Back to Journals » Drug Design, Development and Therapy » Volume 19

The Roles of Forkhead Box O3a (FOXO3a) in Bone and Cartilage Diseases – A Narrative Review

Authors Wu Z, Zhan W, Wu L , Yu L, Xie X, Yu F, Kong W, Bi S, Liu S, Yin G, Zhou J 

Received 5 September 2024

Accepted for publication 20 February 2025

Published 27 February 2025 Volume 2025:19 Pages 1357—1375

DOI https://doi.org/10.2147/DDDT.S494841

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Yan Zhu



Zhenyu Wu,1,2 Wang Zhan,2 Longhuo Wu,3 Luhu Yu,4 Xunlu Xie,5 Fang Yu,6 Weihao Kong,6 Shengrong Bi,6 Shiwei Liu,6 Guoqiang Yin,7 Jianguo Zhou6

1Department of Medical Imaging, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People’s Republic of China; 2First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, People’s Republic of China; 3College of Pharmacy, Gannan Medical University, Ganzhou, 341000, People’s Republic of China; 4Department of Clinical Laboratory, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China; 5Department of Pathology, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China; 6Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China; 7Department of Joint Surgery, Ganzhou Hospital Affiliated to Nanchang University, Ganzhou, 341000, People’s Republic of China

Correspondence: Jianguo Zhou, Email [email protected]

Abstract: Bone and cartilage diseases are significantly associated with musculoskeletal disability. However, no effective drugs are available to cure them. FOXO3a, a member of the FOXO family, has been implicated in cell proliferation, ROS detoxification, autophagy, and apoptosis. The biological functions of FOXO3a can be modulated by post-translational modifications (PTMs), such as phosphorylation and acetylation. Several signaling pathways, such as MAPK, NF-κB, PI3K/AKT, and AMPK/Sirt1 pathways, have been implicated in the development of bone and cartilage diseases by mediating the expression of FOXO3a. In particular, FOXO3a acts as a transcriptional factor in mediating the expression of various genes, such as MnSOD, CAT, BIM, BBC3, and CDK6. FOXO3a plays a critical role in the metabolism of bone and cartilage. In this article, we mainly discussed the biological functions of FOXO3a in bone and cartilage diseases, such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), ankylosing spondylitis (AS), and intervertebral disc degeneration (IDD). FOXO3a can promote osteogenic differentiation, induce osteoblast proliferation, inhibit osteoclast activity, suppress chondrocyte apoptosis, and reduce inflammatory responses. Collectively, up-regulation of FOXO3a expression shows beneficial effects, and FOXO3a has become a potential target for bone and cartilage diseases.

Keywords: FOXO3a, osteoporosis, osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, intervertebral disc degeneration

Graphical Abstract:

Introduction

Bone and cartilage diseases, the main causes of musculoskeletal disability, are characterized by bone and cartilage structural destruction, degradation, and remodeling. Bone and cartilage diseases, such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), ankylosing spondylitis (AS), and intervertebral disc degeneration (IDD), greatly affect patients’ life quality and impose social and economic burden worldwide.1 OP affects appropriately 10% of individuals aged ≥50, with a rise to about 13.6% by the year 2030.2 An estimated 240 million patients in the world have symptomatic OA.3 Extensive research has been explored, and several therapeutic strategies have been used to manage bone and cartilage diseases. For example, several disease-modifying drugs, such as tofacitinib (a Janus kinase inhibitor) and tocilizumab (a non-tumor necrosis factor inhibitor), have been explored for RA treatment. However, no absolute cure is available.4 This might be explained by the lack of a full understanding of the molecular mechanism of bone and cartilage diseases.

The pathological mechanisms of bone and cartilage diseases are regulated by a network of signaling pathways, which disrupt the homeostasis of bone and cartilage by increasing catabolism and decreasing anabolism. A review study reports that MAPK, NF-κB, Wnt/β-catenin, and AMPK/Sirt1 signaling pathways have been implicated in the pathogenesis of bone and cartilage diseases.5 It is reported that the MAPK/NF-κB signaling pathway is involved in osteoclastogenesis in RANKL-treated bone marrow-derived macrophages (BMMs), disrupting the balance between osteoblast-regulated bone formation and osteoclast-controlled bone resorption.6 Similarly, the MAPK/NF-κB signaling pathway plays a crucial role in inflammation and immune modulation, which regulate the pathogenesis of human OA, RA, and SA.7,8 Wnt/β-catenin signaling pathway has been reported to regulate cellular differentiation, proliferation, and apoptosis. The roles of the Wnt/β-catenin signaling pathway in bone and cartilage diseases have been intensively studied in mice.9,10 The AMPK/Sirt1 signaling pathway plays a key role in mediating mitochondrial energy metabolism and oxidative stress, which are involved in the pathological development of mouse bone and cartilage diseases.11,12

The FOX family, a transcription factor superfamily, has a DNA-binding forkhead (FH) domain and contains 19 subclasses of FOXA-FOXS with 50 or more members in mammals.13 The FOXO family in mammals has four members, including FOXO1, FOXO3a, FOXO4, and FOXO6. They share more than 30% of identity in amino acid sequences and appropriately 90% of similarity in their DNA-binding FH domain (Figure 1). FOXO1, FOXO3a, and FOXO4 are commonly expressed, but with different preferences, in a multitude of mammalian tissues and organs. Interestingly, the expression of FOXO6 in human beings is originally and merely found in the brain. However, it is recently reported that FOXO6 can be found in human liver.14 The activities of FOXOs can be mediated by a variety of factors, such as insulin,15 insulin-like growth factor,16 nutrients,17 and oxidative stress.18 For example, oxidative stress can stimulate the expression of FOXOs, which in turn mediate the transcription of genes encoding antioxidants, cell cycle mediators, and pro-apoptotic factors.18 Activation of FOXOs may lead to the induction of cell survival or cell death.19 FOXO3a, also known as FOXO3, has been extensively studied as a critical mediator in stress responses, cellular homeostasis, and lifespan. FOXO3a has been implicated in cell proliferation, ROS detoxification, autophagy, and apoptosis.20,21 Various studies have shown that FOXO3a plays a critical role in bone and cartilage diseases by cross-talking with a network of signaling pathways.22–25 In this article, we mainly discuss the biological activities of FOXO3a in bone and cartilage diseases. FOXO3a shows potential effects, and it becomes a therapeutic target for bone and cartilage diseases. As far as we know, no review articles have been reported to advance the knowledge in this field.

Figure 1 The sequence and functional domains of FOXO family members are shown. Four FOXO family members (FOXO1, FOXO3a, FOXO4, and FOXO6) have more than 30% of identity in amino acid sequences and appropriate 90% of similarity in the FH domain. There are three CRs, including CR1, CR2, and CR3, in four FOXO family members. FOXO3a contains NES1 and NES2 sequences.

The Structure and Regulation of FOXO3a

The Structure of FOXO3a

The members of the FOXO family are characterized by the “wing-like spiral structure”, which includes three intertwined α helices, three β chains, a ring structure, and C-terminus. The FOXO3a protein, with a molecular weight of 71 kDa, has five structural domains, including a highly conserved forkhead wing-turn-helix DNA domain (FH), two nuclear localization sequences (NLS), a nuclear export sequence (NES), and a C-terminal transactivation domain (TAD).26 The FH domain is surrounded by some large and disordered C- and N-terminal regions, which are called intrinsically disordered regions (IDRs). There are some conserved regions (CRs) within IDRs, such as CR1, CR2, and CR327 (Figure 1). Particularly, there are AKT-phosphorylation sites and 14-3-3-binding regions within CR1 and CR2, which play an important role in mediating the stability of the complex composed of FOXO3a and DNA.28 The FH domain is the region responsible for the interaction of FOXO3a with estrogen receptor α (ERα) and p53.29 All FOXO family members can recognize the consensus DNA motifs 5ʹ-GTAAA(T/C)AA-3ʹ and 5ʹ-(C/A)(A/C)AAA(C/T)-3ʹ. The TAD domain is critical for the transactivation of target genes of FOXO3a.30 Therefore, these results show that various domains in the FOXO3a protein exhibit different biological functions, including interaction with mediators and DNA.

The Regulation of FOXO3a

The protein structure of FOXO3a indicates that FOXO3a has various biological functions and mediates various cellular processes, such as proliferation, autophagy, and apoptosis, by regulating the expression of downstream factors. There are binding regions for FOXO3a in the gene promoters of various factors, such as BIM,31 BBC3 (also named as PUMA),32 tumor necrosis factor-related apoptosis-inducing ligand (TRAIL),33 PINK1,34 intercellular cell adhesion molecule-1 (ICAM1),35 CDK6,36 manganese superoxide dismutase (MnSOD),37 and catalase (CAT).37 In peroxide-treated human primary tenocytes cultured in low serum media, FOXO3a can protect against cell apoptosis and inhibit cell proliferation. However, FOXO3a may promote cell apoptosis when cells are cultured in high serum. In particular, proliferating cells are more susceptible to the pro-apoptotic actions of FOXO3a.38 Therefore, these results show that the distinctive functions of FOXO3a are associated with different cellular environments and cell lines.

In addition, the biological functions of FOXO3a may be mediated by post-translational modifications (PTMs), such as phosphorylation (Figure 2) and acetylation.30 Phosphorylation modification is the most critical mechanism in mediating the transcriptional functions of FOXO3a. There are several serine (Ser)/threonine (Thr) residues in the FOXO3a protein that can be phosphorylated. For example, the residue Thr32, Ser253, and Ser315 of FOXO3a can be phosphorylated by AKT in the nucleus (Figure 2). Interestingly, Thr32 and Ser253 phosphorylation of FOXO3a are essential for its binding to 14-3-3 proteins, and this interaction may lead to its cytoplasmic translocation from the nucleus.39,40 However, the deficiency of Ser315 phosphorylation does not seem to affect this interaction.39 AKT can phosphorylate the 14-3-3 protein, induce the dissociation of 14-3-3, and trigger the nuclear translocation of FOXO3a.41

Figure 2 The phosphorylation modification of FOXO3a has been shown. FOXO3a can be phosphorylated by AKT, SGK, AMPK, MST1, MK5, p38, JNK, ERK, and IKK. Phosphorylation at Thr32, Ser253, Ser294, Ser344, Ser425, and Ser644 can induce nuclear export. In contrast, phosphorylation at Ser7, Ser207, Ser212, and Ser574 can induce nuclear translocation. ERK-induced Ser12 phosphorylation and AMPK-induced Ser30 phosphorylation promote the mitochondrial accumulation of FOXO3a.

