Skip to main content

Mesenchymal stem cells and dental implant osseointegration during aging: from mechanisms to therapy

Abstract

Dental implants are widely used to replace missing teeth, providing patients with unparalleled levels of effectiveness, convenience, and affordability. The biological basis for the clinical success of dental implants is osseointegration. Bone aging is a high-risk factor for the reduced osseointegration and survival rates of dental implants. In aged individuals, mesenchymal stem cells (MSCs) in the bone marrow show imbalanced differentiation with a reduction in osteogenesis and an increase in adipogenesis. This leads to impaired osseointegration and implant failure. This review focuses on the molecular mechanisms underlying the dysfunctional differentiation of aged MSCs, which primarily include autophagy, transcription factors, extracellular vesicle secretion, signaling pathways, epigenetic modifications, microRNAs, and oxidative stress. Furthermore, this review addresses the pathological changes in MSCs that affect osseointegration and discusses potential therapeutic interventions to enhance osseointegration by manipulating the mechanisms underlying MSC aging.

Introduction

The rapid increase in the aging population has become a concern worldwide; the global population aged ≥ 65 years is estimated to reach approximately 1.5 billion by 2050 [1]. With a surge in the aged population, an increased number of patients will face dental issues, requiring tooth replacement [2]. Tooth loss not only impairs the ability to chew and grind food but also makes it difficult to speak and modifies the facial features [3]. For decades, dental implants and implant-supported prostheses have been clinically applied as the most effective methods for oral rehabilitation of partially or fully edentulous patients. Notably, the long-term survival rate is 93.3–97% [4, 5]. The prerequisite for clinical success of dental implants is osseointegration, defined as a direct structural and functional connection between the bone and the surface of the implant [6, 7]. Elderly patients undergoing implant surgery are at higher risk compared to younger patients owing to their age-related health conditions [8]. With an increase in age, bone quality and quantity deteriorate, leading to osteoporosis [9]. Senescent bone marrow mesenchymal stem cells (BMMSCs) have impaired osteogenic differentiation and increased adipogenic differentiation abilities and play key roles in bone aging [10]. Differences in bone metabolism and biological properties of MSCs between elderly and young patients could result in differences in osseointegration patterns and success rate. The present review aims to provide an overview of the existing literature on the molecular mechanisms of MSC and bone aging and their effect on osseointegration. Based on this information, possible techniques to enhance osseointegration in aged population are discussed.

Osseointegration and aging

In the 1960s, Professor P. I. Branemark laid the foundation of modern implant dentistry by discovering the phenomenon of direct bone-to-implant contact; he termed it as “osseointegration” [6]. During the same period, Professor Andre Schroeder reported direct bone-to-implant contact and soft tissue reaction to titanium [11, 12]. Osseointegration is the direct structural and functional connection between the dental implant surface and the living bone without intervening soft tissue.

Titanium implants need to go through a cascade of healing events before they are “accepted” by the body tissues. The implantation of biomaterials results in injury and initiation of the inflammatory response regardless of the method of introduction of the biomaterial into the body [13, 14]. An immune response occurs, as biomaterial insertion leads to the disruption of the host’s tissue [15]. Accordingly, osseointegration can be perceived as an immune-modulated inflammatory process, wherein the immune system is locally regulated, thus influencing the whole healing process. The osseointegration process includes several phases post implant placement. Initially, titanium implant surface causes surface protein absorption, followed by coagulation and complement system activation. Then, monocytes are recruited and differentiate to macrophages to modulate the immune response. MSCs are recruited and differentiate to osteoblasts and osteocytes to deposit collagen matrix and form new bone. Finally, the peri-implant interface is completely replaced by a mature lamellar bone [16, 17] (Fig. 1).

Fig. 1
figure 1

Schematic representation of the osseointegration process. Implant placement triggers the following events to initiate osseointegration. Titanium implant surface causes protein adsorption, followed by coagulation and complement system activation. Monocytes differentiate into macrophages and control the immune response; MSCs are recruited in an accurate balance and commit to bone-forming cells, leading to bone formation. Created with http://BioRender.com

Bone aging is characterized by decreased osteogenesis and increased adipogenesis[10]. Whether bone aging is a negative factor in implant osseointegration has gained interest among the research communities. However, studies have reported contrasting results regarding the success and failure of dental implants in both patients and animals with bone aging. It has been reported that patients with advanced age receiving dental implants could have excellent implants survival rates and low periodontal disease index scores. A systematic review and meta-analysis reported high implant survival rates in patients aged over 75 years (97.3% for 1 year and 96.1% for 5 years) [18]. In 1998, 39 older patients who had 190 implants supporting 45 oral prostheses and 43 younger adults who had 184 implants supporting 45 oral prostheses were compared after being monitored for a period of 4–16 years. The data showed an implant success rate of 92.0% for the older group compared to an success rate of 86.5% for the younger group; however, no statistical significance was observed [19]. Moreover, a prospective study concluded that the clinical performance of mandibular two-implant overdentures is equally successful in younger and older patients after 10 years [20]. In contrast, Alsaadi et al. reported a positive relation between osteoporosis and early implant failure in a retrospective study [21]. Similarly, a systemic review concluded that osteoporotic subjects presented higher rates of implant loss [22]. Moreover, Niedermaier et al. studied survival rates of implant-supported dentures in patients with osteoporosis for up to 7 years and revealed increased rates of implant failure in these patients [23]. A multicenter study reported the lowest success rate in older patients after 6 years for 1022 implants placed in 440 patients [24]. Bertl et al. [25] reported that patients ≥ 80 years old showed a higher rate of early implant loss than younger patients. A retrospective, cross-sectional, matched sample study concluded that osteoporosis has a significant influence on peri-implant marginal bone level at the mesial and the distal implant aspect in postmenopausal women [26]. An animal study revealed that age-related estrogen deficiency in rats negatively influences preexisting bone around titanium implants in both the cortical and cancellous bones [27]. Collectively, it can be concluded that the osseointegration process is compromised in aged individuals, which may cause implant failure. Therefore, therapies enhancing the osseointegration process in the aged population should be developed.

Strategies to improve osseointegration in aged individuals

Various methods have been used to improve osseointegration in the aged population. These include systemic and local application of certain drugs and modification of the implant surface through novel techniques, such as varying the topography, applying coatings, or combining both of these. Systemic administration of bone-regulating hormones, such as calcitonin, parathyroid hormone (PTH), and estrogen, significantly improved bone formation and implant anchorage in osteoporotic rats [28,29,30,31]. Strontium ranelate and simvastatin improve titanium implant osseointegration by promoting bone formation and inhibiting bone absorption through various signaling pathways [32]. Currently, the common sand blasting and acid etching strategy is widely applied to increase the surface roughness of implants, which enhances biological properties of MSCs and osseointegration process [33]. In addition to the design of implant surface, it is necessary to promote osteoinduction and inhibit bone resorption locally in osteoporotic patients. Implants coated with bioactive agents, such as anti-osteoporosis drugs [34, 35], bioactive molecules [36, 37], or bioactive inorganic elements [38, 39] with proper controlled release have been reported to be able to improve osseointegration in osteoporotic individuals. Although numerous methods have been developed to achieve favorable osseointegration in osteoporotic conditions, the exact underlying biological mechanisms of how bone aging affects the osseointegration process and possible methods to improve osseointegration based on molecular mechanisms of bone aging have not been summarized before.

MSCs in bone aging

MSCs are mesoderm-derived progenitor cells that can be obtained from a wide range of tissues such as bone marrow, adipose tissue, umbilical cord, muscle, and dental tissue. They have the ability to self-renew and differentiate into various mesodermal cell types, including osteoblasts, adipocytes, and chondrocytes [40]. During osseointegration, followed by MSCs recruitment to the bone remodeling site, these cells proliferate and commit to pre-osteoblasts, further maturing into osteoblasts, which are involved in initial matrix secretion, maturation, and mineralization. At the end of the bone-forming stage, osteoblasts can have one of the following fates: develop into osteocytes embedded in the mineralized bone matrix, inactivate to quiescent bone-lining cells, or undergo apoptosis (Fig. 1) [16, 17, 41]. During bone aging, BMMSCs population declines and show reduced osteogenic differentiation and increased adipogenic differentiation [42, 43]. This could be a possible reason for impaired osseointegration and implant failure. However, the signaling pathways driving this pathological shift remain elusive and are currently under thorough investigation. Understanding the molecular mechanisms governing the dysregulated osteogenesis of aged MSCs is crucial for developing new treatments to promote bone formation and enhance osseointegration. Here, we summarize the pathological and molecular changes governing the biased differentiation of BMMSCs from various aspects, including autophagy, transcription factors, EV secretion, signaling pathways, epigenetic modifications, microRNAs, and oxidative stress. Furthermore, the effects of pathological changes in MSCs on osseointegration and possible interventions at the molecular level, such as modifications of implants and their utility in treatment-based applications, are discussed (Fig. 2).

Fig. 2
figure 2

Schematic representation of the potential uses of the mechanisms of MSC aging in improvement of osseointegration. Molecular changes associated with age-related changes in MSCs from varying perspectives include autophagy, transcription factors, EV secretion, signaling pathways, epigenetic modifications, microRNAs, and oxidative stress. Possible interventions at the molecular level to improve osseointegration include systemic administration, local delivery, and surface modification. Created with http://BioRender.com

Osseointegration and aging: lessons from MSCs

Autophagy

Autophagy is a cellular process through which redundant components, such as proteins and damaged mitochondria, are engulfed and delivered to lysosomes for degradation and recycling to maintain cellular homeostasis [44]. Reduced autophagy is a hallmark of aging in both cells and tissues. The expression of several autophagy-related genes is significantly reduced in various aged cells and tissues compared to that in their younger counterparts [45, 46]. Activation of autophagy by rapamycin, an inhibitor of the mTOR pathway, has been shown to increase the life expectancy of mice when fed them to late in life [47] or for a short interval during mid-life [48]. BMMSCs from aged mice present impaired autophagy compared to those from young mice. Furthermore, stimulation of autophagy by rapamycin not only enhances the osteogenic differentiation of aged BMMSCs in vitro but also restores the bone mineral density of senile osteoporotic aged mice [49].

