Skip to main content

Type 2 diabetic mellitus related osteoporosis: focusing on ferroptosis

Abstract

With the aging global population, type 2 diabetes mellitus (T2DM) and osteoporosis(OP) are becoming increasingly prevalent. Diabetic osteoporosis (DOP) is a metabolic bone disorder characterized by abnormal bone tissue structure and reduced bone strength in patients with diabetes. Studies have revealed a close association among diabetes, increased fracture risk, and disturbances in iron metabolism. This review explores the concept of ferroptosis, a non-apoptotic cell death process dependent on intracellular iron, focusing on its role in DOP. Iron-dependent lipid peroxidation, particularly impacting pancreatic β-cells, osteoblasts (OBs) and osteoclasts (OCs), contributes to DOP. The intricate interplay between iron dysregulation, which comprises deficiency and overload, and DOP has been discussed, emphasizing how excessive iron accumulation triggers ferroptosis in DOP. This concise overview highlights the need to understand the complex relationship between T2DM and OP, particularly ferroptosis. This review aimed to elucidate the pathogenesis of ferroptosis in DOP and provide a prospective for future research targeting interventions in the field of ferroptosis.

Introduction

As the global population ages, two common conditions, namely diabetes mellitus (DM) and osteoporosis (OP), become increasingly prevalent. The 10th edition of the IDF Diabetes Atlas estimates that the prevalence of diabetes in the global population aged 20–79 years is estimated to be 10.5% (536.6 million) by the 2021, and will rise to 12.2% (783.2 million) by 2045 [1]. In China, approximately 30% of the elderly population (aged 60 years and older) had DM in 2020, with over 95% being T2DM [2]. Furthermore, the WHO estimated that more than 200 million women worldwide have OP [3]. In China, an epidemiological survey on OP revealed a prevalence rate of 19.2% among people aged 50 or above, with 32.1% in females and 6.9% in males. In those aged ≥ 65 years, the prevalence rate of OP was 32%, with 51.6% in females and 10.7% in males [4]. Globally, OP causes more than 8.9 million fractures annually, resulting in an osteoporotic fracture every 3 s [5]. One in three women aged > 50 years will experience osteoporotic fractures, as will one in five men aged > 50 years [6]. Osteoporotic fractures (fragility fractures) often lead to severe complications due to minor trauma (equivalent to falling from standing height or lower) [7].

Diabetic osteoporosis (DOP) is a systemic chronic metabolic bone disease characterized by abnormal bone tissue structure and reduced bone strength in patients [8]. Over 90,000 osteoporotic fractures occur globally each year, most of which are associated with DOP and pose a significant threat to human health and socioeconomic factors [5]. As glucolipid homeostasis is disrupted in DM, the unique diabetic microenvironment is characterized by abnormally increased levels of extracellular metabolites. It is well known that OP is one of the common complications of DM. According to related data, approximately 50–66% of patients with DM have decreased bone mineral density (BMD), and approximately 33% of them have been diagnosed with DOP. Over the past decade, extensive investigations of the diabetic microenvironment and mineral homeostasis have identified increased cortical porosity, imbalanced bone metabolism, and distorted bone microarchitecture as the three main characteristics of DOP [910]. Current studies indicate that DM, whether type I or type II, is a contributing factor to increased fracture risk [11], therefore, it is crucial to pay special attention to DOP, particularly in a large population of patients with T2DM. Studies have shown that ferroptosis plays an important role in the development and progression of T2DM and complications [12]. To identify a potential treatment approach and its clinical applications for this larger colony type 2 Diabetic osteoporosis (T2DOP). Therefore, there is a need to further elucidate the relationship between T2DM and OP by focusing on ferroptosis [13].

Ferroptosis is a form of apoptosis that relies on intracellular iron [14]. It is characterized by iron-dependent lipid peroxidation. The depletion of cellular glutathione (GSH) and reduced activity of glutathione peroxidase 4 (GPX4) results in the inability of GPX4 to catalyze the reduction of lipid peroxides in ferroptosis. This leads to the Fe2+ oxidation of lipids through the Fenton reaction, causing the accumulation of reactive oxygen species (ROS) and ultimately promoting cell ferroptosis [15]. Iron deposition in pancreatic β-cells accelerating diabetes progression [16]. Iron-related oxidative processes and ROS accumulation in pancreatic β-cells are closely associated with insulin resistance and insufficient insulin secretion, leading to the pathogenesis of T2DM [17, 18]. Disturbances in iron metabolism, including iron deficiency and overload, are also strongly associated with OP [19, 20]. Excessive iron accumulation mediated by NADPH oxidase 4 (NOX4) can cause iron metabolism in osteoblasts (OBs), leading to OP [21]. DOP progression is accompanied by impaired glucolipid homeostasis and elevated plasma glucolipid metabolites; hence, ferroptosis may play a vital role in DOP pathogenesis [13]. In this review, we comprehensively discuss the research progress of ferroptosis in T2DOP.

Ferroptosis

Ferroptosis is a novel iron-dependent form of programmed cell death (PCD), that is distinct from apoptosis, necrosis, and autophagy [14]. Morphologically, ferroptosis exhibits the following characteristics: (1) cells appear smaller and more rounded, with increased intercellular spacing; (2) the cell membrane remains intact, but there is cytoplasmic hemorrhage and no nuclear condensation; and (3) the number of mitochondria is reduced, with reduced or absent cristae, increased mitochondrial membrane density, and a ruptured outer mitochondrial membrane. Biological features include decreased cystine uptake, decreased GSH consumption, inhibition of cystine/glutamate reverse transport system (Xc system) activity, and abnormal accumulation of iron ions and ROS [22]. The most common lethal factors triggering ferroptosis are intracellular iron overload and excessive lipid peroxidation [23, 24]. And there is a simple map that makes ferroptosis easy to understand (shown in Fig. 1).

Fig. 1
figure 1

Map of ferroptosis

Iron overload is a critical factor for ferroptosis. Excess intracellular iron is exported by ferroportin (FPN) to maintain intracellular iron homeostasis [15]. Overloaded Fe2+ interacts with H2O2 via the Fenton reaction, leading to lipid peroxidation of polyunsaturated fatty acids (PUFAs) ultimately inducing ferroptosis [25]. Additionaly, resarch suggests that excessive autophagy and lysosomal activity promote ferroptosis through iron accumulation and lipid peroxidation. Ferritin (FTH) is a complex of ferritin light chain (FTL) and ferritin heavy chain 1 (FTH1). The degradation of FTH is dependent on autophagy and is termed ferritinophagy [26]. The Fe2 + stored in FTH can also be released into the cytoplasm. Nuclear receptor coactivator 4 (NCOA4) is a selective autophagy receptor that binds to FTH1 and mediates the delivery of intracellular ferritin to autophagosomes, ultimately releasing free iron [23]. Bellellelli et al. have proposed that iron overload initiates iron autophagy, which ultimately triggers ferroptosis [27]. Therefore, NCOA4 levels are key determinants of FTH engulfment flux, which is closely relatedd to ferroptosis.

Lipid peroxidation is thought to be a major factor in ferroptosis beacase lipid peroxides can destabilize cell membranes, thereby promoting membrane rupture [28]. Studies have shown that PUFAs, particularly arachidonic acid (AA) and adrenergic acid (AdA), are highly sensitive to lipid peroxidation and readily react with ROS, leading to lipid peroxidation and ultimately triggering ferroptosis [29]. Ferroptosis cells accumulate oxidized phospholipids, particularly phosphatidylinositol (PL) [30]. Phosphatidylethanolamine (PE), containing AA or AdA, is a critical PL that induces ferroptosis. Excessive ROS damage cellular components, including DNA, because lipid peroxidation directly damages phospholipids in cell and organelle membranes [31]. Acyl-CoA synthetase long-chain family member 4 (ACSL4) is a pivotal enzyme in lipid metabolism. ACSL4 catalyzes the synthesis of long-chain polyunsaturated-CoAs with a preference for AA and AdA, thus facilitating their esterification into phospholipids. PUFA-CoA is then transformed into phospholipid PUFAs (PUFA-PL) through the action of lysophosphatidylcholine acyltransferase 3 (LPCAT3) [32] and subsequently undergoes oxidation by lipoxygenases (LOXs) to produce lipid peroxides (LOOH), ultimately inducing cell ferroptosis [23]. Furthermore, the sensitivity of PUFA to lipid peroxidation is associated with the presence of highly reactive hydrogen atoms in the methylene bridges. Hydroxyl radicals interact directly with PUFA in phospholipid membranes, influencing the formation of lipid peroxides and triggering ferroptosis [33]. Products of lipid peroxidation, such as nucleic acids and proteins, as well as derivatives like 4-Hydroxynonenal (4-HNE) and malondialdehyde (MDA), can damage cells. These derivatives can be used as markers for ferroptosis and lipid peroxidation [34]. Nicotinamide adenine dinucleotide phosphate (NADPH) - cytochrome P450 (CYP450) reductase transfers electrons from NADPH to oxygen, resulting in hydrogen peroxide (H2O2) production, which then reacts with Fe. This generates hydroxyl radicals that disrupt the integrity of cell membranes by causing peroxidation of PUFA chains in membrane phospholipids [35]. Additionally, ferroptosis promotes LOX catalysis, leading to the generation of lipid peroxides in cell membrane phospholipids, which further enhances ferroptosis. FINO2 oxidizes ferrous iron to generate ROS and lipids to produce lipid peroxides [36].

GPX4 is a glutathione-related peroxidase enzyme [37] closely associated with ferroptosis. GSH is a tripeptide synthesized from cysteine, glutamic acid, and glycine [38]. With the catalytic action of selenium-containing cysteine (the active site of GPX4), GSH can reduce LOOH to harmless lipid alcohols [39]. Therefore, selenium deficiency in the serum or cytoplasm may impair GPX4 activity. The depletion of intracellular GSH and decreased GPX4 activity prevent the metabolic reduction of lipid peroxides catalyzed by GPX4, leading to the accumulation of ROS and ultimately promoting ferroptosis [15]. Studies have shown that RAS-selective lethal small molecule 3 (RSL3) contains a chloroacetamide moiety that reacts with nucleophilic amino acid residues on GPX4, resulting in GPX4 inactivation. Thus, RSL3 induces ferroptosis [40]. Ferroptosis inducing (FIN56) depletes coenzyme Q10 (CoQ10), degradates GPX4 and induces cellular ferroptosis, whereas 1,2-dioxin cycloalkane (FINO2) leads to iron oxidation and loss of GPX4 enzyme activity. FIN56 interferes with the mevalonate pathway. It affects the homeostasis of selenium proteins and other antioxidant molecules, which are essential for balancing OCs and OBs [41,42,43].

Additionally, the inhibition of the Xc system can further promote ferroptosis. The Xc system mediates a 1:1 exchange of intracellular glutamate for extracellular cysteine, reducing cysteine inside the cell to synthesize the antioxidant GSH [44]. Small-molecule compounds such as erastin reduce the synthesis of cysteine-dependent reduced GSH by inhibiting the Xc system, leading to intracellular GSH depletion, thereby causing the accumulation of a large number of peroxides and triggering cell ferroptosis. SLC7A11 is a light-chain subunit and a reverse transport protein that comprises the Xc system [45]. The SLC7A11 gene’s flanking region contains a site that perfectly matches the binding sequence of P53 which is a critical tumor suppressor gene. Thus, SLC7A11 is a downstream target of P53’s downstream targets [4647]. P53 activation significantly reduces the expression of SLC7A11, which inhibits the Xc system and promotes ferroptosis [46]. Furthermore, P53 enhances the expression of metabolism-related genes, including spermidine/spermine N1-acetyltransferase 1 (SAT1), a transcriptional target of P53, leading to lipid peroxidation [48]. Research has revealed that multiple antioxidant systems can counteract ferroptosis [49].

(1) Iron responsive element binding protein 2 (IREB2) is an RNA-binding protein that regulates iron homeostasis. It significantly increases the expression of iron metabolism-related genes such as FTH1 and FTL, thereby inhibiting ferroptosis [28].

(2) P53 plays a dual role in regulating ferroptosis [47]. When P53 binds to dipeptidylpeptidase-4 (DPP4) in the nucleus, it inhibits NADPH oxidase and reduces ROS levels, thereby inhibiting ferroptosis [50].

(3) Nuclear factor E2-related factor 2 (Nrf2) is a vital transcription factor that regulates the transcription of antioxidant enzymes [5152]. Under physiological conditions, Nrf2 interacts with Kelch-like ECH-associated protein 1 (Keap1) in the cytoplasm. Keap1 mediates Nrf2 ubiquitination and degradation, maintaining low levels of Nrf2f [53]. Nrf2 activity is tightly regulated by Keap1, a ubiquitin ligase adaptor protein for Cullin3-based E3 ligase. It isolates Nrf2 frpm the cytoplasm, preventing Nrf2 from entering the nucleus, and exerts its transcriptional activity to coordinate the expression of cell-protective genes, thereby maintaining cellular homeostasis. Even when a small amount of free Nrf2 moves to the nucleus, it binds to antioxidant response elements (AREs) and mediates the expression of basal ARE-dependent genes to maintain intracellular homeostasis [5455]. Nrf2 regulates downstream gene transcription and prevents lipid peroxidation and ferroptosis by inducing the synthesis of genes such as SLC7A11 [56]. Keap1-Nrf2 prevents ferroptosis by upregulating the heme oxygenase-1 (HO-1) and FTH1 pathways [57].

(4) Activating transcription factor 4 (ATF4) promotes the transcription of SLC7A11 by binding to amino acid reaction element (AARE) in the SLC7A11 promoter region [58]. ATF4 can inhibit ferroptosis by activating SLC7A11 expression [59].

(5) Dihydroorotate dehydrogenase (DHODH) is a flavin-dependent enzyme located in the mitochondrial inner membrane, catalyzing the fourth step of pyrimidine nucleotide synthesis [60]. DHODH suppresses mitochondrial iron apoptosis by regulating the production of the coenzyme QH2 (CoQH2) in the inner mitochondrial membrane [61].

(6) Mitochondrial ferritin (FtMt) stores iron in mitochondria and protects them from iron-induced oxidative stress [62].

(7) Ferroptosis inhibitory protein 1 (FSP1) is an NADPH-dependent redox enzyme with DNA-binding activity [63]. FSP1 reduces CoQ10 to coenzyme Q10H2 (CoQ10H2) via its redox enzyme activity. CoQ10H2 is a lipophilic, free radical-scavenging antioxidant (RTA). The FSP1-CoQ-NAD(P)H axis inhibits ferroptosis by catalyzing the continuous regeneration of CoQ10 [64].

(8) The mevalonate pathway is a protein isoprenylationpathway. Mevalonate regulates the maturation of selenocysteine-specific tRNA (Trsp), which is required for the synthesis of selenium enzymes, including GPX4. Inhibition of the mevalonate pathway downregulates Trsp and GPX4 synthesis [65]. Recent research has indicated that FSP1 has an antiferroptotic mechanism parallel to the cysteine-GSH-GPX4 axis. FSP1 reduces CoQ10 to decrease lipid radicals, inhibit lipid peroxidation and subsequently ferroptosis [66].

(9) Additionally, lipid peroxidation can be inhibited by ferrostatin-1 (Fer-1), liproxtatin-1 (Lip-1), and vitamin E, which are free radical scavengers that can reduce lipid peroxidation and effectively block ferroptosis [6768].

Ferroptosis in OP

OP is a systemic disorder of bone metabolism characterized by reduced bone mass and structural damage, resulting in increased bone fragility and susceptibility to fracture [69]. The pathophysiology of OP involves a dynamic imbalance between bone resorption and formation, resulting in reduced bone mass and the destruction of trabecular bone, which are closely linked to OBs and Osteoclasts (OCs) [70]. Studies have reported that disturbances in iron metabolism, encompassing both iron deficiency and iron overload, may result in a dynamic imbalance between bone resorption and formation, contributing to OP [20, 71]. Excess Fe is considered toxic in animal models. Mice with iron overload exhibit increased oxidative stress, characterized by elevated ROS levels, and develop OP [72]. Some clinical studies have suggested that appropriate iron chelation therapy can prevent OP [73]. Maintaining iron homeostasis and reducing the levels of ROS can effectively prevent OP. Substantial evidence suggests that the oxidative stress induced by iron overload contributes to the pathophysiology of OP [44]. Consequently, the mechanisms through which iron overload leads to osteoporotic ferroptosis have received increasing attention (shown in Fig. 2).

Fig. 2
figure 2

Osteoporotic ferroptosis

Ferroptosis in BMSCs

The source of OBs is the bone marrow mesenchymal stem cells (BMSCs), which exert their regulatory functions through remodeling. Iron overload-mediated oxidative stress, which increases ROS levels, is toxic to BMSCs, impairs mitochondrial dysfunction, and reduces their mineralization and osteogenic differentiation ability of BMSCs [74].

