Next Article in Journal
Digested Cinnamon (Cinnamomum verum J. Presl) Bark Extract Modulates Claudin-2 Gene Expression and Protein Levels under TNFα/IL-1β Inflammatory Stimulus
Next Article in Special Issue
The Role of mTORC1 Pathway and Autophagy in Resistance to Platinum-Based Chemotherapeutics
Previous Article in Journal
Purinergic Signalling in Physiology and Pathophysiology
Previous Article in Special Issue
Activation of the PI3K/AKT/mTOR Pathway in Cajal–Retzius Cells Leads to Their Survival and Increases Susceptibility to Kainate-Induced Seizures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia

1
State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
2
West China School of Medicine, Sichuan University, Chengdu 610041, China
3
Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9198; https://doi.org/10.3390/ijms24119198
Submission received: 3 April 2023 / Revised: 2 May 2023 / Accepted: 4 May 2023 / Published: 24 May 2023
(This article belongs to the Special Issue Regulation of mTOR Signaling in Human Diseases)

Abstract

:
The interplay between bone and glucose metabolism has highlighted hyperglycemia as a potential risk factor for bone diseases. With the increasing prevalence of diabetes mellitus worldwide and its subsequent socioeconomic burden, there is a pressing need to develop a better understanding of the molecular mechanisms involved in hyperglycemia-mediated bone metabolism. The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that senses extracellular and intracellular signals to regulate numerous biological processes, including cell growth, proliferation, and differentiation. As mounting evidence suggests the involvement of mTOR in diabetic bone disease, we provide a comprehensive review of its effects on bone diseases associated with hyperglycemia. This review summarizes key findings from basic and clinical studies regarding mTOR’s roles in regulating bone formation, bone resorption, inflammatory responses, and bone vascularity in hyperglycemia. It also provides valuable insights into future research directions aimed at developing mTOR-targeted therapies for combating diabetic bone diseases.

1. Introduction

Diabetes mellitus (DM) is a metabolic disorder that has become a worldwide epidemic [1]. It is characterized by hyperglycemia and multisystem complications that can impact patients’ quality of life and impose a significant socioeconomic burden on individuals and society [2]. The most well-documented DM complications include microvascular complications, such as retinopathy and nephropathy, and macrovascular complications, such as cardiovascular disease [3]. In recent decades, it has been increasingly recognized that DM also impairs bone health. Individuals with type 1 diabetes mellitus (T1DM) experience insulinopenia, which attenuates bone anabolism and results in reduced bone mineral density (BMD) [4], and an approximately sevenfold increase in the risk of hip fracture [5]. On the other hand, type 2 diabetes mellitus (T2DM) is associated with normal or high BMD but paradoxically increased fracture risk due to hyperglycemia-induced alterations in organic ingredients and skeletal microarchitecture [6]. Additionally, diabetic patients experience prolonged fracture healing times of approximately 87%, with a higher risk of delayed union, redislocation, and pseudoarthrosis [7,8,9]. With the increasing incidence of DM and the substantial socioeconomic burden it imposes globally, there is a pressing need for an improved understanding of bone metabolism in hyperglycemia.
The mammalian target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine kinase that acts as a central regulator of cellular and organismal growth and homeostasis [10,11]. It integrates various environmental inputs from nutrients and growth factors to regulate a diverse array of physiological processes, including macromolecular synthesis, ribosome biogenesis, cell growth, survival, and autophagy [12]. Initially, mTOR is considered a target of interest in cancer control due to its proliferation control. Later, mounting evidence confirmed that mTOR is particularly important to metabolic balance as well due to its response to nutrients [13]. Indeed, various endocrine disorders including DM and insulin resistance which are induced by aberrant energy homeostasis are often accompanied by deregulated mTOR signaling [10,11,14]. Hence, mTOR has been proposed as a promising therapeutic target for DM treatment [10,13]. Moreover, mTOR dysregulation is also heavily implicated in diabetes-related complications, including nephropathy, heart failure, neuropathy, and diabetic osteoporosis [4,15,16,17]. Regarding bone health, mTOR signaling is crucial to multiple aspects of skeletal development and health. Furthermore, mTOR signaling is crucial to multiple aspects of skeletal development and health [18]. Dysregulation of mTOR pathways renders bone marrow mesenchymal stem cells (BMSCs) unable to proliferate and differentiate properly, leading to bone loss and osteoporosis [18,19,20,21]. Recently, mTOR signaling has emerged as a pivotal regulator in bone metabolism under hyperglycemic conditions [22], making targeting mTOR a plausible approach for treating diabetic bone disorders. Thus, understanding the detailed molecular process of mTOR-regulated bone metabolism in hyperglycemia is critical for developing strategies to combat diabetic bone diseases. Herein, we summarized the current knowledge of mTOR in bone metabolism, with a particular emphasis on its role in diabetic bone disorders and the therapeutic potential of targeting mTOR pathways for bone health in diabetic patients.

2. mTOR Signaling Pathways

2.1. mTOR Complexes

mTOR exists in two structurally and functionally distinct complexes, known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) [12] (Figure 1). mTORC1 comprises five components: mTOR, mammalian lethal with sec-13 protein 8 (mLST8), regulatory-associated protein of mTOR (Raptor), DEP-domain containing mTOR-interacting protein (DEPTOR), and proline-rich AKT substrate of 40 kDa (PRAS40). mTORC2 is composed of six components: mTOR, mLST8, interacting protein 1 (mSIN1), rapamycin-insensitive companion of mTOR (Rictor), protein observed with Rictor 1 and 2 (Protor1/2), and DEPTOR [23,24,25,26]. For both mTORC1 and mTORC2, mLST8 acts as the positive regulator of mTOR activity while DEPTOR has a negative effect. For mTORC1, Raptor is a unique core subunit that modulates mTORC1 subcellular localization and substrate recognition by binding to the mTOR signaling motif, while PRAS40 inhibits mTOR activity [26,27,28]. Rictor is a key component of mTORC2 which is essential for mTORC2 function, while mSin1 and Protor1/2 serve as the regulatory subunits [12].

2.2. mTORC1 Signaling

mTORC1 is a crucial integrator of intracellular and extracellular signals, coordinating various biological processes, including protein synthesis, lipid synthesis, and autophagy [10,29]. The regulation of mTORC1 is well understood. Multiple factors, such as oxygen, energy, stress, and various growth factors, have been identified as activators of mTORC1 signaling by inhibiting the tuberous sclerosis complex (TSC) [18,30]. The TSC complex and the small guanine-5′-triphosphatase (GTPase), known as RAS homolog enriched in the brain (Rheb), are the major components responsible for transducing upstream signal to mTORC1 [31]. Whereas Rheb binds and activates mTORC1 directly, the activation of the TSC complex converts the active GTP-loaded Rheb into its inactive form, thereby negatively regulating mTORC1 activity [32]. Amino acids represent another main upstream stimulator of mTORC1. The availability of amino acids triggers the conversion of Ras-related GTP-binding protein homolog (Rag) family GTPases into their active conformation, which subsequently translocates mTORC1 to the lysosome, where Rheb is anchored and actives mTORC1 [33,34,35,36].
mTORC1 targets a variety of downstream molecules, including p70S6 kinase 1 (S6K1), eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1), sterol regulatory element binding protein (SREBP) and Unc-51-like kinase 1 (ULK1). The promotion of protein synthesis by mTORC1 activation primarily relies on S6K1 and 4EBP1 [37]. mTORC1 phosphorylates S6K1 on its hydrophobic motif and 4EBP1 at multiple sites to enhance the translation efficiency of spliced mRNAs, which further modulates protein synthesis [38,39,40,41]. mTORC1 also regulates de novo lipid synthesis by activating SREBP through both an S6K1-dependent manner and the phosphorylation of an additional substrate, known as Lipin1 [42,43,44]. In addition, mTORC1 inhibits autophagy by blocking ULK1 activation by 5′ adenosine monophosphate-activated protein kinase (AMPK) and phosphorylation of transcription factor EB (TFEB) which regulates gene expression related to lysosomal biogenesis and autophagy [45,46,47,48].

2.3. mTORC2 Signaling

In contrast to the varied upstream signals of mTORC1, the activity of mTORC2 is mainly regulated by growth factors, including insulin, which signals phosphoinositide 3-kinase (PI3K) [49]. The inhibition of mTORC2 catalytic activity by the mSin1 PH domain is relieved upon binding to phosphatidylinositol-3,4,5-triphosphate (PIP3) generated by insulin/PI3K signaling [50]. The presence of insulin also facilitates the association of mTORC2 with ribosomes to enhance mTORC2 activity [51]. Besides, the negative feedback loop between insulin/PI3K signaling and mTORC1 also has a regulatory effect on mTORC2 activity. The phosphorylation of two mTORC1 downstream targets, S6K1 and growth factor receptor-bound protein 10 (Grb10), negatively regulates the insulin signal, thereby inhibiting mTORC2 activity [52,53].
AKT, a crucial effector of insulin/PI3K signaling, acts as the primary downstream target of mTORC2 [54]. Upon phosphorylation and activation, AKT modulates downstream substrates such as Forkhead box O1/3 (FOXO1/3a), glycogen synthase kinase 3 β (GSK3β), and TSC2 to promote cell growth, proliferation, and survival [55,56]. Moreover, mTORC2 regulates ion transport and cell survival through serum/glucocorticoid-regulated kinase 1 (SGK1) [54,57].

3. mTOR in Bone Metabolism

Bone metabolism is a complicated process requiring constant bone formation and resorption, which is regulated by the dynamic interplay between osteoblasts, osteoclasts, and various signaling pathways [58]. Exposure to a diabetic environment has profound adverse effects on bone metabolism, which compromises the structural and functional integrity of the skeletal system and leads to various clinical manifestations. The molecular process behind bone pathogenesis in hyperglycemia has been extensively studied, with particular attention given to mTOR signaling in recent years. In this subsection, we presented a summary of the current knowledge regarding the involvement of mTOR signaling in bone metabolism under high glucose conditions (Figure 2).