Recently, it was revealed that during the process of implant osseointegration, several modifications in the topography of dental implants could promote osseointegration by modulating autophagy of the surrounding cells [50, 51]. Furthermore, metformin treatment promotes implant osseointegration under osteoporotic conditions by increasing autophagy and the osteogenesis of MSCs [52]. Moreover, multifunctional hydrogels fabricated from dynamic crosslinking of synthetic polymers, natural polymers, and silver nanowires to deliver rapamycin significantly improve osseointegration in vivo and restore degenerative cellular properties of BMMSCs in vitro in osteoporotic models by increasing autophagy [53]. Collectively, the up-regulation of autophagy by different methods, including implant modification and drug application, could be a promising mechanism to enhance osseointegration in the aged population.

Transcription factors

Numerous transcription factors have been identified as critical mediators involved in the aging process and osteogenic differentiation [54]. Recent studies have shown that the expression of transcription factors, including FOXP1, MAF, CBFβ, and SATB2, could potentially affect and attenuate senescence-related changes, including reduced bone mineral density, decreased trabecular thickness, and increased bone marrow adipogenic differentiation [55,56,57,58]. In this context, elucidation of the links between transcription factors and osseointegration may aid in understanding bone metabolism, optimizing aging-relevant skeletal disease therapies, and broadening the applications of dental implants.

FOXP1

Forkhead box P1 (FOXP1) is a critical transcription factor that participates in and affects multiple biological processes. It plays a vital role in defining the switch of MSCs from osteogenic to adipogenic differentiation by binding to the proliferator-activated receptor γ2 (PPARγ2) promoter [59, 60]. Li et al. recently reported age-correlated FOXP1 declination in BMMSCs as a cause of up-regulated adiposity and down-regulated bone mass, which may function through the inhibition of p16 [61]. Specifically, conditional ablation of FOXP1 in MSCs is associated with up-regulation of the CEBPβ/δ complex and dysregulation of recombination signal binding protein for immunoglobulin κ J region (RBPjκ), which further results in a reduced regenerative capacity of MSCs in vivo [55]. These studies preliminarily demonstrate the role of FOXP1 in regulating the fate of MSCs and age-dependent bone metabolism [62]. However, further studies are required to elucidate the underlying mechanism connecting FOXP1 and osseointegration.

MAF

MAF bZIP transcription factor (MAF) is a regulatory factor in the development of multiple tissues and immune system. Additionally, it is involved in senescence-dependent MSC differentiation [10]. Nishikawa revealed the age-dependent declination in Maf expression in murine MSCs, which affected the osteoblast/adipocyte bifurcation in MSCs by interfering with the osteogenesis mediator Runx2 and the adipogenesis regulator Pparg. [58]. Furthermore, strontium can attenuate age-related bone loss, characteristic of decreased bone mineral density and trabecular thickness, by targeting Maf [63, 64]. Similarly, Zn2 + /Sr2 + -collagen co-assembly with hydroxyapatite (HA) promotes bone reconstruction by up-regulating Nfatc1/Maf signaling pathway [65]. These results suggest Maf as a promising target to allocate the lineage of MSCs by inhibiting aging-dependent switching of osteoblasts to adipocytes and could be applied in the process of implant osseointegration in individuals with osteoporosis.

CBFβ

Core binding factor subunit beta (CBFβ) serves as a pivotal transcription factor in regulating osteoblast differentiation, bone anabolism, and senescence-related skeletal development by stabilizing and promoting Runx family proteins [56, 66,67,68]. Along with substantial accumulation of bone marrow adipocytes and reduced bone mineral density, Cbfβ expression is dramatically diminished in aged mice, indicating that Cbfβ plays a critical role in age-related bone anabolic metabolism [69]. Furthermore, Wu et al. reported that Cbfβ manipulates osteogenesis/adipogenesis lineage commitment by activating the Wnt10b/β-catenin signaling pathway and inhibiting adipogenesis regulatory gene (c/ebpα) expression [69]. In addition, conditional abrogation of Cbfβ in BMMSCs significantly impairs osteogenic capacity and reduces bone mineral density [69, 70], whereas its overexpression can certainly reverse senescence and adipogenesis mediated by p53/miR-145a [71, 72]. In conclusion, these studies primarily unveil the regulatory role of CBFβ in osteoblast/adipocyte lineage commitment, which could be exploited as a therapeutic target for promoting osseointegration in aged individuals.

SATB2

Special AT-rich sequence-binding protein 2 (SATB2), a nuclear matrix protein, is a pivotal mediator in coordinating stemness maintenance, craniofacial skeletal patterning, and osteogenic differentiation by H19 [73], miR-31 [74, 75], miR-103 [76, 77], miR-140-5p [78], and MALAT1 [79]. Interestingly, SATB2 expression decreases during senescence in alveolar bone-derived BMMSCs [80], through the regulation of Nanog transcription [81, 82]. In parallel with SATB2 declination, the BMMSCs derived from elderly rats exhibit abrogated osteogenesis and up-regulated adipogenesis [83]. However, exogenous overexpression of SATB2 decreases the age-related changes in MSCs, thereby reversing senescence-related alveolar bone loss [81] and facilitating bone reconstruction in critical size mandibular defects [84]. In addition, local delivery of SATB2 significantly accelerates new bone formation and promotes the osseointegration of titanium implants in vivo [57]. Considering this, increased SATB2 expression could be an effective strategy to regulate bone osteointegration and manage age-related bone diseases.

Extracellular vesicles (EVs)

Exosomes

EVs are membrane nano- and micro-vesicles generated by various cells and organs (MSCs [85], DCs [86], B cells [87], mast cells [88], epithelial cells [89]), which are important in cell-to-cell communication [90] and multiple biological processes [91, 92]. Among them, MSC-derived exosomes containing proteins, RNAs, and lipids serve as key mediators of the aging process [93]. The expression pattern of bone marrow exosomes differs significantly between the young and elderly [94]. For instance, exosomal presentation of miR-294 and miR-872-3p [95] is significantly reduced during senescence, while bone marrow expression of miR-335-5p and miR-146a-5p [96] is characteristic in MSC-derived microvesicles secreted by older rats. The expression of exosomal miR-31a-5p, serving as a critical modulator of osteoclastogenesis and bone resorption, is markedly higher in aged MSCs than in young cells. Inhibition of miR-31a-5p expression prevents bone loss and reduces the osteoclastic activity of aged rats in the bone marrow microenvironment [83]. Intercellular transfer of microvesicles from young MSCs rejuvenates aged murine hematopoietic stem cells (HSCs) [97]. In addition, incubation of young MSCs with aged exosomes (including miRNA-183-5p) inhibits MSC differentiation into osteoblasts, thereby inhibiting osteogenesis [98]. In contrast, exosomes highly expressing miRNA-19b-3p from young donors ameliorate the reduced osteogenic differentiation of BMMSCs in aged rats with osteoporosis in vivo [99]. The exosomes derived from young BMMSCs specifically containing CD9, CD63, and TSG101 promote new bone regeneration and osseointegration during distraction osteogenesis (DO) in older rats [100]. Collectively, these results indicate that the exosomes derived from older MSCs are associated with reduced osseointegration. In terms of sustained release and durable efficacy, liposomes [101], nanohydroxyapatite (nHP) [102, 103], tricalcium phosphate (β-TCP) ceramics [104, 105] and hydrogel formulations (e.g., alginate [106, 107], hyaluronic acid [106, 107], poly (lactic-co-glycolic acid) (PLGA) [108,109,110], and polydopamine (PDA) [109, 110]) have been used to deliver MSC-Exos for osteogenesis and osseointegration. Recently, micro/nano-textured hierarchical titanium topography has been shown to be favorable for BMMSC-Exos biogenesis and secretion, as it promotes osseointegration [111]. These findings suggest that remodeling MSC-Exos via surface modification of biomaterials or local delivery may provide a cell-free strategy for enhancing osseointegration (Fig. 3). However, the efficacy of this method requires further validation.

Fig. 3
figure 3

Schematic representation of exosome application from young individuals for improving osseointegration with aging. The expression pattern of bone marrow exosomes differs significantly between the young and elderly. The application process includes MSC-Exo harvest, local application, and implantation. Created with http://BioRender.com

Apoptotic vesicles

Apoptotic vesicles (ApoVs) are a heterogeneous population of EVs generated during apoptosis [112]. Reduction of apoptotic body formation significantly impairs the self-renewal capacity and the balance of BMMSCs between osteogenesis-or-adipogenesis [113]. Young MSCs-derived apoVs effectively rejuvenate aged BMMSCs by enriching the levels of Ras-related protein 7 (Rab7) [112]. Previous studies have shown apoVs to be effective in bone homeostasis, as they regulate bone remodeling [114, 115]. Nevertheless, further investigation on the design and use of MSC-Exos, especially MSC-apoVs, can potentially present a unique opportunity for developing strategies for osseointegration enhancement and bone regeneration in the future [116].

Intracellular signaling pathways

Multiple signaling pathways regulate bone homeostasis and bone cell differentiation. Therefore, targeting signaling pathways to enhance the osseointegration process is feasible. Here, we describe the pathways that are known to be affected by age and result in biased differentiation of MSCs.