Nrf2/Keap1 pathway has been found to be involved in the oxidative stress and inflammatory response. Erastin-induced ferroptosis causes an increase in Nrf2 and a decrease in Keap1 expression in BMSCs, with increased expression of TFR1/FPN1 and decreased expression of GPX4 and SLC7A11. Engeletin (dihydrokaempferol 3-rhamnoside), an effective endogenous antioxidant, inhibits erastin-induced ferroptosis in BMSCs via the Nrf2/Keap1 pathway [75].

The NOP2/Sun RNA methyltransferase 5 (NSUN5) -FTH1/FTL pathway could be a causative event leading to ferroptosis of BMSCs. NSUN5 Recruits TRAP1 to Modify FTH1 and FTL. NSUN5 is downregulated in erastin-induced BMSC ferroptosis by increasing FTH1 and FTL levels, which inhibit ferroptosis [76].

Ferroptosis in OBs

OBs are derived from BMSCs, and their bone formation plays an important role in maintaining the stability of bone structure and balance of bone metabolism [77]. Iron overload can affectt the OBs in various ways.

Iron overload has an inhibitory effect on the differentiation of OBs

(1) Iron overload suppressed OB differentiation. Runt-related transcription factor 2 (Runx2) is a necessary transcription factor for OB differentiation and maturation [78]. Iron overload can downregulate the expression of Runx2 and its targets, such as alkaline phosphatase and osteocalcin, in BMSCs and osteoprogenitor cells, thereby inhibiting OB formation [79].

(2) Additionally, the use of deferoxamine, an iron chelator, or superparamagnetic iron oxide nanoparticles with anti-osteogenic properties, reversed the inhibitory effect of free iron on BMSC differentiation into OBs [80].

Iron overload can affect the activity and function of bone cells

(1) Elevated NOX4 levels have been observed in the osteoporotic mouse and human bone tissues. Iron can directly interact with iron regulatory protein 1 (IRP1) in OBs, leading to IRP1 separation from iron-responsive element (IRE)-like sequences within the NOX4 gene. This promotes the expression of NOX4, resulting in increased ROS production when NADPH is used as a substrate. Accumulation of lipid peroxides causes mitochondrial dysfunction and promotes iron-dependent cell death in OBs [21].

(2) Protection through the Nrf2/HO-1 pathway: Activation of the NRF2/HO-1 pathway can significantly reduce ferritin levels. NRF2 activation initiates cellular defense mechanisms by increasing the expression of antioxidant enzymes, including GPX and superoxide dismutase(SOD). The toxic effects of iron overload on OBs are reduced by reducing oxidative stress, thereby inhibiting ferroptosis and promoting bone growth [81,82,83].

(3) The redox homeostasis of MC3T3-E1 mouse embryonic osteoblasts can be disrupted by iron overload, as manifested by excess intercellular ROS, lipid peroxidation, increased MDA, and decreased SOD and GSH, whereas Fer-1 improves the antioxidant homeostasis of the above peroxides. Iron overload inhibits the protein expression of GPX4, and haem oxygenase 1 (HMOX1), and increases the protein expression of FTH. Iron overload inhibits the osteogenic differentiation and mineralization of MC3T3-E1 cells [84]. Furthermore, iron overload impaired the viability of MC3T3-E1 pre-OB cells and induce apoptosis [85].

(4) Ammonium ferric citrate (FAC) significantly suppresses OBs function, inhibit ferroptosis, and activate the ASK1-p38 pathway, which is associated with anti-osteogenic effects [86]. Ferroptosis in OBs under high glucose/high fat conditions may be associated with the methyltransferase-like 3 (METTL3)/ASK1-p38 pathway. In an OB model, treatment of pre-OB cells (MC3T3-E1) with high glucose and palmitic acid (HGPA) will not only inhibit OBs differentiation and mineralisation, but also induce OBs ferroptosis [87]. Iron overload disrupt OBs activity and function by inducing ferroptosis via the ASK1/p38 pathway.

(5) Abnormalities in iron metabolism can lead to the accumulation of highly reductive free Fe2+ in OBs, causing oxidative stress. Excess Fe2+ ions can generate significant amounts of hydroxyl radicals and other ROS via Fenton reactions. ROS can stimulate the release of cytochrome C from mitochondria and activate apoptotic proteins such as caspase3, ultimately leading to apoptosis in OBs. ROS scavengers, such as N-acetylcysteine (NAC) can restore OBs activity [8889].

(6) Mitochondrial ferritin (FtMt) is a protein that stores ferrous ions in cellular mitochondria. FtMt inhibits the onset of ferroptosis in OBs by reducing oxidative stress induced by excess ferrous ions [63]. FtMt deficiency induces mitochondrial autophagy by activating the ROS/PINK1/Parkin pathway, resulting in the release of more free Fe2 + from the mitochondria, which also occurs through the Fenton reaction and causes OBs ferroptosis [25].

Some studies have shown that inhibiting ferroptosis in OBs through the Nrf2/GPX4 pathway can improve bone formation capacity and microstructure. Vitamin D receptor (VDR) activators (1,25(OH)2D3) suppress ferroptosis in OBs by activating the Nrf2/GPX4 pathway [90]. Melatonin can improve bone microstructure both in vivo and in vitro by inhibiting ferroptosis in OBs. Melatonin reduces ROS levels, increases SLC7A11 levels, and enhances GPX4 activity by opening the NRF2/HO-1 antioxidant pathway, thereby reducing the toxicity of lipid peroxides to protect the biomembrane system and inhibit ferroptosis in OBs [81].

Ferroptosis in OCs

OCs are terminally differentiated multinucleated cells of the monocyte/macrophage lineage with the unique function of resorbing bone matrix [91]. OCs play an important role in bone development and remodeling. The inhibition of bone resorption by OCs is essential for promoting bone matrix formation and bone remodeling [92]. Iron overload may also affecte OCs activity and funtion of OCs. Evidence suggests that FTN autophagy occurs when cells are iron deficient, making them more susceptible to intracellular Fe2+-induced ferroptosis [62, 93]. In addition, mature OCs require more cytoplasmic iron than the other cells. Therefore, OCs are more susceptible to ferroptosis [94]. OC formation and differentiation are associated with significant changes in the cellular iron homeostasis [9596].

(1) A recent study showed that the lack of FPN in myeloid cells leads to iron accumulation and stimulates OC differentiation in mice. Iron overload induces ROS production and promotes OC differentiation [97].

(2) Macrophage-colony stimulating factor (M-CSF) and the receptor activator of nuclear factor κB ligand (RANKL) drive differentiation of mononuclear macrophage lineage cells and BMSCs, which later fuse to form multinucleated cells and further differentiate into mature OCs [98]. Iron overload significantly increases RANKL expression in MLO-Y4 cells (a mouse bone-like cell line), disrupted the RANKL/osteoprotegerin (OPG) balance, and promotes OC differentiation and bone resorption. Iron overload also induces apoptosis in MLO-Y4 and primary bone cells [99].

(3) ROS induced by iron overload has been reported to activate the mitogen-activated protein kinase (MAPK) pathway, which promotes osteoblast differentiation in bone metabolism and resorption [100]. Iron overload induces ROS accumulation, which activates the intracellular MAPK/NrκB/nucleotide-binding oligomerized structural domains, leucine-rich repeat sequences, and pyrin structural domains of 3 (NLRP3) signaling pathway, leading to OCs-mediated bone loss in OP [100].

Studies have shown that OP can be ameliorated by inhibiting the OCs. Artesunate (ARS) compounds inhibit iron uptake-stimulated OCs differentiation by inhibiting iron uptake, leading to ferroptosis [71]. ARS compounds inhibit ROS production and suppress ferroptosis during OCs differentiation by activating p62/Nrf2 signaling [100]. Andrographolide modulates OPG/RANKL signaling and inhibits the TNFα-activated NFκB pathway, which accelerates OBs differentiation. It may also interfere with extracellular signal-regulated kinase (ERK)/MAPK and NF-κB signaling to block RANKL-induced OCs differentiation [101]. Zoledronic acid inhibits FBXO9 (F-box protein 9) expression and increases OCs ferroptosis by triggering FBXO9-mediated ubiquitination and P53 degradation, thereby decreasing p53 protein stability [102].

Ferroptosis in T2DM

The relationship between DM and ferroptosis is significant. T2DM is characterized by impaired pancreatic β-cell function and peripheral insulin resistance [103]. T2DM is characterized by the progressive loss of insulin secretion in the context of insulin resistance. Studies have shown that individuals with DM often exhibit imbalances in iron metabolism and increased serum circulating iron and FTN levels compared with healthy individuals [104105]. Iron overload is considered a contributing factor to the worsening of T2DM [106].

Adipocyte metabolism

This study showed that obesity-associated T2DM entails insulin resistance and loss of β-cell mass. Adipocyte mitochondrial dysfunction increases cell mass during obesity-related insulin resistance. And mitochondrial dysfunction in the adipose tissue is a key determinant of the etiology of type 2 diabetes [107]. Iron, an important regulator of energy metabolism, is primarily found in adipose tissue. Iron deficiency and iron overload are considered important causes of chronic metabolic diseases (such as T2DM or obesity) [108] Studies have shown that mice fed with rich iron diets showed upregulation of IR-related adipokines because an iron-enriched diet induces insulin resistance and hypertriglyceridemia and affects visceral adipose tissue metabolism by a mechanism involving hepcidin up-regulation [109]. FTMT is a mitochondrial matrix protein that chelates iron. Induction of FtMT in adipocytes promotes local ROS damage during obesity. It activates the mitochondrial stress response in adipocytes [108]. This response may induce ferroptosis during adipocyte metabolism, aggravating IR. In obesity, abnormal adipocyte metabolism is related to IR owing to adipocyte mitochondrial dysfunction. However, the association between adipocyte metabolism, ferroptosis, and DOP remains unclear. Obesity may be the most relevant factor affecting adipocyte metabolism.

Liver metabolism

The relationship between iron metabolism, T2DM, and liver disease is complex. Iron main store in hepatocytes which play a dual role in iron metabolism [110]. In addition, hepatocytes secrete an iron regulatory hormone (hepcidin). Insufficient hepcidin expression results in iron overload, which triggers ROS synthesis which may have a relationship between ferroptosis pathogenesis of β cell exhaustion and IR-mediated T2DM [17, 111]. The end-stage liver disease would lead to the massive liver iron overload when the hepcidin-producing liver mass are losing [112]. Meanwhile iron overload may affect the development of liver fibrosis by aggravating lipid peroxidation [113]. Ferroptosis is involved in hepatic fibrosis and leads to liver injury in db/db mice, which may be attributed to increased ROS and lipid peroxidation, as well as iron deposition. Suppression of ferroptosis in hepatocytes by the Nrf2/HO-1 pathway. Moreover, liraglutide, widely used to treat obesity and T2DM, can prevent the occurrence of oxidative stress, iron overload and ferroptosis by activating Nrf2/HO-1, thereby improving liver fibrosis [114115]. Recent studies have revealed an increased level of ferroptosis in hepatic cells during the progression of nonalcoholic fatty liver disease (NAFLD). NAFLD is closely associated with insulin resistance T2DM. And metformin can modulate the xCT/GPX4/ACSL4 axis to alleviate hepatic cell ferroptosis in T2DM-related NAFLD [116]. But the connect of ferroptosis, DOP and liver disease was still to find out. Maybe serious liver disease lead massive liver iron overload that cause ferroptosis of β cell exhaustion and IR-mediated T2DM which would affect DOP [117].

Ferroptosis in β-cells

Ferroptosis is associated with insulin secretion dysfunction in pancreatic β-cells. The function of pancreatic islets can be impaired by proferroptotic factors even before β-cells die [118]. Iron plays a crucial role in pancreatic β-cells insulin secretion. Iron intake is mainly derived from the diet [119]. Iron is taken up by the cell’s divalent metal transporter 1 (DMT-1) in the form of free Fe2+ and the transferrin receptor (TFR) is the main carrier of iron into the cell, binding to iron to form a trivalent iron complex (Tf-Fe3+), which is then transported into tissues [28], and Tf-Fe3+ is taken up by pancreatic β-cells via DMT-1 [120]. Fe2+ is retained in the labile iron pool (LIP) where it is sequestered by ferritin, a unique cytoplasmic iron storage protein [121]. Fe2+ bound to FTN to synthesize iron-dependent proteins in the cytoplasm or mitochondria [122]. Extracellularly, pancreatic β-cells release hepcidin, which induces the internalization of binding transferrin [123124]. Studies have shown that hepcidin improves pancreatic β-cells function by alleviating mitochondrial iron overload [125]. Tf mediates a positive feedback mechanism for iron regulation during glucose-stimulated insulin secretion [126]. Therefore, pancreatic β-cells play an important role in controlling iron homeostasis by avoiding excessive free Fe2+.

Ferroptosis is the process of non-apoptotic cell death due to excessive iron uptake associated with a reduction in mitochondrial volume, and is characterized by iron accumulation, lipid peroxidation, and reduced GPX4 expression [23]. For example, the pathogenesis of hereditary hemochromatosis (HH) involves iron deposition in pancreatic β-cells, leading to pancreatic β-cell death and consequent high glucose (HG) conditon, even DM [127]. As pancreatic β-cells express low levels of antioxidant enzymes such as SOD, GSH peroxidase, and catalase [128], pancreatic β-cells are susceptible to oxidative stress, have relatively low antioxidant capacity, and are prone to ferroptosis. Studies have shown that ferroptosis can be induced in rat’s pancreatic β-cells by silencing GPX4 and exposure to tert-butyl hydroperoxide (tert-BHP). It means GPX4 or Fer-1 effectively attenuates tert-BHP-induced pancreatic β-cell death, and thus, GPX4 is essential for pancreatic β-cell survival [129]. Erastin and RSL3 induce pancreatic β-cell ferroptosis, leading to impaired islet function, whereas Fer-1 restores impaired glucose-stimulated insulin secretion (GSIS) [130]. Cryptochlorohydric acid (CCA) exerts potent antidiabetic effects by activating the cystine/Xc-system/GPX4/Nrf2 pathway and inhibiting NCOA4 to suppress ferroptosis [131]. Ferroptosis is involved in pancreatic injury, glucose tolerance, iron deposition, and diabetic symptoms of T2DM in mice, whereas inhibition of ferroptosis can restore islet function [132]. Thus, the pathogenesis of T2DM is closely linked to pancreatic β-cell ferroptosis.

The mechanisms by which ferroptosis occurs in pancreatic β-cells in the context of T2DM may involve several pathways (shown in Fig. 3):

(1) A deficiency of iron-sulfur clusters synthesis reduces the function of Cdk5-regulatory subunit-associated protein 1-like 1 (Cdkal1). Cdkal1 catalyzes the methylthiolation of N6-threonylcarbamoyl adenosine 37 (t6A37) in cytosolic tRNALysUUU to generate 2-methylthio-N6-threonylcarbamoyl adenosine (ms2t6A37), which is required for accurate translation of lysine codons in proinsulin. Proinsulin is misread because of a deficiency in the synthesis of Fe-S, leading to a decrease in insulin content and secretion, as well as transport to the endoplasmic reticulum, causing endoplasmic reticulum stress [12]. This can trigger endoplasmic reticulum (ER) stress. ER stress can lead to apoptosis of pancreatic β-cells through the activation of apoptosis signal-regulating kinase 1 (ASK1) and the ASK1/P38/CCAAT-enhancer-binding protein homologous protein (CHOP) signaling pathway [133134]. This contributed to the development of T2DM [12]. In addition, Fe2+ accumulation in the mitochondria leads to a deficiency of the Fe-S clusters, resulting in increased ROS levels in the mitochondria, which may subsequently lead to pancreatic β-cells ferroptosis due to the accumulation of lipid peroxides [12].

(2) ACSL4 exists in human and rat pancreatic β-cells and plays a role in the modification of fatty acids (FAs) within insulin secretion granules [135]. In studies involving mice fed a high-fat diet, specific inhibition of ACSL4 led to protection against insulin resistance [136]. Thus, ACSL4 could be an important lipid-metabolizing enzyme associated with pancreatic β-cells ferroptosis, potentially mediating ferroptosis through the generation of lipid peroxidation via PUFAs.

(3) P53 is a tumor suppressor gene that induces cell cycle arrest, apoptosis, and senescence [137]. When β-cells are treated with free fatty acids (FFAs), p53 is activated ROS production is increased [138], and upregulation of p53 in adipose tissue leads to an inflammatory response that results in insulin resistance [139]. The underlying mechanism may be that the activation of p53 severely reduces the protein level of SLC7A11. P53 is closely related to GPX4, the only member of the GPX family that can resist peroxide damage. GPX4 mediates oxidative stress and regulates ferroptosis [140]. P53 severe activation will reduce the SLC7A11 protein levels, which can lead to GPX4 deficiency and increased ROS production in pancreatic β-cells, which may mediate ferroptosis in the Xc-system of pancreatic β-cells, triggering steatosis causing T2DM.

