Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Feb 5.
Published in final edited form as: Curr Osteoporos Rep. 2023 Sep 28;21(6):842–853. doi: 10.1007/s11914-023-00824-4

Inflammatory Processes Affecting Bone Health and Repair

Haydee M Torres 1, Katherine M Arnold 1,2, Manuela Oviedo 1, Jennifer J Westendorf 1,3, Samantha R Weaver 1
PMCID: PMC10842967  NIHMSID: NIHMS1936715  PMID: 37759135

Abstract

Purpose of Review

The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing.

Recent Findings

Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells.

Summary

Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.

Keywords: Inflammatory bone loss, Cytokines, Osteoporosis, Inflammatory osteoclast, Senescence

Introduction

Osteoporosis is an enormous public health burden that affects men and women of all races. About 50% of women and 20% of men in the USA over the age of 50 years will experience an osteoporotic fracture [1]. Osteoporosis is characterized by bone mineral density (BMD) at the hip or lumbar spine that is ≤ 2.5 standard deviations below the mean BMD of a reference population of young adults. The main features of osteoporosis are low bone density, disrupted bone microarchitecture, compromised bone strength, and fracture [1]. Despite effective pharmacological interventions, osteoporosis remains underdiagnosed and undertreated. The osteoporosis treatment gap (defined as the difference between the number of patients that meet treatment indications and the number that receive treatment) is a crisis in patient care worldwide [2, 3]. It is crucial to identify and understand underlying diseases and physiological processes that may contribute to osteoporosis, including inflammation.

Inflammation is the body’s response to pathogens or tissue damage. During an inflammatory response, cells of the innate and adaptive immune systems are recruited and activated by cytokines, such as interferons, interleukins, and chemokines. These inflammatory factors have a direct effect on the differentiation and function of multiple bone cell types, including osteoblasts, osteoclasts, and osteocytes [4, 5]. Many diseases can lead to inflammatory bone loss, including inflammatory bowel disease, spondyloarthritis, systemic lupus erythematosus, and chronic obstructive pulmonary disease, among others [6]. Anti-inflammatory medications can also induce bone loss, most notably glucocorticoids. In recent years, the field of osteoimmunology has developed new techniques and models to interrogate inflammatory processes in the bone. From these discoveries, therapies targeted at blocking cytokine activity and depleting or inhibiting aberrant immune responses are in development to improve bone density and reduce fracture risk [4, 79]. However, it is important not to eliminate all inflammation from the bone microenvironment, as inflammatory processes are crucial for early stages of bone healing and repair.

Inflammation is Critical in Bone Healing

While inflammation is largely associated with negative outcomes on bone, inflammatory processes are also essential for maintaining bone health. Bone healing following fracture takes place in three overlapping phases: the inflammatory phase, the repair phase, and the remodeling phase [10]. Complete healing of the injury is critically dependent on success of the initial inflammatory phase. Acute inflammation following injury peaks within the first 24–48 hours and is generally complete by 7 days. In response to fracture, blood vessels rupture within and around the fracture site, inducing a hematoma. The hematoma is a scaffold for infiltrating macrophages (osteomacs), lymphocytes, fibroblasts, and mast cells, which release pro-inflammatory cytokines (e.g., interleukin (IL)-1, IL-6, tumor necrosis factor (TNF)-α), chemokines, and growth factors [11, 12]. Polymorphonuclear neutrophils and M1-polarized macrophages in this first wave of immune infiltration clear away dead cells and debris. To resolve the initial acute inflammation and allow for the next phases of repair, macrophages polarize from an M1 to an M2 phenotype following stimulation by anti-inflammatory cytokines, including IL-4 and IL-10, while mesenchymal progenitor cells are attracted to the site of injury by TNF-α and CXCL12. This cocktail of pro- and anti-inflammatory cytokines, along with growth factors (TGF-β) and bone morphogenic proteins (BMPs), initiates angiogenesis and osteogensis to form a soft callus. Pro-inflammatory cytokines are more lowly expressed during the repair phase, when the hard callus is formed, but are elevated again during the remodeling phase, when bone is restored to its original shape, structure, and mechanical integrity. Inhibiting inflammatory cytokines such as IL-6 or TNF-α delays fracture healing [13, 14]. Additionally, prolonged use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with a higher risk of bone nonunions and delayed unions [15, 16]. These and other studies highlight the critical role of inflammatory mediators in effective bone healing.

During the inflammatory phase of fracture repair, mesenchymal progenitors are recruited from both the periosteum and the bone marrow to the injury [17]. Periosteal progenitor cells are influenced by the inflammatory milieu [18, 19]. For example, inflammation associated with aging alters effective recruitment and differentiation of periosteal progenitors, delaying bone healing post-fracture [20]. Aged human periosteal progenitors are less proliferative and produce more IL-6 and other inflammatory cytokines than progenitors from younger subjects [21]. In mouse models, progenitor cells from old mice create a pro-inflammatory, degenerative niche that limits effective fracture healing [20]. Bone healing demonstrates that numerous cell types at various stages of differentiation in the microenvironment are all potential targets of inflammatory processes.

The Effects of Inflammation on Bone Cells

Bone Remodeling and the RANK-RANKL-OPG Axis

Inflammatory processes cause bone loss by uncoupling bone formation from resorption. Osteoblasts form bone and couple with bone-resorbing osteoclasts [22] by secreting two cytokines: macrophage colony stimulating factor (M-CSF) and receptor activator of NF-κB (RANK) ligand (RANKL). Osteocytes embedded in the bone matrix are also major producers of RANKL [23]. Upon stimulation with M-CSF and RANKL, myeloid progenitor cells expressing Csf1r (the receptor for M-CSF) and RANK (the receptor for RANKL) activate transcription factors including c-Fos, NF-κB, and NFATc1 to induce osteoclast differentiation. Osteoprotegerin (OPG) is also secreted by osteoblasts and is a soluble decoy receptor for RANKL, antagonizing RANK signaling. RANKL signaling is a potent inducer of osteoclast activity and induces bone loss. The FDA-approved therapeutic denosumab is a monoclonal antibody against RANKL and suppresses osteoclastogenesis to prevent excessive bone resorption [24].

RANK-RANKL signaling is at the crux of osteoimmunology. In addition to regulating osteoclastogenesis in the bone, RANK-RANKL signaling controls maturation and functioning of the thymus, bone marrow, and lymph nodes. In bone, various pro-inflammatory cytokines, including IL-6, IL-17, and TNF-α, induce RANK-RANKL signaling to drive osteoclastic resorption and bone loss [25].

Osteoclasts are the Main Targets of Inflammatory Processes in Bone

Osteoclasts are the main targets of inflammatory processes in the bone microenvironment (Fig. 1) [26]. Osteoclasts are bone-resorbing cells that derive from the monocyte/macrophage lineage following stimulation by M-CSF and RANKL [27, 28]. In inflammatory conditions [29], osteoclasts can arise from other precursors, including inflammatory dendritic cells [30, 31] or arthritis-associated osteoclastogenic macrophages (AtoMs) [32, 33]. Inflammatory cytokines such as TNF-α [3438], IL-1 [3942], IL-6 [43, 44], IL-7 [4547], and IL-17 [4851] induce osteoclast maturation and activity. TNF-α is particularly important in regulating bone resorption and can induce inflammatory osteoclast differentiation independent of the canonical RANKL pathway, ultimately resulting in bone loss [38, 52]. Osteoclastic response to inflammation is sexually dimorphic, with pre-osteoclasts derived from female mice showing enhanced inflammatory pathway activation compared to male pre-osteoclasts [53].

Fig. 1.

Fig. 1

Physiological versus pathological osteoclast maturation and activity. Under normal physiological conditions, osteoclasts work in balance with osteoblasts and osteocytes to remodel bone. Under pathological inflammatory conditions, osteoclasts are derived from multiple sources and aberrantly activated, inducing bone loss. AtOM, arthritis-associated osteoclastogenic macrophages; SASP, senescence-associated secretory phenotype. Figure created with http://Biorender.com.

In human and murine bone, osteoclasts form either pits (round holes in the bone surface) or trenches (formed by osteoclasts migrating across the bone surface while resorbing) [54, 55]. Bone from osteoporotic patients shows a high prevalence of trenches [56], which are correlated with impaired bone strength [57]. Glucocorticoids increase trench formation in human and rabbit bone [5759].

While excessive osteoclast activity induces pathological bone loss, basal osteoclastogenesis and bone resorption is a crucial component of healthy bone remodeling [60, 61]. Inflammatory osteoclast precursors (Ly6ChiCD11bhi) expand during chronic inflammation and generate more active osteoclasts. These inflammatory osteoclast precursors are regulated by TNF-α, as they were rendered ineffective in tnf-α−/− mice [62••]. Mature osteoclasts are also regulated by inflammation. Pathologically activated osteoclasts (PAOCs) have significant proton secretion and motility, leading to a high degree of bone resorption in the presence of IL-1β [41]. Recently, a new class of inflammatory osteoclasts (iOCs) were characterized as Cx3cr1 negative (Cx3cr1neg) or positive (Cx3cr1+). Cx3cr1neg iOCs potently resorb bone, produce pro-inflammatory cytokines, and activate inflammatory bone marrow CD4+ T cells. By contrast, Cx3cr1+ iOCs have an immune-suppressive effect on Cx3cr1neg iOCs in culture [30, 63••, 64]. Identifying classes of inflammatory pre-osteoclasts or osteoclasts is an essential step in developing new therapeutics targeted at only pathologically active, inflammatory osteoclasts [65•].