Wnt signaling

Wnt signaling is essential for differentiation of MSCs and bone metabolism. Recently, it has been shown to be associated with aging. In the canonical Wnt pathway, Wnt proteins bind to their respective dimeric cell surface receptors, comprising seven transmembrane frizzled proteins and the LRP5/6, to activate the signaling cascade by recruiting, phosphorylating, and activating the cytoplasmic protein disheveled (DVL). This in turn inhibits the phosphorylation of β-catenin by the complex (GSK-3, APC, and Axin) and subsequently, promotes the translocation of stabilized β-catenin into the nucleus to stimulate target gene expression [117]. Zhang et al. reported that Wnt/β-catenin signaling is highly activated during MSC aging, leading to DNA damage response and p53/p21 pathway activation by regulating reactive oxygen species (ROS) production [118, 119]. Another study showed that the expression of various Wnt-associated genes decreased in the bone tissues of aged mice compared with that in the tissues of young mice [120]. Kathleen et al. performed RNA-seq of implant-associated tissue from old and young mice one week after implantation and revealed that the expression of several Wnt ligand receptors (Fzd4, 5, 8, and 9) and Wnt-regulated transcription factors (Tcf7l1, Tcf7l2, Tle2, and runx2) decreased in old mice [121]. Wnt-10b is essential to maintain normal bone density and mesenchymal progenitor activity in adult bone. Wnt-10b-/- mice show age-dependent loss of bone mass and a reduced number of MSCs. However, transgenic overexpression of Wnt-10b prevents bone loss in aged mice and enhances osteogenesis in vitro [122]. In addition, ROS activate FoxOs, which in turn, binds β-catenin to reduce its availability for activating osteogenesis-related transcription factors, leading to slow bone formation [123, 124]. A recent study revealed that Gli1 + cells contribute to the formation of new bone around the dental implant and that the ablation of these cells impairs osseointegration. Canonical Wnt signaling plays crucial roles in mediating Gli1 + stem cells [125]. Collectively, Wnt signaling mediates bone aging process by acting at different levels. Thus, modulating Wnt signaling could be a promising direction for improving osseointegration.

Notch pathway

Notch pathway is an essential regulator of bone development. Notch signaling is crucial for the maintenance of MSCs pool under physiological conditions. Disruption of Notch signaling causes bone loss in aged mice and increased trabecular bone mass in adolescent mice [126]. RNA-Seq of peri-implant tissue from young and old mice reveals multiple genes, including Notch ligands (Jag1, Jag2, Dll1, and Dll4), receptors (Notch 3, Notch 4), and downstream transcription factors (Hey1, Hey2, Heyl, and Hes1), in the Notch pathway; the expression of these genes are significantly decreased in old peri-implant tissue. This indicates that Notch signaling is inhibited during aging after implantation [121] and that targeting the Notch pathway may enhance osseointegration in older patients. However, another study showed that the Notch pathway is activated in aged BMMSCs, and inhibition of the Notch signaling by γ-secretase inhibitor restores the osteogenic ability of BMMSCs [127]. Nevertheless, 12-month-old C57BL/6 mice were used as the old group in this study, and only an in vitro study was conducted, whereas the previous study was performed on 21-month-old mice as the old group in vivo. These differences may be responsible for the varying results. Notch activation by Jagged 1 enhances the osteogenic differentiation of MSCs sheet by inhibiting the cellular senescence caused by high density sheet culture [128]. Therefore, Notch pathway could be a key regulator of MSCs and bone aging. However, the role of Notch signaling in osseointegration has not been thoroughly explored. Local aspirin administration enhanced hydroxyapatite-coated titanium implant osseointegration in OVX rats through activation of the Notch pathway in osteoporotic bone [129]. Notch pathway is activated by chemical/nanostructural (modSLA) and micro-roughened (SLA) surfaces that lead to increased osteogenic differentiation after culturing in osteogenic media [130]. In summary, Notch signaling is closely associated with bone and MSC aging, and its modulation might be an effective strategy to enhance osseointegration in the elderly.

NF-κB signaling

The transcription factor nuclear factor κB (NF-κB) is a key regulator of inflammation and bone-modeling process. Increased NF-kB activity is accompanied by increased bone-resorption and decreased bone-formation abilities [131]. The MSCs derived from aged mice show impaired osteogenesis, which is associated with increased NF-κB activity [132]. The proinflammatory cytokines TNF and IL-17 inhibit the osteogenic differentiation of MSCs by activating NF-κB signaling. Inhibition of NF-κB signaling promotes bone regeneration and repair under chronic inflammatory conditions [133]. Prophylactic melatonin administration promotes the osteogenesis of BMMSCs and osteoblasts in vitro and reduces bone resorption and proinflammatory cytokine levels by inhibiting the activation of NF-κB to down-regulate TNF, IL-1β, and IL-6 [134]. These findings provide an unexplored strategy for enhancing osseointegration by inhibiting the NF-κB pathway. Methods such as drug administration to inhibit NF-κB signaling should be further investigated.

Epigenetic modification

The role of epigenetic alterations in aging has become an interesting topic owing to the reversibility of these alterations. This may introduce a scope for developing treatment options for age-related diseases [135]. Epigenetic modifications such as DNA methylation, histone modification, and chromatin remodeling regulate the patterns of gene expression by altering DNA accessibility or chromatin structure without changing the DNA sequence. They are both heritable and reversible and can occur in response to environmental stimuli and intrinsic changes to maintain cell homeostasis and function [136]. It has been widely documented that epigenetic changes play crucial roles in the aging process of MSCs. Several regulatory factors and sites related to DNA methylation have been found to have profound effects on MSC aging. Kalyan et al. compared the methylomes of young and aged human BMMSCs using sequencing-based methods, identified DNA methylation changes associated with aging, and constructed hypo- and hypermethylation-specific regulatory networks [137]. RG108, the inhibitor of DNA methyltransferase (DNMT), can alleviate the senescence of aged human BMMSCs by eliminating ROS and upregulating telomerase traverse transcriptase (TERT) activity, which can repair shortened telomeres [138]. Depletion of the DNA demethylases TET1 and TET2 leads to an osteopenia phenotype in mice. Moreover, it reduces the self-renewal and osteogenic differentiation abilities of BMMSCs by inhibiting demethylation of the P2rX7 promoter and the release of exosomes, leading to the accumulation of miR-297a-5p, miR-297b-5p, and miR-297c-5p in BMMSCs, which inhibits the Runx2 signaling pathway [139]. Histone modifications regulate aging in MSCs by regulating the transcriptional activity of the related genes. For instance, Twist-1 is down-regulated in aged MSCs, whereas overexpression of Twist-1 alleviates human MSC senescence by increasing the recruitment of EZh2, reducing the expression of the Lnk4A/Arf locus, and enhancing the levels of histone H3K27me3 at p16/p14 promoters [140]. In osteoporosis models, epigenetic modifying protein lysine (K)-specific demethylase 5A (KDM5A) is up-regulated, which in turn suppresses runx2 expression by reducing H3K4me3 levels [141]. Similarly, the expression of enhancer of zeste homology 2 (EZH2) increases in osteporotic MSCs. It directly elevates H3K27me3 levels on the promoters of Wnt1, Wnt6, and Wnt10a to silence Wnt signaling. Knockdown of EZH2 and inhibition of H3K27me3 inhibit the repression of Wnt signaling and restore the osteogenic differentiation of osteoporotic MSCs [142]. However, absent, small, or homeotic-like 1 (Ash1l), a histone 3 lysine 4 (H3K4) trimethyltransferase in mice, is downregulated and fails to mediate H3K4me3 recruitment at the transcription start sites of the OSX, Runx2, Sox9, and Cred genes. Silencing of Ash1l impairs osteogenic differentiation and promotes adipogenic differentiation [143]. In addition, microRNAs participate in the epigenetic regulation of MSC aging and osseointegration. This is further elaborated in the following section.

The involvement of epigenetic modifications in osseointegration and their application in functionalizing implant surface or systematically improve osseointegration need to be further investigated. To date, only the effects of microRNA coatings on implant surface have been investigated; DNA methylation and histone modifications have not yet been studied, although they could hold some potential in enhancing the osseointegration process.

MicroRNAs

MicroRNAs (miRNAs) are single-stranded noncoding RNAs (22 nucleotides long) involved in the repression of the expression of target genes via either mRNA degradation or translational inhibition in a broad range of organisms [144]. miRNAs participate in controlling cell function by acting on more than one gene [145]. Several miRNAs are closely related to age-related changes in MSCs and bone aging. miRNA-195 targets the 3′ untranslated region of TERT. During skeletal aging, MiR-196a was found to be up-regulated in MSCs that targeted homeobox B7 (HOXB7). A forced HOXB7 expression in MSCs led to increased cell growth, reduced senescence, and improved osteogenic differentiation [146]. MiR-188 is a crucial regulator of the age-related differentiation imbalance in MSCs. Its expression is elevated in MSCs derived from aged mice and humans, and forced expression of miR-188 causes age-related bone loss and fat accumulation. It directly targets histone deacetylase 9 (HDAC9) and RPTOR- independent companion of MTOR complex 2 (Rictor) [147]. MiR-141 is up-regulated in the bones and MSCs of mice and humans. It inhibits the osteogenesis of MSCs. It targets ZMPSTE24, BMI1, SDF-1, SVCT2, and DLX5, which are known to control MSCs differentiation, migration, and proliferation [148,149,150,151]. Additionally, the expression of microRNAs is reduced in aged MSCs. The expression of MIR17–92 is reduced in old mice, and miR-17 overexpression restores the osteogenesis of old MSCs by regulating Smurf1 [152]. Both microRNA 23a and microRNA 23b are remarkably down-regulated in the MSCs of old mice and humans and are closely related to the imbalanced differentiation of aged MSCs. Overexpression of miR-23a/b promotes the osteogenic differentiation of MSCs, which targets transmembrane protein 64 (TMEM64) [153].

Multiple microRNAs regulate the osseointegration process by controlling the osteogenesis of MSCs [154]. MiRNA and anti-miRNA delivery has been used to improve the osseointegration process by applying them to the implant surface or surrounding tissue of the implant. An miR-21 nanocapsule immobilized on titanium surface via an in situ polymerization method enhanced both the osteogenesis and angiogenesis of MSCs in vitro and improved osseointegration during early stages in vivo [155]. Similarly, Zhen et al. fabricated a titanium (Ti)-based SrHA/miR-21 composite coating via hydrothermal deposition of SrHA, followed by miR-21 nanocapsule immobilization, and revealed that the coating not only increased osteoblast proliferation and differentiation in vitro but also enhanced osseointegration and bone-implant bonding strength in vivo [156]. A microporous Ti implant surface formed by microarc oxidation (MAO) loaded with miR-29b and antimiR-138 via lyophilization enhances the osteogenic differentiation of MSCs and potentially leads to rapid and increased osseointegration of the clinical implant interface [157]. These observations indicate that the application of age-related microRNAs to titanium surface could be a promising direction to enhance osseointegration in the aged population. Further research is necessary for optimizing the process.