(4) NAF-1 is a highly conserved [2Fe-2 S] protein of the NEET protein family localized to mitochondria, that is, the ER and mitochondria-associated membrane junctions. In a rat islet tumor clonal cell model (INS-1) lacking NFA-1, insulin secretion was impaired in the presence of increased lipid peroxidation, GPX4 expression, and mitochondrial death characteristic of ferroptosis. In contrast, combined treatment with ferrostatin-1 (iron chelator deferiprone) and the glutathione precursor N-acetylcysteine promoted mitochondrial and ER structural repair, and reduced mitochondrial destabilizing iron and ROS levels. It also improves the growth of NFA-1-deficient INS-1 cells and restore glucose-stimulated insulin secretion [141]. While the genes encoding NAF-1 are WFS-1 and mutant CISD2, this gene is present in Wolfram syndrome type 2 (WFS-T2), so the pancreatic β-cells ferroptosis due to NAF-1 deficiency may be the mechanism of diabetes associated with WFS-T2.

(5) In response to ROS, the cysteine residue of Keap1 is modified to dissociate between Nrf2 and Keap1 or to activate Nrf2 by activating its phosphorylation by a variety of kinases, thus enhancing the stability of Nrf2 and translocating it to the nucleus. It attaches to the promoter region and activates downstream molecules, including NAD(P)H: quinone oxidoreductase 1 (NQO1), HO-1, GPX4, SLC7A11 [142]. Among the downstream target molecules of Nrf2, GPX4 and SLC7A11 are closely associated with ferroptosis. Therefore, ferroptosis may occur when the antioxidant system collapses if excess ROS leads to peroxidation, which exceeds the antioxidant capacity of the Nrf2 pathway. Nrf2 protects β-cells from metabolic stress-induced oxidative damage. Because Nrf2 is a central regulator of β-cell mass by managing the level of cellular redox. One study found that the contribution of Nrf2 activity to the construction of β-cell mass included the maintenance of insulin levels [143]. When β-cells lack Nrf2 activity, insulin immunostaining and insulin content decrease. And Nrf stimulated the proliferation of the rat INS-1 and primary pancreatic β-cells [144]. In addition, Bollong [145] et al. found that methylglyoxal, a metabolite of glycolysis, covalently modified the Keap1 cysteine C151, leading to dimerization and Nrf2 activation [146]. Methylglyoxal itself can also generate ROS in β-cells, and increased glycolysis activates Nrf2. Thus, the Keap1-Nrf2-ARE signaling pathway is closely related to pancreatic β-cell ferroptosis. Nrf2 activity deficiency, along with diminished GPX4 and SLC7A11 levels among downstream target molecules, lowers the antioxidant capability within pancreatic β-cells. This reduction heightens pancreatic β-cells ferroptosis, contributing to diminished insulin mass or content, which will lead to a decrease in insulin mass or content decrease, which may induce or aggravate T2DM.

(6) The NADPH oxidase (NOX) protein is a membrane-associated multi-subunit enzyme that transfers electrons across biological membranes and is required for superoxide production. Evidence suggests that NOX is a major source of mitochondrial ROS in β-cells. In addition, NOX1, NOX2, NOX4, and a number of other cytoplasmic regulators and their homologs NOXO1 and NOXA1 are expressed in pancreatic β-cells [147,148,149]. Superoxide radicals produced by NOXs can contribute to pancreatic β-cell damage and induce intracellular oxidative stress [150151]. When cells respond to chronic hyperglycemia, the expression of NOX enzymes is upregulated, activating angiotensin II type 1 receptor (AT1R) and increasing superoxide production [152153]. Hyperglycaemia and AT1R-induced pro-inflammatory cytokines in human pancreatic islets lead to impaired insulin secretion and inflammation [154155]. AT1R inhibition selectively downregulates NOX, which in turn inhibits oxidative stress, improving β-cell insulin secretion and reducing β-cell apoptosis [156]. In addition, exposure of β-cells to HG induces an increase in CD36 expression on the plasma membrane, leading to enhanced uptake of FFAs and a robustly upregulated NOX activity mediated by RAC1 induction.

Fig. 3
figure 3

Ferroptosis occurs in pancreatic β-cells

Simultaneously, elevated FFAs’ levels lead to ceramide accumulation and ceramide-induced defects in β-cell function and promote insulin resistance, obesity, and inflammatory cytokines through the involvement of the RAC1-NOX pathway [157158]. NOX4 contains a catalytic subunit that transfers electrons from NADPH to oxygen, resulting in ROS formation [159]. NOX4 has been identified as a mitochondrial targeting sequence and has been shown to be a major source of ROS in various cell types [160]. Ferroptosis is closely associated with ROS, mitochondria, and ROS-generating enzymes such as NOX4, which are known sources of ROS in bone tissue [161162]. Recent studies have shown that both mitochondria and NOX4 are involved in ferroptosis [163164]. NOX4 generates ROS using NADPH as a substrate, and one study showed that the activation of NOX4 leads to the overproduction of lipid peroxides and induces ferroptosis in glioma cells [164]. However, whether NOX4 or other NADPH oxidases cause ferroptosis in β-cells and thus induce or exacerbate diabetes needs further validation.

Ferroptosis in T2DOP

OP can be classified as primary, secondary, or idiopathic. DOP are among the most common secondary OPs [5], and are closely associated with diabetes. Research recommends that blood 25-hydroxyvitamin D (25-OH-VD) levels should be tested in clinically admitted (living in nursing homes or residential care) patients with diabetes at risk of falls and fractures. This helps to rapidly identify treatable causes of falls and fractures. Studies have also found a strong correlation between poor glycemic control and increased bone fragility. The HbA1c threshold was > 9% (75 mmol/mol) for patients with T2DM and > 9% (63 mmol/mol) for patients with T1DM [165]. In addition, research suggests that DM leads to the accumulation of advanced glycation end products (AGEs) in various organs, including bones. In terms of bone structural properties, the accumulation of AGEs in the bone and the crosslinking of collagen fibers (intra- and inter-crosslinks) reduce bone biomechanical properties by increasing material stiffness [166]. Studies in Zucker diabetic Sprague-Dawley (ZDSD) rats, an animal model of T2DM, suggested that although mineralization remains normal, bone strength decreases significantly with the duration of diabetes [167]. HG conditions in T2DM severely impair the biological function of OBs, leading to increased mitochondrial double-layer density, a reduced number of mitochondrial cristae, and accumulation of ROS, and lipid peroxidation. This results in OBs exhibiting oxidative stress and lipid peroxidation, which accelerates apoptosis and autophagy [168]. Lin et al. found significantly elevated serum ferritin levels in a rat model of diabetic OP. SLC7A11 and GPX4 expression is significantly reduced [87]. These findings suggest that the HG environment in T2DM may induce ferroptosis in OBs, disrupting bone homeostasis and leading to OP. Therefore, bone cell ferroptosis may play an important role in T2DOP. This article reviews the relationship between T2DM and OP, and discusses the potential mechanisms underlying ferroptosis in T2DOP.

OP’s ferroptosis in HG environment

OBs’ ferroptosis in HG environment

HG toxicity may directly affect the differentiation and function of OBs and OCs, leading to impaired bone remodeling and OP development. Studies have observed that the differentiation of BMSCs into OBs is inhibited in the serum of patients with T2DM [169]. High blood glucose levels lead to reduced OB capacity and decreased bone mineralization. Research on newly diagnosed patients with T2DM suggests that the HG microenvironment may lead to impaired OB maturation and bone formation [170]. Similarly, iron overload downregulates the expression of RUNX2 and its targets, alkaline phosphatase and osteocalcin, in BMSCs and OBs progenitors, thereby inhibiting OBs formation [79]. HG conditions may also affect iron metabolism in OBs. Excess iron reduced the viability of MC3T3 cells and induced apoptosis. Excessive iron may partially inhibit OB activity and disrupt OB differentiation and mineralization [85]. HG induces ferroptosis in OBs by activating the METTL3/ASK1-p38 signaling pathway [171]. Therefore, abnormal iron metabolism under HG conditions may affect OB differentiation, leading to ferroptosis. There is evidence of ferroptosis in OBs in HG environments. HG/high oxygen conditions and the NFκB pathway promote the expression of NOX4. The expression of NOX4 is negatively regulated by several antioxidant factors [171]. Elevated levels of NOX4 result in the production of ROS and accumulation of lipid peroxidation, leading to morphological and functional impairments in OBs mitochondria and promoting ferroptosis in OBs [21]. Activating Transcription Factor 3 (ATF3), a member of the ATF/Cyclic AMP Response Element-Binding protein (CREB) transcription factor family, plays a crucial role in transcriptional repression and promotion with a dual function of regulating cell apoptosis, proliferation, and the cell cycle. HG conditions induce the upregulation of ATF3, leading to the reduced expression of SLC7A11 and lower intracellular levels of GSH and extracellular glutamate. Therefore, ferroptosis in OBs under HG conditions is mediated by the inhibition of Xc-system activity by ATF3 [172]. Under HG conditions, FtMt overexpression reduced ferroptosis in OBs, whereas FtMt inhibition induced mitochondrial autophagy through the ROS/PINK1/Parkin pathway, resulting in increased ferroptosis [63]. Additionally, amino acids and reducing sugars undergo non-enzymatic reactions in the physiological environment to form AGEs. Prolonged high blood glucose levels lead to the significant accumulation of late-stage AGEs in the body [166]. Research suggests that AGEs promote ferroptosis in OBs, thereby contributing to OP development [173].

OCs’ ferroptosis in HG environment

Several studies have indicated that HG mircoenvironment stimulates the differentiation of OCs, thereby increasing their capacity for bone resorption [174]. Additionally, clinical research has observed elevated levels of tartrate-resistant acid phosphatase in the bloodstream of patients with T2DM, suggesting increased OC activity [175]. In animal models, studies have revealed elevated bone resorption in T2DM rats compared to normal glucose control groups. This indicates higher OC activity in T2DM models [176177]. Under HG conditions, factors including TNF-α, macrophage colony-stimulating factor, receptor activator of RANKL, and vascular endothelial growth factor-A are heightened in diabetic mice. These factors facilitate the differentiation and activation of OCs [178179]. Studies have indicated that the differentiation of bone marrow monocytes into OCs is stimulated by the RANKL, which enhances NOX4 mRNA expression and ROS production. In NOX4-deficient mice, RANKL was ineffective in stimulating the expression of two recognized transcription factors for osteoclast differentiation, namely the nuclear factor of activated T cells cytoplasmic 1 (NFATC1) and activator protein 1; resulting in augmented bone mass and thicker trabecular bone structure in NOX4-deficient mice [180181]. The hyperglycaemic/hyperoxic environment and the NF-κB pathway promote the expression of NOX4, which is negatively regulated by various antioxidant factors [160, 171]. Under HG conditions, the NFκB pathway activation leads to increased NOX4 expression, which may in turn activate OCs differentiation transcription factors via RANKL, thereby increasing OCs differentiation. The build-up of AGEs in the body during the later stages of T2DM stimulates OCs activity, resulting in a higher bone resorption area, and thus increased bone resorption [182]. The presence of AGEs causes oxidative stress responses and triggers the activation of the receptor for AGEs (RAGE), leading to RANKL-mediated OCs formation [183184]. The NFκB also plays a role in oxidative stress and increased OCs activity, while simultaneously decreasing OBs differentiation [185]. As blood glucose levels increase, the levels of additional proteins including AGEs, pro-inflammatory cytokines, and ROS also increase [186]. ROS activation triggers the intracellular MAPK signaling pathway. Activation of ROS/MAPKs/NF-κB/NLRP3 causes OC-mediated bone loss in patients with DOP [174].

Furthermore, in vitro studies using RAW264.7 cells indicate that HG reduces OCs autophagy, thus amplifying OCs formation [187]. Ferritin is responsible for storing excess cellular iron. When cells are iron deficient, they may degraded, a phenomenon commonly referred as “ferritinophagy”. This leads to increased cell sensitivity to ferroptosis caused by Fe2+ in cells [188]. Owing to their high energy demands to function as acid-secreting and absorptive bone cells, OCs are abundant in mitochondria [189]. Therefore, fully developed OCs require higher amount of free intracellular iron than other bone cells. The iron-chelating agent deferoxamine mesylate (DFO) impedes OC formation in vitro [94]. Ni et al. discovered that ferroptosis impacts OCs during the differentiation phase when stimulated by RANKL and overexpressed TfR1, causing a reduction in the iron concentration prompted by the decreased activity of urocanase [190]. Owing to OCs’ iron-dependent nature of OCs, which is similar to that of cancer cells, they are more prone to iron-triggered ferroptosis. Research findings indicate that the artemisinin compound ART enhances the generation of MDA and 4-HNE, resulting in the manifestation of mitochondrial ferroptosis traits and prompting ferroptosis in OCs, which hinders their differentiation [191]. OCs differentiation increases under high glucose conditions, thereby enhancing the bone resorption capacity. Furthermore, it is possible that the OCs in DOP undergo ferroptosis, presenting a promising therapeutic avenue for DOP through the induction of OC death and inhibition of their differentiation.

Insulin resistance and deficiency in OP-related ferroptosis

Razny et al. discovered that insulin resistance results in reduced mRNA expression in OBs generation, increased mRNA expression of OBs ogenesis inhibitors, and suppression of miRNAs, such as miRNA-29b and miRNA-181a. These factors are responsible for promoting OBs generation and osteogenic differentiation of stem cells. Consequently, defects occur in the OBs of patients [192]. Insulin-like growth factor 1 (IGF-1) secreted by pancreatic β-cells plays a crucial role in bone mass and is strongly associated with vertebral fractures. Impaired IGF-1 function, reduced synthesis of active vitamin D, inhibition of bone calcitonin synthesis and secretion, and increased ceramide synthesis can adversely affect the bone microstructure and material properties. Subsequently, bone strength is reduced, and the likelihood of fracture is elevated [193]. As T2DM advances and insulin levels decrease, metabolic disruptions occur in glucose, fat, and protein, resulting in a negative nitrogen balance in the body. This allows the compensatory breakdown of bone collagen and reduces its synthesis, ultimately resulting in decreased bone formation. Additionally, insulin restricts cyclic adenosine monophosphate (cAMP). cAMP motivates bone resorption and reduces bone calcium salt deposition. Insulin deficiency diminishes this inhibitory effect, boosts cAMP activity, and consequently enhances bone resorption, culminating in OP [194]. Osteocalcin is a non-collagenous protein that is mainly synthesized and secreted by OBs. Its primary role is to uphold the rate of bone mineralization, foster bone metabolism, and indicate the level of OBs’ activity [195]. Insulin deficiency may impede OBs from synthesizing osteocalcin, which may decelerate bone mineralization, metabolism, and activity, and reduce bone formation.

In addition, insulin directly facilitates the reabsorption of calcium, phosphate, and magnesium in the renal tubules. When insulin is inadequate, renal tubules reabsorb fewer minerals, resulting in their loss, decreased concentration of calcium in the blood, and the release of calcium from the bones. This leads to reduced bone density and the onset of OP [196]. Research indicates that increased ATF3 expression leads to cell damage, including apoptosis of pancreatic β-cells, the generation of ROS, and high levels of glucose or FA associated with complications in diabetes. High levels of ATF3 are linked to stress-induced apoptosis of pancreatic β-cells [197]. Under HG conditions, ATF3 upregulation inhibits the activity of the Xc-system and causes ferroptosis in OBs [172]. Therefore, ATF3-mediated ferroptosis of pancreatic β-cells occurring with insulin deficiency may be linked to ferroptosis in OBs associated with OP.

OP’s ferroptosis in diabetic microvascular disease

Microvascular diseases in DM are characterized by augmented capillary permeability, thickening of microvascular basement membranes, destruction of endothelial cells, and platelet adhesion and aggregation, culminating in the creation of microthrombi or microvascular occlusion. This can have an impact on multiple organs in the body and diseases, including the diabetic nephropathy(DN), diabetic retinopathy (DR), and diabetic peripheral neuropathy (DPN) [198]. Diabetes complications, such as damage to vascular dilation, vascular calcification and vascular regeneration, can affect the development of OBs progenitor cells within the hematopoietic niche, as well as the transportation of OBs (or periosteal cells) and OCs to the Haversian canals [199]. Impaired blood flow and damage to neovascularization may lead to decreased OBs formation and reduced bone remodeling activity, ultimately resulting in decreased bone mass and delayed fracture healing. Furthermore, numerous studies have indicate that the AGE/RAGE signaling pathway has a significant impact on vascular calcification. In peripheral vascular smooth muscle cells (VSMCs), AGEs activate RAGE that leads to a sequence of signaling pathways involving MAPK, TGF-β, and NF-κB. This leads to the downregulation of VSMC markers and activation of an OBs program, resulting in increased expression of Runx2 and osteocalcin, as well as enhanced alkaline phosphatase activity, ultimately leading to VSMC calcification [200]. Iron overload induces ROS accumulation, which activates intracellular MAPK signaling pathways. ROS/MAPKs/NF-κB/NLRP3 activation results in OCs-mediated bone loss in DOP [174]. Bone vascular calcification is linked to the ROS/MAPKs pathway, which can be activated by iron overload-induced ROS accumulation, affecting OBs funtion or activity. Additionally, AGEs contribute to vascular calcification and also induce ferroptosis in OBs [201]. These findings suggest a potential association between microvascular complications in diabetes and the ferroptosis of DOP.