3.1. mTOR in BMSCs Osteogenesis and Bone Formation

BMSCs represent a multipotent precursor population, playing an essential role in bone homeostasis and formation via their osteogenic differentiation potential. The commitment of BMSCs to either an osteogenic or adipogenic lineage is delicately orchestrated by multiple mechanisms in physiological conditions. Nevertheless, mounting evidence suggests that the shift in BMSC differentiation toward adipogenesis in hyperglycemic conditions is likely associated with altered mTOR signaling activity.
The major pathophysiology of T1DM and T2DM involves insulin deficiency and insulin resistance, respectively. However, insulin and insulin-like growth factor (IGF-1) have been shown to exert bone anabolic effects through modulating mTOR activity [59]. Specifically, treatment with IGF-1 has been found to enhance the osteogenic differentiation of stem cells by significantly increasing the phosphorylation of AKT and p70S6K in a dose-dependent manner [20,59]. This effect was found to be reversed by inhibitors of PI3K and mTOR, such as LY294002 and rapamycin [20]. Furthermore, excessive activation of mTOR induced by the knockdown of TSC2 inhibited insulin sensitivity in osteoblasts, leading to decreased osteogenic markers [60]. These results demonstrate the significant regulatory role of insulin and IGF-1 via the mTOR pathway in bone formation, which may be compromised in diabetic conditions due to reduced insulin stimulation. Indeed, extracts from Rehmanniae Radix Praeparata have been shown to alleviate bone loss and architectural deterioration in diabetic rats by promoting IGF-1 expression and activation of the downstream PI3K/AKT/mTOR pathway [61]. Glutamine, the richest semi-essential amino acid in the human body, signals to mTOR through Rag GTPase-independent mechanisms [62]. Under high glucose conditions, the increased glutamine concentration has been reported to hyperactivate mTORC1, which inhibited mTORC2 activity through the phosphorylation of S6K1, thereby reducing the runt-related transcription factor 2 (RUNX2) expression of murine mesenchymal stem cells (MSCs) along with impaired extracellular matrix calcification [22].
Either acute or chronic hyperglycemia leads to the accumulation of reactive oxygen species (ROS) [63], resulting in oxidative stress that can trigger cell senescence [64,65,66], apoptosis [64,67], and autophagy [65,68]. Such stress has been associated with the onset of DM and the development of diabetic bone complications [69]. As a critical regulator of BMSCs survival and function, the mTOR signaling pathway is involved in ROS-mediated pathological alterations. In osteoblastic MC3T3-E1 cells exposed to high glucose, ROS production significantly inhibited the AKT/mTOR pathway, upregulated autophagy-related genes, and boosted autophagy [68]. Despite its pro-survival role, mTOR-mediated autophagy appears to be a double-edged sword in diabetic impairment of osteogenesis. In hyperglycemia, the autophagy promoted by AMPK phosphorylation and mTOR inhibition partially rescued the compromised BMSCs osteogenic differentiation, and the downregulation of autophagy led to opposite outcomes [70]. Additionally, inhibition of mTOR by rapamycin in BMSCs treated with high glucose showed osteogenic protection and anti-apoptotic effects [65]. However, apoptotic BMSCs isolated from diabetic mice exhibited endoplasmic reticulum stress (ERS), which enhanced autophagy by inhibiting mTOR and induced apoptosis [67]. Besides, excessive ERS caused by glutathione peroxidase 7 (GPx7) knockdown negatively regulated AKT/mTOR activity and inhibited osteogenic differentiation of BMSCs [71]. Similarly, arginine pyrimidine (APMD), a core product of DM, downregulated the PI3K/AKT/mTOR pathway in periodontal cells and activated autophagy, thus promoting periodontal bone destruction [72].
Peroxisome proliferator-activated receptor γ (PPARγ) is an essential nuclear transcription factor for the balance between adipogenesis and osteogenesis of BMSCs. Recent studies have recognized the existence of crosstalk between mTOR and PPARγ during BMSC differentiation. The overexpression of miRNA188 in BMSCs directly inhibited Rictor, thus promoting the activity of PPARγ and enhancing adipogenic differentiation and adipose accumulation within the bone marrow microenvironment [73]. Furthermore, the heightened expression levels of DEPTOR in BMSCs isolated from osteoporotic mice induced by ovariectomy inhibited the nuclear translocation of transcriptional coactivator with a PDZ-binding motif (TAZ) thereby repressing RUNX2 transcription by facilitating PPARγ transcription [74]. These findings highlight the potential involvement of PPARγ in the mTOR-mediated osteogenesis in hyperglycemia, which requires further research for comprehensive elucidation.
Exposure to a hyperglycemic environment has been demonstrated to modify the characteristics of BMSCs and induce senescence [75] with a limited understanding of the contributions of the mTOR pathway [76]. Recent research indicates that reduced mTOR activity plays a critical role in the initiation of BMSCs senescence [77]. Interestingly, although BMSCs senescence stemming from autophagy under high glucose conditions is not affected by further mTOR inhibition using rapamycin [65], activation of the PI3K/AKT/mTOR pathway contributes to BMSCs senescence by inhibiting the Indian hedgehog pathway, and mTOR inhibitors reversed the senescent state of BMSCs [78]. Consistent with this, rapamycin-mediated mTOR inhibition protected BMSCs against oxidative stress-induced senescence, as evidenced by the mitigated senescence phenotype and mitochondrial damage [79].

3.2. mTOR in Osteoclast Formation and Bone Resorption

Osteoclasts are essential for bone resorption as they secrete degradative enzymes and create an acidic environment on the bone surface to demineralize bone tissue [80]. While some studies suggest that a diabetic environment may increase osteoclast differentiation and activity, ultimately leading to reduced bone mass and osteoporosis [81,82,83,84], others indicate that hyperglycemia and advanced glycation end products (AGEs) may inhibit osteoclast activity, with abnormal bone resorption and turnover [85,86,87].
The role of mTOR in regulating osteoclast differentiation and function is complicated [88,89]. The receptor activator of NF-kB ligand/osteoprotegerin/receptor activator of the NF-kB (RANKL/OPG/RANK) system is pivotal for osteoclastogenesis, with RANKL stimulating osteoclast differentiation by binding to RANK receptors on osteoclast precursor cells, while OPG acts as a competitive inhibitor [80]. mTORC1 has been shown to exert a biphasic regulatory effect on RANKL-directed osteoclastogenesis [90,91], with its activity in bone marrow-derived macrophages being activated in the early stages, but inhibited in the later stages following RANKL treatment [92]. Either inhibition of mTORC1 at an early stage or activation at a late stage during osteoclastogenesis suppressed osteoclast differentiation and bone resorption [92]. In contrast, a further study found that inhibiting mTORC1 at the late stage could also enhance osteoclast formation by promoting phosphorylation of the nuclear factor of activated T cells 1 (NFATc1) [91]. OPG inhibits mTOR by suppressing the PI3K/AKT pathway or activating the AMPK pathway, inducing autophagy and diminishing osteoclast viability, differentiation, and bone resorption activity [93,94,95]. Of note, the activation of autophagy does not necessarily suppress osteoclastogenesis. Mounting evidence has indicated that autophagy may be positively correlated with osteoclastic activity [89,96].
The detailed mechanism underlying the regulation of mTOR in osteoclastic activity in hyperglycemia is not well understood. It has been found that hyperglycemia inhibited the AMPK/mTOR/ULK1 pathway, thereby suppressing autophagy and inhibiting the formation and function of osteoclasts [97], whereas activation of the PI3K/AKT/mTOR pathway in diabetic mice reduced the RANKL/OPG ratio of osteoblastic cells, thus alleviating bone resorption [98]. Future studies are needed to obtain a better understanding regarding the involvement of the mTOR pathway in bone resorption in hyperglycemia.

3.3. mTOR in Inflammatory Response

DM is known to alter immune system components and has been considered an inflammatory disease [99]. The inflammatory microenvironment induced by hyperglycemia includes hyperactivated immune cells, increased chemokines and pro-inflammatory factors such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), and altered ratios of T helper 17 (Th17) cells and T regulatory (Treg) cells in the peripheral blood [100,101,102]. These factors exacerbate the suppression of osteoblastic activity [102,103]. mTOR is closely associated with immune and inflammatory responses. Inhibition of mTOR blocked NLR family pyrin domain containing (NLRP3) inflammasome activation and facilitated macrophage polarization toward the M2 subtype [104], which secretes multiple anti-inflammatory factors and promotes bone regeneration [105,106,107]. mTOR also promoted CD4+ T cell differentiation into Th17 cells, thus enhancing the adaptive immune responses [108]. In addition, targeting mTOR has been shown to regulate cytokine production and alleviate the development and severity of several inflammatory diseases, such as spondyloarthritis [109], rosacea [110], and acute gouty arthritis [111].
It has been shown that the high glucose-mediated mTOR phosphorylation in bone marrow-derived macrophages resulted in a senescent phenotype and upregulation of pro-inflammatory factors, ultimately exacerbating macrophage inflammation and periodontal bone destruction [112]. In addition, hyperglycemia was also demonstrated to promote macrophage pyroptosis and pro-inflammatory factor secretion by activating mTOR/4EBP1 and decreasing downstream ULK1 activity and autophagy flux, which in turn, aggravates alveolar bone resorption in periodontitis [113]. On the contrary, activation of the mTOR/AKT pathway inhibited pro-inflammatory factor production in hyperglycemic bone marrow-derived macrophages [114]. The conflicting results might be attributed to the different sources of macrophages. Moreover, the mTOR pathway regulates the responses of myeloid-derived suppressor cells (MDSCs), which restrict immune responses in bone repair [115]. High glucose promoted the differentiation of bone marrow-derived MDSCs into pro-inflammatory M1 macrophages, stimulating the accumulation of abnormal MDSCs in bone tissue [116]. This interference of immunosuppression depends on mTOR activation with phosphorylation of 4EBP1 and S6K1 and could be reversed by an mTOR kinase inhibitor [116]. Since M1 macrophage could inhibit osteoblastogenesis and facilitate apoptosis of MSCs [117,118], diabetes-stimulated M1 macrophage differentiation might be responsible for impaired bone defect regeneration in hyperglycemia. Hence, mTOR seems to positively regulate inflammatory responses in bone tissue and aggravate diabetes-induced bone metabolic aberrations.

3.4. mTOR in Bone Vascularity

The vascular components within the skeletal system play a vital role in regulating bone metabolism by facilitating the necessary supply of nutrients and biochemical factors, mobilizing bone progenitor cells, and balancing osteogenesis and osteolysis [119]. Diseased conditions such as DM have been shown to negatively impact the expression of angiogenesis-related genes in the skeletal system and impair BMSCs and pericyte functionality, leading to pro-angiogenic dysfunction [120,121,122]. T1DM mice demonstrated vascular lesions in bone, especially damage to type H blood vessels, which are responsible for coupling vascularization with osteogenesis [123], while the reduced microvascular blood flow in T2DM patients was notably linked to elevated cortical bone porosity [124]. Besides, inadequate vascularization under high-glucose conditions was detrimental to bone regeneration [125]. Overall, vascularization dysfunction constitutes a significant contributor to abnormal bone metabolism in diabetic conditions.
The mTOR pathway contributes to vascular dysfunction in diabetic bone by affecting endothelial cell (EC) functionality. mTOR activation is necessary for the tube formation of bone marrow-derived endothelial progenitor cells (EPCs) and human umbilical vein endothelial cells (HUVECs) [126,127], which can be inhibited in hyperglycemia through mTOR suppression. Mechanistically, the increased expression of circ-ADAM9 in EPCs induced by high glucose conditions negatively regulated its sponge microRNA-20a-5p, leading to increased autophagy and apoptosis in EPCs through inhibiting mTOR phosphorylation [128]. Additionally, the knockdown of circ-ADAM9 greatly reduced autophagy and apoptosis-associated protein expression in EPCs and increased tissue perfusion rates in diabetic mice [128]. Furthermore, upregulated miR-328 prevented the angiogenesis of HUVECs by suppressing AKT/mTOR pathway in a high-glucose low-serum environment [129], whereas activation of the PI3K/AKT/mTOR pathway effectively counteracts the anti-angiogenic effects of high glucose, promoting diabetic wound healing in vivo [130].
The function of the cellular components in the skeletal system under the diabetic environment is precisely regulated by the diabetic microenvironment (extracellular signals) and the consequent cascade of intracellular signals. Hyperglycemia is the most obvious and prominent extracellular signal. Its induced intracellular glucose metabolism disturbance and the ensuing redox imbalance cause oxidative stress and excessive production of ROS, which sets the cellular foundation for the establishment of skeletal complications [131,132]. At the molecular level, oxidative stress stimulates phosphatase activity, deactivating PI3K and disrupting the intracellular transduction of anabolic insulin signals [133]. Besides, ROS also results in the inactivation and degradation of AMPK by inhibiting its phosphorylation and promoting MG53-mediated ubiquitination [134]. The altered activity of PI3K and AMPK leads to the disruption of the normal intracellular transmission of mTOR signals. According to available findings, alterations in mTOR pathway activity mainly affect the downstream S6K1- and 4EBP1-mediated protein synthesis pathways and the ULK1-mediated autophagy pathway, which in turn leads to disruption of cellular component function and activity, thereby interrupting the balance between bone formation and bone resorption in the diabetic microenvironment.

4. Therapeutic Prospects

Strategies for alleviating the adverse impacts of hyperglycemia on bone metabolism are in urgent demand. The comprehensive regulatory effects of the mTOR pathway on the onset and development of diabetic bone complications render mTOR a potential therapeutic target. Here, we summarized several current mTOR-related drugs for combating skeletal deterioration (Table 1), providing insights for improving bone metabolism in the context of DM.