Oxidative stress

ROS are chemically reactive chemical species containing oxygen, which are mostly generated within the electron transport chain of mitochondria. An increase in ROS levels is highly correlated with aging and damage to cells and tissues [154, 158]. Aging can shift the differentiation preference of MSCs from osteogenic to adipogenic through oxidative stress [159,160,161]. Consistent studies have shown that treatments eliminating ROS through various mechanisms can alleviate bone aging, and promote bone formation. SIRT3, a sirtuin involved in aging and the overexpression of SIRT3 in MSCs, restores the capacity of MSCs to differentiate and reduces oxidative stress [162]. Similarly, rapamycin enhances the osteogenesis of aged MSCs by restoring autophagy and reducing levels of ROS [49]. Recent research has revealed that titanium surfaces coated with ROS-responsive gelatin/chitosan hold the potential to promote the osteogenic differentiation of MSCs and enhance osseointegration by eliminating intracellular ROS in osteoporotic conditions [163]. Therefore, reducing oxidative stress via application of drugs or implant modifications could be a promising strategy for implant treatment in aged population.

Conclusions and prospects

The pathological differentiation of senescent MSCs contributes to bone aging, which is detrimental for the osseointegration process and implant success. In this review, we summarized the current knowledge of molecular mechanisms underlying impaired MSCs differentiation in aging and the methods to counteract this imbalanced differentiation to improve osseointegration. In the field of dental implantology, it's evident that the journey toward achieving better osseointegration is multifaceted. This review provides new perspectives and potential strategies to enhance osseointegration and increase implant survival rate. However, osseointegration process include a cascade of events involving different kinds of cells and we still lack the knowledge of the cellular and molecular mechanisms of osseointegration during aging. Further understanding of these processes will be crucial for developing better strategies to achieve optimal osseointegration. In addition, different methods such as surface modification, systemic or local administration of certain drugs need to be optimized. Also, how to minimize the side effects should be considered in future studies.

Availability of data and materials

Not applicable.

Abbreviations

ApoVs:

Apoptotic vesicles

ASH1L:

Absent, small, or homeotic-like 1

BIC:

Bone-to-implant contact

BLCs:

Bone-lining cells

BMMSCs:

Bone marrow mesenchymal stem cells

CBFβ:

Core binding factor subunit beta

DVL:

Disheveled

EVs:

Extracellular vesicles

EZH2:

Enhancer of zeste homology 2

FOXP1:

Forkhead box P1

H3K4:

Histone 3 lysine 4

HA:

Hydroxyapatite

HDAC9:

Histone deacetylase 9

HOXB7:

Homeobox B7

HSCs:

Hematopoietic stem cells

KDM5A:

Lysine (K)-specific demethylase 5A

MAF:

MAF bZIP transcription factor

MAO:

Microarc oxidation

miRNAs:

MicroRNAs

MSCs:

Mesenchymal stem cells

nHP:

Nanohydroxyapatite

NF-κB:

Nuclear factor κB

PDA:

Polydopamine

PLGA:

Poly (lactic-co-glycolic acid)

PPARγ2:

Proliferator-activated receptor γ2

PTH:

Parathyroid hormone

RBPjκ:

Recombination signal binding protein for immunoglobulin κ J region

ROS:

Reactive oxygen species

SATB2:

Special AT-rich sequence-binding protein 2

SLA:

Sandblasted, large grit, acid-etched

Sr:

Strontium

TERT:

Telomerase traverse transcriptase

TMEM64:

Transmembrane protein 64

References

  1. Lunenfeld B. An aging World–demographics and challenges. Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol. 2008;24(1):1–3.

    Article  Google Scholar 

  2. Kanasi E, Ayilavarapu S, Jones J. The aging population: demographics and the biology of aging. Periodontol 2000. 2016;72(1):13–8.

    Article  PubMed  Google Scholar 

  3. Saintrain MVD, de Souza EHA. Impact of tooth loss on the quality of life. Gerodontology. 2012;29(2):E632–6.

    Article  PubMed  Google Scholar 

  4. Krebs M, Schmenger K, Neumann K, Weigl P, Moser W, Nentwig GH. Long-term evaluation of ANKYLOS(R) dental implants, part i: 20-year life table analysis of a longitudinal study of more than 12,500 implants. Clin Implant Dent Relat Res. 2015;17(Suppl 1):e275–86.

    PubMed  Google Scholar 

  5. Busenlechner D, Fürhauser R, Haas R, Watzek G, Mailath G, Pommer B. Long-term implant success at the Academy for oral implantology: 8-year follow-up and risk factor analysis. J Periodontal Implant Sci. 2014;44(3):102–8.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Branemark PI, Adell R, Breine U, Hansson BO, Lindstrom J, Ohlsson A. Intra-osseous anchorage of dental prostheses. I. Experimental studies. Scand J Plast Reconstr Surg. 1969;3(2):81–100.

    CAS  PubMed  Google Scholar 

  7. Jayesh RS, Dhinakarsamy V. Osseointegration. J Pharm Bioallied Sci. 2015;7(Suppl 1):S226.

    PubMed  PubMed Central  Google Scholar 

  8. Choi YS, Song IS, Kim MY, Lim HK, Chongyang L, Shim JS. Implant stability changes over time following implant placement in elderly patients: a prospective study. Clin Oral Implan Res. 2023;34(6):575–81.

    Article  Google Scholar 

  9. Chandra A, Rajawat J. Skeletal aging and osteoporosis: mechanisms and therapeutics. Int J Mol Sci. 2021;22(7):3553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Infante A, Rodriguez CI. Osteogenesis and aging: lessons from mesenchymal stem cells. Stem Cell Res Ther. 2018;9(1):244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Schroeder A. Gewebsreaktion auf ein titan-hohlzylinderimplantat mit titan-spritzschichtoberfläche (1976).

  12. Schroeder A, van der Zypen E, Stich H, Sutter F. The reactions of bone, connective tissue, and epithelium to endosteal implants with titanium-sprayed surfaces. J Maxillofac Surg. 1981;9:15–25.

    Article  CAS  PubMed  Google Scholar 

  13. Tang LP, Eaton JW. Inflammatory responses to biomaterials. Am J Clin Pathol. 1995;103(4):466–71.

    Article  CAS  PubMed  Google Scholar 

  14. Anderson JM. Inflammatory response to implants. ASAIO Trans. 1988;34(2):101–7.

    Article  CAS  PubMed  Google Scholar 

  15. Gonzalez-Simon AL, Eniola-Adefeso O. Host response to biomaterials. Eng Biomat Regener Med Nov Technol Clin Appl. 2012. https://doi.org/10.1007/978-1-4614-1080-5_6.

    Article  Google Scholar 

  16. Trindade R, Albrektsson T, Tengvall P, Wennerberg A. Foreign body reaction to biomaterials: on mechanisms for buildup and breakdown of osseointegration. Clin Implant Dent Relat Res. 2016;18(1):192–203.

    Article  PubMed  Google Scholar 

  17. Wang YL, Zhang YF, Miron RJ. Health, maintenance, and recovery of soft tissues around implants. Clin Implant Dent R. 2016;18(3):618–34.

    Article  Google Scholar 

  18. Schimmel M, Srinivasan M, McKenna G, Müller F. Effect of advanced age and/or systemic medical conditions on dental implant survival: a systematic review and meta-analysis. Clin Oral Implant Res. 2018;29:311–30.

    Article  Google Scholar 

  19. Bryant SR, Zarb GA. Osseointegration of oral implants in older and younger adults. Int J Oral Maxillofac Implants. 1998;13(4):492–9.

    CAS  PubMed  Google Scholar 

  20. Hoeksema AR, Visser A, Raghoebar GM, Vissink A, Meijer HJA. Influence of age on clinical performance of mandibular two-implant overdentures: a 10-year prospective comparative study. Clin Implant Dent R. 2016;18(4):745–51.

    Article  Google Scholar 

  21. Alsaadi G, Quirynen M, Komarek A, van Steenberghe D. Impact of local and systemic factors on the incidence of oral implant failures, up to abutment connection. J Clin Periodontol. 2007;34(7):610–7.

    Article  PubMed  Google Scholar 

  22. Giro G, Chambrone L, Goldstein A, Rodrigues JA, Zenobio E, Feres M, Figueiredo LC, Cassoni A, Shibli JA. Impact of osteoporosis in dental implants: a systematic review. World J Orthop. 2015;6(2):311–5.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Niedermaier R, Stelzle F, Riemann M, Bolz W, Schuh P, Wachtel H. Implant-supported immediately loaded fixed full-arch dentures: evaluation of implant survival rates in a case cohort of up to 7 years. Clin Implant Dent Relat Res. 2017;19(1):4–19.

    Article  PubMed  Google Scholar 

  24. Brocard D, Barthet P, Baysse E, Duffort JF, Eller P, Justumus P, Marin P, Oscaby F, Simonet T, Benqué E, Brunel G. A multicenter report on 1,022 consecutively placed ITI implants: a 7-year longitudinal study. Int J Oral Max Implants. 2000;15(5):691–700.

    CAS  Google Scholar 

  25. Bertl K, Ebner M, Knibbe M, Pandis N, Kuchler U, Ulm C, Stavropoulos A. How old is old for implant therapy in terms of early implant losses? J Clin Periodontol. 2019;46(12):1282–93.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wagner F, Schuder K, Hof M, Heuberer S, Seemann R, Dvorak G. Does osteoporosis influence the marginal peri-implant bone level in female patients? A cross-sectional study in a matched collective. Clin Implant Dent Relat Res. 2017;19(4):616–23.

    Article  PubMed  Google Scholar 

  27. Duarte PM, Goncalves PF, Casati MZ, Sallum EA, Nociti FH Jr. Age-related and surgically induced estrogen deficiencies may differently affect bone around titanium implants in rats. J Periodontol. 2005;76(9):1496–501.

    Article  CAS  PubMed  Google Scholar 

  28. Qi MC, Zhou XQ, Hu J, Du ZJ, Yang JH, Liu M, Li XM. Oestrogen replacement therapy promotes bone healing around dental implants in osteoporotic rats. Int J Oral Maxillofac Surg. 2004;33(3):279–85.