Serum and glucocorticoid-inducible kinase (SGK) can also phosphorylate FOXO3a at the residues of Thr32, Ser253, and Ser315 (Figure 2). In contrast to AKT, SGK prefers to phosphorylate the residues of Thr32 and Ser253, leading to the nuclear export of FOXO3a.42 It has been reported that p53-mediated SGK activation can promote FOXO3a nuclear export after DNA damage. The crosstalk between p53 and FOXO3a results in FOXO3a phosphorylation and nuclear export in an AKT-independent manner.43 Mammalian sterile 20-like kinase 1 (MST1) can be activated under oxidative stress. The residues Ser207 and Ser212 of FOXO3a can be phosphorylated by MST1, which inhibits the interaction of FOXO3a with 14-3-3 and promotes the nuclear accumulation of FOXO3a.44 Another study reports that MK5 can phosphorylate FOXO3a at Ser215 and increase its nuclear localization and DNA binding activity in U2OS cells.45 It has been reported that AMP-activated protein kinase (AMPK) can phosphorylate at least six residues (Thr179, Ser399, Ser413, Ser555, S588, and Ser626) of FOXO3a (Figure 2). AMPK-mediated phosphorylation does not affect the subcellular localization of FOXO3a but activates the transcriptional activity of FOXO3a.46

The phosphorylation of FOXO3a is also mediated by the MAPK/NF-κB signaling pathway (Figure 2). It has been reported that the residue Ser7 of FOXO3a can be phosphorylated by p38 MAPK, which induces the nuclear re-localization of FOXO3a and promotes its transcriptional activity.47 However, SB202190, a specific inhibitor of p38 MAPK, can also promote FOXO3a nuclear accumulation and transcriptional activity.48 It has been reported that JNK can induce FOXO3a nuclear localization and inhibit the expression of Bcl-2 by phosphorylating the residue Ser574 of FOXO3a.49 The residues Ser294, Ser344, and Ser425 of FOXO3a can be phosphorylated by ERK, which can induce FOXO3a nuclear export and promote MDM2-mediated ubiquitin-proteasome degradation.50 Another study shows that ERK-mediated Ser12 phosphorylation and AMPK-mediated Ser30 phosphorylation are essential for FOXO3a to accumulate in mitochondria in metabolically stressed cancer cells.51 IκB kinase (IKK), a critical factor in the NF-κB signaling, can phosphorylate the residue Ser644 of FOXO3a, induce its nuclear export, and promote its protein degradation.52 IKK-mediated Ser644 phosphorylation is essential for βTrCP-mediated ubiquitin-proteasome degradation of FOXO3a.53 Therefore, these results show that phosphorylation modification at different sites in FOXO3a can lead to its activation, inactivation, or even degradation.

Acetylation and deacetylation are also the PTMs for mediating the biological functions of proteins. FOXO3a is also mediated by CBP/p300-mediated acetylation and Sirt-mediated deacetylation. CBP/p300 can interact with the N-terminus of FOXO3a and induce its acetylation.54 In comparison to the phosphorylation modification, the role of deacetylation modification in mediating the expression of FOXO3a is controversial. It has been reported that Sirt1 and Sirt2 can promote Skp2-mediated ubiquitination and proteasomal degradation of FOXO3a.55 In non-small cell lung cancer (NSCLC) A549 and H1435 cells, Sirt1-mediated deacetylation consistently induces the ubiquitin-proteasome degradation of FOXO3a.56 Sirtinol, a Sirt1-specific inhibitor, can enhance the levels of FOXO3a in NSCLC H1299 cells.57 However, there are amounts of evidence to support that Sirt-mediated acetylation promotes the nuclear localization and activation of FOXO3a.58–60 It has been reported that naringenin can promote the deacetylation and protein expression of FOXO3a. Sirt1 siRNA transfection can abolish the protein expression and acetylated levels of FOXO3a.60 Sirt2 promotes the deacetylation and transactivation activity of FOXO3a in NIH3T3 cells.61 Sirt3 and Sirt5 can also mediate the deacetylation of FOXO3a. It is reported that the nuclear localization of FOXO3a is significantly decreased in Sirt3-knockout mice.62 Overexpression of Sirt3 can decrease the acetylation of FOXO3a and promote the accumulation of FOXO3a in LPS-treated BEAS-2B cells.63 Consistently, Sirt2 and Sirt5 have been reported to promote the nuclear translocation of FOXO3a in oocytes.64 Therefore, these results show that the roles of acetylation/deacetylation modification in FOXO3a still need more investigation.

The biological activity of FOXO3a can also be mediated by the interacting partners, such as estrogen receptors (ERα and ERβ), β-catenin, and Smads. It has been reported that ERα may interact with the N-terminal region (amino acids 1–300) and ERβ binds to the C-terminal region (amino acids 301–673). The interaction between FOXO3a and ER may lead to the inhibition of ER transcriptional activity. Mechanistically, FOXO3a competes with ERs to bind to the ER-binding sites at the promoter of target genes.65 The physical interaction between FOXO3a and β-catenin may result in the suppression of the Wnt/β-catenin signaling pathway and the down-regulation of target gene expression.66 The FOXO3a/p-Smad3/Smad4 complex can initiate the transcriptional expression of the CCNG2 gene.67 FOXO3a and FOXM1 are the two antagonistic factors that exhibit opposite biological activities. FOXO3a competitively interacts with the FH transcriptional response element (FHRE) in the promoter of VEGF and inhibits its transcription.68 Interestingly, FOXO3a can suppress the transcriptional expression of FOXM1 by recruiting HDAC1 and HDAC2.69 Therefore, these results show that the biological functions of FOXO3a can be mediated by PTMs and interactions with regulators, and this mediation for FOXO3a can be involved in bone and cartilage diseases due to the dysregulation of signaling pathways.

The Roles of FOXO3a in OP Development

OP, a bone disorder, has been associated with disrupted bone microarchitecture and decreased bone mass and strength. There are many factors contributing to the pathogenesis of OP. Aging and estrogen deficiency play critical roles in pathological alterations. In particular, menopause-related estrogen deficiency promotes bone metabolic imbalance, which is shown by decreased bone formation and increased bone resorption.70 FOXO3a has been reported to be a crucial factor in mediating osteogenic differentiation and osteoblast apoptosis under oxidative stress in patients with postmenopausal OP (PMOP).71 17β-estradiol (17β-E2), a sex hormone, has been proven to promote osteoblast differentiation and protect against OP. It is reported that 17β-E2 can up-regulate Sirt1 expression, promote autophagy by activating the AMPK/mTOR signaling, and inhibit osteoblast apoptosis by activating FOXO3a expression22 (Table 1). However, estrogen/17β-E2 replacement for disease treatment may cause severe adverse complications. Therefore, these results show that FOXO3a plays a key role in protecting against OP development.

Table 1 FOXO3a Exhibits the Biological Functions of in Different Bone and Cartilage Models

The Roles of FOXO3a in Osteogenic Differentiation

The physiological levels of reactive oxygen species (ROS) might be beneficial for the maintenance of bone homeostasis. However, the imbalance between ROS overproduction and the antioxidant defense system can lead to bone diseases. During cell differentiation, increased mitochondrial biogenesis may produce more ROS.95 Oxidative stress decreases osteogenic differentiation in cell lines during OP development. It has been established that Sirt1 can inhibit oxidative stress-induced osteogenic differentiation inhibition and osteoporotic phenotype. Hydrogen peroxide (H2O2) induces mesenchymal stem cell (MSC) lineage commitment towards adipogenesis instead of osteogenesis by decreasing the expression of Sirt1, which up-regulates the expression of Runx2 by activating FOXO3a72 (Table 1). In Sirt1-transgenic (Sirt1TG) mice, the skeletal size, bone volume, osteoblast number, alkaline phosphatase (ALP) activity, and osteogenesis-related gene expression (collagen I, Runx2, and osteocalcin) are significantly enhanced, compared to those in the wide-type mice. Mechanistically, Sirt1 reduces the acetylation of FOXO3a, increases the levels of FOXO3a and SOD2, and promotes osteogenesis in MSCs73 (Figure 3). One study reports that Sirt1-activated FOXO3a can bind to the FOXO response elements (FRE) in the promoter region of Runx2 and promote its transcriptional expression in human MSCs (hMSCs).96

Figure 3 A schematic diagram shows the regulation of FOXO3a and its role in transcriptional activity. Activated PI3K/AKT can induce FOXO3a phosphorylation and the interaction of p-FOXO3a and 14-3-3, leading to nuclear export. Then, FOXO3a undergoes ubiquitination and proteasomal degradation. IKKs also promote the proteasomal degradation of FOXO3a. However, AMPK and Sirt1 promote FOXO3a nuclear translocation and transcriptional activity in mediating the downstream factors, such as BIM, BBC3, MnSOD, and CD6. Mitochondrial Sirt3 activates FOXO3a, which decreases ROS production by up-regulating MnSOD expression.

Wnt/β-catenin signaling has been implicated in skeleton formation, and dysregulation of the Wnt/β-catenin signaling is associated with the pathological alterations in bone disorders. The role of Wnt/β-catenin signaling in inducing osteogenesis can be regulated by FOXO3a. Interaction of FOXO3a with β-catenin sequesters β-catenin away from the LEF/TCF complex.97 It is interesting to find that high glucose (HG) inhibits osteogenic differentiation of embryonic stem cells by diverting β-catenin from FOXO3a to the LEF/TCF complex and enhancing the activity of AKT in embryonic stem cells (ESCs).74 MiR-155 plays a crucial role in vascular calcification and promotes the expression of osteogenic factors, such as Runx-2 and OPN, in human vascular smooth muscle cells (VSMCs). MiR-155 deficiency promotes cell survival and migration and inhibits cell apoptosis in VSMCs by activating AKT and promoting phosphorylation and degradation of FOXO3a, thus inhibiting vascular calcification.98

Oxidative stress-mediated expression of FOXO3a is also affected by various external factors, such as alcohol administration and cigarette smoke.76,99 Alcohol administration has been associated with delayed fracture healing, due to oxidative stress. It has been reported that treatment of primary rat MSCs with alcohol (50 mm) can down-regulate the expression of osteogenic and chondrogenic lineage markers. Knockdown of FOXO1/3a can rescue the effects of alcohol (50 mm) on primary rat MSCs. However, inhibitors of FOXO1/3a can partially rescue the expression of pro-osteogenic lineage markers. Under oxidative stress, FOXOs bind and sequester β-catenin, which is crucial for the initiation of MSC osteochondral differentiation.76 Cigarette smoke extract (CSE) can significantly reduce bone density, induce aging, and suppress osteogenic differentiation of BMSCs in vivo and in vitro. Mechanistically, CSE induces ROS generation, stimulates oxidative stress, and inhibits mitophagy by mediating the AKT/FOXO3a/Pink1/Parkin axis.99 It has been reported that selenium nanoparticles (SeNPs) are antioxidants that induce hMSC differentiation toward osteogenesis over adipogenesis by activating the JNK/FOXO3a signaling pathway75 (Table 1). However, one study reports that H2O2 may promote osteogenic differentiation of VSMCs by up-regulating the expression of Runx2. Consistently, dietary salt increases the NADPH oxidase 4 (NOX4)-mediated generation of H2O2 by activating the AKT/FOXO3a signaling pathway.100 This discrepancy might be associated with the different cell lines and microenvironment. Therefore, these results show that oxidative stress inhibits osteogenic differentiation and promotes OP development by negatively regulating the expression of FOXO3a.

The Roles of FOXO3a in Osteoblasts

An increase in oxidative stress can be a crucial pathogenic factor for bone loss. It has been demonstrated that H2O2 treatment can cause the inhibition of osteoblast differentiation in MC3T3-E1 cells.101 Nicotinamide (NAM) is the water-soluble form of vitamin B3 and one of the precursors of NAD+ that is a coenzyme in mediating energy metabolism.102 NAM can improve mitochondrial metabolism, reduce mitochondrial oxidative stress, inhibit apoptosis, and enhance osteoblast differentiation by activating the Sirt3/FOXO3a signaling pathway in H2O2-treated MC3T3-E1 cells77 (Figure 3). The tripartite motif (TRIM) family is a class of E3 ubiquitin ligases. It has been reported that TRIM33 promotes osteoblast proliferation and differentiation.103 Interestingly, TRIM3 can physically interact with FOXO3a and suppress its acetylation and degradation in H2O2-treated osteoblasts, leading to the inhibition of osteoblast apoptosis induced by oxidative stress78 (Table 1).