4.1. Inhibition of mTOR Pathway

The activation of AMPK inhibits mTOR, thus stimulating osteogenesis [149,150]. Metformin, a first-line agent for diabetic treatment, is known as an AMPK activator [151]. Multiple clinical studies have found that patients treated with metformin had lower fracture risk [152], and regulation of the AMPK/mTOR pathway might be one of the hidden mechanisms. By activating AMPK and inhibiting mTOR, metformin could regenerate the osteogenesis of ACSssuppressed by high glucose [135]. Besides, metformin-induced AMPK activation could inhibit the overactivation of mTORC1 caused by high glutamine in MSCs and eventually increase RUNX2 expression through the upregulated mTORC2/AKT-473 axis [22]. In addition to promoting osteogenesis, mTOR inhibition by metformin was also found to suppress adipogenesis mediated by PPAR-γ [136]. Interestingly, metformin might also indirectly activate mTOR in MSCs to contribute to osteogenesis. Shen et al. revealed that metformin could promote macrophage M2 polarization, and after coculture with metformin-pretreated M2 macrophages, MSCs exhibited higher PI3K/AKT/mTOR signaling activity and increased osteoblast differentiation and bone formation ability [153]. Despite its promotive effect on osteogenesis, it is concerning that the alteration in mTOR activity induced by metformin might result in the suppressed angiogenic function of BMSCs [154]. As for clinical practice, metformin is an insulin sensitizer with secondary protection against bone loss, thus metformin might not be considered an anti-osteoporotic drug [155]. The optimal dosing regimen for metformin to exert its bone repair function has not been fully understood either [156]. Studies showed that metformin promoted osteogenesis differentiation by regulating the AMPK pathway with a wide dose range from 0.5 μM to 500 μM [157].
Rapamycin, an mTORC1 inhibitor, might serve as a both beneficial and detrimental agent for DM and coexisting skeletal complications. For glucose metabolism, rapamycin administration is effective in promoting insulin secretion and glucose uptake in the short term [158], but chronic treatment may exacerbate hyperglycemia and insulin resistance [159]. In addition, the effects of rapamycin on osteoblastogenesis and osteoclastogenesis in vitro appeared to be dose-dependent, with lower doses promoting osteoblastogenesis and osteoclastogenesis while higher doses inhibited these processes [91,137,160,161]. In terms of osteoclastogenesis, a clinically relevant dose of rapamycin to treat cancer and suppress immune response was considered low by the above standard, thus clinically appropriate rapamycin might positively regulate osteoclastogenesis and facilitate bone resorption [91]. What is more, rapamycin might have negative effects on bone formation in young animals [138]. Therefore, the use of this particular drug in pediatric patients should be extra cautious [66].
However, studies have also found that inhibiting mTOR might lead to the inhibition of osteogenic differentiation. For example, Liraglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA) that could improve diabetic osteoporosis [162], was found to suppress the differentiation of osteoblasts by activating AMPK and inhibiting mTOR [139]. Interestingly, by activating the GLP-1 receptor and upstream PI3K/AKT pathway of mTOR, liraglutide could also promote osteogenic differentiation [163,164] and inhibit the apoptosis of osteoblasts [165] to regenerate bone formation. The activation of the two signaling pathways might result from different dosing regimens, and the precise function of mTOR in liraglutide treatment remains a challenge to be solved.

4.2. Activation of mTOR Pathway

In another way, the activation of PI3K/AKT/mTOR signaling might be beneficial for diabetic patients with bone complications. The activated PI3K/AKT/mTOR signaling pathway has been found to promote proliferation and differentiation [166]. Thus, developing a therapeutic approach to activate this signaling to maintain bone metabolism might be a worthy pursuit. As expected, Rehmannia glutinosa Libosch extracts were found to stimulate PI3K/AKT/mTOR signaling pathway and thus increase the proliferation and differentiation of osteoblastic MC3T3-E1 cells injured by high glucose [61]. In addition, activating upstream PI3K/AKT of mTOR, S-Equol and the combination of exendin-4 (Ex-4) and eldecalcitol (ED-71) could improve diabetic osteoporosis in vivo [98,167]. Furthermore, many other compounds that activate this signaling pathway exerted positive effects on osteoblasts, though lacking evidence proving their effects in the context of diabetes. For example, the activation of the PI3K/AKT/mTOR pathway by Naringin promoted the proliferation and differentiation of osteoblasts [143]. Moreover, activation of the PI3K/AKT/mTOR signaling pathway by tocopherol might cause attenuated ferroptosis in BMSCs and protect the cells from oxidative stress [140]. However, the effects of activating PI3K/AKT/mTOR signaling might be negative in osteoclasts. For example, the activation of the PI3K/AKT/mTOR signaling pathway by cholesterol could inhibit autophagy during osteoclast differentiation, thereby worsening osteoporosis [168].
Given the core position of the mTOR pathway in metabolic regulation and bone turnover, mTOR-targeted therapeutic strategies have the potential to combat diabetes-related bone diseases. In preclinical investigations, the dominant state of mTOR (activation or suppression) and the interaction with other signaling pathways need to be further clarified to better understand the pathology of diabetic bone complications. Besides, since mTOR exerts different impacts on different kinds of cells, how to target the desired cell types precisely is significant as well, to develop a much more precise treatment modality while reducing the side effects. Moreover, whether an mTOR-targeted agent adheres to a certain diabetes state needs to be elucidated to avoid misuse. Current clinical trials have mainly focused on alleviating diabetes and its complications with mTOR inhibitors, especially metformin. However, despite the potential osteogenic effects in preclinical investigations, metformin treatment could not yield optimal outcomes in improving bone metabolism of patients with T2DM, with BMD, trabecular bone score (TBS), and bone turnover marker levels taken into consideration [169,170,171]. Therefore, based on sufficient preclinical evidence, more clinical studies need to be conducted to evaluate the effectiveness of mTOR-targeted agents against diabetic bone diseases. For safety control, routine monitoring of glycemia and cardiovascular and renal function is required. For efficacy measurement, both glycemic and skeletal outcomes should be evaluated, and bone turnover-related parameters should be as thorough as possible, including BMD, TBS, markers representing bone formation and resorption, and assessment of fracture risk in the long term.

5. Conclusions

The mTOR signaling pathway is essential in regulating multiple aspects of skeletal development and homeostasis. The regulatory effects of mTORC1 on bone formation and resorption have long been recognized, and a growing body of evidence demonstrates that mTORC2 is also pivotal for bone physiology. Because of its crucial role in bone metabolism, the dysregulation of mTOR signaling in hyperglycemia is associated with the bone complications of individuals with DM.
We have thoroughly reviewed the current knowledge regarding the involvement of mTOR signaling in bone metabolism in hyperglycemia, including its effects on bone formation, bone resorption, inflammatory responses and bone vascularity. Despite these significant progresses, numerous challenges remain in elucidating the precise role of mTOR in diabetic bone complications and developing targeted therapeutic strategies. Specifically, it is crucial to further investigate the mechanisms by which hyperglycemia disrupts the integration of extracellular and intracellular signals and how this dysregulation affects mTOR pathway activity. Identifying the downstream effectors of mTOR that mediate bone metabolism in hyperglycemia is also a key priority. In addition, given the complex interplay between mTOR signaling and various physiological processes, developing tissue-specific approaches to modulate mTOR pathway activity and investigating potential side effects in non-skeletal organs will be critical for developing an effective therapy for diabetic bone complications.