Summary

T2DM has a close relationship with the development of OP. Changes in the HG microenvironment, insulin resistance, insulin insufficiency, and microangiopathy can exacerbate the progression of OP in patients with T2DM. The HG environment in T2DM may activate bone cells ferroptosis, disturbing bone homeostasis and leading to OP. Furthermore, cell ferroptosis could impact both pancreatic β-cells and bone cells concurrently through numerous pathways, significantly impacting the advancement of T2DM and OP. However, the mechanism underlying ferroptosis in DOP remain unclear. We examined the possible mechanisms of ferroptosis in DOP (shown in Fig. 4).

Fig. 4
figure 4

Ferroptosis in DOP

(1) ATF3/Xc-system/GPX4.

Jang’s research found that ATF3, by binding to ATF3 response elements in its promoter, suppresses the expression of pancreatic duodenal homeobox 1 (PDX-1) in pancreatic β-cells, thereby inhibiting pancreatic β-cells function [202]. These findings suggest that ATF3 play an important role in the onset and development of T2DM. Okamoto et al. reported an increase in ATF3 expression in renal glomeruli and aortic endothelial cells of a DN, rat model, suggesting that ROS-related vascular complications in diabetes may involve ATF-mediated pathological angiogenesis [203]. ATF3 also mediates the Xc-system/GPX4 pathway in OB ferroptosis under HG conditions. Therefore, T2DM may affect DOP progression via the ATF3-mediated signaling pathway.

(2) NRF2/HO-1/GPX4/SLC7A11.

The T2DM microenvironment significantly promotes ferroptosis in bone cells in vitro with increased lipid peroxidation and iron overload. According to RNA sequencing results, HO-1 expression was significantly upregulated in bone cells. HO-1 is critical for DOP ferroptosis [13]. Yang et al. found that there is more lipid peroxidation in vivo in a DOP rat model, suggesting that the HG microenvironment can induce ferroptosis in bone cells. They believe that in the DM microenvironment, HO-1 transcription is activated upstream by NRF2 and c-JUN, leading to the activation of HO-1, which catalyzes haem oxidation and generates a large amount of free iron [13]. In addition, research by Ma supported the theory that HO-1 mediates HG-induced ferroptosis in bone cells [81]. HO-1 activation and ferroptosis are mutually dependent and may lead to an endless cycle of mutual promotion [204]. Targeting ferroptosis (injection of the ferroptosis inhibitor, Fer-1) or HO-1 (injection of zinc protoporphyrin, an HO-1 inhibitor) may reverse ferroptosis in bone cells during DOP by interfering with lipid peroxidation, ultimately reversing bone loss. Among the downstream targets of NRF2, GPX4 and SLC7A11 are important markers of iron deficiency anemia. The importance of NRF2 transcriptional targets in the regulation of lipid peroxidation, inhibition of free iron accumulation, and glutathione synthesis and metabolism, which are all related to ferroptosis, is becoming increasingly clear [33]. MaR1 treatment alters the NRF2 pathway for SLC7A11/GPX4 signaling, protecting OBs from the effects of ferroptosis in T2DM, thereby improving their bone-forming capacity [205]. In T2DOP, HG induces ferroptosis by increasing ROS/lipid peroxidation/GSH depletion. Melatonin significantly reduces iron-deficiency anaemia in vitro and in vivo and increases the osteogenic capacity of MC3T3-E1 cells by activating the Nrf2/HO-1 pathway [81]. Studies have suggested that pancreatic iron overload is involved in the pathogenesis of T2DM. And vitamin D may inhibit ferroptosis in pancreatic β-cells through the NFκB-DMT1 signaling pathway [205]. In addition, the VDR agonist (1,25(OH)2D3) can inhibit ferroptosis in bone cells by activating the Nrf2/GPX4 signaling pathway [90]. Therefore, vitamin D drugs may protect against DOP development.

(3) NOX4/NF-κB/RANKL.

NOX is a major source of mitochondrial ROS in β-cells. The superoxide radicals produced by NOX can damage pancreatic β-cells and induce intracellular oxidative stress [150151]. When cells are exposed to chronic hyperglycemia, NOX expression is upregulated. The NOX downregulation can suppress oxidative stress, thereby improving pancreatic β-cells insulin secretion and reducing β-cells apoptosis [160]. Increased NOX4 expression promotes ferroptosis in OBs [21]. In an HG environment, the NFκB pathway promotes NOX4 expression, activates RANKL, induces excessive TfR1 expression, and significantly reduces iron levels, which can lead to ferroptosis in OBs and promote OCs differentiation. Whether NOX4 or other NADPH oxidases cause ferroptosis in pancreatic β-cells, thereby inducing or exacerbating diabetes, requires further investigation. However, the HG microenvironment of T2DM may increase NOX and affect OP through the NOX4/NF-κB/RANKL pathway.

(4) Fenton reaction and iron autophagy.

Fe2 + interacts with H2O2 through the Fenton reaction, causing lipid peroxidation and inducing ferroptosis in cells [206]. The Fenton reaction generates a large amount of hydroxyl radicals and other ROS, which stimulate the release of cytochrome C from the mitochondria and activate the key apoptosis protein Caspase3, promoting apoptosis of OBs [8889]. Pancreatic β-cells are susceptible to oxidative stress and have a relatively low antioxidant capacity, making them susceptible to ferroptosis. The accumulation of Fe2+ in mitochondria leads to a lack of Fe-S clusters, resulting in increased mitochondrial ROS levels. The subsequent accumulation of lipid peroxidation products can lead to pancreatic β-cells ferroptosis.

Iron autophagy maintains intracellular iron balance under physiological conditions, but excessive activation of ferritinophagy leads to intracellular iron overload. Iron stored in ferritin can also be released into the cytoplasm, which is considered the starting point and ultimately triggers ferroptosis [27]. Studies have suggested that excessive autophagy and lysosomal activity promote ferroptosis through iron accumulation or lipid peroxidation [207]. FtMt suppresses ferroptosis in OBs by reducing the oxidative stress caused by excess iron ions [62]. In addition, OBs ferroptosis is promoted under FtMt deficiency condition. FtMt deficiency activates the ROS/PINK1/Parkin pathway, induces mitochondrial autophagy, and releases free Fe2+ into the cytoplasm, aggravating the Fenton reaction [206]. Therefore, a relationship between the Fenton reaction and iron autophagy have been implicated in OBs ferroptosis.

Therapeutic opportunities

Although the mechanisms of T2DOP ferroptosis have been discussed above, the therapeutic opportunities of T2DOP target these mechanisms to promote cell recovery during ferroptosis. Ferrostatin-1 (Fer-1), a small-molecule drug, is an effective inhibitor of ferroptosis because of its ability to scavenge lipid peroxides. It can target ferroptosis or HO-1 to efficiently rescue osteocyte death in DOP by disrupting the vicious cycle between lipid peroxidation and HO-1 activation, eventually ameliorating trabecular deterioration [12]. Studies have shown that poliumoside (Pol) inhibits HGHF-induced bone degradation and ferroptosis by elevating GSH and decreasing malondialdehyde (MDA) levels, lipid peroxidation, and mitochondrial reactive oxygen species (ROS). Pol effective in treating T2DOP by activating the Nrf2/GPX4 signaling pathway to inhibit ferroptosis, which is beneficial for HGHF-treated BMSCs [208] Curcumin, a tetrahedral framework nucleic acid (tFNA) can also effectively inhibit ferroptosis by activating the NRF2/GPX4 pathway. It can enhance mitochondrial function by activating this pathway, induce the osteogenic differentiation of BMSCs in the diabetic microenvironment, as well as reduce trabecular loss and increase bone formation in a mouse model of DOP [209]. Maresin1 (MaR1), a derivative of one of the primary omega-3 fatty acids, docosahexaenoic acid (DHA), is an endogenous, pro-resolving lipid mediator produced mainly by macrophages. It was found that could enhance the expression levels of NRF2, GPX4 and SLC7A11 to cause the restraint of ferroptosis under hyperglycemic stimulation. MaR1 can activate the NRF2/GPX4 pathway in vivo and in vitro to alleviate high-glucose-induced ferroptosis greatly [82].

In addition, other compounds affect ferroptosis through different pathways. Vitamin K2 (VK2), a fat-soluble vitamin, can decrease the levels of mitochondrial ROS, lipid peroxidation, and malondialdehyde, and increase GSH in BMCS. It also restored bone mass and increased SIRT1 and GPX4 expression. Therefore VK2 may ameliorate T2DOP through the activation of the AMPK/SIRT1 signaling pathway to inhibit ferroptosis [210] An arabinomannan (PAAP-1B), a molecular weight of 14.0 kDa was isolated from Anemarrhena asphodeloides Bge, suppressed ferroptosis in advanced glycation end product-induced osteoblasts by decreasing MDA, mitochondrial ROS levels, and lipid peroxidation, while reversing the downregulation of SLC7A11 and GSH expression [211]. Eldecalcitol (ED-71), a novel active form of vitamin D, can attenuate the over expression of hypoxia-inducible factor 1α (HIF1α) induced by high glucose levels, also increase GPX4 levels. It may improve osteogenic disorders caused by diabetes such as DOP [212].

However, there are still many potential therapeutic opportunities to promote the ability of cells to recover from oxidative stress, which is beneficial for treating T2DOP. Ferroptosis-specific antagonists for the treatment of T2DOP and the lack of research are not listed in this paper. Meanwhile, herbs or their active ingredients (quercetin, curcumin, cryptochlorogenic acid, resveratrol, Platycodonopsis saponin D, Astragaloside IV) can also inhibit ferroptosis [213], which provides a new idea for T2DOP treatment.

In conclusion, T2DM is closely associated with OP development. Ferroptosis affects the progression through multiple pathways. Regulation of ferroptosis in pancreatic β-cells and bone cells may be a new target for the treatment of DOP.

Data availability

Due to its nature as a review article, all references are published articles. The data underlying this article are available in the Pubmed.

Abbreviations

25-OH-VD:

25-hydroxyvitamin D

4-HNE:

4-Hydroxynonenal

AA:

Arachidonic acid

ACSL4:

Acyl-CoA synthetase long-chain family member 4

AdA:

Adrenergic acid

AGEs:

Advanced glycation end products

AREs:

Antioxidant response elements

ARS:

Artesunate

ASK1:

Apoptosis signal-regulating kinase 1

AT1R:

Angiotensin II type 1 receptor

ATF3:

Activating Transcription Factor 3

ATF4:

Activating transcription factor 4

BMSCs:

Bone marrow mesenchymal stem cells

cAMP:

Cyclic adenosine monophosphate

CCA:

Cryptochlorohydric acid

Cdkal1:

Cdk5-regulatory subunit-associated protein 1-like 1

CHOP:

CCAAT-enhancer-binding protein homologous protein

CoQ10:

Coenzyme Q10

CoQ10H2:

Coenzyme Q10H2

CoQH2:

Coenzyme QH2

CREB:

Cyclic AMP Response Element-Binding protein

CYP450:

Cytochrome P450

DFO:

Deferoxamine mesylate

DHODH:

Dihydroorotate dehydrogenase

DM:

Diabetes mellitus

DMT-1:

Divalent metal transporter 1

DN:

Diabetic nephropathy

DOP:

Diabetic osteoporosis

DPN:

Diabetic peripheral neuropathy

DR:

Diabetic retinopathy

ER:

Endoplasmic reticulum

ERK:

Extracellular signal-regulated kinase

FAC:

Ammonium ferric citrate

FAs:

Fatty acids

F-box protein 9:

FBXO9

Fer-1:

Ferrostatin-1

FFAs:

Free fatty acids

FINO2:

1,2-dioxin cycloalkane

FPN:

Ferroportin

FSP1:

Ferroptosis inhibitory protein 1

FTH:

Ferritin

FTH1:

Ferritin heavy chain 1

FTL:

Ferritin light chain

FtMt:

Mitochondrial ferritin

GPX4:

Glutathione peroxidase 4

GSH:

Glutathione

GSIS:

Glucose-stimulated insulin secretion

H2O2:

Hydrogen peroxide

HG:

High glucose

HGPA:

High glucose and palmitic acid

HH:

Hereditary hemochromatosis

HMOX1:

Haem oxygenase 1

IGF-1:

Insulin-like growth factor-1

IRE:

Iron-responsive element

IREB2:

Iron regulatory protein 2

IRP1:

Iron regulatory protein 1

Keap1:

Kelch-like ECH-associated protein 1

LIP:

Labile iron pool

Lip-1:

Liproxtatin-1

LOOH:

Lipid peroxides

LOXs:

Lipoxygenases

LPCAT3:

Lysophosphatidylcholine acyltransferase 3

MAPK:

Mitogen-activated protein kinase

M-CSF:

Macrophage-colony stimulating factor

MDA:

Malondialdehyde

METTL3:

Methyltransferase-like 3

ms2t6A37:

2-methylthio-N6-threonylcarbamoyl adenosine

NAC:

N-acetylcysteine

NADPH:

Nicotinamide adenine dinucleotide phosphate

NCOA4:

Nuclear receptor coactivator 4

NFATC1:

Nuclear factor of activated T cells cytoplasmic 1

NLRP3:

Pyrin structural domains of 3

NOX:

NADPH oxidase

NOX4:

NADPH oxidase 4

NQO1:

NAD(P)H: quinone oxidoreductase 1

Nrf2:

Nuclear factor E2-related factor 2

OB:

Osteoblast

OBs:

Osteoblasts

OC:

Osteoclast

OCs:

Osteoclasts

OP:

Osteoporosis

OPG:

Osteoprotegerin

PCD:

Programmed cell death

PDX-1:

Pancreatic duodenal homeobox 1

PE:

Phosphatidylethanolamine

PL:

Phosphatidylinositol

PUFA-COA:

ACSL4 forms multiple unsaturated fatty acid coenzyme A

PUFA-PL:

Phospholipid PUFAs

PUFAs:

Polyunsaturated fatty acids

RAGE:

Receptor for AGEs

RANKL:

Nuclear factor κB ligand

ROS:

Reactive oxygen species

RSL3:

RAS-selective lethal small molecule 3

Runx2:

Runt-related transcription factor 2

SAT1:

Spermidine/spermine N1-acetyltransferase 1

SOD:

Superoxide dismutase

T2DM:

Type 2 diabetes mellitus

T2DOP:

Type 2 Diabetic osteoporosis

t6A37:

Cdkal1

TEM:

Transmission electron microscopy

tert-BHP:

Ert-butyl hydroperoxide

TFR:

Transferrin receptor

VDR:

Vitamin D receptor

VSMCs:

Vascular smooth muscle cells

WFS-T2:

Wolfram syndrome type 2

WHO:

The World Health Organization

Xc system:

Cystine/glutamate reverse transport system

ZDSD:

Zucker diabetic Sprague-Dawley

References

  1. Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN, Mbanya JC, Pavkov ME, Ramachandaran A, Wild SH, James S, Herman WH, Zhang P, Bommer C, Kuo S, Boyko EJ, Magliano DJ. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119. https://doi.org/10.1016/j.diabres.2021.109119.

    Article  PubMed  Google Scholar 

  2. Li Y, Teng D, Shi X, Qin G, Qin Y, Quan H, Shi B, Sun H, Ba J, Chen B, Du J, He L, Lai X, Li Y, Chi H, Liao E, Liu C, Liu L, Tang X, Tong N, Wang G, Zhang J, Wang Y, Xue Y, Yan L, Yang J, Yang L, Yao Y, Ye Z, Zhang Q, Zhang L, Zhu J, Zhu M, Ning G, Mu Y, Zhao J, Teng W, Shan Z. Prevalence of diabetes recorded in mainland China using 2018 diagnostic criteria from the American Diabetes Association: national cross sectional study. BMJ. 2020;m997. https://doi.org/10.1136/bmj.m997.

  3. Cooper C, Campion G, Melton LJ. Hip fractures in the elderly: a world-wide projection. Osteoporos Int. 1992;2:285–9. https://doi.org/10.1007/BF01623184.

    Article  CAS  PubMed  Google Scholar 

  4. Wang L, Yu W, Yin X, Cui L, Tang S, Jiang N, Cui L, Zhao N, Lin Q, Chen L, Lin H, Jin X, Dong Z, Ren Z, Hou Z, Zhang Y, Zhong J, Cai S, Liu Y, Meng R, Deng Y, Ding X, Ma J, Xie Z, Shen L, Wu W, Zhang M, Ying Q, Zeng Y, Dong J, Cummings SR, Li Z, Xia W. Prevalence of osteoporosis and fracture in China. JAMA Netw Open. 2021;4:e2121106. https://doi.org/10.1001/jamanetworkopen.2021.21106.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Johnell O, Kanis JA. An estimate of the worldwide prevalence and disability associated with osteoporotic fractures. Osteoporos Int. 2006;17:1726–33. https://doi.org/10.1007/s00198-006-0172-4.