JNK mediates the phosphorylation of FOXO3a, the translocation of the cytoplasm into the nucleus, and the transcriptional activity in up-regulating the expression of antioxidant enzymes, Runx2, and ALP.75,104 It has been reported that aortic carboxypeptidase-like protein (ACLP) is high expression in skeletal muscles and promotes bone formation. The recombinant N-terminal mouse ACLP [rmACLP(N)] increases osteoblast survival, phosphorylated p38MAPK, and nuclear translocation of FOXO3a in H2O2-treated MC3T3-E1 cells.79 Cysteine-rich-61 (Cyr61, also known as CCN1) mediates cell adhesion, chemotaxis, vascular restenosis, and angiogenesis. FOXO3a functions as a transcriptional repressor and interacts with the promoter of Cyr61.105 It has been reported that TNFα up-regulates Cyr61 expression and enhances FOXO3a phosphorylation in MC3T3-E1 cells. Overexpression of FOXO3a compromises the effects of TNFα on Cyr61 expression, CCL2 secretion, and macrophage recruitment, leading to attenuation of bone resorption and alleviation of periapical lesions80 (Table 1).

Non-imprinted in Prader-Willi/Angelman syndrome region protein 2 (NIPA2), a highly selective magnesium transporter, has been negatively associated with the development of type 2 diabetes mellitus (T2DM)-related OP. Magnesium is tightly correlated with mitochondrial physiology by mediating the AMPK signaling pathway.106 Knockout of NIPA2 may result in a deficiency of magnesium. NIPA2 positively regulates the osteogenic capacity by inactivating PINK1/Parkin-mediated mitophagy via activation of the PGC1α/FOXO3a signaling pathway in HG-treated human fetal osteoblastic (hFOB1.19) cells.81 Iron overload has been associated with OP pathogenesis. In ferric ammonium citrate (FAC)-treated MC3T3-E1 cells, the expression of dual-specificity phosphatase 14 (DUSP14) is down-regulated. FOXO3a interacts with the promoter of DUSP14 and enhances its expression and increased expression of DUSP14 protects osteoblasts from iron overload.107

In mice with genetic deletion of 25-hydroxyvitamin D-1α hydroxylase (1αOHase−/− mice), down-regulated expression of Sirt1 and FOXO3a, increased oxidative stress, and up-regulated expression of osteocyte senescence-related markers are observed. Overexpression of Sirt1 in 1αOHase−/− mice may reverse these pathological alterations by up-regulating FOXO3a expression.108 1,25-dihydroxyvitamin D3 (1,25-D3) promotes osteoblast activity and bone mineralization by increasing calcium absorption. It has been reported that 1,25-D3 up-regulates FOXO3a expression and promotes its nuclear translocation. However, FOXO3a overexpression decreases calcium uptake and prevents osteoblast differentiation in 1,25-D3-treated MC3T3-E1 cells82 (Table 1).

Osteoblast apoptosis has been associated with OP pathogenesis. Unexpectedly, it is reported that osteoblast differentiation is suppressed in osteoblast-specific Bcl-2-transgenic mice.109 Bcl-2 deficiency may lead to accelerated osteoblast differentiation by enhancing the expression and activities of FOXO3a via AKT inactivation23 (Figure 3). One study reports that fluid shear stress (FSS) may inhibit TNFα-induced apoptosis in MC3T3-E1 cells. Mechanically, FSS activates ERK5, which phosphorylates the downstream factor AKT. Activated AKT induces the phosphorylation of FOXO3a and suppresses its transcriptional activities in regulating the expression of FasL and Bim.110

Endogenous glucocorticoids (GCs) at a low dose (10−8 M) have been shown to enhance osteoblast viability, decrease apoptosis, and promote autophagy by activating the expression of serum- and glucocorticoid-induced kinase-1 (SGK-1), which phosphorylates and inactivates FOXO3a in MC3T3-E1 cells.111 Hepcidin, an endogenous hormone peptide, regulates iron homeostasis. Hepcidin deficiency suppresses osteoblast differentiation but does not affect osteoclast differentiation. In addition, the hepcidin deficiency diverts the binding of β-catenin from TCF4 to FOXO3a. Inhibition of FOXO3a can rescue the loss of bone mass in hepcidin-knockout mice, in which iron is accumulated.112 Therefore, these results show that FOXO3a activation has been associated with osteoblast differentiation, proliferation, and apoptotic inhibition. Many factors, such as oxidative stress and dysregulated signaling pathways, may compromise FOXO3a activation and inhibit osteoblast activity.

The Roles of FOXO3a in Osteoclasts

During the differentiation of macrophages into osteoclasts, the expression of caspase-2 is decreased. Caspase-2 deficiency does not affect the early stage but promotes the late stage of differentiation. Caspase-2 deficiency-mediated osteoclastogenesis is mediated by the overproduction of ROS and the reduction of FOXO3a and its target antioxidants.113 OP-D down-regulates the expression of serum osteoclastic markers, such as CTX-1 and TRAP, by activating the FOXO3a/β-catenin signaling pathway. In addition, OP-D promotes the production of GSH and decreases the generation of ROS and MDA in RAW264.7 cells.114 Therefore, these results show that down-regulation of FOXO3a expression promotes osteoclastogenesis.

The Roles of FOXO3a in Osteocytes

Osteocytes are the orchestrators connecting with osteoblasts and osteoclasts for bone remodeling. An increase in osteocyte apoptosis in bone might be the potential mechanism underlying bone deterioration.115 Persistent hyperglycemia-induced oxidative stress can induce osteocyte apoptosis in diabetes. Adiponectin (APN) and its receptors, such as AdipoR1 and AdipoR2, can be found in osteoblasts, osteoclasts, and chondrocytes. APN up-regulates the expression of Runx-2, osteocalcin, and alkaline phosphatase (ALP) and promotes osteogenic differentiation.116 In addition, APN promotes cellular proliferation and suppresses apoptosis by activating the AMPK/FOXO3a signaling pathway and attenuating oxidative stress in high glucose-treated MLO-Y4 cells.117 Therefore, these results show that FOXO3a activation can inhibit apoptosis and promote proliferation in osteocytes.

The Roles of FOXO3a in OA Development

OA, a progressive and degenerative joint disease, has been associated with chronic inflammation and reduced physical functions. There are more than 250 million patients with OA worldwide,118 and the incidence rate of knee OA in people over 65 years is about 60% in China.119 Particularly, women after menopause have a higher incidence of OA than age-matched men.120 OA decreases the patients’ life quality and has caused a great economic burden on OA patients. Due to no effective drugs to cure it, OA has become a health concern. Chondrocyte, the unique cell type in the cartilage, is responsible for the metabolism of extracellular matrix (ECM) and the homeostasis of cartilage. It has been reported that chondrocyte apoptosis and ECM degradation are the main pathological alterations in OA pathogenesis.121 Matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondins (ADAMTSs) are responsible for degrading collagens and aggrecans, respectively, which are the main components in the cartilage.122

AMPK, a master regulator of energy homeostasis, is the heterotrimeric complex composed of many subunits, such as AMPKα, AMPKβ, and AMPKγ. Activated AMPK can limit oxidative stress, attenuate the pro-catabolic responses, and protect against OA development by up-regulating the expression of FOXO3a and PGC1α in OA chondrocytes24 (Figure 3). Estrogen deficiency has been associated with accelerated bone loss and cartilage damage.123 Interestingly, co-culture of ovariectomy (OVX)-OA subchondral osteoblasts with chondrocytes can significantly decrease the anabolism of chondrocytes and promote the degeneration of chondrocytes by down-regulating the AMPK/FOXO3a signaling pathway. Treatment with AMPK agonist AICAR can partially rescue the catabolic effects of OVX-OA osteoblasts on OA chondrocytes83 (Table 1). Osteonectin (SPARC), a glycoprotein secreted by osteoblasts, can promote mineralization during bone formation. Recently, it has been reported that SPARC may increase the expression of MMPs and angiogenesis. SPARC released from OVX-OA osteoblasts promotes chondrocyte degradation by activating the NF-κB signaling. AICAR rescues SPARC-induced catabolism by activating the AMPK/FOXO3a signaling pathway.83,124

FOXO3a can interact with C/EBPβ and suppress its activity.125 It has been reported that the naturally occurring flavonoid TMF can inhibit chondrocyte apoptosis and suppress ADAMTS5-mediated aggrecan degradation by attenuating C/EBPβ expression via activating the Sirt1/FOXO3a signaling pathway in IL-1β-treated chondrocytes.84 TMF also mediates ABCA1-mediated cholesterol metabolism and protects against cholesterol accumulation-induced chondrocyte apoptosis by activating the Sirt1/FOXO3a signaling pathway in OA chondrocytes.126 DL-3-n-butylphthalide (NBP) is a small molecule isolated from celery seeds. It is reported that NBP can enhance the expression of ECM-related components, such as collagen II, aggrecan, and proteoglycan 4 (PRG4), and alleviate the apoptosis of human OA chondrocytes by inhibiting the PI3K/AKT signaling and up-regulating FOXO3a expression.127

The PH-domain leucine-rich protein phosphatase 1 (PHLPP1), an atypical protein phosphatase, has been involved in negatively mediating the anabolism-related pathways, such as the AKT and PKC pathways. In OA chondrocytes, the expression of PHLPP1 is significantly increased. This might be due to the low levels of CpG methylation on the promoter of PHLPP1.128 It is interesting to find that FOXO3a mediates the expression of PHLPP1. FOXO3a inhibition decreases the transcriptional activity of the Phlpp1 promoter and mRNA levels and increases GAG staining.129 However, FOXO1/3/4 deficient mice demonstrate OA-like alterations, such as cartilage degradation, indicating the important role of FOXO in cartilage homeostasis. Particularly, the knockout of FOXO3a increases the severity of OA in mice.130 MicroRNA has been implicated in OA development. The expression of miR-30b-5p in OA chondrocytes is up-regulated. Overexpression of miR-30b-5p promotes apoptosis and NLRP3 inflammasome activation in IL-1β-treated chondrocytes by degrading Sirt1. Overexpression of Sirt1 compromises the effects of miR-30b-5p on OA chondrocytes by promoting FOXO3a expression85 (Table 1).

Endochondral ossification, a controlled developmental process, has been associated with the switch of chondrocytes at the growth plate from a resting state to an active state where chondrocytes continue to proliferate to hypertrophy. Hypertrophic chondrocytes secrete collagen X and mineralize the ECM. Next, osteoblasts, osteoclasts, and blood vessels invade the ECM to form the primary ossification center. Subsequently, hypertrophic chondrocyte initiates apoptosis.131 FOXO3a is a transcription factor that is involved in this process. Mice with the complete loss of FOXO show an increase in hypertrophic zone length and hyperkyphosis phenotype at a higher age. This suggests a crucial role of FOXO in normal endochondral ossification.132 BMP‑binding endothelial regulator (BMPER), a secreted glycoprotein, directly interacts with BMP4 to modulate its functions. BMPER is a downstream factor of FOXO3a. It has been reported that TMF-activated FOXO3a can inhibit OA chondrocyte hypertrophy by suppressing the BMPER/BMP4 signaling pathway86 (Table 1).

However, there are opposite comments on the roles of FOXO3a in OA pathogenesis. Transglutaminase 2 (TG2), a calcium-dependent enzyme expressed in the cartilage and growth plate, up-regulating the expression of MMP-3 and MMP-13 by promoting the phosphorylation of AMPK and FOXO3a and the nuclear translocation of FOXO3a. This suggests that FOXO3a is responsible for the production of ECM catabolism and the development of OA by up-regulating the expression of MMP-3 and MMP-13.133 The explanation might be associated with the binding of FOXO3a to the promoter of MMP-13 and the activation of its transcriptional expression in VSMCs.134 Therefore, these results show that FOXO3a activation promotes ECM anabolism, inhibits chondrocyte apoptosis, and protects against cartilage degradation.