Author Contributions

Conceptualization, S.W. (Shuangcheng Wang), J.Y. and M.C.; writing—original draft preparation, S.W. (Shuangcheng Wang), J.W., S.W. (Shuangwen Wang) and R.T.; writing—review and editing, J.Y.; visualization, S.W. (Shuangcheng Wang), J.W. and S.W. (Shuangwen Wang); supervision, J.Y., M.C. and Z.Z.; project administration, Z.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lovic, D.; Piperidou, A.; Zografou, I.; Grassos, H.; Pittaras, A.; Manolis, A. The Growing Epidemic of Diabetes Mellitus. Curr. Vasc. Pharmacol. 2020, 18, 104–109. [Google Scholar] [CrossRef] [PubMed]
  2. Oh, S.H.; Ku, H.; Park, K.S. Prevalence and socioeconomic burden of diabetes mellitus in South Korean adults: A population-based study using administrative data. BMC Public Health 2021, 21, 548. [Google Scholar] [CrossRef] [PubMed]
  3. Hygum, K.; Starup-Linde, J.; Langdahl, B.L. Diabetes and bone. Osteoporos. Sarcopenia 2019, 5, 29–37. [Google Scholar] [CrossRef] [PubMed]
  4. Romero-Díaz, C.; Duarte-Montero, D.; Gutiérrez-Romero, S.A.; Mendivil, C.O. Diabetes and Bone Fragility. Diabetes Ther. 2021, 12, 71–86. [Google Scholar] [CrossRef]
  5. Schwartz, A.V.; Lane, N.E. Bone and Joint Complications in Diabetes. In Diabetes in America, 3rd ed.; Cowie, C.C., Casagrande, S.S., Menke, A., Cissell, M.A., Eberhardt, M.S., Meigs, J.B., Gregg, E.W., Knowler, W.C., Barrett-Connor, E., Becker, D.J., et al., Eds.; National Institute of Diabetes and Digestive and Kidney Diseases (US): Bethesda, MD, USA, 2018. [Google Scholar]
  6. Vestergaard, P. Discrepancies in bone mineral density and fracture risk in patients with type 1 and type 2 diabetes—A meta-analysis. Osteoporos. Int. 2007, 18, 427–444. [Google Scholar] [CrossRef]
  7. Jiao, H.; Xiao, E.; Graves, D.T. Diabetes and Its Effect on Bone and Fracture Healing. Curr. Osteoporos. Rep. 2015, 13, 327–335. [Google Scholar] [CrossRef]
  8. Retzepi, M.; Donos, N. The effect of diabetes mellitus on osseous healing. Clin. Oral. Implant. Res. 2010, 21, 673–681. [Google Scholar] [CrossRef]
  9. Wang, H.; Ba, Y.; Xing, Q.; Du, J.L. Diabetes mellitus and the risk of fractures at specific sites: A meta-analysis. BMJ Open 2019, 9, e024067. [Google Scholar] [CrossRef]
  10. Laplante, M.; Sabatini, D.M. mTOR signaling in growth control and disease. Cell 2012, 149, 274–293. [Google Scholar] [CrossRef]
  11. Howell, J.J.; Manning, B.D. mTOR couples cellular nutrient sensing to organismal metabolic homeostasis. Trends Endocrinol. Metab. 2011, 22, 94–102. [Google Scholar] [CrossRef]
  12. Liu, G.Y.; Sabatini, D.M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 183–203. [Google Scholar] [CrossRef] [PubMed]
  13. Swierczynska, M.M.; Hall, M.N. mTOR in Metabolic and Endocrine Disorders. In Molecules to Medicine with mTOR; Maiese, K., Ed.; Academic Press: Boston, MA, USA, 2016; pp. 347–364. [Google Scholar]
  14. Albert, V.; Hall, M.N. mTOR signaling in cellular and organismal energetics. Curr. Opin. Cell Biol. 2015, 33, 55–66. [Google Scholar] [CrossRef] [PubMed]
  15. Abou Daher, A.; Alkhansa, S.; Azar, W.S.; Rafeh, R.; Ghadieh, H.E.; Eid, A.A. Translational Aspects of the Mammalian Target of Rapamycin Complexes in Diabetic Nephropathy. Antioxid. Redox Signal. 2022, 37, 802–819. [Google Scholar] [CrossRef] [PubMed]
  16. Suhara, T.; Baba, Y.; Shimada, B.K.; Higa, J.K.; Matsui, T. The mTOR Signaling Pathway in Myocardial Dysfunction in Type 2 Diabetes Mellitus. Curr. Diabetes Rep. 2017, 17, 38. [Google Scholar] [CrossRef] [PubMed]
  17. Burillo, J.; Marqués, P.; Jiménez, B.; González-Blanco, C.; Benito, M.; Guillén, C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021, 10, 1236. [Google Scholar] [CrossRef] [PubMed]
  18. Chen, J.; Long, F. mTOR signaling in skeletal development and disease. Bone Res. 2018, 6, 1. [Google Scholar] [CrossRef]
  19. Singha, U.K.; Jiang, Y.; Yu, S.; Luo, M.; Lu, Y.; Zhang, J.; Xiao, G. Rapamycin inhibits osteoblast proliferation and differentiation in MC3T3-E1 cells and primary mouse bone marrow stromal cells. J. Cell. Biochem. 2008, 103, 434–446. [Google Scholar] [CrossRef]
  20. Xian, L.; Wu, X.; Pang, L.; Lou, M.; Rosen, C.J.; Qiu, T.; Crane, J.; Frassica, F.; Zhang, L.; Rodriguez, J.P.; et al. Matrix IGF-1 maintains bone mass by activation of mTOR in mesenchymal stem cells. Nat. Med. 2012, 18, 1095–1101. [Google Scholar] [CrossRef]
  21. Lai, P.; Song, Q.; Yang, C.; Li, Z.; Liu, S.; Liu, B.; Li, M.; Deng, H.; Cai, D.; Jin, D.; et al. Loss of Rictor with aging in osteoblasts promotes age-related bone loss. Cell Death Dis. 2016, 7, e2408. [Google Scholar] [CrossRef]
  22. Gayatri, M.B.; Gajula, N.N.; Chava, S.; Reddy, A.B.M. High glutamine suppresses osteogenesis through mTORC1-mediated inhibition of the mTORC2/AKT-473/RUNX2 axis. Cell Death Discov. 2022, 8, 277. [Google Scholar] [CrossRef]
  23. Jhanwar-Uniyal, M.; Wainwright, J.V.; Mohan, A.L.; Tobias, M.E.; Murali, R.; Gandhi, C.D.; Schmidt, M.H. Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. Adv. Biol. Regul. 2019, 72, 51–62. [Google Scholar] [CrossRef] [PubMed]
  24. Srinivas, K.P.; Viji, R.; Dan, V.M.; Sajitha, I.S.; Prakash, R.; Rahul, P.V.; Santhoshkumar, T.R.; Lakshmi, S.; Pillai, M.R. DEPTOR promotes survival of cervical squamous cell carcinoma cells and its silencing induces apoptosis through downregulating PI3K/AKT and by up-regulating p38 MAP kinase. Oncotarget 2016, 7, 24154–24171. [Google Scholar] [CrossRef] [PubMed]
  25. Sancak, Y.; Thoreen, C.C.; Peterson, T.R.; Lindquist, R.A.; Kang, S.A.; Spooner, E.; Carr, S.A.; Sabatini, D.M. PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase. Mol. Cell 2007, 25, 903–915. [Google Scholar] [CrossRef] [PubMed]
  26. Hara, K.; Maruki, Y.; Long, X.; Yoshino, K.; Oshiro, N.; Hidayat, S.; Tokunaga, C.; Avruch, J.; Yonezawa, K. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 2002, 110, 177–189. [Google Scholar] [CrossRef]
  27. Yang, H.; Rudge, D.G.; Koos, J.D.; Vaidialingam, B.; Yang, H.J.; Pavletich, N.P. mTOR kinase structure, mechanism and regulation. Nature 2013, 497, 217–223. [Google Scholar] [CrossRef] [PubMed]
  28. Pearce, L.R.; Huang, X.; Boudeau, J.; Pawłowski, R.; Wullschleger, S.; Deak, M.; Ibrahim, A.F.; Gourlay, R.; Magnuson, M.A.; Alessi, D.R. Identification of Protor as a novel Rictor-binding component of mTOR complex-2. Biochem. J. 2007, 405, 513–522. [Google Scholar] [CrossRef] [PubMed]
  29. Ben-Sahra, I.; Manning, B.D. mTORC1 signaling and the metabolic control of cell growth. Curr. Opin. Cell Biol. 2017, 45, 72–82. [Google Scholar] [CrossRef] [PubMed]
  30. Shimobayashi, M.; Hall, M.N. Making new contacts: The mTOR network in metabolism and signalling crosstalk. Nat. Rev. Mol. Cell Biol. 2014, 15, 155–162. [Google Scholar] [CrossRef]
  31. Tee, A.R.; Manning, B.D.; Roux, P.P.; Cantley, L.C.; Blenis, J. Tuberous Sclerosis Complex Gene Products, Tuberin and Hamartin, Control mTOR Signaling by Acting as a GTPase-Activating Protein Complex toward Rheb. Curr. Biol. 2022, 32, 733–734. [Google Scholar] [CrossRef]
  32. Inoki, K.; Li, Y.; Xu, T.; Guan, K.L. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 2003, 17, 1829–1834. [Google Scholar] [CrossRef]
  33. Sancak, Y.; Peterson, T.R.; Shaul, Y.D.; Lindquist, R.A.; Thoreen, C.C.; Bar-Peled, L.; Sabatini, D.M. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 2008, 320, 1496–1501. [Google Scholar] [CrossRef] [PubMed]
  34. Sancak, Y.; Bar-Peled, L.; Zoncu, R.; Markhard, A.L.; Nada, S.; Sabatini, D.M. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 2010, 141, 290–303. [Google Scholar] [CrossRef] [PubMed]
  35. Bar-Peled, L.; Schweitzer, L.D.; Zoncu, R.; Sabatini, D.M. Ragulator is a GEF for the rag GTPases that signal amino acid levels to mTORC1. Cell 2012, 150, 1196–1208. [Google Scholar] [CrossRef] [PubMed]
  36. Bar-Peled, L.; Chantranupong, L.; Cherniack, A.D.; Chen, W.W.; Ottina, K.A.; Grabiner, B.C.; Spear, E.D.; Carter, S.L.; Meyerson, M.; Sabatini, D.M. A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 2013, 340, 1100–1106. [Google Scholar] [CrossRef] [PubMed]
  37. Holz, M.K.; Ballif, B.A.; Gygi, S.P.; Blenis, J. mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events. Cell 2005, 123, 569–580. [Google Scholar] [CrossRef]
  38. Dorrello, N.V.; Peschiaroli, A.; Guardavaccaro, D.; Colburn, N.H.; Sherman, N.E.; Pagano, M. S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth. Science 2006, 314, 467–471. [Google Scholar] [CrossRef]
  39. Ma, X.M.; Yoon, S.O.; Richardson, C.J.; Jülich, K.; Blenis, J. SKAR links pre-mRNA splicing to mTOR/S6K1-mediated enhanced translation efficiency of spliced mRNAs. Cell 2008, 133, 303–313. [Google Scholar] [CrossRef]
  40. Hsieh, A.C.; Liu, Y.; Edlind, M.P.; Ingolia, N.T.; Janes, M.R.; Sher, A.; Shi, E.Y.; Stumpf, C.R.; Christensen, C.; Bonham, M.J.; et al. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 2012, 485, 55–61. [Google Scholar] [CrossRef]
  41. Thoreen, C.C.; Chantranupong, L.; Keys, H.R.; Wang, T.; Gray, N.S.; Sabatini, D.M. A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 2012, 485, 109–113. [Google Scholar] [CrossRef]
  42. Düvel, K.; Yecies, J.L.; Menon, S.; Raman, P.; Lipovsky, A.I.; Souza, A.L.; Triantafellow, E.; Ma, Q.; Gorski, R.; Cleaver, S.; et al. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol. Cell 2010, 39, 171–183. [Google Scholar] [CrossRef]
  43. Peterson, T.R.; Sengupta, S.S.; Harris, T.E.; Carmack, A.E.; Kang, S.A.; Balderas, E.; Guertin, D.A.; Madden, K.L.; Carpenter, A.E.; Finck, B.N.; et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 2011, 146, 408–420. [Google Scholar] [CrossRef] [PubMed]