Osteoblasts and Osteocytes are Regulated by Inflammatory Signaling

Inflammatory mediators directly and indirectly suppress osteoblast activity to prevent bone formation [4]. In addition to producing RANKL and OPG, which agonize and antagonize, respectively, the RANK signaling cascade, osteoblasts produce pro-inflammatory cytokines [6668]. Cytokines, including IL-6, IL-7, IL-17, and TNF-α, stimulate secretion of RANKL from osteoblasts [69]. Cytokines can also inhibit osteoblasts through mechanisms independent of RANKL. For example, TNF-α inhibits osteoblastogenesis [7072] by repressing Runx2, the master transcription factor regulating osteoblast maturation [73]. TNF-α also induces the expression of Dickkopf-1 (DKK1), inhibiting Wnt signaling that is crucial for osteoblastogenesis [74]. IL-1β impairs osteoblast migration, potentially impairing osteoblast recruitment during early stages of fracture healing [75].

While inflammatory cytokines generally favor bone resorption resulting in bone loss, certain cytokines are osteoblastogenic [9]. For example, while IL-11 was originally characterized pro-osteoclastic [76], more recent reports indicate that IL-11 enhances osteogenesis [77]. IL-11−/− mice have low bone mass due to suppressed osteoblast activity [78, 79]. IL-10−/− mice also have low bone mass [80], as IL-10 induces osteogenesis [81, 82] and inhibits bone resorption [83]. IFN-γ promotes osteoblast differentiation from mesenchymal progenitors [84] and reverses osteoporosis in ovariectomized mice [85] but was shown to stimulate bone loss in vivo [86]. Finally, although the main role of IL-6 is to induce osteoclastogenesis and stimulate bone resorption [87], IL-6 stimulates mesenchymal progenitor cell differentiation towards osteoblasts [88] and can stimulate bone formation in vivo in the presence of a soluble IL-6 receptor [89].

Osteocytes are terminally differentiated osteoblasts and are the most abundant and longest-living cell type in bone [90]. Osteocytes are the main mechanosensing cell in bone and coordinate the action of osteoblasts and osteoclasts on bone surfaces [9193]. Through a process called perilacunar/canalicular remodeling, osteocytes can also resorb their surrounding matrix. Glucocorticoids suppress normal perilacunar remodeling, causing bone degeneration [94]. Osteocytes express pro-inflammatory cytokines including IL-1β, IL-6, and TNF-α [95], which in turn enhance osteocyte RANKL expression, inducing osteoclastogenesis and bone loss [34, 96, 97]. In animal models of periodontitis [98, 99] and inflammatory bowel disease [100], the number of osteocytes expressing RANKL is increased along with greater osteoclast activity. Due to their mechanosensing properties, osteocytes may play a particularly significant role in regulating the inflammatory response to periodontal disease because alveolar surfaces on teeth are under consistent mechanical stress [99]. All bone cells, including osteoclasts, osteoblasts, and osteocytes, are susceptible to the effects of inflammation, highlighting why so many diseases are associated with inflammatory bone loss.

Diseases and Therapeutics Associated with Inflammatory Bone Loss

Chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease (IBD) (e.g., Crohn’s disease and ulcerative colitis), and periodontitis are frequently associated with low BMD [6]. Inflammation also drives bone loss in cystic fibrosis [101], ankylosing spondylitis [102], chronic obstructive pulmonary disease [103], systemic lupus erythematosus [104], and psoriasis [105] (Table 1). Chronic, systemic inflammation characteristic of these diseases increases the risk of fracture.

Table 1.

Inflammatory diseases and therapeutics comorbid with bone loss.

Disease / therapeutic Type of bone loss References
Ankylosing spondylitis Systemic (90)
Chronic obstructive pulmonary disease Systemic (91)
Cystic fibrosis Systemic (89)
Glucocorticoids Systemic (48, 49, 99, 136139, 172)
Inflammatory bowel disease & gut dysbiosis Systemic (122128)
Periodontitis Systemic, local (alveolar) (7, 20, 86, 87, 129135, 165)
Postmenopausal osteoporosis Systemic (109116)
Psoriasis Systemic (93)
Rheumatoid arthritis Systemic, local (focal / periarticular) (20, 117121)
Systemic lupus erythematosus Systemic (92)

Inflammation in Fracture Risk and Bone Mechanical Strength

The central concern associated with inflammatory bone loss is increased susceptibility to fracture. Mechanical integrity of the bone plays a key role in determining the risk of fracture and severity of injury. In addition to the direct effects of inflammatory mediators on bone cells, chronic inflammatory conditions are also associated with immobility, poor nutrition, and low exercise tolerance, which can themselves lead to bone loss [6]. Inflammatory bone loss is associated with an increased fracture risk [4], which is correlated with altered BMD [106], increased cortical porosity [107], and disruption of various architectural components of bone [108, 109]. Using any one measure to assess bone quality and fracture risk is a challenge due to the complex and dynamic nature of bone. As such, none of these measures alone can completely account for fracture risk in inflammatory bone conditions. Recently, more comprehensive measures such as Trabecular Bone Score are improving patient monitoring in multiple inflammation-based chronic conditions including ankylosing spondylitis and glucocorticoid-induced osteoporosis [110113].

Chronic, low-grade inflammation is also associated with aging, so-called inflammaging [114]. In the elderly, fractures are frequent and heal slowly, with more nonunions than young populations [115]. Aged macrophages are more sensitive to inflammatory signals and fail to resolve from the M1 to the M2 phenotype, delaying healing after fracture [116]. In addition, age-related inflammation mediated by NF-κB and TLR signaling reduces the number and function of skeletal progenitor cells, compromising regenerative processes needed for fracture repair [117119]. Recent work has examined the therapeutic potential of optimizing the inflammatory stage of bone healing to prevent impaired fracture healing (e.g., hypertrophic or atrophic nonunions) [10, 114].

Despite the correlation between chronic inflammatory conditions and increased fracture risk, the effects of inflammation on the mechanical integrity of the bone vary significantly. For example, osteoporosis and type 2 diabetes mellitus patients have increased fracture risk. However, while osteoporosis reduces BMD, patients with type 2 diabetes may have normal or even high volumetric BMD, despite suppressed bone formation [120]. These data indicate that different inflammatory conditions alter bone quality through distinct pathways and therefore cannot be monitored or assessed using the same standard measures.

Postmenopausal Osteoporosis

Following menopause, a sharp drop in estrogen levels causes the rate of bone resorption to exceed that of bone formation, resulting in net bone loss. As such, postmenopausal women are at significant risk for osteoporosis [121]. Estrogen inhibits the production of pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, and IL-17 by immune cells, including B cells, T cells, and macrophages [122]. Increased levels of pro-inflammatory cytokines in the serum are associated with low BMD and elevated fracture risk in postmenopausal women [123126]. Reducing systemic inflammation may be considered therapeutic for postmenopausal women. Administration of IL-1β (anakinra) or TNF-α (etanercept) inhibitors following acute estrogen withdrawal in postmenopausal women suppressed the normal rise in bone resorption markers in the serum [127].

Estrogen coordinates multiple organ systems that cross talk to maintain bone. For example, estrogen decline and bone loss in ovariectomized mice induces chronic low-grade inflammation (including elevated IL-17 and TNF-α) mediated by dendritic cell activation of memory T cells via IL-7 and IL-15. Ablating IL15RA in T cells prevented elevations in IL-17 and TNF-α and reversed the ovariectomy-induced bone loss [128••]. These and other studies suggest that the pro-inflammatory immune status of postmenopausal women likely contributes to the direct effects of estrogen withdrawal on bone cells to drive bone loss [122].

Rheumatoid Arthritis

Rheumatoid arthritis stimulates both local (focal or periarticular) and systemic bone loss [129, 130]. Osteitis (inflammation within the bone) is evident in patients with rheumatoid arthritis via MRI and can be used as a predictor for rheumatoid arthritis prognosis and progression [131]. Inflammation in the joint of rheumatic patients involves all compartments, including the synovial membrane, cartilage, and subchondral bone. Osteoporosis and rheumatoid arthritis severity is correlated with circulating levels of IL-6, IL-10, and C-reactive protein (CRP), indicating that local joint inflammation may drive systemic inflammation and bone loss [132]. Several current therapies for rheumatoid arthritis target pro-inflammatory cytokines such as TNF-α or block cytokine-mediated activation of the Janus kinases (JAK)–signal transducer and activator of transcription (STAT) signaling pathways [133].