The Roles of FOXO3a in RA Development

RA, a chronic inflammatory autoimmune disease, is characterized by swollen joints, synovial inflammation, and bone destruction. Genetic and environmental factors have been included in the pathogenesis of RA. However, the exact cause of RA remains unknown. It is well documented that fibroblast-like synoviocytes (FLSs) and synovial tissue-specific cells are the critical effectors in RA development. It has been reported that RA FLSs are responsible for the synthesis of pro-inflammatory cytokines, such as IL-1β, IL-6, and TNFα.135 FOXO3a in the immune system has multiple potential roles in mediating cell cycle and cell apoptosis. FOXO3a deficiency leads to mild systemic autoimmunity, as shown by T-cell hyper-activation and increased cytokine production.136 However, another study reports that the transcript levels of FOXO3a in the peripheral blood mononuclear cells (PBMCs) from RA patients show no significant difference compared with normal controls.137

The expression of FOXO3a is attenuated in RA synovial tissues and lipopolysaccharide (LPS)-treated FLSs. Overexpression of FOXO3a suppresses LPS-induced production of pro-inflammatory cytokines and inhibits collagen-induced arthritis in rats.87,138 Consistently, the decreased expression of FOXO3a is associated with the increased expression of miR-155 in RA synovial tissues. Overexpression of FOXO3a compromises the regulatory activity of miR-155 in up-regulating the expression of IL-1β, IL-6, and TNFα.25 Tripartite motif-containing protein 3 (TRIM3) regulates the innate immune responses and carcinogenesis. FOXO3a can interact with the promoter of TRIM3 and stimulate its activity. In addition, FOXO3a compromises the effects of LPS on TRIM3 expression, protecting against RA development87 (Table 1).

Cyr61 has been implicated in RA pathogenesis. It is reported that Cyr61 is up-regulated in RA synovial tissues and plays a crucial role in the IL-17-regulated proliferation of RA synovial fibroblasts (SFs) by mediating the ανβ5/AKT/NF-κB signaling pathway.139 TNFα up-regulates Cyr61 expression and decreases FOXO3a nuclear translocation. However, FOXO3a overexpression can reverse TNFα-induced Cyr61 expression by directly binding to the promoter of Cyr61 in RASFs. Simvastatin protects collagen-induced inflammatory arthritis by activating the Sirt1/FOXO3a signaling and inhibiting Cyr61 expression.88 FOXO3a suppresses the activation of NF-κB, which is responsible for T-cell hyperactivity. FOXO3a mediates helper T-cell activation and tolerance by inhibiting inflammatory responses.140 In FOXO3a-knockout mice, intraperitoneal injection of serum derived from KRN transgenic mice induces severe inflammatory arthritis. FOXO3a maintains neutrophil vitality, and FOXO3a-deleted neutrophils increase Fas ligand expression and stimulate apoptosis.141

Angiogenesis is one of the critical underlying mechanisms for sustaining a chronic inflammatory state in early RA pathogenesis. IL-1β significantly promotes the expression of angiogenic factors, such as VEGF, and induces synovial angiogenesis. Phospholipase D (PLD) is significantly up-regulated in IL-1β-treated synoviocytes from RA patients. Mechanistically, IL-1β increases the binding of NF-κB and ATF2 to the promoter of PLD and leads to the enhancement of angiogenesis and inflammation. However, activation of FOXO3a induced by PLD inhibitors can abolish IL-1β-induced angiogenesis and inflammation in RA FLSs.89 Consistently, another study supports that inhibition of the PI3K/AKT and MEK/ERK signaling pathways and subsequent activation of FOXO3a can enhance the antiangiogenic effects of resveratrol in human umbilical vein endothelial cells (HUVECs)90 (Table 1). Resveratrol is a natural compound and has been widely used in protecting against cancer, diabetic retinopathy, psoriasis, and cardiovascular diseases. Interestingly, whether resveratrol-mediated FOXO3a activation is associated with angiogenesis inhibition in these diseases still needs to be verified.

Therefore, these results show that FOXO3a activity is attenuated in RA synovial tissues, and increased FOXO3a expression can ameliorate inflammatory responses, inhibit angiogenesis, and protect against RA development.

The Roles of FOXO3a in AS Development

Ankylosing spondylitis (AS), a chronic inflammatory autoimmune disease, may cause joint pain, limited mobility, and even disability. Particularly, low-back pain and morning stiffness are the symptoms at the early stage of AS, and spinal stiffness and deformity may occur at the late stage.142 The global prevalence of AS is 0.2–1.4%, and most patients are young men.143 In the clinic, there are no effective therapeutic strategies to cure AS. In addition, the potential molecular mechanisms in the pathogenesis of AS are complicated and remain unclear.

Earlier studies on AS development have identified human leukocyte antigen (HLA)-B27 as a genetic risk factor for AS. A recent study suggests that HLA-B27 can only account for 20% of overall genetic susceptibility to AS.144 The relationship between FOXO3a gene polymorphisms and AS susceptibility has been explored in the Eastern Chinese Han population. FOXO3a polymorphisms rs12212067 and rs3800232 can be risk factors for AS development.145 DNA methylation plays a critical role in AS pathogenesis. It has been reported that differential methylation has been observed in 19 CpG sites in the promoter of FOXO3a, and the CpG4 and CpG5 islands are significantly hypomethylated in patients with AS. The mRNA expression levels of FOXO3a are decreased and negatively correlated with the methylation of the CpG2 island in AS patients.146

It has been reported that FOXO3a inhibits inflammatory responses and oxidative stress in Jurkat cells, as shown by decreased levels of IL-8, IL-17A, and IL-23 and increased levels of SOD, CAT, and T-AOC. Bioinformatics analysis shows that FOXO3a can bind to the promoters of TGFβ and HO-1 and transcriptionally activate their expression91 (Table 1). T-cell immune imbalance-induced inflammatory responses and tissue damage play a critical role in AS pathogenesis. It has been reported that methyltransferase-like 14 (METTL14)-regulated N6-methyladenosine (m6A) modification in T cells from AS patients is significantly decreased. METTL14 plays a critical role in alleviating inflammatory responses and activating autophagy by up-regulating the downstream factor FOXO3a expression in an m6A-dependent manner92 (Table 1). Therefore, these results show that FOXO3a polymorphism is associated with AS pathogenesis, and up-regulation of FOXO3a expression can inhibit AS development by suppressing inflammatory responses and oxidative stress.

The Roles of FOXO3a in IDD Development

Intervertebral disc (IVD) degeneration (IDD) is a spinal-related disorder with low-back pain that is associated with inflammation, oxidative stress, and catabolism.147 Dysregulation of ECM catabolism in the IVD represents a critical pathophysiological feature of IDD. Zinc homeostasis is important for cellular metabolism. ZIP4, a member belonging to the SLC39A family, is a zinc transporter that plays a critical role in mediating nucleus pulposus (NP) physiological actions. It has been reported that ZIP4 expression is up-regulated in the NP cells of IDD patients. Enhanced expression of ZIP4 is associated with inflammation, oxidative stress, and ECM degradation by up-regulating HDAC4 expression and down-regulating Sirt1 and FOXO3a expression in the NP cells93 (Table 1).

Apoptosis, a fundamental biological process, contributes to the pathogenesis of IDD. Increased apoptosis of NP and annulus fibrosus (AF) cells has been observed in IDD.148 The expression of pro-apoptotic factor Bcl-2-binding component 3 (BBC3, also known as PUMA) is mediated by FOXO3a and up-regulated in IVD cells from IDD patients. Mechanically, CUL4A and CUL4B, the scaffold proteins, recruit DDB1, RBX1, and DCAF6 to assemble a CRL4DCAF6 E3 ligase that ubiquitinates and degrades the transcriptional co-repressors CtBP1/2 and consequently releases FOXO3a. TSC01131, an inhibitor of CRL4DCAF6 E3 ligase, can suppress IVD cell apoptosis mediated by the FOXO3a/BBC3 axis94 (Table 1). The potential activities of leaf extracts from Violina pumpkin (Cucurbita moschata Duch.) on the recovery of degenerated IVD cells have been investigated. The leaf extracts show anabolic effects on bone, as shown by the stimulation of osteoblast differentiation and the inhibition of osteoclast activity.149 Furthermore, the fraction of leaf extracts, consisting almost entirely of p-coumaric acid, enhances the expression of cellular homeostasis and stress response regulators, such as Sirt1, FOXO3a, NRF2, and SOD2.150 Therefore, these results show that FOXO3a expression exhibits protective activity against IDD development.

FOXO3a Becomes a Potential Target for Bone and Cartilage Diseases

FOXO3a has been used as a potential diagnostic indicator for RA. Sixty RA patients and thirty healthy subjects have been included in a study that FOXO3a expression in the serum is correlated with the disease activity of RA.151 Consistently, the genetic variation in FOXO3a may regulate the production of inflammatory cytokines, and it is suggested to predict the prognosis in RA. However, the single-nucleotide polymorphism (SNP) in FOXO3a is not associated with disease susceptibility but related to a milder course of RA.152,153 Interestingly, a non-coding polymorphism in FOXO3a (rs12212067: T > G) at which the minor (G) allele has been identified. Rs12212067 variation decreases LPS-induced production of proinflammatory cytokines, such as TNFα, IL-1β, IL-6, and IL-8, and increases the anti-inflammatory factor IL-10 in peripheral blood mononuclear cells (PBMC).152

FOXO3a has become a potential biomarker to evaluate chondrocyte differentiation. Autologous chondrocyte implantation (ACI), a popular surgical approach for repairing osteochondral defects, is challenged by the de-differentiation of chondrocytes. Interestingly, chondrocytes embedded into the soft collagen and alginate (Col/Alg) hydrogels initiate to re-differentiate, as indicated by up-regulated expression of Col2a1, aggrecan, and FOXO3a.154 Another study also reports that overexpression of FOXO3a promotes the expression of chondrocyte differentiation markers, such as collagen II, aggrecan, and SOX9, and enhances early chondrogenesis in ATDC5 cells or undifferentiated MSCs.155

Aging increases the susceptibility of the body to various chronic degenerative diseases, such as OP and OA. S-adenosyl-L-methionine (SAM) has been reported to promote osteogenic differentiation and ameliorate senescence by activating the PI3K/AKT/FOXO3a signaling pathway in H2O2-treated adipose-derived MSC (ADSC).156 Melatonin suppresses oxidative stress and promotes osteogenic differentiation in hMSCs by activating the AMPK/FOXO3a signaling pathway.157 Resveratrol, an activator of Sirt1, can significantly improve bone microarchitecture, promote osteogenesis, and suppress osteoclastogenesis by up-regulating the expression of FOXO3a and activating the Wnt/β-catenin signaling pathway.158

Recently, it has been reported that several anti-hypertensive drugs, particularly angiotensin-converting enzyme inhibitors, can alleviate bone fracture. Further study shows that angiotensin II causes mitochondrial DNA damage and triggers oxidative stress by inhibiting the Sirt1/FOXO3a signaling pathway in osteoblasts.159 Ophiopogonin D (OP-D), obtained from Radix Ophiopogon japonicus (L.f). Ker-Gawl, acts as an antioxidant in protecting against OP development. Mechanically, OP-D promotes cellular proliferation and up-regulates the expression of osteogenic genes, such as BGLAP, Col1a1, and SPP1, by up-regulating the expression of FOXO3a and promoting the nuclear translocation of FOXO3a and β-catenin in H2O2-treated MC3T3-E1 cells.114

The natural compound 2,3,5,4-tetrahydroxystilbene-2-o‐β‐D‐glucoside (TSG) inhibits oxidative stress and apoptosis by mediating the FOXO3a/β-catenin signaling in H2O2-treated MC3T3-E1 cells.160 Consistently, tanshinol rescues oxidative stress-reduced osteoblast differentiation by inhibiting FOXO3a expression and activating the Wnt/β-catenin signaling in H2O2-treated MC3T3-E1 cells.161 In addition, activation of the PI3K/AKT/FOXO3a and Wnt/β-catenin signaling pathways has been involved in the regulatory activity of different TiO2 nanotubes in promoting osteoblast differentiation under oxidative stress.162

Conclusions

FOXO3a plays a critical role in skeletal biology, as it mediates bone and cartilage events, such as structural destruction, degradation, and remodeling. Particularly, FOXO3a has been implicated in osteogenesis, osteoclastogenesis, chondrogenesis, and apoptosis, which are important events during the pathological development of bone and cartilage diseases. FOXO3a has become a potential target for managing bone and cartilage diseases.