  44. Porstmann, T.; Santos, C.R.; Griffiths, B.; Cully, M.; Wu, M.; Leevers, S.; Griffiths, J.R.; Chung, Y.L.; Schulze, A. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008, 8, 224–236. [Google Scholar] [CrossRef] [PubMed]
  45. Settembre, C.; Zoncu, R.; Medina, D.L.; Vetrini, F.; Erdin, S.; Erdin, S.; Huynh, T.; Ferron, M.; Karsenty, G.; Vellard, M.C.; et al. A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. EMBO J. 2012, 31, 1095–1108. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef]
  47. Martina, J.A.; Chen, Y.; Gucek, M.; Puertollano, R. MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy 2012, 8, 903–914. [Google Scholar] [CrossRef]
  48. Roczniak-Ferguson, A.; Petit, C.S.; Froehlich, F.; Qian, S.; Ky, J.; Angarola, B.; Walther, T.C.; Ferguson, S.M. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci. Signal. 2012, 5, ra42. [Google Scholar] [CrossRef]
  49. Yoon, M.S. The Role of Mammalian Target of Rapamycin (mTOR) in Insulin Signaling. Nutrients 2017, 9, 1176. [Google Scholar] [CrossRef]
  50. Liu, P.; Gan, W.; Chin, Y.R.; Ogura, K.; Guo, J.; Zhang, J.; Wang, B.; Blenis, J.; Cantley, L.C.; Toker, A.; et al. PtdIns(3,4,5)P3-Dependent Activation of the mTORC2 Kinase Complex. Cancer Discov. 2015, 5, 1194–1209. [Google Scholar] [CrossRef]
  51. Zinzalla, V.; Stracka, D.; Oppliger, W.; Hall, M.N. Activation of mTORC2 by association with the ribosome. Cell 2011, 144, 757–768. [Google Scholar] [CrossRef]
  52. Hsu, P.P.; Kang, S.A.; Rameseder, J.; Zhang, Y.; Ottina, K.A.; Lim, D.; Peterson, T.R.; Choi, Y.; Gray, N.S.; Yaffe, M.B.; et al. The mTOR-regulated phosphoproteome reveals a mechanism of mTORC1-mediated inhibition of growth factor signaling. Science 2011, 332, 1317–1322. [Google Scholar] [CrossRef]
  53. Yu, Y.; Yoon, S.O.; Poulogiannis, G.; Yang, Q.; Ma, X.M.; Villén, J.; Kubica, N.; Hoffman, G.R.; Cantley, L.C.; Gygi, S.P.; et al. Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling. Science 2011, 332, 1322–1326. [Google Scholar] [CrossRef]
  54. Sarbassov, D.D.; Guertin, D.A.; Ali, S.M.; Sabatini, D.M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005, 307, 1098–1101. [Google Scholar] [CrossRef]
  55. Guertin, D.A.; Stevens, D.M.; Thoreen, C.C.; Burds, A.A.; Kalaany, N.Y.; Moffat, J.; Brown, M.; Fitzgerald, K.J.; Sabatini, D.M. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev. Cell 2006, 11, 859–871. [Google Scholar] [CrossRef]
  56. Jacinto, E.; Loewith, R.; Schmidt, A.; Lin, S.; Rüegg, M.A.; Hall, A.; Hall, M.N. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat. Cell Biol. 2004, 6, 1122–1128. [Google Scholar] [CrossRef]
  57. García-Martínez, J.M.; Alessi, D.R. mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1). Biochem. J. 2008, 416, 375–385. [Google Scholar] [CrossRef]
  58. Datta, H.K.; Ng, W.F.; Walker, J.A.; Tuck, S.P.; Varanasi, S.S. The cell biology of bone metabolism. J. Clin. Pathol. 2008, 61, 577–587. [Google Scholar] [CrossRef]
  59. Bakker, A.D.; Gakes, T.; Hogervorst, J.M.; de Wit, G.M.; Klein-Nulend, J.; Jaspers, R.T. Mechanical Stimulation and IGF-1 Enhance mRNA Translation Rate in Osteoblasts Via Activation of the AKT-mTOR Pathway. J. Cell. Physiol. 2016, 231, 1283–1290. [Google Scholar] [CrossRef]
  60. Riddle, R.C.; Frey, J.L.; Tomlinson, R.E.; Ferron, M.; Li, Y.; DiGirolamo, D.J.; Faugere, M.C.; Hussain, M.A.; Karsenty, G.; Clemens, T.L. Tsc2 is a molecular checkpoint controlling osteoblast development and glucose homeostasis. Mol. Cell. Biol. 2014, 34, 1850–1862. [Google Scholar] [CrossRef]
  61. Gong, W.; Zhang, N.; Cheng, G.; Zhang, Q.; He, Y.; Shen, Y.; Zhang, Q.; Zhu, B.; Zhang, Q.; Qin, L. Rehmannia glutinosa Libosch Extracts Prevent Bone Loss and Architectural Deterioration and Enhance Osteoblastic Bone Formation by Regulating the IGF-1/PI3K/mTOR Pathway in Streptozotocin-Induced Diabetic Rats. Int. J. Mol. Sci. 2019, 20, 3964. [Google Scholar] [CrossRef]
  62. Meng, D.; Yang, Q.; Wang, H.; Melick, C.H.; Navlani, R.; Frank, A.R.; Jewell, J.L. Glutamine and asparagine activate mTORC1 independently of Rag GTPases. J. Biol. Chem. 2020, 295, 2890–2899. [Google Scholar] [CrossRef]
  63. Volpe, C.M.O.; Villar-Delfino, P.H.; Dos Anjos, P.M.F.; Nogueira-Machado, J.A. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018, 9, 119. [Google Scholar] [CrossRef] [PubMed]
  64. Liang, F.; Luo, Y.F.; Guo, Z.; Qian, Q.; Meng, X.B.; Mo, Z.H. MicroRNA-139-5p mediates BMSCs impairment in diabetes by targeting HOXA9/c-Fos. FASEB J. 2023, 37, e22697. [Google Scholar] [CrossRef] [PubMed]
  65. Chang, T.C.; Hsu, M.F.; Wu, K.K. High glucose induces bone marrow-derived mesenchymal stem cell senescence by upregulating autophagy. PLoS ONE 2015, 10, e0126537. [Google Scholar] [CrossRef] [PubMed]
  66. Teissier, T.; Temkin, V.; Pollak, R.D.; Cox, L.S. Crosstalk Between Senescent Bone Cells and the Bone Tissue Microenvironment Influences Bone Fragility During Chronological Age and in Diabetes. Front. Physiol. 2022, 13, 812157. [Google Scholar] [CrossRef]
  67. Meng, Y.; Ji, J.; Tan, W.; Guo, G.; Xia, Y.; Cheng, C.; Gu, Z.; Wang, Z. Involvement of autophagy in the procedure of endoplasmic reticulum stress introduced apoptosis in bone marrow mesenchymal stem cells from nonobese diabetic mice. Cell Biochem. Funct. 2016, 34, 25–33. [Google Scholar] [CrossRef]
  68. Wang, X.; Feng, Z.; Li, J.; Chen, L.; Tang, W. High glucose induces autophagy of MC3T3-E1 cells via ROS-AKT-mTOR axis. Mol. Cell. Endocrinol. 2016, 429, 62–72. [Google Scholar] [CrossRef]
  69. Kaludercic, N.; Di Lisa, F. Mitochondrial ROS Formation in the Pathogenesis of Diabetic Cardiomyopathy. Front. Cardiovasc. Med. 2020, 7, 12. [Google Scholar] [CrossRef]
  70. Chen, M.; Jing, D.; Ye, R.; Yi, J.; Zhao, Z. PPARβ/δ accelerates bone regeneration in diabetic mellitus by enhancing AMPK/mTOR pathway-mediated autophagy. Stem Cell Res. Ther. 2021, 12, 566. [Google Scholar] [CrossRef]
  71. Hu, X.; Li, B.; Wu, F.; Liu, X.; Liu, M.; Wang, C.; Shi, Y.; Ye, L. GPX7 Facilitates BMSCs Osteoblastogenesis via ER Stress and mTOR Pathway. J. Cell. Mol. Med. 2021, 25, 10454–10465. [Google Scholar] [CrossRef]
  72. Li, S.; Yang, D.; Gao, X.; Yao, S.; Wang, S.; Zhu, J.; Shu, J. Argpyrimidine bonded to RAGE regulates autophagy and cell cycle to cause periodontal destruction. J. Cell. Physiol. 2022, 237, 4460–4476. [Google Scholar] [CrossRef]
  73. Li, C.J.; Cheng, P.; Liang, M.K.; Chen, Y.S.; Lu, Q.; Wang, J.Y.; Xia, Z.Y.; Zhou, H.D.; Cao, X.; Xie, H.; et al. MicroRNA-188 regulates age-related switch between osteoblast and adipocyte differentiation. J. Clin. Investig. 2015, 125, 1509–1522. [Google Scholar] [CrossRef]
  74. Ouyang, Z.; Kang, D.; Li, K.; Liang, G.; Liu, Z.; Mai, Q.; Chen, Q.; Yao, C.; Wei, R.; Tan, X.; et al. DEPTOR exacerbates bone-fat imbalance in osteoporosis by transcriptionally modulating BMSC differentiation. Biomed. Pharmacother. 2022, 151, 113164. [Google Scholar] [CrossRef]
  75. Weng, Z.; Wang, Y.; Ouchi, T.; Liu, H.; Qiao, X.; Wu, C.; Zhao, Z.; Li, L.; Li, B. Mesenchymal Stem/Stromal Cell Senescence: Hallmarks, Mechanisms, and Combating Strategies. Stem Cells Transl. Med. 2022, 11, 356–371. [Google Scholar] [CrossRef]
  76. Rastaldo, R.; Vitale, E.; Giachino, C. Dual Role of Autophagy in Regulation of Mesenchymal Stem Cell Senescence. Front. Cell Dev. Biol. 2020, 8, 276. [Google Scholar] [CrossRef]
  77. Zheng, Y.; Lei, Y.; Hu, C.; Hu, C. p53 regulates autophagic activity in senescent rat mesenchymal stromal cells. Exp. Gerontol. 2016, 75, 64–71. [Google Scholar] [CrossRef]
  78. Al-Azab, M.; Wang, B.; Elkhider, A.; Walana, W.; Li, W.; Yuan, B.; Ye, Y.; Tang, Y.; Almoiliqy, M.; Adlat, S.; et al. Indian Hedgehog regulates senescence in bone marrow-derived mesenchymal stem cell through modulation of ROS/mTOR/4EBP1, p70S6K1/2 pathway. Aging 2020, 12, 5693–5715. [Google Scholar] [CrossRef]
  79. Liu, F.; Yuan, Y.; Bai, L.; Yuan, L.; Li, L.; Liu, J.; Chen, Y.; Lu, Y.; Cheng, J.; Zhang, J. LRRc17 controls BMSC senescence via mitophagy and inhibits the therapeutic effect of BMSCs on ovariectomy-induced bone loss. Redox. Biol. 2021, 43, 101963. [Google Scholar] [CrossRef]
  80. Udagawa, N.; Koide, M.; Nakamura, M.; Nakamichi, Y.; Yamashita, T.; Uehara, S.; Kobayashi, Y.; Furuya, Y.; Yasuda, H.; Fukuda, C.; et al. Osteoclast differentiation by RANKL and OPG signaling pathways. J. Bone Miner. Metab. 2021, 39, 19–26. [Google Scholar] [CrossRef]
  81. Karalazou, P.; Ntelios, D.; Chatzopoulou, F.; Fragou, A.; Taousani, M.; Mouzaki, K.; Galli-Tsinopoulou, A.; Kouidou, S.; Tzimagiorgis, G. OPG/RANK/RANKL signaling axis in patients with type I diabetes: Associations with parathormone and vitamin D. Ital. J. Pediatr. 2019, 45, 161. [Google Scholar] [CrossRef]
  82. An, Y.; Zhang, H.; Wang, C.; Jiao, F.; Xu, H.; Wang, X.; Luan, W.; Ma, F.; Ni, L.; Tang, X.; et al. Activation of ROS/MAPKs/NF-κB/NLRP3 and inhibition of efferocytosis in osteoclast-mediated diabetic osteoporosis. FASEB J. 2019, 33, 12515–12527. [Google Scholar] [CrossRef]
  83. Tian, Y.; Ming, J. Melatonin inhibits osteoclastogenesis via RANKL/OPG suppression mediated by Rev-Erbα in osteoblasts. J. Cell. Mol. Med. 2022, 26, 4032–4047. [Google Scholar] [CrossRef] [PubMed]
  84. Qu, B.; Gong, K.; Yang, H.; Li, Y.; Jiang, T.; Zeng, Z.; Cao, Z.; Pan, X. SIRT1 suppresses high glucose and palmitate-induced osteoclast differentiation via deacetylating p66Shc. Mol. Cell. Endocrinol. 2018, 474, 97–104. [Google Scholar] [CrossRef] [PubMed]
  85. Hu, Z.; Ma, C.; Liang, Y.; Zou, S.; Liu, X. Osteoclasts in bone regeneration under type 2 diabetes mellitus. Acta Biomater. 2019, 84, 402–413. [Google Scholar] [CrossRef] [PubMed]