    Article  CAS  PubMed  Google Scholar 

  6. Kanis JA, Johnell O, Oden A, Sembo I, Redlund-Johnell I, Dawson A, De Laet C, Jonsson B. Long-term risk of osteoporotic fracture in Malmö. Osteoporos Int. 2000;11:669–74. https://doi.org/10.1007/s001980070064.

    Article  CAS  PubMed  Google Scholar 

  7. Siris ES, Adler R, Bilezikian J, Bolognese M, Dawson-Hughes B, Favus MJ, Harris ST, Jan De Beur SM, Khosla S, Lane NE, Lindsay R, Nana AD, Orwoll ES, Saag K, Silverman S, Watts NB. The clinical diagnosis of osteoporosis: a position statement from the National Bone Health Alliance Working Group. Osteoporos Int. 2014;25:1439–43. https://doi.org/10.1007/S00198-014-2655-Z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cortet B, Lucas S, Legroux-Gerot I, Penel G, Chauveau C, Paccou J. Bone disorders associated with diabetes mellitus and its treatments. Joint Bone Spine. 2019;86:315–20. https://doi.org/10.1016/J.JBSPIN.2018.08.002.

    Article  CAS  PubMed  Google Scholar 

  9. Shanbhogue VV, Hansen S, Frost M, Brixen K, Hermann AP. Bone disease in diabetes: another manifestation of microvascular disease? Lancet Diabetes Endocrinol. 2017;5:827–38. https://doi.org/10.1016/S2213-8587(17)30134-1.

    Article  PubMed  Google Scholar 

  10. Kim J-H, Kim AR, Choi YH, Jang S, Woo G-H, Cha J-H, Bak E-J, Yoo Y-J. Tumor necrosis factor-α antagonist diminishes osteocytic RANKL and sclerostin expression in diabetes rats with periodontitis. PLoS ONE. 2017;12:e0189702. https://doi.org/10.1371/journal.pone.0189702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Martinez-Laguna D, Nogues X, Abrahamsen B, Reyes C, Carbonell-Abella C, Diez-Perez A, Prieto-Alhambra D. Excess of all-cause mortality after a fracture in type 2 diabetic patients: a population-based cohort study. Osteoporos Int. 2017;28:2573–81. https://doi.org/10.1007/s00198-017-4096-y.

    Article  CAS  PubMed  Google Scholar 

  12. Miao R, Fang X, Zhang Y, Wei J, Zhang Y, Tian J. Iron metabolism and ferroptosis in type 2 diabetes mellitus and complications: mechanisms and therapeutic opportunities. Cell Death Dis. 2023;14:186. https://doi.org/10.1038/s41419-023-05708-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yang Y, Lin Y, Wang M, Yuan K, Wang Q, Mu P, Du J, Yu Z, Yang S, Huang K, Wang Y, Li H, Tang T. Targeting ferroptosis suppresses osteocyte glucolipotoxicity and alleviates diabetic osteoporosis. Bone Res. 2022;10:26. https://doi.org/10.1038/s41413-022-00198-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B, Stockwell BR. Ferroptosis: An Iron-Dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72. https://doi.org/10.1016/j.cell.2012.03.042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li J, Cao F, Yin H, Huang Z, Lin Z, Mao N, Sun B, Wang G. Ferroptosis: past, present and future. Cell Death Dis. 2020;11:88. https://doi.org/10.1038/s41419-020-2298-2.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Abraham D, Rogers J, Gault P, Kushner JP, McClain DA. Increased insulin secretory capacity but decreased insulin sensitivity after correction of iron overload by phlebotomy in hereditary haemochromatosis. Diabetologia. 2006;49:2546–51. https://doi.org/10.1007/S00125-006-0445-7.

    Article  CAS  PubMed  Google Scholar 

  17. Stancic A, Saksida T, Markelic M, Vucetic M, Grigorov I, Martinovic V, Gajic D, Ivanovic A, Velickovic K, Savic N, Otasevic V. Ferroptosis as a novel determinant of β-Cell death in Diabetic conditions. Oxid Med Cell Longev. 2022;2022. https://doi.org/10.1155/2022/3873420.

  18. Rehman K, Akash MSH. Mechanism of generation of oxidative stress and pathophysiology of type 2 diabetes Mellitus: how are they interlinked? J Cell Biochem. 2017;118:3577–85. https://doi.org/10.1002/JCB.26097.

    Article  CAS  PubMed  Google Scholar 

  19. Okabe H, Suzuki T, Uehara E, Ueda M, Nagai T, Ozawa K. The bone marrow hematopoietic microenvironment is impaired in iron-overloaded mice. Eur J Haematol. 2014;93:118–28. https://doi.org/10.1111/ejh.12309.

    Article  CAS  PubMed  Google Scholar 

  20. Che J, Yang J, Zhao B, Zhang G, Wang L, Peng S, Shang P. The effect of abnormal Iron metabolism on osteoporosis. Biol Trace Elem Res. 2020;195:353–65. https://doi.org/10.1007/s12011-019-01867-4.

    Article  CAS  PubMed  Google Scholar 

  21. Zhang H, Wang A, Li G, Zhai Q, Huang Z, Wang X, Cao Z, Liu L, Liu G, Chen B, Zhu K, Xu Y, Xu Y. Osteoporotic bone loss from excess iron accumulation is driven by NOX4-triggered ferroptosis in osteoblasts. Free Radic Biol Med. 2023;198:123–36. https://doi.org/10.1016/j.freeradbiomed.2023.01.026.

    Article  CAS  PubMed  Google Scholar 

  22. Gao Z, Chen Z, Xiong Z, Liu X. Ferroptosis - a new target of osteoporosis. Exp Gerontol. 2022;165:111836. https://doi.org/10.1016/j.exger.2022.111836.

    Article  CAS  PubMed  Google Scholar 

  23. Latunde-Dada GO. Ferroptosis: role of lipid peroxidation, iron and ferritinophagy. Biochim et Biophys Acta (BBA) - Gen Subj. 2017;1861:1893–900. https://doi.org/10.1016/j.bbagen.2017.05.019.

    Article  CAS  Google Scholar 

  24. Lei P, Bai T, Sun Y. Mechanisms of ferroptosis and relations with regulated cell death: a review. Front Physiol. 2019;10. https://doi.org/10.3389/fphys.2019.00139.

  25. Abe C, Miyazawa T, Miyazawa T. Current use of Fenton reaction in drugs and food. Molecules. 2022;27:5451. https://doi.org/10.3390/molecules27175451.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xie L, Fang B, Zhang C. The role of ferroptosis in metabolic diseases, Biochimica et Biophysica Acta (BBA) - molecular Cell Research. 1870 (2023) 119480. https://doi.org/10.1016/j.bbamcr.2023.119480.

  27. Bellelli R, Federico G, Matte’ A, Colecchia D, Iolascon A, Chiariello M, Santoro M, De Franceschi L, Carlomagno F. NCOA4 Deficiency impairs systemic Iron homeostasis. Cell Rep. 2016;14:411–21. https://doi.org/10.1016/j.celrep.2015.12.065.

    Article  CAS  PubMed  Google Scholar 

  28. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, Noel K, Jiang X, Linkermann A, Murphy ME, Overholtzer M, Oyagi A, Pagnussat GC, Park J, Ran Q, Rosenfeld CS, Salnikow K, Tang D, Torti FM, Torti SV, Toyokuni S, Woerpel KA, Zhang DD. Ferroptosis: a regulated cell death Nexus linking metabolism, Redox Biology, and Disease. Cell. 2017;171:273–85. https://doi.org/10.1016/j.cell.2017.09.021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chang S, Tang M, Zhang B, Xiang D, Li F. Ferroptosis in inflammatory arthritis: a promising future. Front Immunol. 2022;13. https://doi.org/10.3389/fimmu.2022.955069.

  30. Lei G, Zhuang L, Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 2022;22:381–96. https://doi.org/10.1038/s41568-022-00459-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yang WS, Stockwell BR. Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 2016;26:165–76. https://doi.org/10.1016/j.tcb.2015.10.014.

    Article  CAS  PubMed  Google Scholar 

  32. Kagan VE, Mao G, Qu F, Angeli JPF, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, Kapralov AA, Amoscato AA, Jiang J, Anthonymuthu T, Mohammadyani D, Yang Q, Proneth B, Klein-Seetharaman J, Watkins S, Bahar I, Greenberger J, Mallampalli RK, Stockwell BR, Tyurina YY, Conrad M, Bayır H. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90. https://doi.org/10.1038/nchembio.2238.

    Article  CAS  PubMed  Google Scholar 

  33. Yan H, Zou T, Tuo Q, Xu S, Li H, Belaidi AA, Lei P. Ferroptosis: mechanisms and links with diseases. Signal Transduct Target Ther. 2021;6:49. https://doi.org/10.1038/s41392-020-00428-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Feng H, Stockwell BR. Unsolved mysteries: how does lipid peroxidation cause ferroptosis? PLoS Biol. 2018;16:e2006203. https://doi.org/10.1371/journal.pbio.2006203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yan B, Ai Y, Sun Q, Ma Y, Cao Y, Wang J, Zhang Z, Wang X. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell. 2021;81:355–e36910. https://doi.org/10.1016/j.molcel.2020.11.024.

    Article  CAS  PubMed  Google Scholar 

  36. Gaschler MM, Andia AA, Liu H, Csuka JM, Hurlocker B, Vaiana CA, Heindel DW, Zuckerman DS, Bos PH, Reznik E, Ye LF, Tyurina YY, Lin AJ, Shchepinov MS, Chan AY, Peguero-Pereira E, Fomich MA, Daniels JD, Bekish AV, Shmanai VV, Kagan VE, Mahal LK, Woerpel KA, Stockwell BR. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. 2018;14:507–15. https://doi.org/10.1038/s41589-018-0031-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu L, Wang M, Gong N, Tian P, Deng H. Se improves GPX4 expression and SOD activity to alleviate heat-stress-induced ferroptosis-like death in goat mammary epithelial cells. Anim Cells Syst (Seoul). 2021;25:283–95. https://doi.org/10.1080/19768354.2021.1988704.

    Article  CAS  PubMed  Google Scholar 

  38. Forman HJ, Zhang H, Rinna A, Glutathione. Overview of its protective roles, measurement, and biosynthesis. Mol Aspects Med. 2009;30:1–12. https://doi.org/10.1016/j.mam.2008.08.006.

    Article  CAS  PubMed  Google Scholar 

  39. Zhang Y, Xin L, Xiang M, Shang C, Wang Y, Wang Y, Cui X, Lu Y. The molecular mechanisms of ferroptosis and its role in cardiovascular disease. Biomed Pharmacother. 2022;145:112423. https://doi.org/10.1016/j.biopha.2021.112423.

    Article  CAS  PubMed  Google Scholar 

  40. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis, Proceedings of the National Academy of Sciences. 113 (2016). https://doi.org/10.1073/pnas.1603244113.

  41. Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, Brown LM, Valenzuela CA, Wolpaw AJ, Stockwell BR. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12:497–503. https://doi.org/10.1038/nchembio.2079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Moosmann B, Behl C, Selenoproteins C-L. Drugs, and the consequences Revisiting of the Mevalonate Pathway. Trends Cardiovasc Med. 2004;14:273–81. https://doi.org/10.1016/j.tcm.2004.08.003.

    Article  CAS  PubMed  Google Scholar 

  43. Abrams RP, Carroll WL, Woerpel KA. Five-Membered Ring Peroxide selectively initiates ferroptosis in Cancer cells. ACS Chem Biol. 2016;11:1305–12. https://doi.org/10.1021/acschembio.5b00900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Liu P, Wang W, Li Z, Li Y, Yu X, Tu J, Zhang Z. Ferroptosis: a New Regulatory mechanism in osteoporosis. Oxid Med Cell Longev. 2022;2022:1–10. https://doi.org/10.1155/2022/2634431.

    Article  CAS  Google Scholar 

  45. Lu SC. Regulation of glutathione synthesis. Mol Aspects Med. 2009;30:42–59. https://doi.org/10.1016/j.mam.2008.05.005.

    Article  CAS  PubMed  Google Scholar 

  46. Jiang L, Kon N, Li T, Wang S-J, Su T, Hibshoosh H, Baer R, Gu W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57–62. https://doi.org/10.1038/nature14344.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Liu Y, Gu W. p53 in ferroptosis regulation: the new weapon for the old guardian. Cell Death Differ. 2022;29:895–910. https://doi.org/10.1038/s41418-022-00943-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 2019;133:162–8. https://doi.org/10.1016/j.freeradbiomed.2018.05.074.

    Article  CAS  PubMed  Google Scholar 

  49. Ursini F, Bosello Travain V, Cozza G, Miotto G, Roveri A, Toppo S, Maiorino M. A white paper on Phospholipid Hydroperoxide glutathione peroxidase (GPx4) forty years later. Free Radic Biol Med. 2022;188:117–33. https://doi.org/10.1016/j.freeradbiomed.2022.06.227.

    Article  CAS  PubMed  Google Scholar 

  50. Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, Zhong M, Yuan H, Zhang L, Billiar TR, Lotze MT, Zeh HJ, Kang R, Kroemer G, Tang D. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692–704. https://doi.org/10.1016/j.celrep.2017.07.055.

    Article  CAS  PubMed  Google Scholar 

  51. Ma Q. Role of Nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol. 2013;53:401–26. https://doi.org/10.1146/annurev-pharmtox-011112-140320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Cuadrado A, Rojo AI, Wells G, Hayes JD, Cousin SP, Rumsey WL, Attucks OC, Franklin S, Levonen A-L, Kensler TW. Dinkova-Kostova, therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov. 2019;18:295–317. https://doi.org/10.1038/s41573-018-0008-x.

    Article  CAS  PubMed  Google Scholar 

  53. Suzuki T, Yamamoto M. Molecular basis of the Keap1–Nrf2 system. Free Radic Biol Med. 2015;88:93–100. https://doi.org/10.1016/j.freeradbiomed.2015.06.006.

    Article  CAS  PubMed  Google Scholar 

  54. Tu W, Wang H, Li S, Liu Q, Sha H. The anti-inflammatory and anti-oxidant mechanisms of the Keap1/Nrf2/ARE Signaling Pathway in Chronic diseases. Aging Dis. 2019;10:637. https://doi.org/10.14336/AD.2018.0513.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res. 2021;31:107–25. https://doi.org/10.1038/s41422-020-00441-1.

    Article  CAS  PubMed  Google Scholar 

  56. Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019;23:101107. https://doi.org/10.1016/j.redox.2019.101107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, Tang D. Activation of the p62-Keap1‐NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63:173–84. https://doi.org/10.1002/hep.28251.

    Article  CAS  PubMed  Google Scholar 

  58. Sato H, Nomura S, Maebara K, Sato K, Tamba M, Bannai S. Transcriptional control of cystine/glutamate transporter gene by amino acid deprivation. Biochem Biophys Res Commun. 2004;325:109–16. https://doi.org/10.1016/j.bbrc.2004.10.009.

    Article  CAS  PubMed  Google Scholar 

  59. Jiang H, Wang C, Zhang A, Li Y, Li J, Li Z, Yang X, Hou Y. ATF4 protects against sorafenib-induced cardiotoxicity by suppressing ferroptosis. Biomed Pharmacother. 2022;153:113280. https://doi.org/10.1016/j.biopha.2022.113280.

    Article  CAS  PubMed  Google Scholar 

  60. Vyas VK, Ghate M. CoMFA and CoMSIA studies on Aryl Carboxylic Acid Amide derivatives as Dihydroorotate dehydrogenase (DHODH) inhibitors. Curr Comput Aided-Drug Des. 2012;8:271–82. https://doi.org/10.2174/157340912803519598.

    Article  CAS  PubMed  Google Scholar 

  61. Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, Poyurovsky MV, Olszewski K, Gan B. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593:586–90. https://doi.org/10.1038/s41586-021-03539-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang X, Ma H, Sun J, Zheng T, Zhao P, Li H, Yang M. Mitochondrial Ferritin Deficiency promotes Osteoblastic Ferroptosis Via Mitophagy in type 2 Diabetic osteoporosis. Biol Trace Elem Res. 2022;200:298–307. https://doi.org/10.1007/s12011-021-02627-z.

    Article  CAS  PubMed  Google Scholar 

  63. Marshall KR, Gong M, Wodke L, Lamb JH, Jones DJL, Farmer PB, Scrutton NS, Munro AW. The human apoptosis-inducing protein AMID is an oxidoreductase with a modified Flavin Cofactor and DNA binding activity. J Biol Chem. 2005;280:30735–40. https://doi.org/10.1074/jbc.M414018200.