The biological effects of FOXO3a are mediated by PTMs (mainly including phosphorylation and acetylation) or the interacting partners. For example, phosphorylation at different sites of FOXO3a protein may determine its fates: activation and nuclear translocation for transcriptional activity or inactivation for ubiquitin-proteasome degradation. Deacetylation may promote the nuclear accumulation of FOXO3a, while acetylation induces its nuclear export. The PTMs of FOXO3a can be mediated by several signaling pathways, such as MAPK, NF-κB, PI3K/AKT, AMPK, and Sirt1 pathways, which have been implicated in the pathogenesis of OP, OA, RA, AS, and IDD.

Indirectly, FOXO3a can act as a co-activator or co-repressor to affect a variety of downstream factors, exhibiting different biological actions. Interestingly, FOXO3a can act as a transcriptional factor to regulate the expression of its target gene expression. For example, FOXO3a can bind and activate the promoter activity of MnSOD, CAT, BIM, BBC3, and CDK6. These target factors are correlated with oxidative stress, apoptosis, or cell cycle, respectively, and may play different effects on cellular differentiation and functions of osteoblasts, chondrocytes, and FLSs.

However, there are limitations of this review article. Most studies are focusing on the biological activities of FOXO3a under a specific condition, and the research data may be controversial. FOXO3a directly or indirectly regulates the pathological changes of bone and cartilage diseases. The direct roles of FOXO3a in bone and cartilage should be further investigated. Several endogenous or exogenous agents have been explored to treat bone and cartilage diseases by indirectly mediating the expression of FOXO3a. However, drugs directly interacting with FOXO3a and mediating its activities are still absent. Data from conditional FOXO3a-knockout animals in bone and cartilage diseases are still needed. It is important to verify the specific biological functions of FOXO3a in different animal models. More comprehensive protocols are needed to elucidate the roles of FOXO3a in bone and cartilage diseases.

Abbreviations

FOXO3a, Forkhead box O3a; OP, osteoporosis; OA, osteoarthritis; RA, rheumatoid arthritis; AS, ankylosing spondylitis; IDD, intervertebral disc degeneration; PTMs, post-translational modifications; ROS, reactive oxygen species; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κ-B; AMPK, adenosine 5ʹ-monophosphate (AMP)-activated protein kinase; Sirt1, sirtuin 1; ERα, estrogen receptor α; BIM, Bcl-2-like protein 11; BBC3, Bcl2 binding component 3; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand; ICAM1, intercellular cell adhesion molecule-1; MnSOD, manganese superoxide dismutase; CAT, catalase; SGK, Serum and glucocorticoid-inducible kinase; MST1, mammalian sterile 20-like kinase 1; PMOP, postmenopausal osteoporosis; MSC, mesenchymal stem cell; CSE, cigarette smoke extract; NOX4, NADPH oxidase 4; T2DM, type 2 diabetes mellitus; APN, adiponectin; OCN, osteocalcin; ALP, alkaline phosphatase; ECM, extracellular matrix; MMPs, Matrix metalloproteinases; ADAMTSs, a disintegrin and metalloproteinase with thrombospondins; FLSs, fibroblast-like synoviocytes; LPS, lipopolysaccharide.

Data Sharing Statement

The data used to support the findings of this study are included within the article.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This study was financially supported by the Science and Technology research project of the Education Department of Jiangxi Province (GJJ2201451), Ganzhou Municipal “Science and Technology + National Regional Medical Center” Joint Project (2023LNS17449, 2022-RC1328, and 2022-YB1395).

Disclosure

The authors declare no conflict of interests.

References

1. Faeed M, Ghiasvand M, Fareghzadeh B, Taghiyar L. Osteochondral organoids: current advances, applications, and upcoming challenges. Stem Cell Res Ther. 2024;15(1):183. doi:10.1186/s13287-024-03790-5

2. Harris K, Zagar CA, Lawrence KV. Osteoporosis: common questions and answers. Am Fam Physician. 2023;107(3):238–246.

3. Allen KD, Thoma LM, Golightly YM. Epidemiology of osteoarthritis. Osteoarthritis Cartilage. 2022;30(2):184–195. doi:10.1016/j.joca.2021.04.020

4. Xu D, Wu L, Chen Q. Editorial: bone and cartilage diseases-the role and potential of natural products. Front Pharmacol. 2022;13:938303. doi:10.3389/fphar.2022.938303

5. Gan J, Deng X, Le Y, Lai J, Liao X. The development of Naringin for use against bone and cartilage disorders. Molecules. 2023;28(9):3716. doi:10.3390/molecules28093716

6. Li X, Deng W, Tang K, et al. Sophoraflavanone G Inhibits RANKL-induced osteoclastogenesis via MAPK/NF-κB signaling pathway. Mol Biotechnol. 2024. doi:10.1007/s12033-024-01185-8

7. Wu Z, Liu L. The protective activity of genistein against bone and cartilage diseases. Front Pharmacol. 2022;13:1016981. doi:10.3389/fphar.2022.1016981

8. Shi LH, Lam SHM, So H, Meng H, Tam LS. Impact of inflammation and anti-inflammatory therapies on the incidence of major cardiovascular events in patients with ankylosing spondylitis: a population-based study. Semin Arthritis Rheum. 2024;67:152477. doi:10.1016/j.semarthrit.2024.152477

9. Zhang Y, Chen H, Wu J, et al. Deficiency of Cbfβ in articular cartilage leads to osteoarthritis-like phenotype through Hippo/Yap, TGFβ, and Wnt/β-catenin signaling pathways. Int J Biol Sci. 2024;20(6):1965–1977. doi:10.7150/ijbs.90250

10. Wang X, Zou C, Hou C, et al. Extracellular vesicles from bone marrow mesenchymal stem cells alleviate osteoporosis in mice through USP7-mediated YAP1 protein stability and the Wnt/β-catenin pathway. Biochem Pharmacol. 2023;217:115829. doi:10.1016/j.bcp.2023.115829

11. Zhang X, Zhang D, Zhao H, et al. gCTRP3 inhibits oophorectomy-induced osteoporosis by activating the AMPK/SIRT1/Nrf2 signaling pathway in mice. Mol Med Rep. 2024;30(2). doi:10.3892/mmr.2024.13257

12. Zhou H, Zou L, Ren H, et al. Cathelicidin-BF regulates the AMPK/SIRT1/NF-κB pathway to ameliorate murine osteoarthritis: in vitro and in vivo studie. Int Immunopharmacol. 2024;134:112201. doi:10.1016/j.intimp.2024.112201

13. Spreitzer E, Alderson TR, Bourgeois B, et al. FOXO transcription factors differ in their dynamics and intra/intermolecular interactions. Curr Res Struct Biol. 2022;4:118–133. doi:10.1016/j.crstbi.2022.04.001

14. Chen HY, Chen YM, Wu J, et al. Expression of FOXO6 is associated with oxidative stress level and predicts the prognosis in hepatocellular cancer: a comparative study. Medicine. 2016;95(21):e3708. doi:10.1097/MD.0000000000003708

15. Bhardwaj G, Penniman CM, Klaus K, et al. Transcriptomic regulation of muscle mitochondria and calcium signaling by Insulin/IGF-1 receptors depends on FoxO transcription factors. Front Physiol. 2021;12:779121. doi:10.3389/fphys.2021.779121

16. Bhardwaj G, Penniman CM, Jena J, et al. Insulin and IGF-1 receptors regulate complex I-dependent mitochondrial bioenergetics and supercomplexes via FoxOs in muscle. J Clin Invest. 2021;131(18). doi:10.1172/JCI146415

17. Rubio B, Mora C, Pintado C, et al. The nutrient sensing pathways FoxO1/3 and mTOR in the heart are coordinately regulated by central leptin through PPARβ/δ. Implications in cardiac remodeling. Metabolism. 2021;115:154453. doi:10.1016/j.metabol.2020.154453

18. Zuo M, Tong R, He X, et al. FOXO signaling pathway participates in oxidative stress-induced histone deacetylation. Free Radic Res. 2023;57(1):47–60. doi:10.1080/10715762.2023.2190862

19. van der Horst A, Burgering BM. Stressing the role of FoxO proteins in lifespan and disease. Nat Rev Mol Cell Biol. 2007;8(6):440–450. doi:10.1038/nrm2190

20. Fasano C, Disciglio V, Bertora S, Lepore Signorile M, Simone C. FOXO3a from the nucleus to the mitochondria: a round trip in cellular stress response. Cells. 2019;8(9):1110. doi:10.3390/cells8091110

21. Webb AE, Brunet A. FOXO transcription factors: key regulators of cellular quality control. Trends Biochem Sci. 2014;39(4):159–169. doi:10.1016/j.tibs.2014.02.003

22. Wang Y, Mei R, Hao S, et al. Up-regulation of SIRT1 induced by 17beta-estradiol promotes autophagy and inhibits apoptosis in osteoblasts. Aging. 2021;13(20):23652–23671. doi:10.18632/aging.203639

23. Moriishi T, Kawai Y, Komori H, et al. Bcl2 deficiency activates FoxO through Akt inactivation and accelerates osteoblast differentiation. PLoS One. 2014;9(1):e86629. doi:10.1371/journal.pone.0086629

24. Zhao X, Petursson F, Viollet B, Lotz M, Terkeltaub R, Liu-Bryan R. Peroxisome proliferator-activated receptor γ coactivator 1α and FoxO3A mediate chondroprotection by AMP-activated protein kinase. Arthritis Rheumatol. 2014;66(11):3073–3082. doi:10.1002/art.38791

25. Wang Y, Feng T, Duan S, et al. miR-155 promotes fibroblast-like synoviocyte proliferation and inflammatory cytokine secretion in rheumatoid arthritis by targeting FOXO3a. Exp Ther Med. 2020;19(2):1288–1296. doi:10.3892/etm.2019.8330

26. Liu Y, Ao X, Ding W, et al. Critical role of FOXO3a in carcinogenesis. Mol Cancer. 2018;17(1):104. doi:10.1186/s12943-018-0856-3

27. Obsil T, Obsilova V. Structure/function relationships underlying regulation of FOXO transcription factors. Oncogene. 2008;27(16):2263–2275. doi:10.1038/onc.2008.20

28. Senf SM, Sandesara PB, Reed SA, Judge AR. p300 Acetyltransferase activity differentially regulates the localization and activity of the FOXO homologues in skeletal muscle. Am J Physiol Cell Physiol. 2011;300(6):C1490–1501. doi:10.1152/ajpcell.00255.2010

29. Morelli C, Lanzino M, Garofalo C, et al. Akt2 inhibition enables the forkhead transcription factor FoxO3a to have a repressive role in estrogen receptor alpha transcriptional activity in breast cancer cells. Mol Cell Biol. 2010;30(3):857–870. doi:10.1128/MCB.00824-09