  86. He, H.; Liu, R.; Desta, T.; Leone, C.; Gerstenfeld, L.C.; Graves, D.T. Diabetes causes decreased osteoclastogenesis, reduced bone formation, and enhanced apoptosis of osteoblastic cells in bacteria stimulated bone loss. Endocrinology 2004, 145, 447–452. [Google Scholar] [CrossRef]
  87. Dong, W.; Qi, M.; Wang, Y.; Feng, X.; Liu, H. Zoledronate and high glucose levels influence osteoclast differentiation and bone absorption via the AMPK pathway. Biochem. Biophys. Res. Commun. 2018, 505, 1195–1202. [Google Scholar] [CrossRef]
  88. Chen, X.; Chen, W.; Aung, Z.M.; Han, W.; Zhang, Y.; Chai, G. LY3023414 inhibits both osteogenesis and osteoclastogenesis through the PI3K/Akt/GSK3 signalling pathway. Bone Jt. Res. 2021, 10, 237–249. [Google Scholar] [CrossRef] [PubMed]
  89. Fu, L.; Wu, W.; Sun, X.; Zhang, P. Glucocorticoids Enhanced Osteoclast Autophagy Through the PI3K/Akt/mTOR Signaling Pathway. Calcif. Tissue Int. 2020, 107, 60–71. [Google Scholar] [CrossRef]
  90. Hiraiwa, M.; Ozaki, K.; Yamada, T.; Iezaki, T.; Park, G.; Fukasawa, K.; Horie, T.; Kamada, H.; Tokumura, K.; Motono, M.; et al. mTORC1 Activation in Osteoclasts Prevents Bone Loss in a Mouse Model of Osteoporosis. Front. Pharmacol. 2019, 10, 684. [Google Scholar] [CrossRef]
  91. Huynh, H.; Wan, Y. mTORC1 impedes osteoclast differentiation via calcineurin and NFATc1. Commun. Biol. 2018, 1, 29. [Google Scholar] [CrossRef]
  92. Bae, S.; Oh, B.; Tsai, J.; Park, P.S.U.; Greenblatt, M.B.; Giannopoulou, E.G.; Park-Min, K.H. The crosstalk between MYC and mTORC1 during osteoclastogenesis. Front. Cell Dev. Biol. 2022, 10, 920683. [Google Scholar] [CrossRef]
  93. Zhao, H.; Sun, Z.; Ma, Y.; Song, R.; Yuan, Y.; Bian, J.; Gu, J.; Liu, Z. Antiosteoclastic bone resorption activity of osteoprotegerin via enhanced AKT/mTOR/ULK1-mediated autophagic pathway. J. Cell. Physiol. 2020, 235, 3002–3012. [Google Scholar] [CrossRef] [PubMed]
  94. Tong, X.; Gu, J.; Song, R.; Wang, D.; Sun, Z.; Sui, C.; Zhang, C.; Liu, X.; Bian, J.; Liu, Z. Osteoprotegerin inhibit osteoclast differentiation and bone resorption by enhancing autophagy via AMPK/mTOR/p70S6K signaling pathway in vitro. J. Cell. Biochem. 2019, 120, 1630–1642. [Google Scholar] [CrossRef] [PubMed]
  95. Tong, X.; Zhang, C.; Wang, D.; Song, R.; Ma, Y.; Cao, Y.; Zhao, H.; Bian, J.; Gu, J.; Liu, Z. Suppression of AMP-activated protein kinase reverses osteoprotegerin-induced inhibition of osteoclast differentiation by reducing autophagy. Cell Prolif. 2020, 53, e12714. [Google Scholar] [CrossRef] [PubMed]
  96. Montaseri, A.; Giampietri, C.; Rossi, M.; Riccioli, A.; Del Fattore, A.; Filippini, A. The Role of Autophagy in Osteoclast Differentiation and Bone Resorption Function. Biomolecules 2020, 10, 1398. [Google Scholar] [CrossRef] [PubMed]
  97. Cai, Z.Y.; Yang, B.; Shi, Y.X.; Zhang, W.L.; Liu, F.; Zhao, W.; Yang, M.W. High glucose downregulates the effects of autophagy on osteoclastogenesis via the AMPK/mTOR/ULK1 pathway. Biochem. Biophys. Res. Commun. 2018, 503, 428–435. [Google Scholar] [CrossRef] [PubMed]
  98. Xu, Z.; Xu, J.; Li, S.; Cui, H.; Zhang, G.; Ni, X.; Wang, J. S-Equol enhances osteoblastic bone formation and prevents bone loss through OPG/RANKL via the PI3K/Akt pathway in streptozotocin-induced diabetic rats. Front. Nutr. 2022, 9, 986192. [Google Scholar] [CrossRef]
  99. Donath, M.Y.; Shoelson, S.E. Type 2 diabetes as an inflammatory disease. Nat. Rev. Immunol. 2011, 11, 98–107. [Google Scholar] [CrossRef]
  100. Shahen, V.A.; Gerbaix, M.; Koeppenkastrop, S.; Lim, S.F.; McFarlane, K.E.; Nguyen, A.N.L.; Peng, X.Y.; Weiss, N.B.; Brennan-Speranza, T.C. Multifactorial effects of hyperglycaemia, hyperinsulinemia and inflammation on bone remodelling in type 2 diabetes mellitus. Cytokine Growth Factor Rev. 2020, 55, 109–118. [Google Scholar] [CrossRef]
  101. Olson, N.C.; Doyle, M.F.; de Boer, I.H.; Huber, S.A.; Jenny, N.S.; Kronmal, R.A.; Psaty, B.M.; Tracy, R.P. Associations of Circulating Lymphocyte Subpopulations with Type 2 Diabetes: Cross-Sectional Results from the Multi-Ethnic Study of Atherosclerosis (MESA). PLoS ONE 2015, 10, e0139962. [Google Scholar] [CrossRef]
  102. Cifuentes-Mendiola, S.E.; Solis-Suarez, D.L.; Martínez-Dávalos, A.; Godínez-Victoria, M.; García-Hernández, A.L. CD4+ T-cell activation of bone marrow causes bone fragility and insulin resistance in type 2 diabetes. Bone 2022, 155, 116292. [Google Scholar] [CrossRef]
  103. Newman, H.; Shih, Y.V.; Varghese, S. Resolution of inflammation in bone regeneration: From understandings to therapeutic applications. Biomaterials 2021, 277, 121114. [Google Scholar] [CrossRef]
  104. Qing, L.; Fu, J.; Wu, P.; Zhou, Z.; Yu, F.; Tang, J. Metformin induces the M2 macrophage polarization to accelerate the wound healing via regulating AMPK/mTOR/NLRP3 inflammasome singling pathway. Am. J. Transl. Res. 2019, 11, 655–668. [Google Scholar]
  105. Jamalpoor, Z.; Asgari, A.; Lashkari, M.H.; Mirshafiey, A.; Mohsenzadegan, M. Modulation of Macrophage Polarization for Bone Tissue Engineering Applications. Iran. J. Allergy Asthma Immunol. 2018, 17, 398–408. [Google Scholar] [CrossRef]
  106. Horwood, N.J. Macrophage Polarization and Bone Formation: A review. Clin. Rev. Allergy Immunol. 2016, 51, 79–86. [Google Scholar] [CrossRef]
  107. Wang, Y.; Smith, W.; Hao, D.; He, B.; Kong, L. M1 and M2 macrophage polarization and potentially therapeutic naturally occurring compounds. Int. Immunopharmacol. 2019, 70, 459–466. [Google Scholar] [CrossRef]
  108. Wang, P.; Zhang, Q.; Tan, L.; Xu, Y.; Xie, X.; Zhao, Y. The Regulatory Effects of mTOR Complexes in the Differentiation and Function of CD4+ T Cell Subsets. J. Immunol. Res. 2020, 2020, 3406032. [Google Scholar] [CrossRef]
  109. Chen, S.; van Tok, M.N.; Knaup, V.L.; Kraal, L.; Pots, D.; Bartels, L.; Gravallese, E.M.; Taurog, J.D.; van de Sande, M.; van Duivenvoorde, L.M.; et al. mTOR Blockade by Rapamycin in Spondyloarthritis: Impact on Inflammation and New Bone Formation in vitro and in vivo. Front. Immunol. 2019, 10, 2344. [Google Scholar] [CrossRef]
  110. Deng, Z.; Chen, M.; Liu, Y.; Xu, S.; Ouyang, Y.; Shi, W.; Jian, D.; Wang, B.; Liu, F.; Li, J.; et al. A positive feedback loop between mTORC1 and cathelicidin promotes skin inflammation in rosacea. EMBO Mol. Med. 2021, 13, e13560. [Google Scholar] [CrossRef]
  111. Zhang, X.J.; Shang, K.; Pu, Y.K.; Wang, Q.; Wang, T.T.; Zou, Y.; Wang, Y.M.; Xu, Y.J.; Li, X.L.; Zhang, R.H.; et al. Leojaponin inhibits NLRP3 inflammasome activation through restoration of autophagy via upregulating RAPTOR phosphorylation. J. Ethnopharmacol. 2021, 278, 114322. [Google Scholar] [CrossRef]
  112. Wang, Q.; Nie, L.; Zhao, P.; Zhou, X.; Ding, Y.; Chen, Q.; Wang, Q. Diabetes fuels periodontal lesions via GLUT1-driven macrophage inflammaging. Int. J. Oral Sci. 2021, 13, 11. [Google Scholar] [CrossRef]
  113. Zhao, Z.; Ming, Y.; Li, X.; Tan, H.; He, X.; Yang, L.; Song, J.; Zheng, L. Hyperglycemia Aggravates Periodontitis via Autophagy Impairment and ROS-Inflammasome-Mediated Macrophage Pyroptosis. Int. J. Mol. Sci. 2023, 24, 6309. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, C.; Sun, L.; Ma, H.; Peng, J.; Zhen, Y.; Duan, K.; Liu, G.; Ding, W.; Zhao, Y. The phenotype and functional alterations of macrophages in mice with hyperglycemia for long term. J. Cell. Physiol. 2012, 227, 1670–1679. [Google Scholar] [CrossRef]
  115. Kawai, H.; Oo, M.W.; Tsujigiwa, H.; Nakano, K.; Takabatake, K.; Sukegawa, S.; Nagatsuka, H. Potential role of myeloid-derived suppressor cells in transition from reaction to repair phase of bone healing process. Int. J. Med. Sci. 2021, 18, 1824–1830. [Google Scholar] [CrossRef]
  116. Li, Y.; Xu, Y.; Liu, X.; Yan, X.; Lin, Y.; Tan, Q.; Hou, Y. mTOR inhibitor INK128 promotes wound healing by regulating MDSCs. Stem Cell Res. Ther. 2021, 12, 170. [Google Scholar] [CrossRef] [PubMed]
  117. Kang, M.; Huang, C.C.; Lu, Y.; Shirazi, S.; Gajendrareddy, P.; Ravindran, S.; Cooper, L.F. Bone regeneration is mediated by macrophage extracellular vesicles. Bone 2020, 141, 115627. [Google Scholar] [CrossRef] [PubMed]
  118. Qi, Y.; Zhu, T.; Zhang, T.; Wang, X.; Li, W.; Chen, D.; Meng, H.; An, S. M1 macrophage-derived exosomes transfer miR-222 to induce bone marrow mesenchymal stem cell apoptosis. Lab. Investig. 2021, 101, 1318–1326. [Google Scholar] [CrossRef] [PubMed]
  119. Hofbauer, L.C.; Busse, B.; Eastell, R.; Ferrari, S.; Frost, M.; Müller, R.; Burden, A.M.; Rivadeneira, F.; Napoli, N.; Rauner, M. Bone fragility in diabetes: Novel concepts and clinical implications. Lancet Diabetes Endocrinol. 2022, 10, 207–220. [Google Scholar] [CrossRef]
  120. Peng, J.; Hui, K.; Hao, C.; Peng, Z.; Gao, Q.X.; Jin, Q.; Lei, G.; Min, J.; Qi, Z.; Bo, C.; et al. Low bone turnover and reduced angiogenesis in streptozotocin-induced osteoporotic mice. Connect. Tissue Res. 2016, 57, 277–289. [Google Scholar] [CrossRef] [PubMed]
  121. Mangialardi, G.; Ferland-McCollough, D.; Maselli, D.; Santopaolo, M.; Cordaro, A.; Spinetti, G.; Sambataro, M.; Sullivan, N.; Blom, A.; Madeddu, P. Bone marrow pericyte dysfunction in individuals with type 2 diabetes. Diabetologia 2019, 62, 1275–1290. [Google Scholar] [CrossRef]
  122. Ribot, J.; Denoeud, C.; Frescaline, G.; Landon, R.; Petite, H.; Pavon-Djavid, G.; Bensidhoum, M.; Anagnostou, F. Experimental Type 2 Diabetes Differently Impacts on the Select Functions of Bone Marrow-Derived Multipotent Stromal Cells. Cells 2021, 10, 268. [Google Scholar] [CrossRef]
  123. Hu, X.F.; Xiang, G.; Wang, T.J.; Ma, Y.B.; Zhang, Y.; Yan, Y.B.; Zhao, X.; Wu, Z.X.; Feng, Y.F.; Lei, W. Impairment of type H vessels by NOX2-mediated endothelial oxidative stress: Critical mechanisms and therapeutic targets for bone fragility in streptozotocin-induced type 1 diabetic mice. Theranostics 2021, 11, 3796–3812. [Google Scholar] [CrossRef] [PubMed]