    Article  CAS  PubMed  Google Scholar 

  29. Duarte PM, Cesar-Neto JB, Sallum AW, Sallum EA, Nociti FH Jr. Effect of estrogen and calcitonin therapies on bone density in a lateral area adjacent to implants placed in the tibiae of ovariectomized rats. J Periodontol. 2003;74(11):1618–24.

    Article  CAS  PubMed  Google Scholar 

  30. Gabet Y, Muller R, Levy J, Dimarchi R, Chorev M, Bab I, Kohavi D. Parathyroid hormone 1–34 enhances titanium implant anchorage in low-density trabecular bone: a correlative micro-computed tomographic and biomechanical analysis. Bone. 2006;39(2):276–82.

    Article  CAS  PubMed  Google Scholar 

  31. Jiang L, Zhang W, Wei L, Zhou Q, Yang G, Qian N, Tang Y, Gao Y, Jiang X. Early effects of parathyroid hormone on vascularized bone regeneration and implant osseointegration in aged rats. Biomaterials. 2018;179:15–28.

    Article  CAS  PubMed  Google Scholar 

  32. Apostu D, Lucaciu O, Lucaciu GD, Crisan B, Crisan L, Baciut M, Onisor F, Baciut G, Câmpian RS, Bran S. Systemic drugs that influence titanium implant osseointegration. Drug Metab Rev. 2017;49(1):92–104.

    Article  CAS  PubMed  Google Scholar 

  33. Chakravorty N, Jaiprakash A, Ivanovski S, Xiao Y. Scaffolds, Implant surface modifications and osseointegration. Biomater Implants Scaffolds. 2017. https://doi.org/10.1007/978-3-662-53574-5_4.

    Article  Google Scholar 

  34. Lai M, Yan XF, Jin ZY. The response of bone cells to titanium surfaces modified by simvastatin-loaded multilayered films. J Biomat Sci-Polym E. 2018;29(15):1895–908.

    Article  CAS  Google Scholar 

  35. Tang JH, Yan DY, Chen L, Shen ZJ, Wang BZ, Weng SJ, Wu ZY, Xie ZJ, Fang KH, Hong CX, Xie J, Yang L, Shen LY. Enhancement of local bone formation on titanium implants in osteoporotic rats by biomimetic multilayered structures containing parathyroid hormone (PTH)-related protein. Biomed Mater. 2020;15(4):045011.

    Article  CAS  PubMed  Google Scholar 

  36. Jurczak P, Witkowska J, Rodziewicz-Motowidlo S, Lach S. Proteins, peptides and peptidomimetics as active agents in implant surface functionalization. Adv Colloid interface Sci. 2020;276:102083.

    Article  CAS  PubMed  Google Scholar 

  37. Wang J, He XT, Xu XY, Yin Y, Li X, Bi CS, Hong YL, Chen FM. Surface modification plasmid-mediated pLAMA3-CM gene transfection promotes the attachment of gingival epithelial cells to titanium sheets and improves biological sealing at the transmucosal sites of titanium implants. J Mater Chem B. 2019;7(46):7415–27.

    Article  CAS  PubMed  Google Scholar 

  38. Xia LG, Xie YT, Fang B, Wang XH, Lin KL. In situ modulation of crystallinity and nano-structures to enhance the stability and osseointegration of hydroxyapatite coatings on Ti-6Al-4V implants. Chem Eng J. 2018;347:711–20.

    Article  CAS  Google Scholar 

  39. Lin GF, Zhou C, Lin MN, Xu AT, He FM. Strontium-incorporated titanium implant surface treated by hydrothermal reactions promotes early bone osseointegration in osteoporotic rabbits. Clin Oral Implants Res. 2019;30(8):777–90.

    Article  PubMed  Google Scholar 

  40. Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119(11):2204–13.

    Article  PubMed  Google Scholar 

  41. Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21(2):115–37.

    CAS  PubMed  Google Scholar 

  42. Corrado A, Cici D, Rotondo C, Maruotti N, Cantatore FP. Molecular basis of bone aging. Int J Mol Sci. 2020;21(10):3679.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sanghani-Kerai A, Osagie-Clouard L, Blunn G, Coathup M. The influence of age and osteoporosis on bone marrow stem cells from rats. Bone Joint Res. 2018;7(4):289–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Aman Y, Schmauck-Medina T, Hansen M, Morimoto RI, Simon AK, Bjedov I, Palikaras K, Simonsen A, Johansen T, Tavernarakis N, Rubinsztein DC, Partridge L, Kroemer G, Labbadia J, Fang EF. Autophagy in healthy aging and disease. Nat Aging. 2021;1(8):634–50.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Kaushik S, Arias E, Kwon H, Lopez NM, Athonvarangkul D, Sahu S, Schwartz GJ, Pessin JE, Singh R. Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO Rep. 2012;13(3):258–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ott C, Konig J, Hohn A, Jung T, Grune T. Macroautophagy is impaired in old murine brain tissue as well as in senescent human fibroblasts. Redox Biol. 2016;10:266–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller RA. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460(7253):392–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bitto A, Ito TK, Pineda VV, LeTexier NJ, Huang HZ, Sutlief E, Tung H, Vizzini N, Chen B, Smith K, Meza D, Yajima M, Beyer RP, Kerr KF, Davis DJ, Gillespie CH, Snyder JM, Treuting PM, Kaeberlein M. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. elife. 2016;5:e16351.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Ma Y, Qi M, An Y, Zhang L, Yang R, Doro DH, Liu W, Jin Y. Autophagy controls mesenchymal stem cell properties and senescence during bone aging. Aging Cell. 2018;17(1):e12709.

    Article  PubMed  Google Scholar 

  50. Wang H, Ma Y, Li J, Zhou C, Xu A, Xu Y, He F. Modulating autophagy by strontium-doped micro/nano rough titanium surface for promotion of osteogenesis and inhibition of osteoclastogenesis. Colloids Surf B Biointerfaces. 2022;210:112246.

    Article  CAS  PubMed  Google Scholar 

  51. Li L, Yang S, Xu L, Li Y, Fu Y, Zhang H, Song J. Nanotopography on titanium promotes osteogenesis via autophagy-mediated signaling between YAP and beta-catenin. Acta Biomater. 2019;96:674–85.

    Article  PubMed  Google Scholar 

  52. Lin J, Xu R, Shen X, Jiang H, Du S. Metformin promotes the osseointegration of titanium implants under osteoporotic conditions by regulating BMSCs autophagy, and osteogenic differentiation. Biochem Biophys Res Commun. 2020;531(2):228–35.

    Article  CAS  PubMed  Google Scholar 

  53. Li Z, Zhao Y, Wang Z, Ren M, Wang X, Liu H, Lin Q, Wang J. Engineering multifunctional hydrogel-integrated 3D printed bioactive prosthetic interfaces for osteoporotic osseointegration. Adv Healthc Mater. 2022;11(11):e2102535.

    Article  PubMed  Google Scholar 

  54. Chan WCW, Tan Z, To MKT, Chan D. Regulation and role of transcription factors in osteogenesis. Int J Mol Sci. 2021;22(11):5445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Li H, Liu P, Xu S, Li Y, Dekker JD, Li B, Fan Y, Zhang Z, Hong Y, Yang G, Tang T, Ren Y, Tucker HO, Yao Z, Guo X. FOXP1 controls mesenchymal stem cell commitment and senescence during skeletal aging. J Clin Invest. 2017;127(4):1241–53.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Lim KE, Park NR, Che X, Han MS, Jeong JH, Kim SY, Park CY, Akiyama H, Kim JE, Ryoo HM, Stein JL. Core binding factor β of osteoblasts maintains cortical bone mass via stabilization of Runx2 in mice. J Bone Miner Res. 2015;30(4):715–22.

    Article  CAS  PubMed  Google Scholar 

  57. Yan SG, Zhang J, Tu QS, Ye JH, Luo E, Schuler M, Kim MS, Griffin T, Zhao J, Duan XJ, Cochran DJ, Murray D, Yang PS, Chen J. Enhanced osseointegration of titanium implant through the local delivery of transcription factor SATB2. Biomaterials. 2011;32(33):8676–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nishikawa K, Nakashima T, Takeda S, Isogai M, Hamada M, Kimura A, Kodama T, Yamaguchi A, Owen MJ, Takahashi S, Takayanagi H. Maf promotes osteoblast differentiation in mice by mediating the age-related switch in mesenchymal cell differentiation. J Clin Invest. 2010;120(10):3455–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhao H, Zhou W, Yao Z, Wan Y, Cao J, Zhang L, Zhao J, Li H, Zhou R, Li B, Wei G, Zhang Z, French CA, Dekker JD, Yang Y, Fisher SE, Tucker HO, Guo X. Foxp1/2/4 regulate endochondral ossification as a suppresser complex. Dev Biol. 2015;398(2):242–54.

    Article  CAS  PubMed  Google Scholar 

  60. Wang FS, Chen YS, Ko JY, Kuo CW, Ke HJ, Hsieh CK, Wang SY, Kuo PC, Jahr H, Lian WS. Bromodomain protein BRD4 accelerates glucocorticoid dysregulation of bone mass and marrow adiposis by modulating H3K9 and Foxp1. Cells. 2020;9(6):1500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lin H, Sohn J, Shen H, Langhans MT, Tuan RS. Bone marrow mesenchymal stem cells: aging and tissue engineering applications to enhance bone healing. Biomaterials. 2019;203:96–110.

    Article  CAS  PubMed  Google Scholar 

  62. Xu J, Wang K, Zhang Z, Xue D, Li W, Pan Z. The role of forkhead box family in bone metabolism and diseases. Front Pharmacol. 2021;12:772237.

    Article  CAS  PubMed  Google Scholar 

  63. Saidak Z, Hay E, Marty C, Barbara A, Marie PJ. Strontium ranelate rebalances bone marrow adipogenesis and osteoblastogenesis in senescent osteopenic mice through NFATc/Maf and Wnt signaling. Aging Cell. 2012;11(3):467–74.