30. Dong Z, Guo Z, Li H, et al. FOXO3a-interacting proteins’ involvement in cancer: a review. Mol Biol Rep. 2024;51(1):196. doi:10.1007/s11033-023-09121-w

31. Vogiatzi P, De Falco G, Claudio PP, Giordano A. How does the human RUNX3 gene induce apoptosis in gastric cancer? Latest data, reflections and reactions. Cancer Biol Ther. 2006;5(4):371–374. doi:10.4161/cbt.5.4.2748

32. Yan J, Yang S, Tian H, Zhang Y, Zhao H. Copanlisib promotes growth inhibition and apoptosis by modulating the AKT/FoxO3a/PUMA axis in colorectal cancer. Cell Death Dis. 2020;11(11):943. doi:10.1038/s41419-020-03154-w

33. Allen JE, Krigsfeld G, Mayes PA, et al. Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects. Sci Transl Med. 2013;5(171):171ra117. doi:10.1126/scitranslmed.3004828

34. Gupta P, Sharma G, Lahiri A, Barthwal MK. FOXO3a acetylation regulates PINK1, mitophagy, inflammasome activation in murine palmitate-conditioned and diabetic macrophages. J Leukoc Biol. 2022;111(3):611–627. doi:10.1002/JLB.3A0620-348RR

35. Chen H, Tang X, Han TL, et al. Potential role of FoxO3a in the regulation of trophoblast development and pregnancy complications. J Cell Mol Med. 2021;25(9):4363–4372. doi:10.1111/jcmm.16499

36. Liu J, Duan Z, Guo W, et al. Targeting the BRD4/FOXO3a/CDK6 axis sensitizes AKT inhibition in luminal breast cancer. Nat Commun. 2018;9(1):5200. doi:10.1038/s41467-018-07258-y

37. Guo Y, Cai X, Lu H, et al. 17β-Estradiol promotes apoptosis of HepG2 cells caused by oxidative stress by increasing Foxo3a phosphorylation. Front Pharmacol. 2021;12:607379. doi:10.3389/fphar.2021.607379

38. Poulsen RC, Carr AJ, Hulley PA. Cell proliferation is a key determinant of the outcome of FOXO3a activation. Biochem Biophys Res Commun. 2015;462(1):78–84. doi:10.1016/j.bbrc.2015.04.112

39. Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999;96(6):857–868. doi:10.1016/S0092-8674(00)80595-4

40. Brunet A, Kanai F, Stehn J, et al. 14-3-3 transits to the nucleus and participates in dynamic nucleocytoplasmic transport. J Cell Biol. 2002;156(5):817–828. doi:10.1083/jcb.200112059

41. Sunayama J, Tsuruta F, Masuyama N, Gotoh Y. JNK antagonizes Akt-mediated survival signals by phosphorylating 14-3-3. J Cell Biol. 2005;170(2):295–304. doi:10.1083/jcb.200409117

42. Brunet A, Park J, Tran H, Hu LS, Hemmings BA, Greenberg ME. Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol. 2001;21(3):952–965. doi:10.1128/MCB.21.3.952-965.2001

43. You H, Jang Y, You-ten AI, et al. p53-dependent inhibition of FKHRL1 in response to DNA damage through protein kinase SGK1. Proc Natl Acad Sci U S A. 2004;101(39):14057–14062. doi:10.1073/pnas.0406286101

44. Yasuda K, Ueda Y, Ozawa M, Matsuda T, Kinashi T. Enhanced cytotoxic T-cell function and inhibition of tumor progression by Mst1 deficiency. FEBS Lett. 2016;590(1):68–75. doi:10.1002/1873-3468.12045

45. Kress TR, Cannell IG, Brenkman AB, et al. The MK5/PRAK kinase and Myc form a negative feedback loop that is disrupted during colorectal tumorigenesis. Mol Cell. 2011;41(4):445–457. doi:10.1016/j.molcel.2011.01.023

46. Greer EL, Oskoui PR, Banko MR, et al. The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J Biol Chem. 2007;282(41):30107–30119. doi:10.1074/jbc.M705325200

47. Ho KK, McGuire VA, Koo CY, et al. Phosphorylation of FOXO3a on Ser-7 by p38 promotes its nuclear localization in response to doxorubicin. J Biol Chem. 2012;287(2):1545–1555. doi:10.1074/jbc.M111.284224

48. Germani A, Matrone A, Grossi V, et al. Targeted therapy against chemoresistant colorectal cancers: inhibition of p38α modulates the effect of cisplatin in vitro and in vivo through the tumor suppressor FoxO3A. Cancer Lett. 2014;344(1):110–118. doi:10.1016/j.canlet.2013.10.035

49. Li Z, Zhao J, Tikhanovich I, et al. Serine 574 phosphorylation alters transcriptional programming of FOXO3 by selectively enhancing apoptotic gene expression. Cell Death Differ. 2016;23(4):583–595. doi:10.1038/cdd.2015.125

50. Yang JY, Zong CS, Xia W, et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat Cell Biol. 2008;10(2):138–148. doi:10.1038/ncb1676

51. Celestini V, Tezil T, Russo L, et al. Uncoupling FoxO3A mitochondrial and nuclear functions in cancer cells undergoing metabolic stress and chemotherapy. Cell Death Dis. 2018;9(2):231. doi:10.1038/s41419-018-0336-0

52. Hu MC, Lee DF, Xia W, et al. IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell. 2004;117(2):225–237. doi:10.1016/S0092-8674(04)00302-2

53. Su JL, Cheng X, Yamaguchi H, et al. FOXO3a-dependent mechanism of E1A-induced chemosensitization. Cancer Res. 2011;71(21):6878–6887. doi:10.1158/0008-5472.CAN-11-0295

54. Mahmud DL, Deb DK, Platanias LC, Uddin S, Wickrema A. Phosphorylation of forkhead transcription factors by erythropoietin and stem cell factor prevents acetylation and their interaction with coactivator p300 in erythroid progenitor cells. Oncogene. 2002;21(10):1556–1562. doi:10.1038/sj.onc.1205230

55. Wang F, Chan CH, Chen K, Guan X, Lin HK, Tong Q. Deacetylation of FOXO3 by SIRT1 or SIRT2 leads to Skp2-mediated FOXO3 ubiquitination and degradation. Oncogene. 2012;31(12):1546–1557. doi:10.1038/onc.2011.347

56. Song S, Tang H, Quan W, Shang A, Ling C. Estradiol initiates the immune escape of non-small cell lung cancer cells via ERβ/SIRT1/FOXO3a/PD-L1 axis. Int Immunopharmacol. 2022;107:108629. doi:10.1016/j.intimp.2022.108629

57. Fong Y, Lin YC, Wu CY, et al. The antiproliferative and apoptotic effects of sirtinol, a sirtuin inhibitor on human lung cancer cells by modulating Akt/β-catenin-Foxo3a axis. Sci World J. 2014;2014:937051. doi:10.1155/2014/937051

58. Bordbari S, Mörchen B, Pylaeva E, et al. SIRT1-mediated deacetylation of FOXO3a transcription factor supports pro-angiogenic activity of interferon-deficient tumor-associated neutrophils. Int J Cancer. 2022;150(7):1198–1211. doi:10.1002/ijc.33871

59. Chen L, Li S, Zhu J, et al. Mangiferin prevents myocardial infarction-induced apoptosis and heart failure in mice by activating the Sirt1/FoxO3a pathway. J Cell Mol Med. 2021;25(6):2944–2955. doi:10.1111/jcmm.16329

60. Wang J, Wu R, Hua Y, Ling S, Xu X. Naringenin ameliorates vascular senescence and atherosclerosis involving SIRT1 activation. J Pharm Pharmacol. 2023;75(8):1021–1033. doi:10.1093/jpp/rgad053

61. Wang F, Nguyen M, Qin FX, Tong Q. SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell. 2007;6(4):505–514. doi:10.1111/j.1474-9726.2007.00304.x

62. Kim HS, Patel K, Muldoon-Jacobs K, et al. SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell. 2010;17(1):41–52. doi:10.1016/j.ccr.2009.11.023

63. Wu Z, Wang Y, Lu S, Yin L, Dai L. SIRT3 alleviates sepsis-induced acute lung injury by inhibiting pyroptosis via regulating the deacetylation of FoxO3a. Pulm Pharmacol Ther. 2023;82:102244.

64. Xu D, Wu L, Jiang X, et al. SIRT2 inhibition results in meiotic arrest, mitochondrial dysfunction, and disturbance of redox homeostasis during bovine oocyte maturation. Int J Mol Sci. 2019;20(6).

65. Zou Y, Tsai WB, Cheng CJ, et al. Forkhead box transcription factor FOXO3a suppresses estrogen-dependent breast cancer cell proliferation and tumorigenesis. Breast Cancer Res. 2008;10(1):R21. doi:10.1186/bcr1872

66. 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):27298–27305. doi:10.1074/jbc.M702811200

67. Fu G, Peng C. Nodal enhances the activity of FoxO3a and its synergistic interaction with Smads to regulate cyclin G2 transcription in ovarian cancer cells. Oncogene. 2011;30(37):3953–3966. doi:10.1038/onc.2011.127

68. Karadedou CT, Gomes AR, Chen J, et al. FOXO3a represses VEGF expression through FOXM1-dependent and -independent mechanisms in breast cancer. Oncogene. 2012;31(14):1845–1858. doi:10.1038/onc.2011.368

69. Osei-Sarfo K, Gudas LJ. Retinoids induce antagonism between FOXO3A and FOXM1 transcription factors in human oral squamous cell carcinoma (OSCC) cells. PLoS One. 2019;14(4):e0215234. doi:10.1371/journal.pone.0215234

70. Eastell R, Szulc P. Use of bone turnover markers in postmenopausal osteoporosis. Lancet Diabetes Endocrinol. 2017;5(11):908–923. doi:10.1016/S2213-8587(17)30184-5

71. Ren ZQ, Wang YF, Ao GF, et al. Overall adjustment acupuncture for postmenopausal osteoporosis (PMOP): a study protocol for a randomized sham-controlled trial. Trials. 2020;21(1):465. doi:10.1186/s13063-020-04435-7

72. 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):786–796. doi:10.1111/jcmm.13356

73. Sun W, Qiao W, Zhou B, et al. Overexpression of Sirt1 in mesenchymal stem cells protects against bone loss in mice by FOXO3a deacetylation and oxidative stress inhibition. Metabolism. 2018;88:61–71. doi:10.1016/j.metabol.2018.06.006

74. Dienelt A, Keller KC, Zur Nieden NI. High glucose impairs osteogenic differentiation of embryonic stem cells via early diversion of beta-catenin from Forkhead box O to T cell factor interaction. Birth Defects Res. 2022;114(16):1056–1074. doi:10.1002/bdr2.2085

75. Fatima S, Alfrayh R, Alrashed M, Alsobaie S, Ahmad R, Mahmood A. Selenium nanoparticles by moderating oxidative stress promote differentiation of mesenchymal stem cells to osteoblasts. Int J Nanomed. 2021;16:331–343. doi:10.2147/IJN.S285233

76. Sharieh F, Eby JM, Roper PM, Callaci JJ. Ethanol inhibits mesenchymal stem cell osteochondral lineage differentiation due in part to an activation of forkhead box protein O-specific signaling. Alcohol Clin Exp Res. 2020;44(6):1204–1213. doi:10.1111/acer.14337