  124. Samakkarnthai, P.; Sfeir, J.G.; Atkinson, E.J.; Achenbach, S.J.; Wennberg, P.W.; Dyck, P.J.; Tweed, A.J.; Volkman, T.L.; Amin, S.; Farr, J.N.; et al. Determinants of Bone Material Strength and Cortical Porosity in Patients with Type 2 Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2020, 105, e3718–e3729. [Google Scholar] [CrossRef] [PubMed]
  125. Caliaperoumal, G.; Souyet, M.; Bensidhoum, M.; Petite, H.; Anagnostou, F. Type 2 diabetes impairs angiogenesis and osteogenesis in calvarial defects: MicroCT study in ZDF rats. Bone 2018, 112, 161–172. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, J.; Zhang, H.; Chen, Y.; Fu, J.; Lei, Y.; Sun, J.; Tang, B. Platelet-derived growth factor D promotes the angiogenic capacity of endothelial progenitor cells. Mol. Med. Rep. 2019, 19, 125–132. [Google Scholar] [CrossRef]
  127. Liu, Z.; Li, Y.; Yang, J.; Huang, J.; Luo, C.; Zhang, J.; Yan, W.; Ao, Y. Bone morphogenetic protein 9 enhances osteogenic and angiogenic responses of human amniotic mesenchymal stem cells cocultured with umbilical vein endothelial cells through the PI3K/AKT/m-TOR signaling pathway. Aging 2021, 13, 24829–24849. [Google Scholar] [CrossRef]
  128. Tian, D.; Xiang, Y.; Tang, Y.; Ge, Z.; Li, Q.; Zhang, Y. Circ-ADAM9 targeting PTEN and ATG7 promotes autophagy and apoptosis of diabetic endothelial progenitor cells by sponging mir-20a-5p. Cell Death Dis. 2020, 11, 526. [Google Scholar] [CrossRef]
  129. Zou, Y.; Wu, F.; Liu, Q.; Deng, X.; Hai, R.; He, X.; Zhou, X. Downregulation of miRNA-328 promotes the angiogenesis of HUVECs by regulating the PIM1 and AKT/mTOR signaling pathway under high glucose and low serum condition. Mol. Med. Rep. 2020, 22, 895–905. [Google Scholar] [CrossRef]
  130. Wei, P.; Zhong, C.; Yang, X.; Shu, F.; Xiao, S.; Gong, T.; Luo, P.; Li, L.; Chen, Z.; Zheng, Y.; et al. Exosomes derived from human amniotic epithelial cells accelerate diabetic wound healing via PI3K-AKT-mTOR-mediated promotion in angiogenesis and fibroblast function. Burn. Trauma 2020, 8, tkaa020. [Google Scholar] [CrossRef]
  131. Wronka, M.; Krzemińska, J.; Młynarska, E.; Rysz, J.; Franczyk, B. The Influence of Lifestyle and Treatment on Oxidative Stress and Inflammation in Diabetes. Int. J. Mol. Sci. 2022, 23, 15743. [Google Scholar] [CrossRef]
  132. Kim, H.J.; Han, S.J.; Kim, D.J.; Jang, H.C.; Lim, S.; Choi, S.H.; Kim, Y.H.; Shin, D.H.; Kim, S.H.; Kim, T.H.; et al. Effects of valsartan and amlodipine on oxidative stress in type 2 diabetic patients with hypertension: A randomized, multicenter study. Korean J. Intern. Med. 2017, 32, 497–504. [Google Scholar] [CrossRef]
  133. Langlais, P.; Yi, Z.; Finlayson, J.; Luo, M.; Mapes, R.; De Filippis, E.; Meyer, C.; Plummer, E.; Tongchinsub, P.; Mattern, M.; et al. Global IRS-1 phosphorylation analysis in insulin resistance. Diabetologia 2011, 54, 2878–2889. [Google Scholar] [CrossRef] [PubMed]
  134. Jiang, P.; Ren, L.; Zhi, L.; Yu, Z.; Lv, F.; Xu, F.; Peng, W.; Bai, X.; Cheng, K.; Quan, L.; et al. Negative regulation of AMPK signaling by high glucose via E3 ubiquitin ligase MG53. Mol. Cell 2021, 81, 629–637.e5. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, M.; Yang, B.; Peng, S.; Xiao, J. Metformin Rescues the Impaired Osteogenesis Differentiation Ability of Rat Adipose-Derived Stem Cells in High Glucose by Activating Autophagy. Stem Cells Dev. 2021, 30, 1017–1027. [Google Scholar] [CrossRef] [PubMed]
  136. Chen, S.C.; Brooks, R.; Houskeeper, J.; Bremner, S.K.; Dunlop, J.; Viollet, B.; Logan, P.J.; Salt, I.P.; Ahmed, S.F.; Yarwood, S.J. Metformin suppresses adipogenesis through both AMP-activated protein kinase (AMPK)-dependent and AMPK-independent mechanisms. Mol. Cell. Endocrinol. 2017, 440, 57–68. [Google Scholar] [CrossRef] [PubMed]
  137. Lee, K.W.; Yook, J.Y.; Son, M.Y.; Kim, M.J.; Koo, D.B.; Han, Y.M.; Cho, Y.S. Rapamycin promotes the osteoblastic differentiation of human embryonic stem cells by blocking the mTOR pathway and stimulating the BMP/Smad pathway. Stem Cells Dev. 2010, 19, 557–568. [Google Scholar] [CrossRef]
  138. Phornphutkul, C.; Lee, M.; Voigt, C.; Wu, K.Y.; Ehrlich, M.G.; Gruppuso, P.A.; Chen, Q. The effect of rapamycin on bone growth in rabbits. J. Orthop. Res. 2009, 27, 1157–1161. [Google Scholar] [CrossRef]
  139. Hu, X.K.; Yin, X.H.; Zhang, H.Q.; Guo, C.F.; Tang, M.X. Liraglutide attenuates the osteoblastic differentiation of MC3T3-E1 cells by modulating AMPK/mTOR signaling. Mol. Med. Rep. 2016, 14, 3662–3668. [Google Scholar] [CrossRef]
  140. Lan, D.; Yao, C.; Li, X.; Liu, H.; Wang, D.; Wang, Y.; Qi, S. Tocopherol attenuates the oxidative stress of BMSCs by inhibiting ferroptosis through the PI3k/AKT/mTOR pathway. Front. Bioeng. Biotechnol. 2022, 10, 938520. [Google Scholar] [CrossRef]
  141. Ferroni, L.; Gardin, C.; Dolkart, O.; Salai, M.; Barak, S.; Piattelli, A.; Amir-Barak, H.; Zavan, B. Pulsed electromagnetic fields increase osteogenetic commitment of MSCs via the mTOR pathway in TNF-α mediated inflammatory conditions: An in-vitro study. Sci. Rep. 2018, 8, 5108. [Google Scholar] [CrossRef]
  142. Karner, C.M.; Lee, S.Y.; Long, F. Bmp Induces Osteoblast Differentiation through both Smad4 and mTORC1 Signaling. Mol. Cell. Biol. 2017, 37, e00253-16. [Google Scholar] [CrossRef]
  143. Ge, X.; Zhou, G. Protective effects of naringin on glucocorticoid-induced osteoporosis through regulating the PI3K/Akt/mTOR signaling pathway. Am. J. Transl. Res. 2021, 13, 6330–6341. [Google Scholar] [PubMed]
  144. Zhou, H.; Jiao, G.; Dong, M.; Chi, H.; Wang, H.; Wu, W.; Liu, H.; Ren, S.; Kong, M.; Li, C.; et al. Orthosilicic Acid Accelerates Bone Formation in Human Osteoblast-Like Cells Through the PI3K-Akt-mTOR Pathway. Biol. Trace Elem. Res. 2019, 190, 327–335. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, Z.; Zhang, X.; Zhao, D.; Liu, B.; Wang, B.; Yu, W.; Li, J.; Yu, X.; Cao, F.; Zheng, G.; et al. TGF-β1 promotes the osteoinduction of human osteoblasts via the PI3K/AKT/mTOR/S6K1 signalling pathway. Mol. Med. Rep. 2019, 19, 3505–3518. [Google Scholar] [CrossRef] [PubMed]
  146. Zhao, B.; Xiong, Y.; Zhang, Y.; Jia, L.; Zhang, W.; Xu, X. Rutin promotes osteogenic differentiation of periodontal ligament stem cells through the GPR30-mediated PI3K/AKT/mTOR signaling pathway. Exp. Biol. Med. 2020, 245, 552–561. [Google Scholar] [CrossRef] [PubMed]
  147. Jiang, Y.; Luo, W.; Wang, B.; Yi, Z.; Gong, P.; Xiong, Y. 1α,25-Dihydroxyvitamin D3 ameliorates diabetes-induced bone loss by attenuating FoxO1-mediated autophagy. J. Biol. Chem. 2021, 296, 100287. [Google Scholar] [CrossRef]
  148. Mizerska-Kowalska, M.; Sławińska-Brych, A.; Kaławaj, K.; Żurek, A.; Pawińska, B.; Rzeski, W.; Zdzisińska, B. Betulin Promotes Differentiation of Human Osteoblasts In Vitro and Exerts an Osteoinductive Effect on the hFOB 1.19 Cell Line Through Activation of JNK, ERK1/2, and mTOR Kinases. Molecules 2019, 24, 2637. [Google Scholar] [CrossRef]
  149. Day, E.A.; Ford, R.J.; Steinberg, G.R. AMPK as a Therapeutic Target for Treating Metabolic Diseases. Trends Endocrinol. Metab. 2017, 28, 545–560. [Google Scholar] [CrossRef]
  150. Agarwal, S.; Bell, C.M.; Rothbart, S.B.; Moran, R.G. AMP-activated Protein Kinase (AMPK) Control of mTORC1 Is p53- and TSC2-independent in Pemetrexed-treated Carcinoma Cells. J. Biol. Chem. 2015, 290, 27473–27486. [Google Scholar] [CrossRef]
  151. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef]
  152. Zhang, Y.S.; Zheng, Y.D.; Yuan, Y.; Chen, S.C.; Xie, B.C. Effects of Anti-Diabetic Drugs on Fracture Risk: A Systematic Review and Network Meta-Analysis. Front. Endocrinol. 2021, 12, 735824. [Google Scholar] [CrossRef]
  153. Shen, M.; Yu, H.; Jin, Y.; Mo, J.; Sui, J.; Qian, X.; Chen, T. Metformin Facilitates Osteoblastic Differentiation and M2 Macrophage Polarization by PI3K/AKT/mTOR Pathway in Human Umbilical Cord Mesenchymal Stem Cells. Stem Cells Int. 2022, 2022, 9498876. [Google Scholar] [CrossRef] [PubMed]
  154. Montazersaheb, S.; Kabiri, F.; Saliani, N.; Nourazarian, A.; Avci, Ç.B.; Rahbarghazi, R.; Nozad Charoudeh, H. Prolonged incubation with Metformin decreased angiogenic potential in human bone marrow mesenchymal stem cells. Biomed. Pharmacother. 2018, 108, 1328–1337. [Google Scholar] [CrossRef] [PubMed]
  155. McCarthy, A.D.; Cortizo, A.M.; Sedlinsky, C. Metformin revisited: Does this regulator of AMP-activated protein kinase secondarily affect bone metabolism and prevent diabetic osteopathy. World J. Diabetes 2016, 7, 122–133. [Google Scholar] [CrossRef] [PubMed]
  156. Zhu, M.; Zhao, Z.; Xu, H.H.K.; Dai, Z.; Yu, K.; Xiao, L.; Schneider, A.; Weir, M.D.; Oates, T.W.; Bai, Y.; et al. Effects of Metformin Delivery via Biomaterials on Bone and Dental Tissue Engineering. Int. J. Mol. Sci. 2022, 23, 15905. [Google Scholar] [CrossRef]
  157. Al Jofi, F.E.; Ma, T.; Guo, D.; Schneider, M.P.; Shu, Y.; Xu, H.H.K.; Schneider, A. Functional organic cation transporters mediate osteogenic response to metformin in human umbilical cord mesenchymal stromal cells. Cytotherapy 2018, 20, 650–659. [Google Scholar] [CrossRef]
  158. Krebs, M.; Brunmair, B.; Brehm, A.; Artwohl, M.; Szendroedi, J.; Nowotny, P.; Roth, E.; Fürnsinn, C.; Promintzer, M.; Anderwald, C.; et al. The Mammalian target of rapamycin pathway regulates nutrient-sensitive glucose uptake in man. Diabetes 2007, 56, 1600–1607. [Google Scholar] [CrossRef]
  159. Houde, V.P.; Brûlé, S.; Festuccia, W.T.; Blanchard, P.G.; Bellmann, K.; Deshaies, Y.; Marette, A. Chronic rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue. Diabetes 2010, 59, 1338–1348. [Google Scholar] [CrossRef]