Inflammatory Bowel Disease

Intestinal inflammation characteristic of IBD induces bone loss through secretion of pro-inflammatory cytokines including IL-1, IL-6, and TNF-α. Patients (particularly postmenopausal patients) with IBD are at risk for osteoporosis and fracture, primarily due to insufficient calcium, vitamin D, and vitamin K absorption [134]. The intestinal microbiota is also disrupted in IBD, which can lead to inflammatory bone loss [135137]. For example, deletion of toll-like receptor 9 (TLR9) alters the gut microbiota to induce low-grade chronic inflammation and bone resorption [138]. New therapies for osteoporosis based on modulation of the gut microbiota and the immuno-inflammatory response are under investigation, including modulation of T helper 17 (Th17) cells [139, 140].

Periodontitis

Periodontitis is inflammation of the tissue around the teeth and is associated with both local (alveolar) and systemic bone loss, as well as fracture [4, 141, 142]. Upon bacterial infection, there is sustained production of pro-inflammatory cytokines, causing a local immune reaction that stimulates osteoclastogenesis. Recent single-cell RNA sequencing experiments showed that, in response to acute inflammation, periodontal mesenchymal progenitor cells differentiate into osteoblasts, presumably to balance the osteoclastogenic response to inflammation [143••]. Ultimately, sustained periodontal inflammation causes lesions in the bone that painfully expose tooth roots and cause tooth loss. Like rheumatoid arthritis and IBD, patients with periodontitis have elevated serum levels of various pro-inflammatory factors, including TNF-α, IL-1, and IL-6 [144]. Recruiting regulatory T cells (Tregs) to the site of inflammation prevents periodontal bone loss in both murine and canine models [145].

In periodontitis and other chronic conditions, inflammation may be the key mechanistic link between the tissue-specific disease and systemic bone loss [146]. This is exemplified by a study in which inducing periodontitis in mice caused epigenetic changes to hematopoietic progenitor cells in the bone marrow as well as production of myeloid cells. Transplanting the periodontitis-induced bone marrow to naïve recipients increased the recipients’ inflammatory response and the disease severity of induced inflammatory arthritis [147••]. Therefore, an immune system “trained” to respond to inflammatory cues may predispose individuals to inflammatory comorbidities, including bone loss.

Glucocorticoids

Glucocorticoids reduce inflammation and are primary drugs for treatment of chronic inflammatory diseases, including those that induce bone loss. Ironically, glucocorticoid-induced osteoporosis is common and is associated with fracture risk in a dose-dependent manner [148]. Glucocorticoids both inhibit bone formation and stimulate bone resorption, with direct effects on osteoclasts, osteoblasts, and osteocytes [149]. Careful monitoring of patients using glucocorticoids with high fracture risk is warranted and usually necessitates co-treatment with anti-osteoporotic drugs, such as bisphosphonates [150, 151].

Inflammation in Bone Pain

In addition to increasing risk of fracture, inflammatory processes can also induce bone pain, significantly affecting quality of life. The periosteum, bone marrow, and the Haversian canals of cortical bone are highly innervated with sensory and sympathetic nerve fibers of the peripheral nervous system (PNS) that influence bone homeostasis and are sensitive to inflammatory stimuli [152, 153]. Two major ascending (or afferent) routes relay signals from the PNS to the central nervous system. The first consists of somatosensory neurons in the dorsal root ganglia that signal from the skeleton, skin, or muscle to the brain via the dorsal horn of the spinal cord about temperature, pain, and injury [154]. The second route consists of sensory ganglia that receive peripheral sensory stimuli in the cranial and vagal pathways. Afferent sensory nerve endings within the skeleton are responsible for detecting and transmitting the quality of pain to the brain.

During skeletal stress (i.e., mechanical distortion, local acidosis produced by proton release from osteoclasts and inflammatory cells, or an increase in intramedullary pressure), osteoblasts, osteoclasts, and immune cells produce growth factors and inflammatory mediators that affect sensory neuron activation, sprouting, and neuropeptide release. Activated small- and medium-sized neurons release neurotransmitters that act on skeletal cells such as osteoblasts, osteoclasts, and bone marrow adipocytes [155]. The release of these mediators at the site of stress can sensitize nerve endings, making Aδ-fibers and C-fibers easier to depolarize and thus highly sensitive to nociceptive stimuli [156]. Inflammatory mediators such as histamine, serotonin, bradykinin, and prostaglandin E2 can also cause sensitization of nociceptor sensory nerve endings [157159]. While excessive pain profoundly affects quality of life, pain is also crucially important to alert the body of injury or disease. In this way, inflammatory signaling in bone serves an essential role to maintain overall health.

New Targets for Alleviating Inflammatory Bone Loss

Modulation of the Osteo-immune Axis

Tissue engineering strategies are leveraging the osteo-immune axis to resolve chronic bone loss and enhance bone healing. Various efforts in skeletal regenerative medicine are targeted at modulating mesenchymal progenitor cells (MPC) [160162]. Application of MPCs from various sources, including adipose tissue, bone marrow, or dental pulp, aids in fracture repair in part by inhibiting chronic inflammation [162]. Pre-conditioning MPCs with inflammatory stimuli can enhance their osteogenic capabilities [10]. MPCs hold great promise in orthopedic therapies, but their success has been limited thus far in clinical trials. Improving MPC survival and response to a harsh inflammatory environment is key to their use in future clinical applications to aid bone healing or prevent bone loss [160].

Additional efforts are targeted at modulating the osteo-immune axis by delivering anti-inflammatory cytokines. Anti-inflammatory cytokines such as IL-4 prevent bone loss and accelerate bone formation in animal models of chronic inflammation and bone healing by polarizing macrophages to the M2 phenotype [163166]. The method of releasing these cytokines in the local bone environment, for example, in accelerating or aiding healing following fracture, is dependent on multiple factors. The appropriate scaffold must be used, taking into consideration the timing, dose, and duration of release [167, 168]. In addition, the appropriate cytokine or combination of cytokines must be considered. For example, IL-10 is generally considered pro-osteogenic at low concentrations, whereas higher concentrations of IL-10 have been shown to inhibit osteogenesis [81]. Additionally, circulating levels of IL-10 are negatively correlated with bone regeneration following traumatic orthopedic injury [169]. These results highlight that the concentration, timing, and type of injury all need to be considered when targeting the inflammatory axis to maintain bone health.

Alleviating Cellular Senescence-Induced Inflammation

One of the hallmarks of cellular senescence is production of inflammatory factors [170]. Senescent cells are beneficial in normal physiological functions, such as wound healing, when senescent cells attract and activate immune cells to initiate tissue remodeling [171]. However, chronic senescent cell accumulation contributes to a variety of pathologies across the lifespan, and particularly in aging [170]. Accumulation of senescent cells during aging is, in part, attributable to impaired surveillance of immune cells and chronic systemic inflammation [172]. Classic features of senescent cells include enlarged and flattened cell morphology, highly condensed chromatin, growth arrest, resistance to apoptosis, and development of a senescence-associated secretory phenotype (SASP). The SASP is composed of pro-inflammatory cytokines, chemokines, growth factors, and matrix-degrading proteases, and its release has a profound effect on the local environment surrounding the senescent cell.

Senescent cells induce local and systemic pro-inflammatory reactions that negatively impact bone [173]. Senescent cells and their associated SASP factors are present in the bone microenvironment of old mice [174], and reducing the systemic senescent cell burden prevents age-related bone loss [175, 176]. In an aged murine model of periodontal disease, senescent osteocytes exacerbate chronic inflammation, suppressing alveolar bone regeneration [177••]. Osteocyte senescence is also implicated in diabetic bone loss [178] and focal bone loss following radiation [179] in rodent models. Senescent osteocytes in aged human bone tissue induce inflammation that inhibits bone formation [180•].

Inflammatory senescence has negative effects on young bone as well. Glucocorticoids are widely prescribed for chronic inflammatory childhood illnesses, and glucocorticoid-induced osteoporosis is the most common form of secondary pediatric osteoporosis [181]. Glucocorticoid treatment of young mice induces vascular endothelial cell senescence in the metaphysis of long bone, and inhibiting this senescence improves angiogenesis and osteogenesis [182]. Glucocorticoids also induce senescence of adult bone marrow adipocytes, causing secondary senescence in surrounding bone tissue and inducing bone loss [183]. Eliminating senescent cells systemically [184] or in bone marrow adipocytes [183] prevents glucocorticoid-induced bone loss. Clearing transient senescent cells that accumulate following fracture also improves bone healing [185].

Conclusions

Inflammatory processes play a critical role in regulating bone homeostasis and disease. Systemic bone loss induced by inflammatory diseases, including rheumatoid arthritis, IBD, and periodontitis, contributes significantly to the worldwide osteoporosis crisis. While pro-inflammatory cytokines such as TNF-α and IL-1 have largely negative effects, inducing aberrant osteoclastogenesis and bone loss, pro-osteogenic cytokines such as IL-4 and IL-11 target osteoblasts to induce osteogenesis. New advances in osteoimmunology have isolated subtypes of bone cells that are the major players in inflammatory bone loss, including inflammatory osteoclasts and senescent osteocytes. Identification of inflammation-specific processes is crucial to identify therapies that can improve pathological inflammatory bone loss, while maintaining physiological bone turnover.