    Article  CAS  PubMed  Google Scholar 

  64. Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, Mourão A, Buday K, Sato M, Wanninger J, Vignane T, Mohana V, Rehberg M, Flatley A, Schepers A, Kurz A, White D, Sauer M, Sattler M, Tate EW, Schmitz W, Schulze A, O’Donnell V, Proneth B, Popowicz GM, Pratt DA, Angeli JPF, Conrad M. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693–8. https://doi.org/10.1038/s41586-019-1707-0.

    Article  CAS  PubMed  Google Scholar 

  65. Lin Z, Yang X, Guan L, Qin L, Ding J, Zhou L. The link between ferroptosis and airway inflammatory diseases: a novel target for treatment. Front Mol Biosci. 2022;9. https://doi.org/10.3389/fmolb.2022.985571.

  66. Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, Bassik MC, Nomura DK, Dixon SJ, Olzmann JA. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688–92. https://doi.org/10.1038/s41586-019-1705-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Krümmel B, von Hanstein A-S, Plötz T, Lenzen S, Mehmeti I. Differential effects of saturated and unsaturated free fatty acids on ferroptosis in rat β-cells. J Nutr Biochem. 2022;106:109013. https://doi.org/10.1016/j.jnutbio.2022.109013.

    Article  CAS  PubMed  Google Scholar 

  68. Ni Z, Li Y, Song D, Ding J, Mei S, Sun S, Cheng W, Yu J, Zhou L, Kuang Y, Li M, Cai Z, Yu C. Iron-overloaded follicular fluid increases the risk of endometriosis-related infertility by triggering granulosa cell ferroptosis and oocyte dysmaturity. Cell Death Dis. 2022;13:579. https://doi.org/10.1038/s41419-022-05037-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lane NE. Epidemiology, etiology, and diagnosis of osteoporosis. Am J Obstet Gynecol. 2006;194:S3–11. https://doi.org/10.1016/j.ajog.2005.08.047.

    Article  CAS  PubMed  Google Scholar 

  70. Armas LAG, Recker RR. Pathophysiology of osteoporosis. Endocrinol Metab Clin North Am. 2012;41:475–86. https://doi.org/10.1016/j.ecl.2012.04.006.

    Article  CAS  PubMed  Google Scholar 

  71. D’Amelio P, Cristofaro MA, Tamone C, Morra E, Di Bella S, Isaia G, Grimaldi A, Gennero L, Gariboldi A, Ponzetto A, Pescarmona GP, Isaia GC. Role of iron metabolism and oxidative damage in postmenopausal bone loss. Bone. 2008;43:1010–5. https://doi.org/10.1016/j.bone.2008.08.107.

    Article  CAS  PubMed  Google Scholar 

  72. Tsay J, Yang Z, Ross FP, Cunningham-Rundles S, Lin H, Coleman R, Mayer-Kuckuk P, Doty SB, Grady RW, Giardina PJ, Boskey AL, Vogiatzi MG. Bone loss caused by iron overload in a murine model: importance of oxidative stress. Blood. 2010;116:2582–9. https://doi.org/10.1182/blood-2009-12-260083.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Jeney V. Clinical impact and Cellular mechanisms of Iron Overload-Associated Bone loss. Front Pharmacol. 2017;8. https://doi.org/10.3389/fphar.2017.00077.

  74. Li X, Zeng J, Liu Y, Liang M, Liu Q, Li Z, Zhao X, Chen D. Inhibitory effect and mechanism of action of Quercetin and Quercetin Diels-Alder Anti-dimer on Erastin-Induced ferroptosis in bone marrow-derived mesenchymal stem cells. Antioxid (Basel). 2020;9. https://doi.org/10.3390/antiox9030205.

  75. Huang L, Bian M, Lu S, Wang J, Yu J, Jiang L, Zhang J. Engeletin alleviates erastin-induced oxidative stress and protects against ferroptosis via Nrf2/Keap1 pathway in bone marrow mesenchymal stem cells. Tissue Cell. 2023;82:102040. https://doi.org/10.1016/j.tice.2023.102040.

    Article  CAS  PubMed  Google Scholar 

  76. Liu J, Ren Z, Yang L, Zhu L, Li Y, Bie C, Liu H, Ji Y, Chen D, Zhu M, Kuang W. The NSUN5-FTH1/FTL pathway mediates ferroptosis in bone marrow-derived mesenchymal stem cells. Cell Death Discov. 2022;8:99. https://doi.org/10.1038/s41420-022-00902-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kim J-M, Lin C, Stavre Z, Greenblatt MB, Shim J-H. Osteoblast-osteoclast communication and Bone Homeostasis. Cells. 2020;9:2073. https://doi.org/10.3390/cells9092073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kawane T, Qin X, Jiang Q, Miyazaki T, Komori H, A. C, dos K. V, Sakane C, Matsuo Y, Nagai K, Maeno T, Date Y, Nishimura R, Komori T. Runx2 is required for the proliferation of osteoblast progenitors and induces proliferation by regulating Fgfr2 and Fgfr3. Sci Rep. 2018;8:13551. https://doi.org/10.1038/s41598-018-31853-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Balogh E E., Tolnai, Nagy B, Nagy B, Balla G, Balla J, Jeney V. Iron overload inhibits osteogenic commitment and differentiation of mesenchymal stem cells via the induction of ferritin. Biochim et Biophys Acta (BBA) - Mol Basis Disease. 2016;1862:1640–9. https://doi.org/10.1016/j.bbadis.2016.06.003.

  80. Chen Y-C, Hsiao J-K, Liu H-M, Lai I-Y, Yao M, Hsu S-C, Ko B-S, Chen Y-C, Yang C-S, Huang D-M. The inhibitory effect of superparamagnetic iron oxide nanoparticle (Ferucarbotran) on osteogenic differentiation and its signaling mechanism in human mesenchymal stem cells. Toxicol Appl Pharmacol. 2010;245:272–9. https://doi.org/10.1016/j.taap.2010.03.011.

    Article  CAS  PubMed  Google Scholar 

  81. Ma H, Wang X, Zhang W, Li H, Zhao W, Sun J, Yang M. Melatonin suppresses Ferroptosis Induced by high glucose via activation of the Nrf2/HO-1 signaling pathway in type 2 Diabetic osteoporosis. Oxid Med Cell Longev. 2020;2020:1–18. https://doi.org/10.1155/2020/9067610.

    Article  CAS  Google Scholar 

  82. Zhang Z, Ji C, Wang Y-N, Liu S, Wang M, Xu X, Zhang D. Maresin1 suppresses high-glucose-Induced ferroptosis in Osteoblasts via NRF2 activation in type 2 Diabetic osteoporosis. Cells. 2022;11:2560. https://doi.org/10.3390/cells11162560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Song X, Long D. Nrf2 and ferroptosis: a New Research Direction for neurodegenerative diseases. Front Neurosci. 2020;14. https://doi.org/10.3389/fnins.2020.00267.

  84. Jiang Z, Wang H, Qi G, Jiang C, Chen K, Yan Z. Iron overload-induced ferroptosis of osteoblasts inhibits osteogenesis and promotes osteoporosis: an in vitro and in vivo study. IUBMB Life. 2022;74:1052–69. https://doi.org/10.1002/iub.2656.

    Article  CAS  PubMed  Google Scholar 

  85. Feng Y, He P-Y, Kong W-D, Cen W-J, Wang P-L, Liu C, Zhang W, Li S-S, Jiang J-W. Apoptosis-promoting properties of mir-3074-5p in MC3T3-E1 cells under iron overload conditions. Cell Mol Biol Lett. 2021;26:37. https://doi.org/10.1186/s11658-021-00281-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Hengwu Z, Wenjuan W, Shengwu C. Iron overload downregulates osteoblast function via ASK1-p38 pathway-mediated ferroptosis. J China Med Univ. 2021;50. https://doi.org/10.12007/j.issn.0258-4646.2021.06.011.

  87. Lin Y, Shen X, Ke Y, Lan C, Chen X, Liang B, Zhang Y, Yan S. Activation of osteoblast ferroptosis via the METTL3/ASK1-p38 signaling pathway in high glucose and high fat (HGHF)‐induced diabetic bone loss. FASEB J. 2022;36. https://doi.org/10.1096/fj.202101610R.

  88. Liu M, Wu X, Cui Y, Liu P, Xiao B, Zhang X, Zhang J, Sun Z, Song M, Shao B, Li Y. Mitophagy and apoptosis mediated by ROS participate in AlCl3-induced MC3T3-E1 cell dysfunction. Food Chem Toxicol. 2021;155:112388. https://doi.org/10.1016/j.fct.2021.112388.

    Article  CAS  PubMed  Google Scholar 

  89. Kalpage HA, Bazylianska V, Recanati MA, Fite A, Liu J, Wan J, Mantena N, Malek MH, Podgorski I, Heath EI, Vaishnav A, Edwards BF, Grossman LI, Sanderson TH, Lee I, Huttemann M. Tissue-specific regulation of cytochrome c by post‐translational modifications: respiration, the mitochondrial membrane potential, ROS, and apoptosis. FASEB J. 2019;33:1540–53. https://doi.org/10.1096/fj.201801417R.

    Article  CAS  PubMed  Google Scholar 

  90. Xu P, Lin B, Deng X, Huang K, Zhang Y, Wang N. VDR activation attenuates osteoblastic ferroptosis and senescence by stimulating the Nrf2/GPX4 pathway in age-related osteoporosis. Free Radic Biol Med. 2022;193:720–35. https://doi.org/10.1016/j.freeradbiomed.2022.11.013.

    Article  CAS  PubMed  Google Scholar 

  91. Datta HK, Ng WF, Walker JA, Tuck SP, Varanasi SS. The cell biology of bone metabolism. J Clin Pathol. 2008;61:577–87. https://doi.org/10.1136/jcp.2007.048868.

    Article  CAS  PubMed  Google Scholar 

  92. Charles JF, Aliprantis AO. Osteoclasts: more than ‘bone eaters’. Trends Mol Med. 2014;20:449–59. https://doi.org/10.1016/j.molmed.2014.06.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zarjou A, Jeney V, Arosio P, Poli M, Zavaczki E, Balla G, Balla J. Ferritin ferroxidase activity: a potent inhibitor of osteogenesis. J Bone Miner Res. 2010;25:164–72. https://doi.org/10.1359/jbmr.091002.

    Article  CAS  PubMed  Google Scholar 

  94. Ishii K, Fumoto T, Iwai K, Takeshita S, Ito M, Shimohata N, Aburatani H, Taketani S, Lelliott CJ, Vidal-Puig A, Ikeda K. Coordination of PGC-1β and iron uptake in mitochondrial biogenesis and osteoclast activation. Nat Med. 2009;15:259–66. https://doi.org/10.1038/nm.1910..

    Article  CAS  PubMed  Google Scholar 

  95. Balogh E, Paragh G, Jeney V. Influence of Iron on Bone Homeostasis. Pharmaceuticals. 2018;11:107. https://doi.org/10.3390/ph11040107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhang J, Zhao H, Yao G, Qiao P, Li L, Wu S. Therapeutic potential of iron chelators on osteoporosis and their cellular mechanisms. Biomed Pharmacother. 2021;137:111380. https://doi.org/10.1016/j.biopha.2021.111380.

    Article  CAS  PubMed  Google Scholar 

  97. Wang L, Fang B, Fujiwara T, Krager K, Gorantla A, Li C, Feng JQ, Jennings ML, Zhou J, Aykin-Burns N, Zhao H. Deletion of ferroportin in murine myeloid cells increases iron accumulation and stimulates osteoclastogenesis in vitro and in vivo. J Biol Chem. 2018;293:9248–64. https://doi.org/10.1074/jbc.RA117.000834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Elson A, Anuj A, Barnea-Zohar M, Reuven N. The origins and formation of bone-resorbing osteoclasts. Bone. 2022;164:116538. https://doi.org/10.1016/j.bone.2022.116538.

    Article  CAS  PubMed  Google Scholar 

  99. Yang J, Dong D, Luo X, Zhou J, Shang P, Zhang H. Iron Overload-Induced Osteocyte apoptosis stimulates osteoclast differentiation through increasing osteocytic RANKL production in Vitro. Calcif Tissue Int. 2020;107:499–509. https://doi.org/10.1007/s00223-020-00735-x.

    Article  CAS  PubMed  Google Scholar 

  100. Zhang Y, Huang X, Qi B, Sun C, Sun K, Liu N, Zhu L, Wei X. Ferroptosis and musculoskeletal diseases: Iron Maiden cell death may be a promising therapeutic target. Front Immunol. 2022;13. https://doi.org/10.3389/fimmu.2022.972753.

  101. Yongyun C, Jingwei Z, Zhiqing L, Wenxiang C, Huiwu L. Andrographolide stimulates osteoblastogenesis and bone formation by inhibiting nuclear factor kappa-Β signaling both in vivo and in vitro. J Orthop Translat. 2019;19:47–57. https://doi.org/10.1016/j.jot.2019.02.001.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Qu X, Sun Z, Wang Y, Ong H.S. Zoledronic acid promotes osteoclasts ferroptosis by inhibiting FBXO9-mediated p53 ubiquitination and degradation. PeerJ. 2021;9:e12510. https://doi.org/10.7717/peerj.12510.

  103. 2. Classification and diagnosis of diabetes: standards of Medical Care in Diabetes—2021. Diabetes Care. 2021;44:S15–33. https://doi.org/10.2337/dc21-S002.

    Article  Google Scholar 

  104. Basuli D, Stevens RG, Torti FM, Torti SV. Epidemiological associations between iron and cardiovascular disease and diabetes. Front Pharmacol. 2014. https://doi.org/10.3389/fphar.2014.00117.

    Article  Google Scholar 

  105. Simcox JA, McClain DA, Iron, Risk D. Cell Metab. 2013;17:329–41. https://doi.org/10.1016/j.cmet.2013.02.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Canturk Z, Çetinarslan B, Tarkun İ, Zafer Canturk N. Serum ferritin levels in poorly- and well‐controlled diabetes Mellitus. Endocr Res. 2003;29:299–306. https://doi.org/10.1081/ERC-120025037.

    Article  CAS  PubMed  Google Scholar 

  107. Rodríguez-Pérez C, Vrhovnik P, González-Alzaga B, Fernández MF, Martin-Olmedo P, Olea N, Fiket Ž, Kniewald G, Arrebola JP. Socio-demographic, lifestyle, and dietary determinants of essential and possibly-essential trace element levels in adipose tissue from an adult cohort. Environ Pollut. 2018;236:878–88. https://doi.org/10.1016/j.envpol.2017.09.093.

    Article  CAS  PubMed  Google Scholar 

  108. Kusminski CM, Ghaben AL, Morley TS, Samms RJ, Adams AC, An Y, Johnson JA, Joffin N, Onodera T, Crewe C, Holland WL, Gordillo R, Scherer PE. A Novel Model of Diabetic complications: Adipocyte mitochondrial dysfunction triggers massive β-Cell Hyperplasia. Diabetes. 2020;69:313–30. https://doi.org/10.2337/db19-0327.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Dongiovanni P, Ruscica M, Rametta R, Recalcati S, Steffani L, Gatti S, Girelli D, Cairo G, Magni P, Fargion S, Valenti L. Dietary iron overload induces visceral adipose tissue insulin resistance. Am J Pathol. 2013;182:2254–63. https://doi.org/10.1016/j.ajpath.2013.02.019.

    Article  CAS  PubMed  Google Scholar 

  110. Pietrangelo A. Iron and the liver. Liver Int. 2016;36(Suppl 1):116–23. https://doi.org/10.1111/liv.13020.

    Article  CAS  PubMed  Google Scholar 

  111. Ambachew S, Biadgo B. Hepcidin in Iron Homeostasis: diagnostic and therapeutic implications in type 2 diabetes Mellitus patients. Acta Haematol. 2017;138:183–93. https://doi.org/10.1159/000481391.

    Article  CAS  PubMed  Google Scholar 

  112. Fenton H, Torbenson M, Vivekanandan P, Yeh MM, Hart J, Ferrell L. Marked iron in liver explants in the absence of major hereditary hemochromatosis gene defects: a risk factor for cardiac failure. Transplantation. 2009;87:1256–60. https://doi.org/10.1097/TP.0b013e31819f2280.

    Article  PubMed  Google Scholar 

  113. Anderson ER, Shah YM. Iron Homeostasis in the liver. Compr Physiol. Wiley; 2013. pp. 315–30. https://doi.org/10.1002/cphy.c120016.