    Article  CAS  PubMed  Google Scholar 

  64. Zhou C, Chen YQ, Zhu YH, Lin GF, Zhang LF, Liu XC, He FM. Antiadipogenesis and osseointegration of strontium-doped implant surfaces. J Dent Res. 2019;98(7):795–802.

    Article  CAS  PubMed  Google Scholar 

  65. Zhong Z, Wu X, Wang Y, Li M, Li Y, Liu X, Zhang X, Lan Z, Wang J, Du Y, Zhang S. Zn/Sr dual ions-collagen co-assembly hydroxyapatite enhances bone regeneration through procedural osteo-immunomodulation and osteogenesis. Bioact Mater. 2022;10:195–206.

    CAS  PubMed  Google Scholar 

  66. Qin X, Jiang Q, Matsuo Y, Kawane T, Komori H, Moriishi T, Taniuchi I, Ito K, Kawai Y, Rokutanda S, Izumi S, Komori T. Cbfb regulates bone development by stabilizing Runx family proteins. J Bone Miner Res. 2015;30(4):706–14.

    Article  CAS  PubMed  Google Scholar 

  67. Morita K, Noura M, Tokushige C, Maeda S, Kiyose H, Kashiwazaki G, Taniguchi J, Bando T, Yoshida K, Ozaki T, Matsuo H, Ogawa S, Liu PP, Nakahata T, Sugiyama H, Adachi S, Kamikubo Y. Autonomous feedback loop of RUNX1-p53-CBFB in acute myeloid leukemia cells. Sci Rep. 2017;7(1):16604.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Qi J, Singh S, Hua WK, Cai Q, Chao SW, Li L, Liu H, Ho Y, McDonald T, Lin A, Marcucci G, Bhatia R, Huang WJ, Chang CI, Kuo YH. HDAC8 inhibition specifically targets Inv(16) acute myeloid leukemic stem cells by restoring p53 acetylation. Cell Stem Cell. 2015;17(5):597–610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Wu M, Wang Y, Shao J-Z, Wang J, Chen W, Li YP. Cbfβ governs osteoblast− adipocyte lineage commitment through enhancing β-catenin signaling and suppressing adipogenesis gene expression. Proc Natl Acad Sci. 2017;114(38):10119–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wu M, Li C, Zhu G, Wang Y, Jules J, Lu Y, McConnell M, Wang YJ, Shao JZ, Li YP, Chen W. Deletion of core-binding factor beta (Cbfbeta) in mesenchymal progenitor cells provides new insights into Cbfbeta/Runxs complex function in cartilage and bone development. Bone. 2014;65:49–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Xia C, Jiang T, Wang Y, Chen X, Hu Y, Gao Y. The p53/miR-145a axis promotes cellular senescence and inhibits osteogenic differentiation by targeting Cbfb in mesenchymal stem cells. Front Endocrinol (Lausanne). 2020;11:609186.

    Article  PubMed  Google Scholar 

  72. Lee J, Cha H, Park TH, Park JH. Enhanced osteogenic differentiation of human mesenchymal stem cells by direct delivery of Cbfbeta protein. Biotechnol Bioeng. 2020;117(9):2897–910.

    Article  CAS  PubMed  Google Scholar 

  73. Chen S, Liu D, Zhou Z, Qin S. Role of long non-coding RNA H19 in the development of osteoporosis. Mol Med. 2021;27(1):122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Deng Y, Wu S, Zhou H, Bi X, Wang Y, Hu Y, Gu P, Fan X. Effects of a miR-31, Runx2, and Satb2 regulatory loop on the osteogenic differentiation of bone mesenchymal stem cells. Stem Cells Dev. 2013;22(16):2278–86.

    Article  CAS  PubMed  Google Scholar 

  75. Ge J, Guo S, Fu Y, Zhou P, Zhang P, Du Y, Li M, Cheng J, Jiang H. Dental follicle cells participate in tooth eruption via the RUNX2-MiR-31-SATB2 loop. J Dent Res. 2015;94(7):936–44.

    Article  CAS  PubMed  Google Scholar 

  76. Lv H, Yang H, Wang Y. Effects of miR-103 by negatively regulating SATB2 on proliferation and osteogenic differentiation of human bone marrow mesenchymal stem cells. PLoS One. 2020;15(5):e0232695.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Shen H, Lu C, Shi J, Li H, Si J, Shen G. Satb2 expression in Foxc1-promoted osteogenic differentiation of MC3T3-E1 cells is negatively regulated by microRNA-103-3p. Acta Biochim Biophys Sin (Shanghai). 2019;51(6):588–97.

    Article  CAS  PubMed  Google Scholar 

  78. Bi HU, Wang D, Liu X, Wang G, Wu X. Long non-coding RNA H19 promotes osteogenic differentiation of human bone marrow-derived mesenchymal stem cells by regulating microRNA-140-5p/SATB2 axis. J Biosci. 2020;45:56.

    Article  CAS  PubMed  Google Scholar 

  79. Yang X, Yang J, Lei P, Wen T. LncRNA MALAT1 shuttled by bone marrow-derived mesenchymal stem cells-secreted exosomes alleviates osteoporosis through mediating microRNA-34c/SATB2 axis. Aging (Albany NY). 2019;11(20):8777–91.

    Article  CAS  PubMed  Google Scholar 

  80. Dobreva G, Chahrour M, Dautzenberg M, Chirivella L, Kanzler B, Farinas I, Karsenty G, Grosschedl R. SATB2 is a multifunctional determinant of craniofacial patterning and osteoblast differentiation. Cell. 2006;125(5):971–86.

    Article  CAS  PubMed  Google Scholar 

  81. Xu R, Fu Z, Liu X, Xiao T, Zhang P, Du Y, Yuan H, Cheng J, Jiang H. Transplantation of osteoporotic bone marrow stromal cells rejuvenated by the overexpression of SATB2 prevents alveolar bone loss in ovariectomized rats. Exp Gerontol. 2016;84:71–9.

    Article  CAS  PubMed  Google Scholar 

  82. Zhou PP, Wu G, Zhang P, Xu RY, Ge J, Fu Y, Zhang YC, Du YF, Ye JH, Cheng J, Jiang HB. SATB2-Nanog axis links age-related intrinsic changes of mesenchymal stem cells from craniofacial bone. Aging-Us. 2016;8(9):2006–22.

    Article  CAS  Google Scholar 

  83. Xu R, Shen X, Si Y, Fu Y, Zhu W, Xiao T, Fu Z, Zhang P, Cheng J, Jiang H. MicroRNA-31a-5p from aging BMSCs links bone formation and resorption in the aged bone marrow microenvironment. Aging Cell. 2018;17(4):e12794.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Dong W, Zhang P, Fu Y, Ge J, Cheng J, Yuan H, Jiang H. Roles of SATB2 in site-specific stemness, autophagy and senescence of bone marrow mesenchymal stem cells. J Cell Physiol. 2015;230(3):680–90.

    Article  CAS  PubMed  Google Scholar 

  85. Tan SHS, Wong JRY, Sim SJY, Tjio CKE, Wong KL, Chew JRJ, Hui JHP, Toh WS. Mesenchymal stem cell exosomes in bone regenerative strategies-a systematic review of preclinical studies. Mater Today Bio. 2020;7:100067.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Thery C, Duban L, Segura E, Veron P, Lantz O, Amigorena S. Indirect activation of naive CD4(+) T cells by dendritic cell-derived exosomes. Nat Immunol. 2002;3(12):1156–62.

    Article  CAS  PubMed  Google Scholar 

  87. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183(3):1161–72.

    Article  CAS  PubMed  Google Scholar 

  88. Ekstrom K, Valadi H, Sjostrand M, Malmhall C, Bossios A, Eldh M, Lotvall J. Characterization of mRNA and microRNA in human mast cell-derived exosomes and their transfer to other mast cells and blood CD34 progenitor cells. J Extracell Vesicles. 2012;1:18389.

    Article  Google Scholar 

  89. van Niel G, Raposo G, Candalh C, Boussac M, Hershberg R, Cerf-Bensussan N, Heyman M. Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterology. 2001;121(2):337–49.

    Article  PubMed  Google Scholar 

  90. Mathieu M, Martin-Jaular L, Lavieu G, Thery C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17.

    Article  CAS  PubMed  Google Scholar 

  91. Boriachek K, Islam MN, Moller A, Salomon C, Nguyen NT, Hossain MSA, Yamauchi Y, Shiddiky MJA. Biological functions and current advances in isolation and detection strategies for exosome nanovesicles. Small. 2018;14(6):1702153.

    Article  Google Scholar 

  92. Qin YH, Sun RX, Wu CL, Wang L, Zhang CQ. Exosome: a novel approach to stimulate bone regeneration through regulation of osteogenesis and angiogenesis. Int J Mol Sci. 2016;17(5):712.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Qin W, Dallas SL. Exosomes and extracellular RNA in muscle and bone aging and crosstalk. Curr Osteoporos Rep. 2019;17(6):548–59.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Neri S, Borzi RM. Molecular mechanisms contributing to mesenchymal stromal cell aging. Biomolecules. 2020;10(2):340.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Wang Y, Fu B, Sun X, Li D, Huang Q, Zhao W, Chen X. Differentially expressed microRNAs in bone marrow mesenchymal stem cell-derived microvesicles in young and older rats and their effect on tumor growth factor-beta1-mediated epithelial-mesenchymal transition in HK2 cells. Stem Cell Res Ther. 2015;6:185.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Lei Q, Liu T, Gao F, Xie H, Sun L, Zhao AQ, Ren WX, Guo H, Zhang LM, Wang HX, Chen ZC, Guo AY, Li QB. Microvesicles as potential biomarkers for the identification of senescence in human mesenchymal stem cells. Theranostics. 2017;7(10):2673–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Kulkarni R, Bajaj M, Ghode S, Jalnapurkar S, Limaye L, Kale VP. Intercellular transfer of microvesicles from young mesenchymal stromal cells rejuvenates aged murine hematopoietic stem cells. Stem Cells. 2018;36(3):420–33.

    Article  CAS  PubMed  Google Scholar 

  98. Davis C, Dukes A, Drewry M, Helwa I, Johnson MH, Isales CM, Hill WD, Liu Y, Shi X, Fulzele S, Hamrick MW. MicroRNA-183-5p increases with age in bone-derived extracellular vesicles, suppresses bone marrow stromal (Stem) cell proliferation, and induces stem cell senescence. Tissue Eng Part A. 2017;23(21–22):1231–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Xun J, Li C, Liu M, Mei Y, Zhou Q, Wu B, Xie F, Liu Y, Dai R. Serum exosomes from young rats improve the reduced osteogenic differentiation of BMSCs in aged rats with osteoporosis after fatigue loading in vivo. Stem Cell Res Ther. 2021;12(1):424.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Jia Y, Qiu S, Xu J, Kang Q, Chai Y. Exosomes secreted by young mesenchymal stem cells promote new bone formation during distraction osteogenesis in older rats. Calcif Tissue Int. 2020;106(5):509–17.

    Article  CAS  PubMed  Google Scholar 

  101. Hu Y, Li X, Zhang Q, Gu Z, Luo Y, Guo J, Wang X, Jing Y, Chen X, Su J. Exosome-guided bone targeted delivery of Antagomir-188 as an anabolic therapy for bone loss. Bioact Mater. 2021;6(9):2905–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Qayoom I, Teotia AK, Kumar A. Nanohydroxyapatite based ceramic carrier promotes bone formation in a femoral neck canal defect in osteoporotic rats. Biomacromolecules. 2020;21(2):328–37.

    Article  CAS  PubMed  Google Scholar 

  103. Zhang Y, Xie Y, Hao Z, Zhou P, Wang P, Fang S, Li L, Xu S, Xia Y. Umbilical mesenchymal stem cell-derived exosome-encapsulated hydrogels accelerate bone repair by enhancing angiogenesis. ACS Appl Mater Interfaces. 2021;13(16):18472–87.

    Article  CAS  PubMed  Google Scholar 

  104. Zhang JY, Liu XL, Li HY, Chen CY, Hu B, Niu X, Li Q, Zhao BZ, Xie ZP, Wang Y. Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway. Stem Cell Res Therapy. 2016;7:1–14.

    Article  Google Scholar 

  105. Qi X, Zhang J, Yuan H, Xu Z, Li Q, Niu X, Hu B, Wang Y, Li X. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells repair critical-sized bone defects through enhanced angiogenesis and osteogenesis in osteoporotic rats. Int J Biol Sci. 2016;12(7):836–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Pishavar E, Luo HR, Naserifar M, Hashemi M, Toosi S, Atala A, Ramakrishna S, Behravan J. Advanced hydrogels as exosome delivery systems for osteogenic differentiation of MSCs: application in bone regeneration. Int J Mol Sci. 2021;22(12):6203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Yang S, Zhu B, Yin P, Zhao L, Wang Y, Fu Z, Dang R, Xu J, Zhang J, Wen N. Integration of human umbilical cord mesenchymal stem cells-derived exosomes with hydroxyapatite-embedded hyaluronic acid-alginate hydrogel for bone regeneration. ACS Biomater Sci Eng. 2020;6(3):1590–602.

    Article  CAS  PubMed  Google Scholar 

  108. Kang Y, Xu C, Meng L, Dong X, Qi M, Jiang D. Exosome-functionalized magnesium-organic framework-based scaffolds with osteogenic, angiogenic and anti-inflammatory properties for accelerated bone regeneration. Bioact Mater. 2022;18:26–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Gao Y, Yuan Z, Yuan X, Wan Z, Yu Y, Zhan Q, Zhao Y, Han J, Huang J, Xiong C, Cai Q. Bioinspired porous microspheres for sustained hypoxic exosomes release and vascularized bone regeneration. Bioact Mater. 2022;14:377–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Li W, Liu Y, Zhang P, Tang Y, Zhou M, Jiang W, Zhang X, Wu G, Zhou Y. Tissue-engineered bone immobilized with human adipose stem cells-derived exosomes promotes bone regeneration. ACS Appl Mater Interfaces. 2018;10(6):5240–54.

    Article  CAS  PubMed  Google Scholar 

  111. Zhang Z, Xu R, Yang Y, Liang C, Yu X, Liu Y, Wang T, Yu Y, Deng F. Micro/nano-textured hierarchical titanium topography promotes exosome biogenesis and secretion to improve osseointegration. J Nanobiotechnol. 2021;19(1):78.

    Article  CAS  Google Scholar 

  112. Lei FC, Huang ZQ, Ou QM, Li JQ, Liu MQ, Ma L, Tan LP, Lin ZM, Kou XX. Apoptotic vesicles rejuvenate mesenchymal stem cells via Rab7-mediated autolysosome formation and alleviate bone loss in aging mice. Nano Res. 2022. https://doi.org/10.1007/s12274-022-4709-4.

    Article  Google Scholar 

  113. Liu D, Kou X, Chen C, Liu S, Liu Y, Yu W, Yu T, Yang R, Wang R, Zhou Y, Shi S. Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors. Cell Res. 2018;28(9):918–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. 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(22):8682.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Ma Q, Liang M, Wu Y, Ding N, Duan L, Yu T, Bai Y, Kang F, Dong S, Xu J, Dou C. Mature osteoclast-derived apoptotic bodies promote osteogenic differentiation via RANKL-mediated reverse signaling. J Biol Chem. 2019;294(29):11240–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Li X, Liu Y, Liu X, Du J, Bhawal UK, Xu J, Guo L, Liu Y. Advances in the therapeutic effects of apoptotic bodies on systemic diseases. Int J Mol Sci. 2022;23(15):8202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Nusse R, Clevers H. Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017;169(6):985–99.

    Article  CAS  PubMed  Google Scholar 

  118. Zhang DY, Wang HJ, Tan YZ. Wnt/beta-catenin signaling induces the aging of mesenchymal stem cells through the DNA damage response and the p53/p21 pathway. PLoS One. 2011;6(6):e21397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Zhang DY, Pan Y, Zhang C, Yan BX, Yu SS, Wu DL, Shi MM, Shi K, Cai XX, Zhou SS, Wang JB, Pan JP, Zhang LH. Wnt/beta-catenin signaling induces the aging of mesenchymal stem cells through promoting the ROS production. Mol Cell Biochem. 2013;374(1–2):13–20.

    Article  CAS  PubMed  Google Scholar 

  120. Rauner M, Sipos W, Pietschmann P. Age-dependent Wnt gene expression in bone and during the course of osteoblast differentiation. Age (Dordr). 2008;30(4):273–82.

    Article  PubMed  Google Scholar 

  121. Turajane K, Ji G, Chinenov Y, Chao M, Ayturk U, Greenblatt MB, Ivashkiv LB, Bostrom MP, Yang X. Age-related impairment of implant osseointegration is associated with immune activation and suppression of angiogenic, notch, and Wnt pathways. bioRxiv (2020).

  122. Stevens JR, Miranda-Carboni GA, Singer MA, Brugger SM, Lyons KM, Lane TF. Wnt10b deficiency results in age-dependent loss of bone mass and progressive reduction of mesenchymal progenitor cells. J Bone Miner Res. 2010;25(10):2138–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Essers MAG, de Vries-Smits LMM, Barker N, Polderman PE, Burgering BMT, Korswagen HC. Functional interaction between beta-catenin and FOXO in oxidative stress signaling. Science. 2005;308(5725):1181–4.

    Article  CAS  PubMed  Google Scholar 

  124. Almeida M, Han L, Martin-Millan M, O’Brien CA, Manolagas SC. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting beta-catenin from T cell factor- to forkhead box O-mediated transcription. J Biol Chem. 2007;282(37):27298–305.

    Article  CAS  PubMed  Google Scholar 

  125. Yi Y, Stenberg W, Luo W, Feng JQ, Zhao H. Alveolar bone marrow Gli1+ stem cells support implant osseointegration. J Dent Res. 2022;101(1):73–82.

    Article  CAS  PubMed  Google Scholar 

  126. Hilton MJ, Tu X, Wu X, Bai S, Zhao H, Kobayashi T, Kronenberg HM, Teitelbaum SL, Ross FP, Kopan R. Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation. Nat Med. 2008;14(3):306–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Tang Z, Wei J, Yu Y, Zhang J, Liu L, Tang W, Long J, Zheng X, Jing W. γ-Secretase inhibitor reverts the Notch signaling attenuation of osteogenic differentiation in aged bone marrow mesenchymal stem cells. Cell Biol Int. 2016;40(4):439–47.

    Article  CAS  PubMed  Google Scholar 

  128. Tian Y, Xu Y, Xue T, Chen L, Shi B, Shu B, Xie C, Max Morandi M, Jaeblon T, Marymont JV, Dong Y. Notch activation enhances mesenchymal stem cell sheet osteogenic potential by inhibition of cellular senescence. Cell Death Dis. 2017;8(2):e2595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Tao Z, Zhou W, Wu X, Lu H, Ma N, Li Y, Zhang R, Yang M, Xu HG. Local administration of aspirin improves osseointegration of hydroxyapatite-coated titanium implants in ovariectomized rats through activation of the Notch signaling pathway. J Biomater Appl. 2020;34(7):1009–18.

    Article  CAS  PubMed  Google Scholar 

  130. Chakravorty N, Hamlet S, Jaiprakash A, Crawford R, Oloyede A, Alfarsi M, Xiao Y, Ivanovski S. Pro-osteogenic topographical cues promote early activation of osteoprogenitor differentiation via enhanced TGFbeta, Wnt, and Notch signaling. Clin Oral Implants Res. 2014;25(4):475–86.

    Article  PubMed  Google Scholar 

  131. Jimi E, Takakura N, Hiura F, Nakamura I, Hirata-Tsuchiya S. The role of NF-κB in physiological bone development and inflammatory bone diseases: is NF-κB inhibition “Killing Two Birds with One Stone”? Cells. 2019;8(12):1636.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lin TH, Gibon E, Loi F, Pajarinen J, Cordova LA, Nabeshima A, Lu L, Yao Z, Goodman SB. Decreased osteogenesis in mesenchymal stem cells derived from the aged mouse is associated with enhanced NF-kappaB activity. J Orthop Res. 2017;35(2):281–8.