Funding

This work was supported by NIH K99AR080745 (SRW), T32DK007352 (HMT), R21AR82134 (JJW), T32AR056950 (MO), the Mayo Clinic Robert and Arlene Kogod Center on Aging (SRW), and the Mayo Clinic Graduate School of Biomedical Sciences (KMA).

Footnotes

Conflict of Interest The authors declare no competing interests.

Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, et al. The clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022;33(10):2049–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Khosla S, Cauley JA, Compston J, Kiel DP, Rosen C, Saag KG, et al. Addressing the crisis in the treatment of osteoporosis: a path forward. J Bone Miner Res. 2017;32(3):424–30. [DOI] [PubMed] [Google Scholar]
  • 3.Roux C, Briot K. The crisis of inadequate treatment in osteoporosis. Lancet Rheumatol. 2020;2(2):e110–9. [DOI] [PubMed] [Google Scholar]
  • 4.Redlich K, Smolen JS. Inflammatory bone loss: pathogenesis and therapeutic intervention. Nat Rev Drug Discov. 2012;11(3):234–50. [DOI] [PubMed] [Google Scholar]
  • 5.Brylka LJ, Schinke T. Chemokines in physiological and pathological bone remodeling. Front Immunol. 2019;10:2182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hardy R, Cooper MS. Bone loss in inflammatory disorders. Journal of Endocrinology. 2009;201(3):309–20. [DOI] [PubMed] [Google Scholar]
  • 7.Pavanelli ALR, de Menezes BS, Pereira EBB, de Souza Morais FA, Cirelli JA, de Molon RS. Pharmacological therapies for the management of inflammatory bone resorption in periodontal disease: a review of preclinical studies. Biomed Res Int. 2022;2022:5832009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li G, Li Z, Li L, Liu S, Wu P, Zhou M, et al. Stem cell-niche engineering via multifunctional hydrogel potentiates stem cell therapies for inflammatory bone loss. Adv Funct Mater. 2023;33(2):2209466. [Google Scholar]
  • 9.Epsley S, Tadros S, Farid A, Kargilis D, Mehta S, Rajapakse CS. The effect of inflammation on bone. Front Physiol. 2021;11:511799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Maruyama M, Rhee C, Utsunomiya T, Zhang N, Ueno M, Yao Z, et al. Modulation of the inflammatory response and bone healing. Front Endocrinol (Lausanne). 2020;11:386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Batoon L, Millard SM, Raggatt LJ, Pettit AR. Osteomacs and bone regeneration. Curr Osteoporos Rep. 2017;15(4):385–95. [DOI] [PubMed] [Google Scholar]
  • 12.Walters G, Pountos I, Giannoudis PV. The cytokines and microenvironment of fracture haematoma: current evidence. J Tissue Eng Regen Med. 2018;12(3):e1662–77. [DOI] [PubMed] [Google Scholar]
  • 13.Yang X, Ricciardi BF, Hernandez-Soria A, Shi Y, Pleshko Camacho N, Bostrom MP. Callus mineralization and maturation are delayed during fracture healing in interleukin-6 knockout mice. Bone. 2007;41(6):928–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gerstenfeld L, Cho T-J, Kon T, Aizawa T, Tsay A, Fitch J, et al. Impaired fracture healing in the absence of TNF-α signaling: the role of TNF-α in endochondral cartilage resorption. J Bone Mineral Res. 2003;18(9):1584–92. [DOI] [PubMed] [Google Scholar]
  • 15.Al Farii H, Farahdel L, Frazer A, Salimi A, Bernstein M. The effect of NSAIDs on postfracture bone healing: a meta-analysis of randomized controlled trials. OTA Int. 2021;4(2):e092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wheatley BM, Nappo KE, Christensen DL, Holman AM, Brooks DI, Potter BK. Effect of NSAIDs on bone healing rates: a meta-analysis. JAAOS J Amer Acad Orthop Surg. 2019;27(7):e330–6. [DOI] [PubMed] [Google Scholar]
  • 17.Lukač N, Katavić V, Novak S, Šućur A, Filipović M, Kalajzić I, et al. What do we know about bone morphogenetic proteins and osteochondroprogenitors in inflammatory conditions? Bone. 2020;137:115403. [DOI] [PubMed] [Google Scholar]
  • 18.Perrin S, Colnot C. Periosteal skeletal stem and progenitor cells in bone regeneration. Curr Osteoporos Rep. 2022;20(5):334–43. [DOI] [PubMed] [Google Scholar]
  • 19.Kushioka J, Chow SK, Toya M, Tsubosaka M, Shen H, Gao Q, et al. Bone regeneration in inflammation with aging and cell-based immunomodulatory therapy. Inflamm Regen. 2023;43(1):29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ambrosi TH, Marecic O, McArdle A, Sinha R, Gulati GS, Tong X, et al. Aged skeletal stem cells generate an inflammatory degenerative niche. Nature. 2021;597(7875):256–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ferretti C, Lucarini G, Andreoni C, Salvolini E, Bianchi N, Vozzi G, et al. Human periosteal derived stem cell potential: the impact of age. Stem Cell Rev Rep. 2015;11(3):487–500. [DOI] [PubMed] [Google Scholar]
  • 22.Durdan MM, Azaria RD, Weivoda MM. Novel insights into the coupling of osteoclasts and resorption to bone formation. Semin Cell Dev Biol. 2022;123:4–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Xiong J, O’Brien CA. Osteocyte RANKL: new insights into the control of bone remodeling. J Bone Miner Res. 2012;27(3):499–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kendler DL, Cosman F, Stad RK, Ferrari S. Denosumab in the treatment of osteoporosis: 10 years later: a narrative review. Adv Ther. 2022;39(1):58–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ono T, Hayashi M, Sasaki F, Nakashima T. RANKL biology: bone metabolism, the immune system, and beyond. Inflamm Regen. 2020;40(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mbalaviele G, Novack DV, Schett G, Teitelbaum SL. Inflammatory osteolysis: a conspiracy against bone. J Clin Investig. 2017;127(6):2030–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Walsh MC, Takegahara N, Kim H, Choi Y. Updating osteoimmunology: regulation of bone cells by innate and adaptive immunity. Nat Rev Rheumatol. 2018;14(3):146–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature. 2003;423(6937):337–42. [DOI] [PubMed] [Google Scholar]
  • 29.Hascoët E, Blanchard F, Blin-Wakkach C, Guicheux J, Lesclous P, Cloitre A. New insights into inflammatory osteoclast precursors as therapeutic targets for rheumatoid arthritis and periodontitis. Bone Res. 2023;11(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Madel M-B, Ibáñez L, Wakkach A, de Vries TJ, Teti A, Apparailly F, et al. Immune function and diversity of osteoclasts in normal and pathological conditions. Front Immunol. 2019;10:1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lapérine O, Blin-Wakkach C, Guicheux J, Beck-Cormier S, Lesclous P. Dendritic-cell-derived osteoclasts: a new game changer in bone-resorption-associated diseases. Drug Discov Today. 2016;21(9):1345–54. [DOI] [PubMed] [Google Scholar]
  • 32.Hasegawa T, Kikuta J, Sudo T, Matsuura Y, Matsui T, Simmons S, et al. Identification of a novel arthritis-associated osteoclast precursor macrophage regulated by FoxM1. Nat Immunol. 2019;20(12):1631–43. [DOI] [PubMed] [Google Scholar]
  • 33.Agemura T, Hasegawa T, Yari S, Kikuta J, Ishii M. Arthritis-associated osteoclastogenic macrophage, AtoM, as a key player in pathological bone erosion. Inflamm Regen. 2022;42(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Marahleh A, Kitaura H, Ohori F, Kishikawa A, Ogawa S, Shen W-R, et al. TNF-α directly enhances osteocyte RANKL expression and promotes osteoclast formation. Front Immunol. 2019;10:2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Luo G, Li F, Li X, Wang ZG, Zhang B. TNF-α and RANKL promote osteoclastogenesis by upregulating RANK via the NF-κB pathway. Mol Med Rep. 2018;17(5):6605–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lam J, Takeshita S, Barker JE, Kanagawa O, Ross FP, Teitelbaum SL. TNF-alpha induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand. J Clin Invest. 2000;106(12):1481–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Fuller K, Murphy C, Kirstein B, Fox SW, Chambers TJ. TNFα potently activates osteoclasts, through a direct action independent of and strongly synergistic with RANKL. Endocrinology. 2002;143(3):1108–18. [DOI] [PubMed] [Google Scholar]
  • 38.Xia Y, Inoue K, Du Y, Baker SJ, Reddy EP, Greenblatt MB, et al. TGFβ reprograms TNF stimulation of macrophages towards a non-canonical pathway driving inflammatory osteoclastogenesis. Nat Commun. 2022;13(1):3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kim JH, Jin HM, Kim K, Song I, Youn BU, Matsuo K, et al. The mechanism of osteoclast differentiation induced by IL-11. J Immunol. 2009;183(3):1862–70. [DOI] [PubMed] [Google Scholar]
  • 40.Lee YM, Fujikado N, Manaka H, Yasuda H, Iwakura Y. IL-1 plays an important role in the bone metabolism under physiological conditions. Int Immunol. 2010;22(10):805–16. [DOI] [PubMed] [Google Scholar]