  114. Deng C, Cao J, Han J, Li J, Li Z, Shi N, He J. Liraglutide activates the Nrf2/HO-1 antioxidant pathway and protects brain nerve cells against cerebral ischemia in Diabetic rats. Comput Intell Neurosci. 2018;2018:3094504. https://doi.org/10.1155/2018/3094504.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Song J-X, An J-R, Chen Q, Yang X-Y, Jia C-L, Xu S, Zhao Y, Ji E-S. Liraglutide attenuates hepatic iron levels and ferroptosis in db/db mice. Bioengineered. 2022;13:8334–48. https://doi.org/10.1080/21655979.2022.2051858.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zhang T, Wang M-Y, Wang G-D, Lv Q-Y, Huang Y-Q, Zhang P, Wang W, Zhang Y, Bai Y-P, Guo L-Q. Metformin improves nonalcoholic fatty liver disease in db/db mice by inhibiting ferroptosis. Eur J Pharmacol. 2024;966:176341. https://doi.org/10.1016/j.ejphar.2024.176341.

    Article  CAS  PubMed  Google Scholar 

  117. Chen J, Li X, Ge C, Min J, Wang F. The multifaceted role of ferroptosis in liver disease. Cell Death Differ. 2022;29:467–80. https://doi.org/10.1038/s41418-022-00941-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sha W, Hu F, Xi Y, Chu Y, Bu S. Mechanism of Ferroptosis and its role in type 2 diabetes Mellitus. J Diabetes Res. 2021;2021:1–10. https://doi.org/10.1155/2021/9999612.

    Article  CAS  Google Scholar 

  119. Hansen JB, Moen IW, Mandrup-Poulsen T. Iron: the hard player in diabetes pathophysiology, Acta Physiologica. 210 (2014) 717–732. Distribution of DMT 1 within the human glandular system., Histol Histopathol. 18 (2003) https://doi.org/10.1111/apha.12256.

  120. Koch RO, Zoller H, Theuri I, Obrist P, Egg G, Strohmayer W, Vogel W, Weiss G. 1095–101. https://doi.org/10.14670/HH-18.1095.

  121. Marku A, Galli A, Marciani P, Dule N, Perego C, Castagna M. Iron Metabolism in pancreatic Beta-cell function and dysfunction. Cells. 2021;10:2841. https://doi.org/10.3390/cells10112841.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Gkouvatsos K, Papanikolaou G, Pantopoulos K. Regulation of iron transport and the role of transferrin. Biochim et Biophys Acta (BBA) - Gen Subj. 2012;1820:188–202. https://doi.org/10.1016/j.bbagen.2011.10.013.

    Article  CAS  Google Scholar 

  123. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, Ganz T, Kaplan J. Hepcidin regulates Cellular Iron Efflux by binding to Ferroportin and Inducing its internalization. Science. 1979;306(2004):2090–3. https://doi.org/10.1126/science.1104742.

    Article  CAS  Google Scholar 

  124. Kulaksiz H, Fein E, Redecker P, Stremmel W, Adler G, Cetin Y. Pancreatic β-cells express hepcidin, an iron-uptake regulatory peptide. J Endocrinol. 2008;197:241–9. https://doi.org/10.1677/JOE-07-0528.

    Article  CAS  PubMed  Google Scholar 

  125. Shu T, Lv Z, Xie Y, Tang J, Mao X. Hepcidin as a key iron regulator mediates glucotoxicity-induced pancreatic β-cell dysfunction. Endocr Connect. 2019;8:150–61. https://doi.org/10.1530/EC-18-0516.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Aigner E, Felder TK, Oberkofler H, Hahne P, Auer S, Soyal S, Stadlmayr A, Schwenoha K, Pirich C, Hengster P, Datz C, Patsch W. Glucose acts as a regulator of serum iron by increasing serum hepcidin concentrations. J Nutr Biochem. 2013;24:112–7. https://doi.org/10.1016/j.jnutbio.2012.02.017.

    Article  CAS  PubMed  Google Scholar 

  127. Soe-Lin S, Apte SS, Andriopoulos B, Andrews MC, Schranzhofer M, Kahawita T, Garcia-Santos D, Ponka P. Nramp1 promotes efficient macrophage recycling of iron following erythrophagocytosis in vivo. Proc Natl Acad Sci. 2009;106:5960–5. https://doi.org/10.1073/pnas.0900808106.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Wang J, Wang H. Oxidative stress in pancreatic Beta cell regeneration. Oxid Med Cell Longev. 2017;2017:1–9. https://doi.org/10.1155/2017/1930261.

    Article  CAS  Google Scholar 

  129. Krümmel B, Plötz T, Jörns A, Lenzen S, Mehmeti I. The central role of glutathione peroxidase 4 in the regulation of ferroptosis and its implications for pro-inflammatory cytokine-mediated beta-cell death, Biochimica et Biophysica Acta (BBA) - molecular basis of Disease. 1867 (2021) 166114. https://doi.org/10.1016/j.bbadis.2021.166114.

  130. Bruni A, Pepper AR, Pawlick RL, Gala-Lopez B, Gamble AF, Kin T, Seeberger K, Korbutt GS, Bornstein SR, Linkermann A, Shapiro AMJ. Ferroptosis-inducing agents compromise in vitro human islet viability and function. Cell Death Dis. 2018;9:595. https://doi.org/10.1038/s41419-018-0506-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Zhou Y. The Protective effects of Cryptochlorogenic Acid on β-Cells function in diabetes in vivo and vitro via inhibition of Ferroptosis. Diabetes Metab Syndr Obes Volume 13 (2020) 1921–31. https://doi.org/10.2147/DMSO.S249382.<\/p>

    Article  CAS  Google Scholar 

  132. Li D, Jiang C, Mei G, Zhao Y, Chen L, Liu J, Tang Y, Gao C, Yao P. Quercetin alleviates ferroptosis of pancreatic β cells in type 2 diabetes. Nutrients. 2020;12:2954. https://doi.org/10.3390/nu12102954.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Yamaguchi K, Takeda K, Kadowaki H, Ueda I, Namba Y, Ouchi Y, Nishitoh H, Ichijo H. Involvement of ASK1–p38 pathway in the pathogenesis of diabetes triggered by pancreatic ß cell exhaustion. Biochim et Biophys Acta (BBA) - Gen Subj. 2013;1830:3656–63. https://doi.org/10.1016/j.bbagen.2013.01.029.

    Article  CAS  Google Scholar 

  134. Deng L, Mo M-Q, Zhong J, Li Z, Li G, Liang Y. Iron overload induces islet β cell ferroptosis by activating ASK1/P-P38/CHOP signaling pathway, PeerJ. 11 (2023) e15206. https://doi.org/10.7717/peerj.15206.

  135. Ansari IH, Longacre MJ, Stoker SW, Kendrick MA, O’Neill LM, Zitur LJ, Fernandez LA, Ntambi JM, MacDonald MJ. Characterization of Acyl-CoA synthetase isoforms in pancreatic beta cells: gene silencing shows participation of ACSL3 and ACSL4 in insulin secretion. Arch Biochem Biophys. 2017;618:32–43. https://doi.org/10.1016/j.abb.2017.02.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Killion EA, Reeves AR, El Azzouny MA, Yan Q-W, Surujon D, Griffin JD, Bowman TA, Wang C, Matthan NR, Klett EL, Kong D, Newman JW, Han X, Lee M-J, Coleman RA, Greenberg AS. A role for long-chain acyl-CoA synthetase-4 (ACSL4) in diet-induced phospholipid remodeling and obesity-associated adipocyte dysfunction. Mol Metab. 2018;9:43–56. https://doi.org/10.1016/j.molmet.2018.01.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Liu Y, Tavana O, Gu W. p53 modifications: exquisite decorations of the powerful guardian. J Mol Cell Biol. 2019;11:564–77. https://doi.org/10.1093/jmcb/mjz060.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Yuan H, Zhang X, Huang X, Lu Y, Tang W, Man Y, Wang S, Xi J, Li J. Oxidase 2-Derived reactive Oxygen species Mediate FFAs-Induced Dysfunction and apoptosis of β-Cells via JNK, p38 MAPK and p53 pathways. PLoS ONE. 2010;5:e15726. https://doi.org/10.1371/journal.pone.0015726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Minamino T, Orimo M, Shimizu I, Kunieda T, Yokoyama M, Ito T, Nojima A, Nabetani A, Oike Y, Matsubara H, Ishikawa F, Komuro I. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med. 2009;15:1082–7. https://doi.org/10.1038/nm.2014.

    Article  CAS  PubMed  Google Scholar 

  140. Brigelius-Flohé R, Maiorino M. Glutathione peroxidases, Biochimica et Biophysica Acta (BBA) - general subjects. 1830 (2013) 3289–303. https://doi.org/10.1016/j.bbagen.2012.11.020.

  141. Ommati MM, Amjadinia A, Mousavi K, Azarpira N, Jamshidzadeh A, Heidari R. N-acetyl cysteine treatment mitigates biomarkers of oxidative stress in different tissues of bile duct ligated rats. Stress. 2021;24:213–28. https://doi.org/10.1080/10253890.2020.1777970.

    Article  CAS  PubMed  Google Scholar 

  142. Pasupuleti VR, Arigela CS, Gan SH, Salam SKN, Krishnan KT, Rahman NA, Jeffree MS. A Review on Oxidative Stress, Diabetic Complications, and the Roles of Honey Polyphenols, Oxid Med Cell Longev. 2020 (2020) 1–16. https://doi.org/10.1155/2020/8878172.

  143. Uruno A, Furusawa Y, Yagishita Y, Fukutomi T, Muramatsu H, Negishi T, Sugawara A, Kensler TW, Yamamoto M. The Keap1-Nrf2 system prevents onset of diabetes Mellitus. Mol Cell Biol. 2013;33:2996–3010. https://doi.org/10.1128/MCB.00225-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Baumel-Alterzon S, Katz LS, Brill G, Jean-Pierre C, Li Y, Tse I, Biswal S, Garcia-Ocaña A, Scott DK. Nrf2 regulates β-Cell Mass by suppressing β-Cell death and promoting β-Cell proliferation. Diabetes. 2022;71:989–1011. https://doi.org/10.2337/db21-0581.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Bollong MJ, Lee G, Coukos JS, Yun H, Zambaldo C, Chang JW, Chin EN, Ahmad I, Chatterjee AK, Lairson LL, Schultz PG, Moellering RE. A metabolite-derived protein modification integrates glycolysis with KEAP1–NRF2 signalling. Nature. 2018;562:600–4. https://doi.org/10.1038/s41586-018-0622-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Bo J, Xie S, Guo Y, Zhang C, Guan Y, Li C, Lu J, Meng QH. Methylglyoxal impairs insulin secretion of pancreatic β -Cells through increased production of ROS and mitochondrial dysfunction mediated by Upregulation of UCP2 and MAPKs. J Diabetes Res. 2016;2016:1–14. https://doi.org/10.1155/2016/2029854.

    Article  CAS  Google Scholar 

  147. Oliveira HR, Verlengia R, Carvalho CRO, Britto LRG, Curi R, Carpinelli AR. Pancreatic β-Cells Express Phagocyte-Like NAD(P)H oxidase, diabetes. 52 (2003) 1457–63. https://doi.org/10.2337/diabetes.52.6.1457.

  148. Uchizono Y, Takeya R, Iwase M, Sasaki N, Oku M, Imoto H, Iida M, Sumimoto H. Expression of isoforms of NADPH oxidase components in rat pancreatic islets. Life Sci. 2006;80:133–9. https://doi.org/10.1016/j.lfs.2006.08.031.

    Article  CAS  PubMed  Google Scholar 

  149. Morgan D, Oliveira-Emilio HR, Keane D, Hirata AE, Santos da Rocha M, Bordin S, Curi R, Newsholme P, Carpinelli AR. Glucose, palmitate and pro-inflammatory cytokines modulate production and activity of a phagocyte-like NADPH oxidase in rat pancreatic islets and a clonal beta cell line. Diabetologia. 2007;50:359–69. https://doi.org/10.1007/s00125-006-0462-6.

    Article  CAS  PubMed  Google Scholar 

  150. Newsholme P, Morgan D, Rebelato E, Oliveira-Emilio HC, Procopio J, Curi R, Carpinelli A. Insights into the critical role of NADPH oxidase(s) in the normal and dysregulated pancreatic beta cell. Diabetologia. 2009;52:2489–98. https://doi.org/10.1007/s00125-009-1536-z.

    Article  CAS  PubMed  Google Scholar 

  151. Syed I, Kyathanahalli CN, Jayaram B, Govind S, Rhodes CJ, Kowluru RA, Kowluru A. Increased phagocyte-like NADPH oxidase and ROS generation in type 2 Diabetic ZDF rat and human islets. Diabetes. 2011;60:2843–52. https://doi.org/10.2337/db11-0809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Lastra G, Manrique C. The expanding role of oxidative stress, Renin Angiotensin System, and β-Cell dysfunction in the Cardiometabolic syndrome and type 2 diabetes Mellitus. Antioxid Redox Signal. 2007;9:943–54. https://doi.org/10.1089/ars.2007.1615.

    Article  CAS  PubMed  Google Scholar 

  153. Lupi R, Del Guerra S, Bugliani M, Boggi U, Mosca F, Torri S, Del Prato S, Marchetti P. The direct effects of the angiotensin-converting enzyme inhibitors, zofenoprilat and enalaprilat, on isolated human pancreatic islets. Eur J Endocrinol. 2006;154:355–61. https://doi.org/10.1530/eje.1.02086.

    Article  CAS  PubMed  Google Scholar 

  154. Chu KY, Lau T, Carlsson P-O, Leung PS. Angiotensin II type 1 receptor blockade improves β-Cell function and glucose tolerance in a mouse model of type 2 diabetes. Diabetes. 2006;55:367–74. https://doi.org/10.2337/diabetes.55.02.06.db05-1022.

    Article  CAS  PubMed  Google Scholar 

  155. Sauter NS, Thienel C, Plutino Y, Kampe K, Dror E, Traub S, Timper K, Bédat B, Pattou F, Kerr-Conte J, Jehle AW, Böni-Schnetzler M, Donath MY. Angiotensin II induces Interleukin-1β–Mediated islet inflammation and β-Cell dysfunction independently of Vasoconstrictive effects, diabetes. 64 (2015) 1273–83. https://doi.org/10.2337/db14-1282.

  156. Chu KY, Leung PS, Angiotensin II. Type 1 receptor antagonism mediates uncoupling protein 2-Driven oxidative stress and ameliorates pancreatic islet β-Cell function in Young Type 2 Diabetic mice. Antioxid Redox Signal. 2007;9:869–78. https://doi.org/10.1089/ars.2007.1590.

    Article  CAS  PubMed  Google Scholar 

  157. Karunakaran U, Elumalai S, Moon JS, Won KC. CD36 dependent redoxosomes promotes ceramide-mediated pancreatic β-cell failure via p66Shc activation. Free Radic Biol Med. 2019;134:505–15. https://doi.org/10.1016/j.freeradbiomed.2019.02.004.

    Article  CAS  PubMed  Google Scholar 

  158. Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, Karin M, Hotamisligil GS. A central role for JNK in obesity and insulin resistance. Nature. 2002;420:333–6. https://doi.org/10.1038/nature01137.

    Article  CAS  PubMed  Google Scholar 

  159. Geiszt M, Kopp JB, Várnai P, Leto TL. Identification of Renox, an NAD(P)H oxidase in kidney, Proceedings of the National Academy of Sciences. 97 (2000) 8010–8014. https://doi.org/10.1073/pnas.130135897.

  160. Gorin Y, Block K. Nox4 and diabetic nephropathy: with a friend like this, who needs enemies? Free Radic Biol Med. 2013;61:130–42. https://doi.org/10.1016/j.freeradbiomed.2013.03.014.

    Article  CAS  PubMed  Google Scholar 

  161. Zheng C-X, Sui B-D, Qiu X-Y, Hu C-H, Jin Y. Mitochondrial regulation of stem cells in bone homeostasis. Trends Mol Med. 2020;26:89–104. https://doi.org/10.1016/j.molmed.2019.04.008.

    Article  CAS  PubMed  Google Scholar 

  162. Atashi F, Modarressi A, Pepper MS. The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review. Stem Cells Dev. 2015;24:1150–63. https://doi.org/10.1089/scd.2014.0484.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Wang Z, Ding Y, Wang X, Lu S, Wang C, He C, Wang L, Piao M, Chi G, Luo Y, Ge P. Pseudolaric acid B triggers ferroptosis in glioma cells via activation of Nox4 and inhibition of xCT. Cancer Lett. 2018;428:21–33. https://doi.org/10.1016/j.canlet.2018.04.021.

    Article  CAS  PubMed  Google Scholar 

  164. Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB, Jiang X. Role of Mitochondria in Ferroptosis. Mol Cell. 2019;73:354–e3633. https://doi.org/10.1016/j.molcel.2018.10.042.

    Article  CAS  PubMed  Google Scholar 

  165. Li C-I, Liu C-S, Lin W-Y, Meng N-H, Chen C-C, Yang S-Y, Chen H-J, Lin C-C, Li T-C. Glycated hemoglobin level and risk of hip fracture in older people with type 2 diabetes: a competing risk analysis of Taiwan Diabetes Cohort Study. J Bone Miner Res. 2015;30:1338–46. https://doi.org/10.1002/jbmr.2462.

