104478

AMA Style

Song Y, Jiao Y, Liu Y, Guo L. Role of Masticatory Force in Modulating Jawbone Immunity and Bone Homeostasis: A Review. International Journal of Molecular Sciences. 2025; 26(10):4478. https://doi.org/10.3390/ijms26104478

Chicago/Turabian Style

Song, Yue, Yao Jiao, Yitong Liu, and Lijia Guo. 2025. "Role of Masticatory Force in Modulating Jawbone Immunity and Bone Homeostasis: A Review" International Journal of Molecular Sciences 26, no. 10: 4478. https://doi.org/10.3390/ijms26104478

APA Style

Song, Y., Jiao, Y., Liu, Y., & Guo, L. (2025). Role of Masticatory Force in Modulating Jawbone Immunity and Bone Homeostasis: A Review. International Journal of Molecular Sciences, 26(10), 4478. https://doi.org/10.3390/ijms26104478

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