  • 41.Shiratori T, Kyumoto-Nakamura Y, Kukita A, Uehara N, Zhang J, Koda K, et al. IL-1β Induces pathologically activated osteoclasts bearing extremely high levels of resorbing activity: a possible pathological subpopulation of osteoclasts, accompanied by suppressed expression of kindlin-3 and talin-1. J Immunol. 2018;200(1):218–28. [DOI] [PubMed] [Google Scholar]
  • 42.Wei S, Kitaura H, Zhou P, Ross FP, Teitelbaum SL. IL-1 mediates TNF-induced osteoclastogenesis. J Clin Investig. 2005;115(2):282–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wong PK, Quinn JM, Sims NA, van Nieuwenhuijze A, Campbell IK, Wicks IP. Interleukin-6 modulates production of T lymphocyte-derived cytokines in antigen-induced arthritis and drives inflammation-induced osteoclastogenesis. Arthritis Rheum. 2006;54(1):158–68. [DOI] [PubMed] [Google Scholar]
  • 44.Harmer D, Falank C, Reagan MR. Interleukin-6 interweaves the bone marrow microenvironment, bone loss, and multiple myeloma. Front Endocrinol. 2019;9:788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kim J-H, Sim JH, Lee S, Seol MA, Ye S-K, Shin HM, et al. Interleukin-7 induces osteoclast formation via STAT5, independent of receptor activator of NF-kappaB ligand. Front Immunol. 2017;8:1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Weitzmann MN, Cenci S, Rifas L, Brown C, Pacifici R. Interleukin-7 stimulates osteoclast formation by up-regulating the T-cell production of soluble osteoclastogenic cytokines. Blood. 2000;96(5):1873–8. [PubMed] [Google Scholar]
  • 47.Toraldo G, Roggia C, Qian W-P, Pacifici R, Weitzmann MN. IL-7 induces bone loss in vivo by induction of receptor activator of nuclear factor κB ligand and tumor necrosis factor α from T cells. Proc National Acad Sci. 2003;100(1):125–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lee Y. The role of interleukin-17 in bone metabolism and inflammatory skeletal diseases. BMB Rep. 2013;46(10):479–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Song L, Tan J, Wang Z, Ding P, Tang Q, Xia M, et al. Interleukin-17A facilitates osteoclast differentiation and bone resorption via activation of autophagy in mouse bone marrow macrophages. Mol Med Rep. 2019;19(6):4743–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Adamopoulos IE, Chao C-c, Geissler R, Laface D, Blumenschein W, Iwakura Y, et al. Interleukin-17A upregulates receptor activator of NF-κB on osteoclast precursors. Arthritis Res Ther. 2010;12(1): R29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tang H, Zhu S, Chen K, Yuan S, Hu J, Wang H. IL-17A regulates autophagy and promotes osteoclast differentiation through the ERK/mTOR/Beclin1 pathway. PLOS ONE. 2023;18(2):e0281845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami M, Kotake S, et al. Tumor necrosis factor alpha stimulates osteoclast differentiation by a mechanism independent of the ODF/RANKL-RANK interaction. J Exp Med. 2000;191(2):275–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mun SH, Jastrzebski S, Kalinowski J, Zeng S, Oh B, Bae S, et al. Sexual dimorphism in differentiating osteoclast precursors demonstrates enhanced inflammatory pathway activation in female cells. J Bone Mineral Res. 2021;36(6):1104–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Merrild DMH, Pirapaharan DC, Andreasen CM, Kjærsgaard-Andersen P, Møller AMJ, Ding M, et al. Pit- and trench-forming osteoclasts: a distinction that matters. Bone Res. 2015;3(1):15032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Borggaard XG, Pirapaharan DC, Delaissé JM, Søe K. Osteoclasts’ ability to generate trenches rather than pits depends on high levels of active cathepsin K and efficient clearance of resorption products. Int J Mol Sci. 2020;21(16):5924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Delaisse J-M, Søe K, Andersen TL, Rojek AM, Marcussen N. The mechanism switching the osteoclast from short to long duration bone resorption. Front Cell Dev Biol. 2021;9:644503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Vanderoost J, Søe K, Merrild DM, Delaissé JM, van Lenthe GH. Glucocorticoid-induced changes in the geometry of osteoclast resorption cavities affect trabecular bone stiffness. Calcif Tissue Int. 2013;92(3):240–50. [DOI] [PubMed] [Google Scholar]
  • 58.Søe K, Delaissé JM. Glucocorticoids maintain human osteoclasts in the active mode of their resorption cycle. J Bone Miner Res. 2010;25(10):2184–92. [DOI] [PubMed] [Google Scholar]
  • 59.Harrison KD, Hiebert BD, Panahifar A, Andronowski JM, Ashique AM, King GA, et al. Cortical bone porosity in rabbit models of osteoporosis. J Bone Miner Res. 2020;35(11):2211–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Saftig P, Hunziker E, Wehmeyer O, Jones S, Boyde A, Rommerskirch W, et al. Impaired osteoclastic bone resorption leads to osteopetrosis in cathepsin-K-deficient mice. Proc National Acad Sci. 1998;95(23):13453–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Li J, Sarosi I, Yan X-Q, Morony S, Capparelli C, Tan H-L, et al. RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc National Acad Sci. 2000;97(4):1566–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.••.Meirow Y, Jovanovic M, Zur Y, Habib J, Colombo DF, Twaik N, et al. Specific inflammatory osteoclast precursors induced during chronic inflammation give rise to highly active osteoclasts associated with inflammatory bone loss. Bone Res. 2022;10(1):36. [DOI] [PMC free article] [PubMed] [Google Scholar]; Defined subsets of inflammatory osteoclast precursors that are induced during chronic inflammation.
  • 63.••.Madel M-B, Ibáñez L, Ciucci T, Halper J, Rouleau M, Boutin A, et al. Dissecting the phenotypic and functional heterogeneity of mouse inflammatory osteoclasts by the expression of Cx3cr1. eLife. 2020;9:e54493. [DOI] [PMC free article] [PubMed] [Google Scholar]; Identified a subset of osteoclasts that are potently resorptive in response to inflammation.
  • 64.Ibáñez L, Abou-Ezzi G, Ciucci T, Amiot V, Belaïd N, Obino D, et al. Inflammatory osteoclasts prime TNFα-producing CD4(+) T cells and express CX(3) CR1. J Bone Miner Res. 2016;31(10):1899–908. [DOI] [PubMed] [Google Scholar]
  • 65.•.Madel M-B, Halper J, Ibáñez L, Claire L, Rouleau M, Boutin A, et al. Specific targeting of inflammatory osteoclastogenesis by the probiotic yeast S. boulardii CNCM I-745 reduces bone loss in osteoporosis. eLife. 2023;12:e82037. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrated that inflammatory osteoclasts can be specifically inhibited in vivo.
  • 66.Marriott I, Gray DL, Tranguch SL, Fowler VG Jr, Stryjewski M, Scott Levin L, et al. Osteoblasts express the inflammatory cytokine interleukin-6 in a murine model of Staphylococcus aureus osteomyelitis and infected human bone tissue. Am J Pathol. 2004;164(4):1399–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dapunt U, Giese T, Stegmaier S, Moghaddam A, Hänsch GM. The osteoblast as an inflammatory cell: production of cytokines in response to bacteria and components of bacterial biofilms. BMC0 Musculoskelet Disord. 2016;17(1):243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Greenfield EM, Gornik SA, Horowitz MC, Donahue HJ, Shaw SM. Regulation of cytokine expression in osteoblasts by parathyroid hormone: rapid stimulation of interleukin-6 and leukemia inhibitory factor mRNA. J Bone Miner Res. 1993;8(10):1163–71. [DOI] [PubMed] [Google Scholar]
  • 69.Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of osteoclast differentiation by cytokine networks. Immune Netw. 2018;18(1):e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gilbert L, He X, Farmer P, Boden S, Kozlowski M, Rubin J, et al. Inhibition of osteoblast differentiation by tumor necrosis factor-α*. Endocrinology. 2000;141(11):3956–64. [DOI] [PubMed] [Google Scholar]
  • 71.Xin W, Wang X, Zhang W, Zhu H, Dong R, Zhang J. Tumor necrosis factor-α inhibits bone marrow stem cell differentiation into osteoblasts by downregulating microRNA-34a expression. Ann Clin Lab Sci. 2019;49(3):324–9. [PubMed] [Google Scholar]
  • 72.Du D, Zhou Z, Zhu L, Hu X, Lu J, Shi C, et al. TNF-α suppresses osteogenic differentiation of MSCs by accelerating P2Y2 receptor in estrogen-deficiency induced osteoporosis. Bone. 2018;117:161–70. [DOI] [PubMed] [Google Scholar]
  • 73.Kaneki H, Guo R, Chen D, Yao Z, Schwarz EM, Zhang YE, et al. Tumor necrosis factor promotes Runx2 degradation through upregulation of Smurf1 and Smurf2 in osteoblasts. J Biol Chem. 2006;281(7):4326–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Diarra D, Stolina M, Polzer K, Zwerina J, Ominsky MS, Dwyer D, et al. Dickkopf-1 is a master regulator of joint remodeling. Nat Med. 2007;13(2):156–63. [DOI] [PubMed] [Google Scholar]