    Article  CAS  PubMed  Google Scholar 

  166. Lecka-Czernik B. Diabetes, bone and glucose-lowering agents: basic biology. Diabetologia. 2017;60:1163–9. https://doi.org/10.1007/s00125-017-4269-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Creecy A, Uppuganti S, Merkel AR, O’Neal D, Makowski AJ, Granke M, Voziyan P, Nyman JS. Changes in the fracture resistance of bone with the progression of type 2 diabetes in the ZDSD rat. Calcif Tissue Int. 2016;99:289–301. https://doi.org/10.1007/s00223-016-0149-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Bao J, Yan Y, Zuo D, Zhuo Z, Sun T, Lin H, Han Z, Zhao Z, Yu H. Iron metabolism and ferroptosis in diabetic bone loss: from mechanism to therapy. Front Nutr. 2023;10. https://doi.org/10.3389/fnut.2023.1178573.

  169. Deng X, Xu M, Shen M, Cheng J. Effects of type 2 Diabetic serum on proliferation and osteogenic differentiation of mesenchymal stem cells. J Diabetes Res. 2018;2018:1–9. https://doi.org/10.1155/2018/5765478.

    Article  CAS  Google Scholar 

  170. Li H, Wen Y, Liu P, Zhang L, Zhang X, Liu Y, Ma B, Kuang H, Wang J, Song L. Characteristics of bone metabolism in postmenopausal women with newly diagnosed type 2 diabetes mellitus. Clin Endocrinol (Oxf). 2021;95:430–8. https://doi.org/10.1111/cen.14501.

    Article  CAS  PubMed  Google Scholar 

  171. Pendyala S, Moitra J, Kalari S, Kleeberger SR, Zhao Y, Reddy SP, Garcia JGN, Natarajan V. Nrf2 regulates hyperoxia-induced Nox4 expression in human lung endothelium: identification of functional antioxidant response elements on the Nox4 promoter. Free Radic Biol Med. 2011;50:1749–59. https://doi.org/10.1016/j.freeradbiomed.2011.03.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding H-F, Zhang J, Wang H, Chen X, Yan C. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc–. Cell Death Differ. 2020;27:662–75. https://doi.org/10.1038/s41418-019-0380-z.

    Article  CAS  PubMed  Google Scholar 

  173. Franke S, Rüster C, Pester J, Hofmann G, Oelzner P, Wolf G. Advanced glycation end products affect growth and function of osteoblasts. Clin Exp Rheumatol. 2011;29:650–60.

    CAS  PubMed  Google Scholar 

  174. An Y, Zhang H, Wang C, Jiao F, Xu H, Wang X, Luan W, Ma F, Ni L, Tang X, Liu M, Guo W, Yu L. 3 and inhibition of efferocytosis in osteoclast‐mediated diabetic osteoporosis. FASEB J. 2019;33:12515–27. https://doi.org/10.1096/fj.201802805RR. Activation of ROS/MAPK < scp > s /NF- κ B/NLRP

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Suzuki K, Kurose T, Takizawa M, Maruyama M, Ushikawa K, Kikuyama M, Sugimoto C, Seino Y, Nagamatsu S, Ishida H. Osteoclastic function is accelerated in male patients with type 2 diabetes mellitus: the preventive role of osteoclastogenesis inhibitory factor/osteoprotegerin (OCIF/OPG) on the decrease of bone mineral density. Diabetes Res Clin Pract. 2005;68:117–25. https://doi.org/10.1016/j.diabres.2004.08.006.

    Article  CAS  PubMed  Google Scholar 

  176. Liu R, Bal HS, Desta T, Krothapalli N, Alyassi M, Luan Q, Graves DT. Diabetes enhances Periodontal Bone loss through enhanced resorption and diminished bone formation. J Dent Res. 2006;85:510–4. https://doi.org/10.1177/154405910608500606.

    Article  CAS  PubMed  Google Scholar 

  177. Alblowi J, Tian C, Siqueira MF, Kayal RA, McKenzie E, Behl Y, Gerstenfeld L, Einhorn TA, Graves DT. Chemokine expression is upregulated in chondrocytes in diabetic fracture healing. Bone. 2013;53:294–300. https://doi.org/10.1016/j.bone.2012.12.006.

    Article  CAS  PubMed  Google Scholar 

  178. Kayal RA, Tsatsas D, Bauer MA, Allen B, Al-Sebaei MO, Kakar S, Leone CW, Morgan EF, Gerstenfeld LC, Einhorn TA, Graves DT. Diminished bone formation during Diabetic Fracture Healing is related to the premature resorption of Cartilage Associated with increased osteoclast activity. J Bone Miner Res. 2007;22:560–8. https://doi.org/10.1359/jbmr.070115.

    Article  CAS  PubMed  Google Scholar 

  179. Wu Y-Y, Xiao E, Graves DT. Diabetes mellitus related bone metabolism and periodontal disease. Int J Oral Sci. 2015;7:63–72. https://doi.org/10.1038/ijos.2015.2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Werner SL, Sharma R, Woodruff K, Horn D, Harris SE, Gorin Y, Lee D, Hua R, Gu S, Fajardo RJ, Habib SL, Jiang JX. CSF-1 in Osteocytes inhibits Nox4‐mediated oxidative stress and promotes normal bone homeostasis, JBMR Plus. 4 (2020). https://doi.org/10.1002/jbm4.10080.

  181. Goettsch C, Babelova A, Trummer O, Erben RG, Rauner M, Rammelt S, Weissmann N, Weinberger V, Benkhoff S, Kampschulte M, Obermayer-Pietsch B, Hofbauer LC, Brandes RP, Schröder K. NADPH oxidase 4 limits bone mass by promoting osteoclastogenesis. J Clin Invest. 2013;123:4731–8. https://doi.org/10.1172/JCI67603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Dong XN, Qin A, Xu J, Wang X. In situ accumulation of advanced glycation endproducts (AGEs) in bone matrix and its correlation with osteoclastic bone resorption. Bone. 2011;49:174–83. https://doi.org/10.1016/j.bone.2011.04.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Ding K-H, Wang Z-Z, Hamrick MW, Deng Z-B, Zhou L, Kang B, Yan S-L, She J-X, Stern DM, Isales CM, Mi Q-S. Disordered osteoclast formation in RAGE-deficient mouse establishes an essential role for RAGE in diabetes related bone loss. Biochem Biophys Res Commun. 2006;340:1091–7. https://doi.org/10.1016/j.bbrc.2005.12.107.

    Article  CAS  PubMed  Google Scholar 

  184. Xie J, Méndez JD, Méndez-Valenzuela V, Aguilar-Hernández MM. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal. 2013;25:2185–97. https://doi.org/10.1016/j.cellsig.2013.06.013.

    Article  CAS  PubMed  Google Scholar 

  185. Abdollahi M, Larijani B, Rahimi R, Salari P. Role of oxidative stress in osteoporosis. Therapy 2. 2005;787–96. https://doi.org/10.2217/14750708.2.5.787.

  186. Jiao H, Xiao E, Graves DT. Diabetes and its Effect on Bone and Fracture Healing. Curr Osteoporos Rep. 2015;13:327–35. https://doi.org/10.1007/s11914-015-0286-8.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Cai Z, Yang B, Shi Y, Zhang W, 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;503:428–35. https://doi.org/10.1016/j.bbrc.2018.04.052.

    Article  CAS  PubMed  Google Scholar 

  188. Qin Y, Qiao Y, Wang D, Tang C, Yan G. Ferritinophagy and ferroptosis in cardiovascular disease: mechanisms and potential applications. Biomed Pharmacother. 2021;141:111872. https://doi.org/10.1016/j.biopha.2021.111872.

    Article  CAS  PubMed  Google Scholar 

  189. Arnett TR, Orriss IR. Metabolic properties of the osteoclast. Bone. 2018;115:25–30. https://doi.org/10.1016/j.bone.2017.12.021.

    Article  CAS  PubMed  Google Scholar 

  190. Ni S, Yuan Y, Qian Z, Zhong Z, Lv T, Kuang Y, Yu B. Hypoxia inhibits RANKL-induced ferritinophagy and protects osteoclasts from ferroptosis. Free Radic Biol Med. 2021;169:271–82. https://doi.org/10.1016/j.freeradbiomed.2021.04.027.

    Article  CAS  PubMed  Google Scholar 

  191. Jin Y, Wu S, Zhang L, Yao G, Zhao H, Qiao P, Zhang J. Artesunate inhibits osteoclast differentiation by inducing ferroptosis and prevents iron overload-induced bone loss. Basic Clin Pharmacol Toxicol. 2023;132:144–53. https://doi.org/10.1111/bcpt.13817.

    Article  CAS  PubMed  Google Scholar 

  192. Razny U, Polus A, Goralska J, Zdzienicka A, Gruca A, Kapusta M, Biela M, Dembinska-Kiec A, Solnica B, Malczewska-Malec M. Effect of insulin resistance on whole blood mRNA and microRNA expression affecting bone turnover. Eur J Endocrinol. 2019;181:525–37. https://doi.org/10.1530/EJE-19-0542.

    Article  CAS  PubMed  Google Scholar 

  193. Epstein S, LeRoith D. Diabetes and fragility fractures — a burgeoning epidemic? Bone. 2008;43:3–6. https://doi.org/10.1016/j.bone.2008.03.017.

    Article  CAS  PubMed  Google Scholar 

  194. Siddappa R, Mulder W, Steeghs I, van de Klundert C, Fernandes H, Liu J, Arends R, van Blitterswijk C, de Boer J. cAMP/PKA signaling inhibits osteogenic differentiation and bone formation in Rodent models. Tissue Eng Part A. 2009;15:2135–43. https://doi.org/10.1089/ten.tea.2008.0512.

    Article  CAS  PubMed  Google Scholar 

  195. DeFranco DJ, Glowacki J, Cox KA, Lian JB. Normal bone particles are preferentially resorbed in the presence of osteocalcin-deficient bone particlesIn vivo. Calcif Tissue Int. 1991;49:43–50. https://doi.org/10.1007/BF02555901.

    Article  CAS  PubMed  Google Scholar 

  196. Irwin R, Lin HV, Motyl KJ, McCabe LR. Normal bone density obtained in the absence of insulin receptor expression in bone, endocrinology. 147 (2006) 5760–7. https://doi.org/10.1210/en.2006-0700.

  197. Zhao Y, Du Y, Gao Y, Xu Z, Zhao D, Yang M. ATF3 Regulates Osteogenic Function by Mediating Osteoblast Ferroptosis in Type 2 Diabetic Osteoporosis, Dis Markers. 2022 (2022) 1–17. https://doi.org/10.1155/2022/9872243.

  198. Li Y, Liu Y, Liu S, Gao M, Wang W, Chen K, Huang L, Liu Y. Diabetic vascular diseases: molecular mechanisms and therapeutic strategies. Signal Transduct Target Ther. 2023;8:152. https://doi.org/10.1038/s41392-023-01400-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Lafage-Proust M-H, Roche B, Langer M, Cleret D, Vanden Bossche A, Olivier T, Vico L. Assessment of bone vascularization and its role in bone remodeling. Bonekey Rep. 2015;4. https://doi.org/10.1038/bonekey.2015.29.

  200. Tanikawa T, Okada Y, Tanikawa R, Tanaka Y. Advanced Glycation End products induce calcification of vascular smooth muscle cells through RAGE/p38 MAPK. J Vasc Res. 2009;46:572–80. https://doi.org/10.1159/000226225.

    Article  CAS  PubMed  Google Scholar 

  201. Ge W, Jie J, Yao J, Li W, Cheng Y, Lu W. Advanced glycation end products promote osteoporosis by inducing ferroptosis in osteoblasts. Mol Med Rep. 2022;25:140. https://doi.org/10.3892/mmr.2022.12656.

  202. Jang MK, Park HJ, Jung MH. ATF3 represses PDX-1 expression in pancreatic β-cells. Biochem Biophys Res Commun. 2011;412:385–90. https://doi.org/10.1016/j.bbrc.2011.07.108.

  203. Okamoto A, Iwamoto Y, Maru Y. Oxidative stress-responsive transcription factor ATF3 potentially mediates Diabetic Angiopathy. Mol Cell Biol. 2006;26:1087–97. https://doi.org/10.1128/MCB.26.3.1087-1097.2006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Kajarabille L-D. Programmed cell-death by Ferroptosis: antioxidants as Mitigators. Int J Mol Sci. 2019;20:4968. https://doi.org/10.3390/ijms20194968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Zhao Y, Mei G, Zhou F, Kong B, Chen L, Chen H, Wang L, Tang Y, Yao P. Vitamin D decreases pancreatic iron overload in type 2 diabetes through the NF-κB-DMT1 pathway. J Nutr Biochem. 2022;99:108870. https://doi.org/10.1016/j.jnutbio.2021.108870.

    Article  CAS  PubMed  Google Scholar 

  206. Litton E, Lim J. Iron Metabolism: an emerging therapeutic target in critical illness. Crit Care. 2019;23:81. https://doi.org/10.1186/s13054-019-2373-1.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang D. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 2020;66:89–100. https://doi.org/10.1016/j.semcancer.2019.03.002.

    Article  CAS  PubMed  Google Scholar 

  208. Xu C-Y, Xu C, Xu Y-N, Du S-Q, Dai Z-H, Jin S-Q, Zheng G, Xie C-L, Fang W-L. Poliumoside protects against type 2 diabetes-related osteoporosis by suppressing ferroptosis via activation of the Nrf2/GPX4 pathway. Phytomedicine. 2024;125:155342. https://doi.org/10.1016/j.phymed.2024.155342.

    Article  CAS  PubMed  Google Scholar 

  209. Li Y, Cai Z, Ma W, Bai L, Luo E, Lin Y. A DNA tetrahedron-based ferroptosis-suppressing nanoparticle: superior delivery of curcumin and alleviation of diabetic osteoporosis. Bone Res. 2024;12:14. https://doi.org/10.1038/s41413-024-00319-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Jin C, Tan K, Yao Z, Lin B, Zhang D, Chen W-K, Mao S, Zhang W, Chen L, Lin Z, Weng S, Bai B, Zheng W, Zheng G, Wu Z, Yang L. A novel anti-osteoporosis mechanism of VK2: interfering with ferroptosis via AMPK/SIRT1 Pathway in type 2 Diabetic osteoporosis. J Agric Food Chem. 2023;71:2745–61. https://doi.org/10.1021/acs.jafc.2c05632.

    Article  CAS  PubMed  Google Scholar 

  211. Lin B, Deng X, Xu P, Ye Q, Zhao G, Ye M, Wang N. Structural characterization and anti-osteoporosis effect of an arabinomannan from Anemarrhena asphodeloides Bge. Int J Biol Macromol. 2023;231:123324. https://doi.org/10.1016/j.ijbiomac.2023.123324.

    Article  CAS  PubMed  Google Scholar 

  212. Wang M, Liu Y, Gui H, Ma G, Li B, Zhang Z, Yu G, Wu A, Xu X, Zhang D, editors. ED-71 ameliorates bone regeneration in type 2 diabetes by reducing ferroptosis in osteoblasts via the HIF1α pathway, Eur J Pharmacol 969 (2024) 176303. https://doi.org/10.1016/j.ejphar.2023.176303.

  213. Xie D, Li K, Feng R, Xiao M, Sheng Z, Xie Y. Ferroptosis and traditional Chinese medicine for type 2 diabetes Mellitus. Diabetes Metab Syndr Obes. 2023;16:1915–30. https://doi.org/10.2147/DMSO.S412747.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Thanks to all authors for their contributions and funding support.

Funding

This research did not receive any specific grant from public, commercial, or not-for-profit sponsoring bodies.

Author information

Authors and Affiliations

Authors

Contributions

Yili Chen, Luyuan Chen and Haizhou Wang provided guidance throughout the manuscript writing process. Yili Chen and Wen Zhao performed the literature search and drafted the original manuscript. Yili Chen and An Hu created the graphs and proofread the manuscript. Wen Zhao and An Hu contributed to the manuscript drafting. Others provided valuable suggestions and critically revised the manuscript.

Corresponding authors

Correspondence to Luyuan Chen or Haizhou Wang.

Ethics declarations

Ethics approval and consent to participate

The paper has obtained consent for participation from the Guangdong Provincial Hospital of Traditional Chinese Medicine, as shown in Supplementary Material 2. And due to its nature as a review article, it does not involve relevant ethical considerations.

Consent for publication

The article has been approved for publication by all authors, shown in the Supplementary Material 2.

Competing interests

The author confirms that no generative AI or AI-assisted technologies were used to assist in writing during the preparation of this work. The authors declare that they have no known competing financial interests or personal relationships that could have influenced the work reported in this paper.

Additional information

Publisher’s Note

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

Electronic supplementary material

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Zhao, W., Hu, A. et al. Type 2 diabetic mellitus related osteoporosis: focusing on ferroptosis. J Transl Med 22, 409 (2024). https://doi.org/10.1186/s12967-024-05191-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05191-x

Keywords