  160. Xing, Y.; Liu, C.; Zhou, L.; Li, Y.; Wu, D. Osteogenic effects of rapamycin on bone marrow mesenchymal stem cells via inducing autophagy. J. Orthop. Surg. Res. 2023, 18, 129. [Google Scholar] [CrossRef]
  161. Isomoto, S.; Hattori, K.; Ohgushi, H.; Nakajima, H.; Tanaka, Y.; Takakura, Y. Rapamycin as an inhibitor of osteogenic differentiation in bone marrow-derived mesenchymal stem cells. J. Orthop. Sci. 2007, 12, 83–88. [Google Scholar] [CrossRef]
  162. Mabilleau, G.; Pereira, M.; Chenu, C. Novel skeletal effects of glucagon-like peptide-1 (GLP-1) receptor agonists. J. Endocrinol. 2018, 236, R29–R42. [Google Scholar] [CrossRef]
  163. Gao, L.; Li, S.L.; Li, Y.K. Liraglutide Promotes the Osteogenic Differentiation in MC3T3-E1 Cells via Regulating the Expression of Smad2/3 Through PI3K/Akt and Wnt/β-Catenin Pathways. DNA Cell Biol. 2018, 37, 1031–1043. [Google Scholar] [CrossRef] [PubMed]
  164. Wu, X.; Li, S.; Xue, P.; Li, Y. Liraglutide, a glucagon-like peptide-1 receptor agonist, facilitates osteogenic proliferation and differentiation in MC3T3-E1 cells through phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), extracellular signal-related kinase (ERK)1/2, and cAMP/protein kinase A (PKA) signaling pathways involving β-catenin. Exp. Cell Res. 2017, 360, 281–291. [Google Scholar] [CrossRef] [PubMed]
  165. Wu, X.; Li, S.; Xue, P.; Li, Y. Liraglutide Inhibits the Apoptosis of MC3T3-E1 Cells Induced by Serum Deprivation through cAMP/PKA/β-Catenin and PI3K/AKT/GSK3β Signaling Pathways. Mol. Cells 2018, 41, 234–243. [Google Scholar] [CrossRef] [PubMed]
  166. Dibble, C.C.; Cantley, L.C. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol. 2015, 25, 545–555. [Google Scholar] [CrossRef] [PubMed]
  167. Lu, Y.; Liu, S.; Yang, P.; Kou, Y.; Li, C.; Liu, H.; Li, M. Exendin-4 and eldecalcitol synergistically promote osteogenic differentiation of bone marrow mesenchymal stem cells through M2 macrophages polarization via PI3K/AKT pathway. Stem Cell Res. Ther. 2022, 13, 113. [Google Scholar] [CrossRef]
  168. Jiang, C.; Wang, Y.; Zhang, M.; Xu, J. Cholesterol inhibits autophagy in RANKL-induced osteoclast differentiation through activating the PI3K/AKT/mTOR signaling pathway. Mol. Biol. Rep. 2022, 49, 9217–9229. [Google Scholar] [CrossRef]
  169. Stage, T.B.; Christensen, M.H.; Jørgensen, N.R.; Beck-Nielsen, H.; Brøsen, K.; Gram, J.; Frost, M. Effects of metformin, rosiglitazone and insulin on bone metabolism in patients with type 2 diabetes. Bone 2018, 112, 35–41. [Google Scholar] [CrossRef]
  170. Nordklint, A.K.; Almdal, T.P.; Vestergaard, P.; Lundby-Christensen, L.; Jørgensen, N.R.; Boesgaard, T.W.; Breum, L.; Gade-Rasmussen, B.; Sneppen, S.B.; Gluud, C.; et al. Effect of Metformin vs. Placebo in Combination with Insulin Analogues on Bone Markers P1NP and CTX in Patients with Type 2 Diabetes Mellitus. Calcif. Tissue Int. 2020, 107, 160–169. [Google Scholar] [CrossRef]
  171. Nordklint, A.K.; Almdal, T.P.; Vestergaard, P.; Lundby-Christensen, L.; Boesgaard, T.W.; Breum, L.; Gade-Rasmussen, B.; Sneppen, S.B.; Gluud, C.; Hemmingsen, B.; et al. The effect of metformin versus placebo in combination with insulin analogues on bone mineral density and trabecular bone score in patients with type 2 diabetes mellitus: A randomized placebo-controlled trial. Osteoporos. Int. 2018, 29, 2517–2526. [Google Scholar] [CrossRef]
Figure 1. Overview of the mTOR signaling pathway and its associated regulators and functions.
Figure 1. Overview of the mTOR signaling pathway and its associated regulators and functions.
Ijms 24 09198 g001
Figure 2. Implication of mTOR signaling in diabetic bone complications. Diabetes bone complications are characterized by imbalance between bone formation and resorption as well as immune and vascular alteration. This figure depicts how mTOR signaling is involved in these cellular abnormalities and contributes to the destructive changes in the diabetic skeletal system. The arrows in the yellow boxes indicate that the levels of the corresponding compounds rise or fall in the diabetic environment.
Figure 2. Implication of mTOR signaling in diabetic bone complications. Diabetes bone complications are characterized by imbalance between bone formation and resorption as well as immune and vascular alteration. This figure depicts how mTOR signaling is involved in these cellular abnormalities and contributes to the destructive changes in the diabetic skeletal system. The arrows in the yellow boxes indicate that the levels of the corresponding compounds rise or fall in the diabetic environment.
Ijms 24 09198 g002
Table 1. Potential mTOR related agents in diabetes bone complications treatment.
Table 1. Potential mTOR related agents in diabetes bone complications treatment.
AgentsTarget Cell/Tissue
(Environment)
TargetMain FindingRef.
Inhibition of mTOR
Metforminadipose-derived stem cells (ASCs)
(40 mM high glucose culture environment)
mTOR0.1 mM metformin reversed the osteogenesis inhibition of ASCs caused by high glucose via inhibiting mTOR and upregulating autophagy[135]
C3H10T1/2 MSCs
(10% FCS plus either IID or PIO)
(10% FCS plus AGD)
mTOR/p70Metformin increased RUNX2 expression and inhibited PPARγ activity in MSCs through the suppression of the mTOR/p70S6K signaling pathway, thereby decreasing adipogenesis[136]
C3H10T1/2 MSCs
(high-glucose conditions with glutamine)
AMPK/mTORActivation of AMPK by metformin inhibited high glutamine-induced mTORC1 hyperactivation and rescues RUNX2 through the mTORC2/AKT-473 axis[22]
Rapamycinhuman embryonic stem cells (hESCs)
(mouse embryonic fibroblast-conditioned medium/serum-free medium)
mTORRapamycin functioned as a potent stimulator of osteoblastic differentiation of hESCs by modulating mTOR and BMP/Smad signaling[137]
Five-week-old New Zealand White rabbitsmTORDirect infusion of rapamycin into proximal tibial growth plates decreased the size of the growth plate and inhibited overall long bone growth[138]
PPARβ/δ Agonistrat BMSCs
(high glucose environment)
SD rats
(1% streptozotocin injected)
AMPK/mTORPPARβ/δ agonist promoted osteogenic differentiation of rat BMSCs through activating AMPK/mTOR-regulated autophagy and improved bone regeneration in type 1 diabetic rats.[70]
LiraglutideMC3T3-E1
(DMEM medium)
AMPK/mTORLiraglutide reduced the differentiation of MC3T3-E1 osteoblasts by regulating AMPK/mTOR pathway[139]
Activation of mTOR signaling
Rehmannia glutinosa Libosch ExtractsMC3T3-E1
(high glucose α-MEM medium)
Wistar rats
(high-fat diet and streptozotocin injection)
IGF-1/
PI3K/mTOR
The extracts increased the proliferation and differentiation of osteoblastic MC3T3-E1 cells injured by high glucose by activating the IGF-1/PI3K/mTOR pathway. Rehmanniae Radix Praeparata could prevent bone loss in type 2 diabetic rats.[61]
Tocopherolrat BMSCs
(treated with H2O2)
PI3K/AKT/
mTOR
Tocopherol protected rat BMSCs from oxidative stress damage by activating PI3K/AKT/mTOR pathway[140]
Pulsed Electromagnetic Fields (PEMFs)MSCs
(treated with 0.1 mg/mL of TNFα)
mTORPEMF increased the expression of osteogenic markers and promoted osteogenic differentiation of MSCs under TNF-α-mediated inflammatory conditions via mTOR activation[141]
BMP-2BMSCs
(α-MEM medium)
mTORBMP-2 activated mTOR signaling pathway and downstream genes regulating protein anabolism to induce osteoblast differentiation[142]
NaringinOsteoblasts cultured from the differentiated BMSCs
(DMEM medium)
PI3K/AKT/
mTOR
Naringin promoted proliferation and differentiation of osteoblasts by activating PI3K/AKT/mTOR pathway[143]
Orthosilicic AcidMG-63 and U2-OS
(DMEM medium)
PI3K/AKT/
mTOR
Orthosilicic acid promoted osteogenesis in vitro by activating PI3K/AKT/mTOR signaling pathway[144]
Transforming growth factor beta 1 (TGF-β1)hFOB 1.19
(DMEM medium)
PI3K/AKT/
mTOR/S6K1
TGF-β1 induced the survival, osteogenic differentiation and migration of human hFOB 1.19 osteoblasts by activating the PI3K/AKT/mTOR/S6K1 pathway[145]
RutinPeriodontal ligament stem cells (PDLSCs)
(α-MEM medium)
PI3K/AKT/
mTOR
Rutin increased proliferation and osteogenic differentiation of PDLSCs through G protein-coupled receptor 30 (GPR30)-mediated PI3K/AKT/mTOR signal transduction[146]
1α,25-Dihydroxyvitamin D3 (1,25D)Wild type mice
(high-fat diet and streptozotocin injection)
Osteoblasts
(high glucose environment)
PI3K/AKT/
FoxO1,
Sesn3/AMPK/
mTORC1
1,25D could reverse dysfunctional bone metabolism in type 2 diabetic mice through attenuating autophagy, by activating PI3K/AKT signaling, inhibiting FoxO1 and Sesn3/AMPK, and upregulating mTORC1.[147]
Betulin (BET)hFOB 1.19
(osteogenic medium and basal medium)
mTORBET increased the expression level of osteogenic differentiation markers and promoted mineralization by activating mitogen-activated protein kinases (MAPKs) and mTOR[148]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, S.; Wang, J.; Wang, S.; Tao, R.; Yi, J.; Chen, M.; Zhao, Z. mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia. Int. J. Mol. Sci. 2023, 24, 9198. https://doi.org/10.3390/ijms24119198

AMA Style

Wang S, Wang J, Wang S, Tao R, Yi J, Chen M, Zhao Z. mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia. International Journal of Molecular Sciences. 2023; 24(11):9198. https://doi.org/10.3390/ijms24119198

Chicago/Turabian Style

Wang, Shuangcheng, Jiale Wang, Shuangwen Wang, Ran Tao, Jianru Yi, Miao Chen, and Zhihe Zhao. 2023. "mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia" International Journal of Molecular Sciences 24, no. 11: 9198. https://doi.org/10.3390/ijms24119198

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

Article Metrics

Back to TopTop