  • 75.Hengartner NE, Fiedler J, Ignatius A, Brenner RE. IL-1β inhibits human osteoblast migration. Mol Med. 2013;19(1):36–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Girasole G, Passeri G, Jilka RL, Manolagas SC. Interleukin-11: a new cytokine critical for osteoclast development. J Clin Invest. 1994;93(4):1516–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sims NA, Jenkins BJ, Nakamura A, Quinn JM, Li R, Gillespie MT, et al. Interleukin-11 receptor signaling is required for normal bone remodeling. J Bone Miner Res. 2005;20(7):1093–102. [DOI] [PubMed] [Google Scholar]
  • 78.Dong B, Hiasa M, Higa Y, Ohnishi Y, Endo I, Kondo T, et al. Osteoblast/osteocyte-derived interleukin-11 regulates osteogenesis and systemic adipogenesis. Nat Commun. 2022;13(1):7194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kido S, Kuriwaka-Kido R, Imamura T, Ito Y, Inoue D, Matsumoto T. Mechanical stress induces Interleukin-11 expression to stimulate osteoblast differentiation. Bone. 2009;45(6):1125–32. [DOI] [PubMed] [Google Scholar]
  • 80.Dresner-Pollak R, Gelb N, Rachmilewitz D, Karmeli F, Weinreb M. Interleukin 10-deficient mice develop osteopenia, decreased bone formation, and mechanical fragility of long bones. Gastroenterology. 2004;127(3):792–801. [DOI] [PubMed] [Google Scholar]
  • 81.Chen E, Liu G, Zhou X, Zhang W, Wang C, Hu D, et al. Concentration-dependent, dual roles of IL-10 in the osteogenesis of human BMSCs via P38/MAPK and NF-κB signaling pathways. Faseb J. 2018;32(9):4917–29. [DOI] [PubMed] [Google Scholar]
  • 82.Xiong Y, Yan C, Chen L, Endo Y, Sun Y, Zhou W, et al. IL-10 induces MC3T3-E1 cells differentiation towards osteoblastic fate in murine model. J Cell Mol Med. 2020;24(1):1076–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhang Q, Chen B, Yan F, Guo J, Zhu X, Ma S, et al. Interleukin-10 inhibits bone resorption: a potential therapeutic strategy in periodontitis and other bone loss diseases. Biomed Res Int. 2014;2014:284836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Tang M, Tian L, Luo G, Yu X. Interferon-gamma-mediated osteoimmunology. Front Immunol. 2018;9:1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Duque G, Huang DC, Dion N, Macoritto M, Rivas D, Li W, et al. Interferon-γ plays a role in bone formation in vivo and rescues osteoporosis in ovariectomized mice. J Bone Miner Res. 2011;26(7):1472–83. [DOI] [PubMed] [Google Scholar]
  • 86.Gao Y, Grassi F, Ryan MR, Terauchi M, Page K, Yang X, et al. IFN-gamma stimulates osteoclast formation and bone loss in vivo via antigen-driven T cell activation. J Clin Invest. 2007;117(1):122–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Steeve KT, Marc P, Sandrine T, Dominique H, Yannick F. IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev. 2004;15(1):49–60. [DOI] [PubMed] [Google Scholar]
  • 88.Taguchi Y, Yamamoto M, Yamate T, Lin SC, Mocharla H, DeTogni P, et al. Interleukin-6-type cytokines stimulate mesenchymal progenitor differentiation toward the osteoblastic lineage. Proc Assoc Am Physicians. 1998;110(6):559–74. [PubMed] [Google Scholar]
  • 89.McGregor NE, Murat M, Elango J, Poulton IJ, Walker EC, Crimeen-Irwin B, et al. IL-6 exhibits both cis- and trans-signaling in osteocytes and osteoblasts, but only trans-signaling promotes bone formation and osteoclastogenesis. J Biol Chem. 2019;294(19):7850–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Metzger CE, Narayanan SA. The role of osteocytes in inflammatory bone loss. Front Endocrinol. 2019;10:285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Choi JUA, Kijas AW, Lauko J, Rowan AE. The mechanosensory role of osteocytes and implications for bone health and disease states. Front Cell Dev Biol. 2022;9:770143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Qin L, Liu W, Cao H, Xiao G. Molecular mechanosensors in osteocytes. Bone. Research 2020;8(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Delgado-Calle J, Bellido T. The osteocyte as a signaling cell. Physiol Rev. 2022;102(1):379–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Fowler TW, Acevedo C, Mazur CM, Hall-Glenn F, Fields AJ, Bale HA, et al. Glucocorticoid suppression of osteocyte perilacunar remodeling is associated with subchondral bone degeneration in osteonecrosis. Sci Rep. 2017;7(1):44618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Pathak JL, Bakker AD, Luyten FP, Verschueren P, Lems WF, Klein-Nulend J, et al. Systemic inflammation affects human osteocyte-specific protein and cytokine expression. Calcif Tissue Int. 2016;98(6):596–608. [DOI] [PubMed] [Google Scholar]
  • 96.Wu Q, Zhou X, Huang D, Ji Y, Kang F. IL-6 Enhances osteocyte-mediated osteoclastogenesis by promoting jak2 and rankl activity in vitro. Cell Physiol Biochem. 2017;41(4):1360–9. [DOI] [PubMed] [Google Scholar]
  • 97.Kitaura H, Marahleh A, Ohori F, Noguchi T, Shen WR, Qi J, et al. Osteocyte-related cytokines regulate osteoclast formation and bone resorption. Int J Mol Sci. 2020;21(14):5169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Graves DT, Alshabab A, Albiero ML, Mattos M, Corrêa JD, Chen S, et al. Osteocytes play an important role in experimental periodontitis in healthy and diabetic mice through expression of RANKL. J Clin Periodontol. 2018;45(3):285–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Huang X, Xie M, Xie Y, Mei F, Lu X, Li X, et al. The roles of osteocytes in alveolar bone destruction in periodontitis. J Trans Med. 2020;18(1):479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Metzger CE, Narayanan A, Zawieja DC, Bloomfield SA. Inflammatory bowel disease in a rodent model alters osteocyte protein levels controlling bone turnover. J Bone Miner Res. 2017;32(4):802–13. [DOI] [PubMed] [Google Scholar]
  • 101.Fonseca Ó, Gomes MS, Amorim MA, Gomes AC. Cystic fibrosis bone disease: the interplay between CFTR dysfunction and chronic inflammation. Biomolecules. 2023;13(3):425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Briot K, Roux C. Inflammation, bone loss and fracture risk in spondyloarthritis. RMD Open. 2015;1(1):e000052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zhang L, Sun Y. Muscle-bone crosstalk in chronic obstructive pulmonary disease. Front Endocrinol (Lausanne). 2021;12:724911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rella V, Rotondo C, Altomare A, Cantatore FP, Corrado A. Bone involvement in systemic lupus erythematosus. Int J Mol Sci. 2022;23(10):5804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Uluçkan Ö, Jimenez M, Karbach S, Jeschke A, Graña O, Keller J, et al. Chronic skin inflammation leads to bone loss by IL-17–mediated inhibition of Wnt signaling in osteoblasts. Sci Trans Med. 2016;8(330):330ra37–ra37. [DOI] [PubMed] [Google Scholar]
  • 106.Marshall D, Johnell O, Wedel H. Meta-analysis of how well measures of bone mineral density predict occurrence of osteoporotic fractures. BMJ. 1996;312(7041):1254–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Cirovic A, Jadzic J, Djukic D, Djonic D, Zivkovic V, Nikolic S, et al. Increased cortical porosity, reduced cortical thickness, and reduced trabecular and cortical microhardness of the superolateral femoral neck confer the increased hip fracture risk in individuals with type 2 diabetes. Calcif Tissue Int. 2022;111(5):457–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Villette CC, Zhang J, Phillips ATM. Influence of femoral external shape on internal architecture and fracture risk. Biomech Model Mechanobiol. 2020;19(4):1251–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Whittier DE, Samelson EJ, Hannan MT, Burt LA, Hanley DA, Biver E, et al. Bone microarchitecture phenotypes identified in older adults are associated with different levels of osteoporotic fracture risk. J Bone Miner Res. 2022;37(3):428–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Shevroja E, Cafarelli FP, Guglielmi G, Hans D. DXA parameters, Trabecular Bone Score (TBS) and bone mineral density (BMD), in fracture risk prediction in endocrine-mediated secondary osteoporosis. Endocrine. 2021;74(1):20–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Florez H, Hernandez-Rodriguez J, Muxi A, Carrasco JL, Prieto-Gonzalez S, Cid MC, et al. Trabecular bone score improves fracture risk assessment in glucocorticoid-induced osteoporosis. Rheumatology (Oxford). 2020;59(7):1574–80. [DOI] [PubMed] [Google Scholar]
  • 112.Zuchowski P, Dura M, Jeka D, Waszczak-Jeka M. The applicability of trabecular bone score for osteoporosis diagnosis in ankylosing spondylitis. Rheumatol Int. 2022;42(5):839–46. [DOI] [PubMed] [Google Scholar]