    Article  CAS  PubMed  Google Scholar 

  133. Chang J, Liu F, Lee M, Wu B, Ting K, Zara JN, Soo C, Al Hezaimi K, Zou W, Chen X. NF-κB inhibits osteogenic differentiation of mesenchymal stem cells by promoting β-catenin degradation. Proc Natl Acad Sci. 2013;110(23):9469–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Wu X, Qiao S, Wang W, Zhang Y, Shi J, Zhang X, Gu W, Zhang X, Li Y, Ding X. Melatonin prevents peri-implantitis via suppression of TLR4/NF-κB. Acta Biomater. 2021;134:325–36.

    Article  CAS  PubMed  Google Scholar 

  135. Brunet A, Berger SL. Epigenetics of aging and aging-related disease. J Gerontol A Biol Sci Med Sci. 2014;69:S17-20.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Loscalzo J, Handy DE. Epigenetic modifications: basic mechanisms and role in cardiovascular disease (2013 Grover Conference series). Pulm Circ. 2014;4(2):169–74.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Pasumarthy KK, Doni Jayavelu N, Kilpinen L, Andrus C, Battle SL, Korhonen M, Lehenkari P, Lund R, Laitinen S, Hawkins RD. Methylome analysis of human bone marrow MSCs reveals extensive age- and culture-induced changes at distal regulatory elements. Stem Cell Rep. 2017;9(3):999–1015.

    Article  CAS  Google Scholar 

  138. Oh YS, Jeong SG, Cho GW. Anti-senescence effects of DNA methyltransferase inhibitor RG108 in human bone marrow mesenchymal stromal cells. Biotechnol Appl Biochem. 2015;62(5):583–90.

    Article  CAS  PubMed  Google Scholar 

  139. Yang R, Yu T, Kou X, Gao X, Chen C, Liu D, Zhou Y, Shi S. Tet1 and Tet2 maintain mesenchymal stem cell homeostasis via demethylation of the P2rX7 promoter. Nat Commun. 2018;9(1):2143.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Cakouros D, Isenmann S, Cooper L, Zannettino A, Anderson P, Glackin C, Gronthos S. Twist-1 induces Ezh2 recruitment regulating histone methylation along the Ink4A/Arf locus in mesenchymal stem cells. Mol Cell Biol. 2012;32(8):1433–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Wang C, Wang J, Li J, Hu G, Shan S, Li Q, Zhang X. KDM5A controls bone morphogenic protein 2-induced osteogenic differentiation of bone mesenchymal stem cells during osteoporosis. Cell Death Dis. 2016;7(8):e2335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Jing H, Liao L, An Y, Su X, Liu S, Shuai Y, Zhang X, Jin Y. Suppression of EZH2 prevents the shift of osteoporotic MSC fate to adipocyte and enhances bone formation during osteoporosis. Mol Ther. 2016;24(2):217–29.

    Article  CAS  PubMed  Google Scholar 

  143. Yin B, Yu F, Wang C, Li B, Liu M, Ye L. Epigenetic control of mesenchymal stem cell fate decision via histone methyltransferase Ash1l. Stem Cells. 2019;37(1):115–27.

    Article  CAS  PubMed  Google Scholar 

  144. Pritchard CC, Cheng HH, Tewari M. MicroRNA profiling: approaches and considerations. Nat Rev Genet. 2012;13(5):358–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–8.

    Article  CAS  PubMed  Google Scholar 

  146. Candini O, Spano C, Murgia A, Grisendi G, Veronesi E, Piccinno MS, Ferracin M, Negrini M, Giacobbi F, Bambi F, Horwitz EM, Conte P, Paolucci P, Dominici M. Mesenchymal progenitors aging highlights a miR-196 switch targeting HOXB7 as master regulator of proliferation and osteogenesis. Stem Cells. 2015;33(3):939–50.

    Article  CAS  PubMed  Google Scholar 

  147. Li CJ, Cheng P, Liang MK, Chen YS, Lu Q, Wang JY, Xia ZY, Zhou HD, Cao X, Xie H, Liao EY, Luo XH. MicroRNA-188 regulates age-related switch between osteoblast and adipocyte differentiation. J Clin Invest. 2015;125(4):1509–22.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Fariyike B, Singleton Q, Hunter M, Hill WD, Isales CM, Hamrick MW, Fulzele S. Role of MicroRNA-141 in the aging musculoskeletal system: a current overview. Mech Ageing Dev. 2019;178:9–15.

    Article  CAS  PubMed  Google Scholar 

  149. Sangani R, Pandya CD, Bhattacharyya MH, Periyasamy-Thandavan S, Chutkan N, Markand S, Hill WD, Hamrick M, Isales C, Fulzele S. Knockdown of SVCT2 impairs in-vitro cell attachment, migration and wound healing in bone marrow stromal cells. Stem Cell Res. 2014;12(2):354–63.

    Article  CAS  PubMed  Google Scholar 

  150. Periyasamy-Thandavan S, Burke J, Mendhe B, Kondrikova G, Kolhe R, Hunter M, Isales CM, Hamrick MW, Hill WD, Fulzele S. MicroRNA-141-3p negatively modulates SDF-1 expression in age-dependent pathophysiology of human and murine bone marrow stromal cells. J Gerontol A Biol Sci Med Sci. 2019;74(9):1368–74.

    Article  CAS  PubMed  Google Scholar 

  151. Potter ML, Hill WD, Isales CM, Hamrick MW, Fulzele S. MicroRNAs are critical regulators of senescence and aging in mesenchymal stem cells. Bone. 2021;142:115679.

    Article  CAS  PubMed  Google Scholar 

  152. Liu W, Qi M, Konermann A, Zhang L, Jin F, Jin Y. The p53/miR-17/Smurf1 pathway mediates skeletal deformities in an age-related model via inhibiting the function of mesenchymal stem cells. Aging (Albany NY). 2015;7(3):205–18.

    Article  CAS  PubMed  Google Scholar 

  153. Guo Q, Chen Y, Guo L, Jiang T, Lin Z. miR-23a/b regulates the balance between osteoblast and adipocyte differentiation in bone marrow mesenchymal stem cells. Bone Res. 2016;4:16022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Sartori EM, Neves AM, Magro-Filho O, Mendonca DBS, Krebsbach PH, Cooper LF, Mendonca G. The role of MicroRNAs in the osseointegration process. Int J Oral Maxillofac Implants. 2019;34(2):397–410.

    Article  PubMed  Google Scholar 

  155. Geng Z, Yu YM, Li ZY, Ma LL, Zhu SL, Liang YQ, Cui ZD, Wang J, Yang XJ, Liu CS. miR-21 promotes osseointegration and mineralization through enhancing both osteogenic and osteoclastic expression. Mat Sci Eng C-Mater. 2020;111:110785.

    Article  CAS  Google Scholar 

  156. Geng Z, Wang X, Zhao J, Li Z, Ma L, Zhu S, Liang Y, Cui Z, He H, Yang X. The synergistic effect of strontium-substituted hydroxyapatite and microRNA-21 on improving bone remodeling and osseointegration. Biomater Sci. 2018;6(10):2694–703.

    Article  CAS  PubMed  Google Scholar 

  157. Wu K, Song W, Zhao L, Liu M, Yan J, Andersen MO, Kjems J, Gao S, Zhang Y. MicroRNA functionalized microporous titanium oxide surface by lyophilization with enhanced osteogenic activity. ACS Appl Mater Interfaces. 2013;5(7):2733–44.

    Article  CAS  PubMed  Google Scholar 

  158. Denu RA, Hematti P. Effects of oxidative stress on mesenchymal stem cell biology. Oxid Med Cell Longev. 2016;2016:2989076.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Kim M, Kim C, Choi YS, Kim M, Park C, Suh YS. Age-related alterations in mesenchymal stem cells related to shift in differentiation from osteogenic to adipogenic potential: implication to age-associated bone diseases and defects. Mech Ageing Dev. 2012;133(5):215–25.

    Article  PubMed  Google Scholar 

  160. Lin CH, Li NT, Cheng HS, Yen ML. Oxidative stress induces imbalance of adipogenic/osteoblastic lineage commitment in mesenchymal stem cells through decreasing SIRT1 functions. J Cell Mol Med. 2018;22(2):786–96.

    Article  CAS  PubMed  Google Scholar 

  161. Almeida M, Han L, Martin-Millan M, Plotkin LI, Stewart SA, Roberson PK, Kousteni S, O’Brien CA, Bellido T, Parfitt AM, Weinstein RS, Jilka RL, Manolagas SC. Skeletal involution by age-associated oxidative stress and its acceleration by loss of sex steroids. J Biol Chem. 2007;282(37):27285–97.

    Article  CAS  PubMed  Google Scholar 

  162. Lombard DB, Alt FW, Cheng HL, Bunkenborg J, Streeper RS, Mostoslavsky R, Kim J, Yancopoulos G, Valenzuela D, Murphy A, Yang Y, Chen Y, Hirschey MD, Bronson RT, Haigis M, Guarente LP, Farese RV, Weissman S, Verdin E, Schwer B. Mammalian sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation. Mol Cell Biol. 2007;27(24):8807–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Chen MH, Li MH, Wei YJ, Xue CC, Chen MW, Fei Y, Tan L, Luo Z, Cai KY, Hu Y. ROS-activatable biomimetic interface mediates in-situ bioenergetic remodeling of osteogenic cells for osteoporotic bone repair. Biomaterials. 2022;291:121878.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The work was supported by the National Natural Science Foundation of China (Grant number 82101644, 82271026, and 82100985) and Postdoctoral Research Foundation of China (Grant number 2020M671757). The funding body played no role in the design of the study, data collection, analysis, interpretation and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

YM, SYW, and HW drafted the manuscript and contributed equally. FMH, HMW, and YJM supervised the work and provided the comments. All the authors revised the manuscript and have read and agreed to the final version of the manuscript.

Corresponding authors

Correspondence to Huiming Wang, Yingjie Mao or Fuming He.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, Y., Wang, S., Wang, H. et al. Mesenchymal stem cells and dental implant osseointegration during aging: from mechanisms to therapy. Stem Cell Res Ther 14, 382 (2023). https://doi.org/10.1186/s13287-023-03611-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-023-03611-1

Keywords