77. Yoon H, Park SG, Kim HJ, et al. Nicotinamide enhances osteoblast differentiation through activation of the mitochondrial antioxidant defense system. Exp Mol Med. 2023;55(7):1531–1543. doi:10.1038/s12276-023-01041-w

78. Zou DB, Mou Z, Wu W, Liu H. TRIM33 protects osteoblasts from oxidative stress-induced apoptosis in osteoporosis by inhibiting FOXO3a ubiquitylation and degradation. Aging Cell. 2021;20(7):e13367. doi:10.1111/acel.13367

79. Kim H, Kim MJ, Moon SA, et al. Aortic carboxypeptidase-like protein, a putative myokine, stimulates the differentiation and survival of bone-forming osteoblasts. FASEB J. 2023;37(8):e23104. doi:10.1096/fj.202300140R

80. Lin LD, Lin SK, Chao YL, et al. Simvastatin suppresses osteoblastic expression of Cyr61 and progression of apical periodontitis through enhancement of the transcription factor Forkhead/winged helix box protein O3a. J Endod. 2013;39(5):619–625. doi:10.1016/j.joen.2012.12.014

81. Zhao W, Zhang W, Ma H, Yang M. NIPA2 regulates osteoblast function by modulating mitophagy in type 2 diabetes osteoporosis. Sci Rep. 2020;10(1):3078. doi:10.1038/s41598-020-59743-4

82. Tang KC, Pan W, Doschak MR, Alexander RT. Increased FoxO3a expression prevents osteoblast differentiation and matrix calcification. Bone Rep. 2019;10:100206. doi:10.1016/j.bonr.2019.100206

83. Jiang A, Xu P, Yang Z, et al. Increased Sparc release from subchondral osteoblasts promotes articular chondrocyte degeneration under estrogen withdrawal. Osteoarthritis Cartilage. 2023;31(1):26–38. doi:10.1016/j.joca.2022.08.020

84. Wang Z, Shi W, Wu L, et al. TMF inhibits extracellular matrix degradation by regulating the C/EBPβ/ADAMTS5 signaling pathway in osteoarthritis. Biomed Pharmacother. 2024;174:116501. doi:10.1016/j.biopha.2024.116501

85. Xu H, Zhang J, Shi X, Li X, Zheng C. NF-κB inducible miR-30b-5p aggravates joint pain and loss of articular cartilage via targeting SIRT1-FoxO3a-mediated NLRP3 inflammasome. Aging. 2021;13(16):20774–20792. doi:10.18632/aging.203466

86. Huang J, Ren Q, Jiao L, et al. TMF suppresses chondrocyte hypertrophy in osteoarthritic cartilage by mediating the FOXO3a/BMPER pathway. Exp Ther Med. 2024;28(1):283. doi:10.3892/etm.2024.12571

87. Wang M, Wu J, Zhou E, Chang X, Gan J, Cheng T. Forkhead box o3a suppresses lipopolysaccharide-stimulated proliferation and inflammation in fibroblast-like synoviocytes through regulating tripartite motif-containing protein 3. J Cell Physiol. 2019;234(11):20139–20148. doi:10.1002/jcp.28615

88. Kok SH, Lin LD, Hou KL, et al. Simvastatin inhibits cysteine-rich protein 61 expression in rheumatoid arthritis synovial fibroblasts through the regulation of sirtuin-1/FoxO3a signaling. Arthritis Rheum. 2013;65(3):639–649. doi:10.1002/art.37807

89. Kang DW, Park MK, Oh HJ, et al. Phospholipase D1 has a pivotal role in interleukin-1β-driven chronic autoimmune arthritis through regulation of NF-κB, hypoxia-inducible factor 1α, and FoxO3a. Mol Cell Biol. 2013;33(14):2760–2772. doi:10.1128/MCB.01519-12

90. Srivastava RK, Unterman TG, Shankar S. FOXO transcription factors and VEGF neutralizing antibody enhance antiangiogenic effects of resveratrol. Mol Cell Biochem. 2010;337(1–2):201–212. doi:10.1007/s11010-009-0300-5

91. Xu S, Zhang X, Ma Y, et al. FOXO3a alleviates the inflammation and oxidative stress via regulating TGF-β and HO-1 in ankylosing spondylitis. Front Immunol. 2022;13:935534. doi:10.3389/fimmu.2022.935534

92. Chen Y, Wu Y, Fang L, et al. METTL14-m6A-FOXO3a axis regulates autophagy and inflammation in ankylosing spondylitis. Clin Immunol. 2023;257:109838. doi:10.1016/j.clim.2023.109838

93. Shen M, Li K, Wang L, et al. ZIP4 upregulation aggravates nucleus pulposus cell degradation by promoting inflammation and oxidative stress by mediating the HDAC4-FoxO3a axis. Aging. 2024;16(1):685–700. doi:10.18632/aging.205412

94. Tseng C, Han Y, Lv Z, et al. The CRL4(DCAF6) E3 ligase ubiquitinates CtBP1/2 to induce apoptotic signalling and promote intervertebral disc degeneration. J Mol Med. 2023;101(1–2):171–181. doi:10.1007/s00109-022-02277-1

95. Li Q, Gao Z, Chen Y, Guan MX. The role of mitochondria in osteogenic, adipogenic and chondrogenic differentiation of mesenchymal stem cells. Protein Cell. 2017;8(6):439–445. doi:10.1007/s13238-017-0385-7

96. Tseng PC, Hou SM, Chen RJ, et al. Resveratrol promotes osteogenesis of human mesenchymal stem cells by upregulating RUNX2 gene expression via the SIRT1/FOXO3A axis. J Bone Miner Res. 2011;26(10):2552–2563. doi:10.1002/jbmr.460

97. McClelland Descalzo DL, Satoorian TS, Walker LM, Sparks NR, Pulyanina PY, Zur Nieden NI. Glucose-induced oxidative stress reduces proliferation in embryonic stem cells via FOXO3A/β-catenin-dependent transcription of p21(cip1). Stem Cell Rep. 2016;7(1):55–68. doi:10.1016/j.stemcr.2016.06.006

98. Li Y, Sun W, Saaoud F, et al. MiR155 modulates vascular calcification by regulating Akt-FOXO3a signalling and apoptosis in vascular smooth muscle cells. J Cell Mol Med. 2021;25(1):535–548. doi:10.1111/jcmm.16107

99. Xiang K, Ren M, Liu F, et al. Tobacco toxins trigger bone marrow mesenchymal stem cells aging by inhibiting mitophagy. Ecotoxicol Environ Saf. 2024;277:116392. doi:10.1016/j.ecoenv.2024.116392

100. Ying KE, Feng W, Ying WZ, et al. Dietary salt initiates redox signaling between endothelium and vascular smooth muscle through NADPH oxidase 4. Redox Biol. 2022;52:102296. doi:10.1016/j.redox.2022.102296

101. Chen F, Zhang X, Chen S, et al. 5-(3ʹ,4ʹ-dihydroxyphenyl)-γ-valerolactone, a microbiota metabolite of flavan-3-ols, activates SIRT1-mediated autophagy to attenuate H2O2-induced inhibition of osteoblast differentiation in MC3T3-E1 cells. Free Radic Biol Med. 2023;208:309–318. doi:10.1016/j.freeradbiomed.2023.08.018

102. Srivastava S. Emerging therapeutic roles for NAD(+) metabolism in mitochondrial and age-related disorders. Clin Transl Med. 2016;5(1):25. doi:10.1186/s40169-016-0104-7

103. Guo J, Qin W, Xing Q, et al. TRIM33 is essential for osteoblast proliferation and differentiation via BMP pathway. J Cell Physiol. 2017;232(11):3158–3169. doi:10.1002/jcp.25769

104. Dhoke NR, Geesala R, Das A. Low oxidative stress-mediated proliferation via JNK-FOXO3a-Catalase signaling in transplanted adult stem cells promotes wound tissue regeneration. Antioxid Redox Signal. 2018;28(11):1047–1065. doi:10.1089/ars.2016.6974

105. Lee HY, Chung JW, Youn SW, et al. Forkhead transcription factor FOXO3a is a negative regulator of angiogenic immediate early gene CYR61, leading to inhibition of vascular smooth muscle cell proliferation and neointimal hyperplasia. Circ Res. 2007;100(3):372–380. doi:10.1161/01.RES.0000257945.97958.77

106. Ha BG, Moon DS, Kim HJ, Shon YH. Magnesium and calcium-enriched deep-sea water promotes mitochondrial biogenesis by AMPK-activated signals pathway in 3T3-L1 preadipocytes. Biomed Pharmacother. 2016;83:477–484. doi:10.1016/j.biopha.2016.07.009

107. Xia D, Wu J, Xing M, et al. Iron overload threatens the growth of osteoblast cells via inhibiting the PI3K/AKT/FOXO3a/DUSP14 signaling pathway. J Cell Physiol. 2019;234(9):15668–15677. doi:10.1002/jcp.28217

108. Chen H, Hu X, Yang R, et al. SIRT1/FOXO3a axis plays an important role in the prevention of mandibular bone loss induced by 1,25(OH)(2)D deficiency. Int J Biol Sci. 2020;16(14):2712–2726. doi:10.7150/ijbs.48169

109. Moriishi T, Maruyama Z, Fukuyama R, et al. Overexpression of Bcl2 in osteoblasts inhibits osteoblast differentiation and induces osteocyte apoptosis. PLoS One. 2011;6(11):e27487. doi:10.1371/journal.pone.0027487

110. Bin G, Bo Z, Jing W, et al. Fluid shear stress suppresses TNF-α-induced apoptosis in MC3T3-E1 cells: involvement of ERK5-AKT-FoxO3a-Bim/FasL signaling pathways. Exp Cell Res. 2016;343(2):208–217. doi:10.1016/j.yexcr.2016.03.014

111. Yu XH, Xu XM, Zhang SX. Low-dose dexamethasone promotes osteoblast viability by activating autophagy via the SGK1/FOXO3a signaling pathway. Cell Biol Int. 2023;47(3):669–678. doi:10.1002/cbin.11971

112. Li G, Zhang H, Wu J, et al. Hepcidin deficiency causes bone loss through interfering with the canonical Wnt/β-catenin pathway via Forkhead box O3a. J Orthop Translat. 2020;23:67–76. doi:10.1016/j.jot.2020.03.012

113. Callaway DA, Riquelme MA, Sharma R, Lopez-Cruzan M, Herman BA, Jiang JX. Caspase-2 modulates osteoclastogenesis through down-regulating oxidative stress. Bone. 2015;76:40–48. doi:10.1016/j.bone.2015.03.006

114. Huang Q, Gao B, Wang L, et al. Ophiopogonin D: a new herbal agent against osteoporosis. Bone. 2015;74:18–28. doi:10.1016/j.bone.2015.01.002

115. Maycas M, McAndrews KA, Sato AY, et al. PTHrP-derived peptides restore bone mass and strength in diabetic mice: additive effect of mechanical loading. J Bone Miner Res. 2017;32(3):486–497. doi:10.1002/jbmr.3007

116. Chen T, Wu YW, Lu H, Guo Y, Tang ZH. Adiponectin enhances osteogenic differentiation in human adipose-derived stem cells by activating the APPL1-AMPK signaling pathway. Biochem Biophys Res Commun. 2015;461(2):237–242. doi:10.1016/j.bbrc.2015.03.168

117. Zeng Y, Liang H, Guo Y, Feng Y, Yao Q. Adiponectin regulates osteocytic MLO-Y4 cell apoptosis in a high-glucose environment through the AMPK/FoxO3a signaling pathway. J Cell Physiol. 2021;236(10):7088–7096. doi:10.1002/jcp.30381

118. Hunter DJ, Schofield D, Callander E. The individual and socioeconomic impact of osteoarthritis. Nat Rev Rheumatol. 2014;10(7):437–441. doi:10.1038/nrrheum.2014.44

119. Huang D, Liu YQ, Liang LS, et al. The Diagnosis and Therapy of Degenerative Knee Joint Disease: expert Consensus from the Chinese Pain Medicine Panel. Pain Res Manag. 2018;2018:2010129. doi:10.1155/2018/2010129

120. watt FE. Hand osteoarthritis, menopause and menopausal hormone therapy. Maturitas. 2016;83:13–18. doi:10.1016/j.maturitas.2015.09.007

121. Thielen NGM, van der Kraan PM, van Caam APM. TGFβ/BMP Signaling Pathway in Cartilage Homeostasis. Cells. 2019;8(9):969. doi:10.3390/cells8090969

122. Wang T, He C. Pro-inflammatory cytokines: the link between obesity and osteoarthritis. Cytokine Growth Factor Rev. 2018;44:38–50. doi:10.1016/j.cytogfr.2018.10.002

123. Castañeda S, Largo R, Calvo E, Bellido M, Gómez-Vaquero C, Herrero-Beaumont G. Effects of estrogen deficiency and low bone mineral density on healthy knee cartilage in rabbits. J Orthop Res. 2010;28(6):812–818. doi:10.1002/jor.21054

124. Castañeda S, Vicente-Rabaneda EF. Disentangling the molecular interplays between subchondral bone and articular cartilage in estrogen deficiency-induced osteoarthritis. Osteoarthritis Cartilage. 2023;31(1):6–8. doi:10.1016/j.joca.2022.09.006

125. Marković J, Uskoković A, Grdović N, et al. Identification of transcription factors involved in the transcriptional regulation of the CXCL12 gene in rat pancreatic insulinoma Rin-5F cell line. Biochem Cell Biol. 2015;93(1):54–62. doi:10.1139/bcb-2014-0104

126. Peng F, Huang X, Shi W, et al. 5,7,3ʹ,4ʹ-tetramethoxyflavone ameliorates cholesterol dysregulation by mediating SIRT1/FOXO3a/ABCA1 signaling in osteoarthritis chondrocytes. Future Med Chem. 2021;13(24):2153–2166. doi:10.4155/fmc-2021-0247

127. Zhang Y, Dai J, Yan L, et al. DL-3-N-Butylphthalide promotes cartilage extracellular matrix synthesis and inhibits osteoarthritis development by regulating FoxO3a. Oxid Med Cell Longev. 2022;2022:9468040. doi:10.1155/2022/9468040

128. Bradley EW, Carpio LR, McGee-Lawrence ME, et al. Phlpp1 facilitates post-traumatic osteoarthritis and is induced by inflammation and promoter demethylation in human osteoarthritis. Osteoarthritis Cartilage. 2016;24(6):1021–1028. doi:10.1016/j.joca.2015.12.014

129. Castillejo Becerra CM, Mattson AM, Molstad DHH, Lorang IM, Westendorf JJ, Bradley EW. DNA methylation and FoxO3a regulate PHLPP1 expression in chondrocytes. J Cell Biochem. 2018;119(9):7470–7478. doi:10.1002/jcb.27056

130. Matsuzaki T, Alvarez-Garcia O, Mokuda S, et al. FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis. Sci Transl Med. 2018;10(428). doi:10.1126/scitranslmed.aan0746

131. Michigami T. Regulatory mechanisms for the development of growth plate cartilage. Cell Mol Life Sci. 2013;70(22):4213–4221. doi:10.1007/s00018-013-1346-9

132. Eelen G, Verlinden L, Maes C, et al. Forkhead box O transcription factors in chondrocytes regulate endochondral bone formation. J Steroid Biochem Mol Biol. 2016;164:337–343. doi:10.1016/j.jsbmb.2015.07.015

133. Han MS, Jung YK, Kim GW, Han S. Transglutaminase-2 regulates Wnt and FoxO3a signaling to determine the severity of osteoarthritis. Sci Rep. 2020;10(1):13228. doi:10.1038/s41598-020-70115-w

134. Yu H, Fellows A, Foote K, et al. FOXO3a (Forkhead Transcription Factor O Subfamily Member 3a) links vascular smooth muscle cell apoptosis, matrix breakdown, atherosclerosis, and vascular remodeling through a novel pathway involving MMP13 (Matrix Metalloproteinase 13). Arterioscler Thromb Vasc Biol. 2018;38(3):555–565. doi:10.1161/ATVBAHA.117.310502

135. Zheng Y, Wei K, Jiang P, et al. Macrophage polarization in rheumatoid arthritis: signaling pathways, metabolic reprogramming, and crosstalk with synovial fibroblasts. Front Immunol. 2024;15:1394108. doi:10.3389/fimmu.2024.1394108

136. Peng SL. Interactions of Fox proteins with inflammatory transcription-factor pathways. Expert Rev Clin Immunol. 2006;2(6):869–876. doi:10.1586/1744666X.2.6.869

137. Kuo CC, Lin SC. Altered FOXO1 transcript levels in peripheral blood mononuclear cells of systemic lupus erythematosus and rheumatoid arthritis patients. Mol Med. 2007;13(11–12):561–566. doi:10.2119/2007-00021.Kuo

138. Ludikhuize J, de Launay D, Groot D, et al. Inhibition of forkhead box class O family member transcription factors in rheumatoid synovial tissue. Arthritis Rheum. 2007;56(7):2180–2191. doi:10.1002/art.22653

139. Lin J, Zhou Z, Huo R, et al. Cyr61 induces IL-6 production by fibroblast-like synoviocytes promoting Th17 differentiation in rheumatoid arthritis. J Immunol. 2012;188(11):5776–5784. doi:10.4049/jimmunol.1103201

140. Lin L, Hron JD, Peng SL. Regulation of NF-kappaB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a. Immunity. 2004;21(2):203–213. doi:10.1016/j.immuni.2004.06.016

141. Jonsson H, Allen P, Peng SL. Inflammatory arthritis requires Foxo3a to prevent Fas ligand-induced neutrophil apoptosis. Nat Med. 2005;11(6):666–671. doi:10.1038/nm1248

142. Wei Y, Zhang S, Shao F, Sun Y. Ankylosing spondylitis: from pathogenesis to therapy. Int Immunopharmacol. 2025;145:113709. doi:10.1016/j.intimp.2024.113709

143. Dean LE, Jones GT, MacDonald AG, Downham C, Sturrock RD, Macfarlane GJ. Global prevalence of ankylosing spondylitis. Rheumatology. 2014;53(4):650–657. doi:10.1093/rheumatology/ket387

144. Chen Y, Yang H, Xu S, et al. Association analysis of B7-H3 and B7-H4 gene single nucleotide polymorphisms in susceptibility to ankylosing spondylitis in eastern Chinese Han population. Int J Immunogenet. 2021;48(6):500–509. doi:10.1111/iji.12559

145. Xu S, Pan Z, Huang L, et al. Association of FOXO3a gene polymorphisms and ankylosing spondylitis susceptibility in Eastern Chinese Han population. Gene. 2021;800:145832. doi:10.1016/j.gene.2021.145832

146. Xu S, Zhang X, Wang X, et al. DNA methylation and transcription of the FOXO3a gene are associated with ankylosing spondylitis. Clin Exp Med. 2023;23(2):483–493. doi:10.1007/s10238-022-00831-2

147. Shi S, Ou X, Liu C, Li R, Zheng Q, Hu L. Nanotechnology-enhanced pharmacotherapy for intervertebral disc degeneration treatment. Int J Nanomed. 2024;19:14043–14058. doi:10.2147/IJN.S500364

148. Zhang F, Zhao X, Shen H, Zhang C. Molecular mechanisms of cell death in intervertebral disc degeneration. Int J Mol Med. 2016;37(6):1439–1448. doi:10.3892/ijmm.2016.2573

149. Lambertini E, Penolazzi L, Pellielo G, et al. Pro-osteogenic properties of violina pumpkin (Cucurbita moschata) leaf extracts: data from in vitro human primary cell cultures. Nutrients. 2021;13(8):2633. doi:10.3390/nu13082633

150. Lambertini E, Penolazzi L, Notarangelo MP, et al. Pro‑differentiating compounds for human intervertebral disc cells are present in Violina pumpkin leaf extracts. Int J Mol Med. 2023;51(5). doi:10.3892/ijmm.2023.5242

151. Wang B, Huang X, Lin J. Serum COX-2 and FOXO3a in patients with rheumatoid arthritis and correlation with disease activity. Exp Ther Med. 2020;20(2):910–916. doi:10.3892/etm.2020.8779

152. Lee JC, Espéli M, Anderson CA, et al. Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell. 2013;155(1):57–69. doi:10.1016/j.cell.2013.08.034

153. van Steenbergen HW, Rantapää-Dahlqvist S, van Nies JA, et al. Does a genetic variant in FOXO3A predict a milder course of rheumatoid arthritis? Arthritis Rheumatol. 2014;66(6):1678–1681. doi:10.1002/art.38405

154. Roncada T, Blunn G, Roldo M. Collagen and alginate hydrogels support chondrocytes redifferentiation in vitro without supplementation of exogenous growth factors. ACS Omega. 2024;9(19):21388–21400. doi:10.1021/acsomega.4c01675

155. Yuan S, Zhang L, Ji L, et al. FoxO3a cooperates with RUNX1 to promote chondrogenesis and terminal hypertrophic of the chondrogenic progenitor cells. Biochem Biophys Res Commun. 2022;589:41–47. doi:10.1016/j.bbrc.2021.12.008

156. Shang L, Li X, Ding X, et al. S-Adenosyl-l-Methionine alleviates the senescence of MSCs through the PI3K/AKT/FOXO3a signaling pathway. Stem Cells. 2024;42(5):475–490. doi:10.1093/stmcls/sxae010

157. Lee S, Le NH, Kang D. Melatonin alleviates oxidative stress-inhibited osteogenesis of human bone marrow-derived mesenchymal stem cells through AMPK activation. Int J Med Sci. 2018;15(10):1083–1091. doi:10.7150/ijms.26314

158. Elseweidy MM, El-Swefy SE, Shaheen MA, Baraka NM, Hammad SK. Effect of resveratrol and mesenchymal stem cell monotherapy and combined treatment in management of osteoporosis in ovariectomized rats: role of SIRT1/FOXO3a and Wnt/β-catenin pathways. Arch Biochem Biophys. 2021;703:108856. doi:10.1016/j.abb.2021.108856

159. Li Y, Shen G, Yu C, et al. Angiotensin II induces mitochondrial oxidative stress and mtDNA damage in osteoblasts by inhibiting SIRT1–FoxO3a–MnSOD pathway. Biochem Biophys Res Commun. 2014;455(1–2):113–118. doi:10.1016/j.bbrc.2014.10.123

160. Zhang J, Li S, Wei L, et al. Protective effects of 2,3,5,4-tetrahydroxystilbene-2-o-β-D-glucoside against osteoporosis: current knowledge and proposed mechanisms. Int J Rheum Dis. 2018;21(8):1504–1513. doi:10.1111/1756-185X.13357

161. Yang Y, Su Y, Wang D, et al. Tanshinol attenuates the deleterious effects of oxidative stress on osteoblastic differentiation via Wnt/FoxO3a signaling. Oxid Med Cell Longev. 2013;2013:351895. doi:10.1155/2013/351895

162. Yu Y, Shen X, Luo Z, et al. Osteogenesis potential of different titania nanotubes in oxidative stress microenvironment. Biomaterials. 2018;167:44–57. doi:10.1016/j.biomaterials.2018.03.024

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.