  • 113.Tamaki J, Iki M, Sato Y, Winzenrieth R, Kajita E, Kagamimori S, et al. Does Trabecular Bone Score (TBS) improve the predictive ability of FRAX((R)) for major osteoporotic fractures according to the Japanese Population-based Osteoporosis (JPOS) cohort study? J Bone Miner Metab. 2019;37(1):161–70. [DOI] [PubMed] [Google Scholar]
  • 114.Kushioka J, Chow SK-H, Toya M, Tsubosaka M, Shen H, Gao Q, et al. Bone regeneration in inflammation with aging and cell-based immunomodulatory therapy. Inflamm Regen. 2023;43(1):29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Clark D, Nakamura M, Miclau T, Marcucio R. Effects of Aging on Fracture Healing. Curr Osteoporos Rep. 2017;15(6):601–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Pajarinen J, Lin T, Gibon E, Kohno Y, Maruyama M, Nathan K, et al. Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials. 2019;196:80–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Josephson AM, Bradaschia-Correa V, Lee S, Leclerc K, Patel KS, Muinos Lopez E, et al. Age-related inflammation triggers skeletal stem/progenitor cell dysfunction. Proc National Acad Sci. 2019;116(14):6995–7004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lopez EM, Leclerc K, Ramsukh M, Parente PEL, Patel K, Aranda CJ, et al. Modulating the systemic and local adaptive immune response after fracture improves bone regeneration during aging. Bone. 2022;157:116324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Josephson AM, Leclerc K, Remark LH, Lopeź EM, Leucht P. Systemic NF-κB-mediated inflammation promotes an aging phenotype in skeletal stem/progenitor cells. Aging (Albany NY). 2021;13(10):13421–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Piccoli A, Cannata F, Strollo R, Pedone C, Leanza G, Russo F, et al. Sclerostin regulation, microarchitecture, and advanced glycation end-products in the bone of elderly women with type 2 diabetes. J Bone Miner Res. 2020;35(12):2415–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Khosla S, Oursler MJ, Monroe DG. Estrogen and the skeleton. Trends Endocrinol Metab. 2012;23(11):576–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin Cell Dev Biol. 2022;123:14–21. [DOI] [PubMed] [Google Scholar]
  • 123.Ilesanmi-Oyelere BL, Schollum L, Kuhn-Sherlock B, McConnell M, Mros S, Coad J, et al. Inflammatory markers and bone health in postmenopausal women: a cross-sectional overview. Immun Ageing. 2019;16:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Damani JJ, De Souza MJ, Strock NCA, Koltun KJ, Williams NI, Weaver C, et al. Associations between inflammatory mediators and bone outcomes in postmenopausal women: a cross-sectional analysis of baseline data from the Prune Study. J Inflamm Res. 2023;16:639–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Al-Daghri NM, Aziz I, Yakout S, Aljohani NJ, Al-Saleh Y, Amer OE, et al. Inflammation as a contributing factor among postmenopausal Saudi women with osteoporosis. Medicine (Baltimore). 2017;96(4):e5780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Chow SK, Chim YN, Wang JY, Wong RM, Choy VM, Cheung WH. Inflammatory response in postmenopausal osteoporotic fracture healing. Bone Joint Res. 2020;9(7):368–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Charatcharoenwitthaya N, Khosla S, Atkinson EJ, McCready LK, Riggs BL. Effect of blockade of TNF-alpha and interleukin-1 action on bone resorption in early postmenopausal women. J Bone Miner Res. 2007;22(5):724–9. [DOI] [PubMed] [Google Scholar]
  • 128.••.Cline-Smith A, Axelbaum A, Shashkova E, Chakraborty M, Sanford J, Panesar P, et al. Ovariectomy activates chronic low-grade inflammation mediated by memory t cells, which promotes osteoporosis in mice. J Bone Miner Res. 2020;35(6):1174–87. [DOI] [PMC free article] [PubMed] [Google Scholar]; Described how estrogen depletion induces low-grade inflammation, specifically affecting a subset of memory T-cells, to induce bone loss.
  • 129.Sakthiswary R, Uma Veshaaliini R, Chin K-Y, Das S, Sirasanagandla SR. Pathomechanisms of bone loss in rheumatoid arthritis. Front Med. 2022;9:962969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Goldring SR, Gravallese EM. Mechanisms of bone loss in inflammatory arthritis: diagnosis and therapeutic implications. Arthritis Res. 2000;2(1):33–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rubin DA. MRI and ultrasound of the hands and wrists in rheumatoid arthritis. I. Imaging findings. Skelet Radiol 2019;48(5):677–95. [DOI] [PubMed] [Google Scholar]
  • 132.Yong W, Hongbin L, Jing W, Jing Z, Ning T, Lijie B. Associations of changes in serum inflammatory factors, MMP-3, 25(OH) D and intestinal flora with osteoporosis and disease activity in rheumatoid arthritis patients. Clin Lab. 2020;66(12). 10.7754/Clin.Lab.2020.200242. [DOI] [PubMed] [Google Scholar]
  • 133.Wu C-Y, Yang H-Y, Lai J-H. Potential therapeutic targets beyond cytokines and Janus kinases for autoimmune arthritis. Biochemical Pharmacology. 2023;213:115622. [DOI] [PubMed] [Google Scholar]
  • 134.Chedid VG, Kane SV. Bone health in patients with inflammatory bowel diseases. J Clin Densitom. 2020;23(2):182–9. [DOI] [PubMed] [Google Scholar]
  • 135.Yu M, Malik Tyagi A, Li JY, Adams J, Denning TL, Weitzmann MN, et al. PTH induces bone loss via microbial-dependent expansion of intestinal TNF(+) T cells and Th17 cells. Nat Commun. 2020;11(1):468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Yu M, Pal S, Paterson CW, Li JY, Tyagi AM, Adams J, et al. Ovariectomy induces bone loss via microbial-dependent trafficking of intestinal TNF+ T cells and Th17 cells. J Clin Invest. 2021;131(4):e143137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Chen Y, Wang X, Zhang C, Liu Z, Li C, Ren Z. Gut microbiota and bone diseases: a growing partnership. Front Microbiol. 2022;13:877776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ding P, Tan Q, Wei Z, Chen Q, Wang C, Qi L, et al. Toll-like receptor 9 deficiency induces osteoclastic bone loss via gut microbiota-associated systemic chronic inflammation. Bone Res. 2022;10(1):42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Lyu Z, Hu Y, Guo Y, Liu D. Modulation of bone remodeling by the gut microbiota: a new therapy for osteoporosis. Bone Res. 2023;11(1):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhu L, Hua F, Ding W, Ding K, Zhang Y, Xu C. The correlation between the Th17/Treg cell balance and bone health. Immun Ageing. 2020;17(1):30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Luo X, Wan Q, Cheng L, Xu R. Mechanisms of bone remodeling and therapeutic strategies in chronic apical periodontitis. Front Cell Infect Microbiol. 2022;12:908859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Hong S-J, Yang B-E, Yoo D-M, Kim S-J, Choi H-G, Byun S-H. Analysis of the relationship between periodontitis and osteoporosis/fractures: a cross-sectional study. BMC Oral Health. 2021;21(1):125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.••.Fan Y, Lyu P, Bi R, Cui C, Xu R, Rosen CJ, et al. Creating an atlas of the bone microenvironment during oral inflammatory-related bone disease using single-cell profiling. eLife. 2023;12:e82537. [DOI] [PMC free article] [PubMed] [Google Scholar]; Developed a single-cell atlas of alveolar bone in healthy and inflammatory disease states. Showed that mesenchymal progenitor cells have osteogenic potential in response to acute periodontal inflammation.
  • 144.Górska R, Gregorek H, Kowalski J, Laskus-Perendyk A, Syczewska M, Madaliński K. Relationship between clinical parameters and cytokine profiles in inflamed gingival tissue and serum samples from patients with chronic periodontitis. J Clin Periodontol. 2003;30(12):1046–52. [DOI] [PubMed] [Google Scholar]
  • 145.Glowacki AJ, Yoshizawa S, Jhunjhunwala S, Vieira AE, Garlet GP, Sfeir C, et al. Prevention of inflammation-mediated bone loss in murine and canine periodontal disease via recruitment of regulatory lymphocytes. Proc National Acad Sci. 2013;110(46):18525–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yu B, Wang C-Y. Osteoporosis and periodontal diseases – an update on their association and mechanistic links. Periodontol 2000. 2022;89(1):99–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.••.Li X, Wang H, Yu X, Saha G, Kalafati L, Ioannidis C, et al. Maladaptive innate immune training of myelopoiesis links inflammatory comorbidities. Cell. 2022;185(10):1709–27.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrated that systemic inflammation alters progenitor cell epigenetic programming, increasing production of myeloid cells prepared to respond to inflammation. This work shows that inflammatory comorbidities may be a result of impaired innate immune training.
  • 148.Compston J. Glucocorticoid-induced osteoporosis: an update. Endocrine. 2018;61(1):7–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hardy RS, Zhou H, Seibel MJ, Cooper MS. Glucocorticoids and bone: consequences of endogenous and exogenous excess and replacement therapy. Endocrine Rev. 2018;39(5):519–48. [DOI] [PubMed] [Google Scholar]
  • 150.Güler-Yüksel M, Hoes JN, Bultink IEM, Lems WF. Glucocorticoids, inflammation and bone. Calcif Tissue Int. 2018;102(5):592–606. [DOI] [PubMed] [Google Scholar]
  • 151.Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, et al. 2017 American College of Rheumatology guideline for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Rheumatol. 2017;69(8):1521–37. [DOI] [PubMed] [Google Scholar]
  • 152.Steverink JG, Oostinga D, van Tol FR, van Rijen MHP, Mackaaij C, Verlinde-Schellekens SAMW, et al. Sensory innervation of human bone: an immunohistochemical study to further understand bone pain. J Pain. 2021;22(11):1385–95. [DOI] [PubMed] [Google Scholar]
  • 153.Pongratz G, Straub RH. The sympathetic nervous response in inflammation. Arthritis Res Ther. 2014;16(6):504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lv X, Gao F, Cao X. Skeletal interoception in bone homeostasis and pain. Cell Metab. 2022;34(12):1914–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Brazill JM, Beeve AT, Craft CS, Ivanusic JJ, Scheller EL. Nerves in bone: evolving concepts in pain and anabolism. J Bone Miner Res. 2019;34(8):1393–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Cook AD, Christensen AD, Tewari D, McMahon SB, Hamilton JA. Immune cytokines and their receptors in inflammatory pain. Trends in Immunology. 2018;39(3):240–55. [DOI] [PubMed] [Google Scholar]
  • 157.Nencini S, Ringuet M, Kim D-H, Chen Y-J, Greenhill C, Ivanusic JJ. Mechanisms of nerve growth factor signaling in bone nociceptors and in an animal model of inflammatory bone pain. Molec Pain. 2017;13:1744806917697011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Nencini S, Ringuet M, Kim D-H, Greenhill C, Ivanusic JJ. GDNF, neurturin, and artemin activate and sensitize bone afferent neurons and contribute to inflammatory bone pain. J Neurosci. 2018;38(21):4899–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Enomoto M, Mantyh PW, Murrell J, Innes JF, Lascelles BDX. Anti-nerve growth factor monoclonal antibodies for the control of pain in dogs and cats. Vet Rec. 2019;184(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Noronha NdC, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JLM, Covas DT, et al. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther. 2019;10(1):131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Han Y, Yang J, Fang J, Zhou Y, Candi E, Wang J, et al. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Sig Transduct Target Ther. 2022;7(1):92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kangari P, Talaei-Khozani T, Razeghian-Jahromi I, Razmkhah M. Mesenchymal stem cells: amazing remedies for bone and cartilage defects. Stem Cell Res Ther. 2020;11(1):492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Rao AJ, Nich C, Dhulipala LS, Gibon E, Valladares R, Zwingen-berger S, et al. Local effect of IL-4 delivery on polyethylene particle induced osteolysis in the murine calvarium. J Biomed Mater Res A. 2013;101(7):1926–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Sato T, Pajarinen J, Behn A, Jiang X, Lin TH, Loi F, et al. The effect of local IL-4 delivery or CCL2 blockade on implant fixation and bone structural properties in a mouse model of wear particle induced osteolysis. J Biomed Mater Res A. 2016;104(9):2255–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Zhang J, Shi H, Zhang N, Hu L, Jing W, Pan J. Interleukin-4-loaded hydrogel scaffold regulates macrophages polarization to promote bone mesenchymal stem cells osteogenic differentiation via TGF-β1/Smad pathway for repair of bone defect. Cell Proliferation. 2020;53(10):e12907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zhao DW, Ren B, Wang HW, Zhang X, Yu MZ, Cheng L, et al. 3D-printed titanium implant combined with interleukin 4 regulates ordered macrophage polarization to promote bone regeneration and angiogenesis. Bone Joint Res. 2021;10(7):411–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Zhu G, Zhang T, Chen M, Yao K, Huang X, Zhang B, et al. Bone physiological microenvironment and healing mechanism: basis for future bone-tissue engineering scaffolds. Bioact Mater. 2021;6(11):4110–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Wang Y, Feng Z, Liu X, Yang C, Gao R, Liu W, et al. Titanium alloy composited with dual-cytokine releasing polysaccharide hydrogel to enhance osseointegration via osteogenic and macrophage polarization signaling pathways. Regen Biomater. 2022;9:rbac003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Cheng A, Vantucci CE, Krishnan L, Ruehle MA, Kotanchek T, Wood LB, et al. Early systemic immune biomarkers predict bone regeneration after trauma. Proc Natl Acad Sci USA. 2021;118(8):e2017889118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–96. [DOI] [PubMed] [Google Scholar]
  • 172.Ovadya Y, Landsberger T, Leins H, Vadai E, Gal H, Biran A, et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat Commun. 2018;9(1):5435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Zhu R, Wan H, Yang H, Song M, Chai Y, Yu B. The role of senescence-associated secretory phenotype in bone loss. Front Cell Dev Biol. 2022;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Farr JN, Fraser DG, Wang H, Jaehn K, Ogrodnik MB, Weivoda MM, et al. Identification of senescent cells in the bone microenvironment. J Bone Miner Res. 2016;31(11):1920–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Farr JN, Xu M, Weivoda MM, Monroe DG, Fraser DG, Onken JL, et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med. 2017;23(9):1072–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Farr JN, Saul D, Doolittle ML, Kaur J, Rowsey JL, Vos SJ, et al. Local senolysis in aged mice only partially replicates the benefits of systemic senolysis. J Clin Invest. 2023;133(8):e162519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.••.Aquino-Martinez R, Eckhardt BA, Rowsey JL, Fraser DG, Khosla S, Farr JN, et al. Senescent cells exacerbate chronic inflammation and contribute to periodontal disease progression in old mice. J Periodontol. 2021;92(10):1483–95. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrated that senescent osteocytes exacerbate chronic inflammation and cause alveolar bone destruction in periodontitis.
  • 178.Eckhardt BA, Rowsey JL, Thicke BS, Fraser DG, O’Grady KL, Bondar OP, et al. Accelerated osteocyte senescence and skeletal fragility in mice with type 2 diabetes. JCI Insight. 2020;5(9):e135236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Chandra A, Lagnado AB, Farr JN, Monroe DG, Park S, Hachfeld C, et al. Targeted reduction of senescent cell burden alleviates focal radiotherapy-related bone loss. J Bone Miner Res. 2020;35(6):1119–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.•.Wang Z, Zhang X, Cheng X, Ren T, Xu W, Li J, et al. Inflammation produced by senescent osteocytes mediates age-related bone loss. Frontiers in Immunology. 2023;14:1114006. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrated that proteins associated with inflammation were increased in elderly human bone tissue which was enriched for senescent osteocytes.
  • 181.Wan M, Gray-Gaillard EF, Elisseeff JH. Cellular senescence in musculoskeletal homeostasis, diseases, and regeneration. Bone Res. 2021;9(1):41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Liu X, Chai Y, Liu G, Su W, Guo Q, Lv X, et al. Osteoclasts protect bone blood vessels against senescence through the angiogenin/plexin-B2 axis. Nat Commun. 2021;12(1):1832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Liu X, Gu Y, Kumar S, Amin S, Guo Q, Wang J, et al. Oxylipin-PPARγ-initiated adipocyte senescence propagates secondary senescence in the bone marrow. Cell Metab. 2023;35(4):667–84. e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wang T, Yang L, Liang Z, Wang L, Su F, Wang X, et al. Targeting cellular senescence prevents glucocorticoid-induced bone loss through modulation of the DPP4-GLP-1 axis. Sig Transduct Target Ther. 2021;6(1):143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Saul D, Monroe DG, Rowsey JL, Kosinsky RL, Vos SJ, Doolittle ML, et al. Modulation of fracture healing by the transient accumulation of senescent cells. eLife. 2021;10:e69958. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES